Abstract
Aims
This study evaluated the safety and pharmacokinetics of naringenin in healthy adults, consuming a whole orange (Citrus Sinensis) extract.
Methods
In a single ascending dose randomized crossover trial, 18 adults ingested 150mg (NAR150), 300mg (NAR300), 600mg (NAR600), and 900mg (NAR900) doses of naringenin or placebo. Each dose or placebo was followed by a wash-out period of at least one week. Blood safety markers were evaluated pre-dose and 24 hours post-dose. Adverse events were recorded. Serum naringenin concentrations were measured before and over 24 hours following ingestion of placebo, NAR150, and NAR600. Four and 24-hour serum measurements were obtained after placebo, NAR300, and NAR900 ingestion. Data were analyzed using a mixed effects linear model.
Results
There were no relevant adverse events or changes in blood safety markers following ingestion of all naringenin doses. The pharmacokinetic parameters were: Maximal concentration: 15.76±7.88μM (NAR150) and 48.45±7.88μM (NAR600); Time to peak: 3.17±0.74h (NAR150) and 2.41±0.74h (NAR600); Area under the 24-hour concentration-time curve: 67.61±24.36μM×h (NAR150) and 199.06±24.36μM×h (NAR600); and Apparent oral clearance: 10.21±2.34L/h (NAR150) and 13.70±2.34L/h (NAR 600). Naringenin half-life was 3.0h (NAR150) and 2.65h (NAR600). After NAR300 ingestion, serum concentrations were 10.67±5.74μM (4h) and 0.35±0.30μM (24h). After NAR900 ingestion, serum concentrations were 43.11±5.26μM (4h) and 0.24±0.30μM (24h).
Conclusions
Ingestion of 150 to 900mg doses of naringenin is safe in healthy adults, and serum concentrations are proportional to the dose administered. Since naringenin (8μM) is effective in primary human adipocytes, ingestion of 300mg naringenin twice/day will likely elicit a physiologic effect.
Introduction
Dietary flavonoids are polyphenolic compounds present in many commonly consumed plant foods 1. Naringenin belongs to a subgroup of flavonoids known as flavanones, and is found predominantly in citrus fruits and tomato 2,3. The therapeutic potential of naringenin in modulating glucose and lipid metabolism has garnered substantial interest. In rodent studies and in vitro models, naringenin reduces diet-induced weight gain and improves glucose and lipid metabolism 4–7. Although most of the effects of naringenin have been shown in the liver, muscle, and islet cells, increases in energy expenditure and activation of brown fat have been demonstrated in mice fed a high-fat diet supplemented with naringenin 8. Our in vitro studies in differentiated human subcutaneous adipose-derived stem cells from overweight and obese female donors 9 show that naringenin stimulates mRNA and protein expression of uncoupling protein 1 (UCP1), glucose transporter type 4 (GLUT4), and carbohydrate responsive element binding protein (ChREBP), key determinants of thermogenesis, whole body insulin sensitivity, and glucose homeostasis 10,11. In prospective studies, high flavanone intake from oranges and grapefruits is associated with a cardioprotective effect, particularly, a reduction in the risk of ischemic stroke 12–14.
Flavanones occur naturally as glycosides which means that they are bound to different sugars. Hydrolysis of the sugar moieties from the flavanone glycosides naringin and narirutin by colonic bacteria releases the aglycone naringenin, and this is the rate-limiting step in the absorption of naringenin 15. After absorption, flavonoids are rapidly conjugated in the intestines or liver and appear in circulation as glucuronides or sulphoglucoronides 16–18. Ingestion of naringin or narirutin in 400 mL to 1L of orange juice, or segments from one medium fresh orange produce < 1 μM of naringenin in circulation 15,16,19,20, far short of the 8–10 μM concentration found to elicit physiologic effects in human adipocytes 9. Therefore, we conducted a Phase 1 study in healthy adults to evaluate the safety, tolerability, and pharmacokinetics of naringenin in the free aglycone form, which can be readily absorbed from the small intestine.
Grapefruit juice has been shown to increase the oral availability of certain drugs. The interaction with grapfruit causes an inhibition of first pass metabolism of the drug mainly catalyzed by an intestinal cytochrome P450 (CYP) 3A421,22. Furanocoumarin derivatives isolated from grapefruit were identified as the clinically active constituents responsible for the inhibition 23. Moreover, it was determined that naringin and naringenin are not the primary inhibitors in grapfruit juice 24. While grapefruit contains a high content of furanocoumarin derivatives, sweet oranges contain a very small amount of these derivatives 25. An extract was prepared from whole sweet oranges (Citrus Sinensis) and we hypothesized that oral administration of the whole orange extract which contained naringenin along with its native plant material would enhance its bioavailability in humans. We evaluated the safety and tolerability of single ascending doses of naringenin in healthy adults and determined its pharmacokinetic profile in serum.
Methods
Study Design
Subjects were enrolled in a placebo-controlled, double-blind, single ascending dose crossover trial to determine the safety, tolerability, and pharmacokinetics of naringenin at the Pennington Biomedical Research Center (PBRC). The maximum recommended starting dose for naringenin was estimated at approximately 135 mg since no adverse events were reported at this dose in a previous study 26. Due to the short elimination half-life (T½) of naringenin delivered in the free aglycone form (T½= 2.31 ± 0.4 h) 26, we hypothesized that multiple dosing up to 600 mg would be necessary for effective duration of action. The dose escalation scheme recommended for first in human studies is rapid dose escalation initially (maximum recommended starting dose [MRSD] x 2) until the estimated effective dose (600 mg) is reached and then at a more cautious escalation (for example, MRSD x 1.5) 27. Therefore, we evaluated the safety of 150, 300, 600, and 900 mg doses of naringenin. Each 150 mg dose capsule (NAR 150) contained 536 mg of the extract (28% naringenin, determined in quantitative analysis). The 300 mg (NAR 300), 600 mg (NAR 600), and 900 mg (NAR 900) naringenin doses were provided in two, four, and six capsules respectively. The placebo capsules contained microcrystalline cellulose and were similar in appearance.
The study design is represented in Table 1. Nine subjects were enrolled in each of two cohorts. Each subject in the cohorts was randomly assigned one of the relevant treatment sequences in order of enrollment. For example, the first three enrolled subjects in Cohort 1 were each randomly assigned to one of three distinct treatment sequences (Sequence 1, 2, or 3), as were enrollees 4–6 and 7–9. As a result, three subjects in each cohort were randomly assigned to each of the three treatment sequences.
Table 1.
Ascending Oral Dose Schedule
Visit 1 | Visit 2 | Visit 3 | |
---|---|---|---|
Cohort 1 | |||
Sequence 1 (n=3) | NAR 150 | Placebo | NAR 300 |
Sequence 2 (n=3) | NAR 150 | NAR 300 | Placebo |
Sequence 3 (n=3) | Placebo | NAR 150 | NAR 300 |
Cohort 2 | |||
Sequence 4 (n=3) | NAR 600 | Placebo | NAR 900 |
Sequence 5 (n=3) | NAR 600 | NAR 900 | Placebo |
Sequence 6 (n=3) | Placebo | NAR 600 | NAR 900 |
Note: In Cohort 1, the first three enrolled subjects were randomly assigned to Sequence 1, 2, or 3. The same assignment was followed for the next two blocks of three subjects for a total of nine subjects. Subjects were similarly assigned in Cohort 2.
To maintain the blind, all subjects in Cohort 1 received two capsules at each visit which comprised either two placebo capsules, one placebo and one NAR 150 capsule or two NAR 150 capsules depending on the randomization sequence. A similar process was employed in Cohort 2 and each subject received six capsules at each visit. Capsules were dispensed by the PBRC pharmacist according to the randomization scheme prepared by the study biostatistician. All other study staff and investigators were blinded to the treatment assignment. Each treatment was followed by a washout period of at least one week. The study was approved by the Institutional Review Board of PBRC. All procedures were in accordance with PBRC’s ethical standards. Subjects provided written informed consent. The trial was registered on ClinicaTrials.gov with registration number NCT03582553. Recruitment for the study commenced in May 2018 and the study was completed in October 2018.
Subjects
Eighteen male and female subjects, 18 years of age or older were enrolled. Subjects were eligible if they presented with a BMI between 20 and 35 kg/m2 and no known history of diseases requiring regular medications. Subjects were excluded for: (i) chronic use of medications, except over the counter medications and supplements that they agreed to stop 72 hours prior to the test day; (ii) fasting glucose > 126 mg/dL; (iii) current pregnancy or lactation; (iv) clinically significant gastrointestinal malabsorption syndromes such as chronic diarrhea, celiac disease or malabsorption from bariatric surgery within one month of the study; (v) reported history of substance abuse or alcoholism, or significant psychiatric disorder that would interfere with the ability to complete the study; (vi) smoking; (vii) and (vii) having citrus allergies. Subjects were provided with a list of foods containing flavanones and were instructed to refrain from consuming these foods for at least 36 hours prior to each test day. A diagram illustrating subject flow through the study is presented in the Supplementary Appendix (Figure 1).
Clinical Assessments
Each eligible subject completed three visits to the clinic. All testing was performed in the morning after an eight-hour overnight fast where only water was permitted. Upon arrival weight and vital signs (blood pressure, heart rate, and temperature) were measured. Blood was drawn for a chemistry panel (glucose, creatinine, potassium, uric acid, albumin, calcium, magnesium, creatine phosphokinase, alanine aminotransferase, alkaline phosphatase, aspartate aminotransferase, blood urea nitrogen, total bilirubin, iron, cholesterol [total, high density lipoprotein, low density lipoprotein], and triglycerides) and complete blood count before ingestion of each naringenin dose, or placebo. The subject returned 24 hours after ingestion of naringenin or placebo for a repeat chemistry panel and complete blood count, to assess the safety of naringenin. Adverse events (AE) were recorded following administration of each dose or placebo and 24 hours later. The definition and classification of AEs is presented in the Supplementary Appendix.
Pharmacokinetic Testing
An intravenous line was placed and blood was drawn prior to ingestion of placebo and NAR 150 or NAR 600, and at 2, 3, 3.5, 4, 4.5, 6, 8, and 12 hours post-dose. Subjects consumed a standard lunch five hours after ingestion of the naringenin dose or placebo. A standard dinner was also provided at the end of 12 hours. Subjects were discharged and returned to the Center the next morning, for a 24-hour post-dose measurement (Visit 1). Complete pharmacokinetic profile including the maximal concentration achieved (Cmax) time to peak (Tmax), area under the 24-hour serum concentration-time curve (AUC0 – 24h), apparent oral clearance (Clearanceoral), and T½ was evaluated.
At Visit 3, subjects consumed the placebo, and NAR 300 or NAR 900 with a standard snack, which permitted evaluation of serum naringenin concentration in a fed state. Blood was drawn at four hours post-dose. Subjects were discharged and returned to the Center the next morning and a 24-hour post-dose blood sample was collected.
Quantitative Analysis of Extract
Quantitative analysis of naringenin was performed at the Rutgers University Botanical Center, New Jersey. Naringenin and naringin were purchased from Cayman Chemical Company (Ann Arbor, MI). Whole sweet oranges (Citrus Sinensis) were subjected to an aqueous and ethanolic extraction process, dried, milled, and provided as a fine powder by Green Chem/Gencor Lifestage Solutions (Irvine, CA). The extract can be ordered from Gencor™ (https://gencorpacific.com/contact)Methodological details are provided in the Supplementary Appendix.
Quantitative Analysis of Serum Naringenin
Serum naringenin concentrations were measured at the University of Tennessee, Knoxville. Serum concentrations were measured after hydrolysis of samples with β-glucoronidase/sulphatase 15,16,20,26. For each sample, a 200 μL serum aliquot was enzymatically unconjugated in a 2 mL Eppendorf tube as previously described 28. Data was analyzed using the MAVEN software package 29,30. Methodological details are provided in the Supplementary Appendix.
Statistical Analysis
The primary aim of this Phase 1 study was to evaluate the effect of single oral administration of naringenin at four escalating doses (150, 300, 600, and 900 mg) on safety and tolerability in healthy volunteers. The secondary objective was to evaluate the 24-hour pharmacokinetic profile of naringenin in serum at 150 and 600 mg doses, and the four and 24-hour serum concentrations at 300 and 900 mg doses. As comparative efficacy of the doses was not the primary issue, power to detect effects was not specifically evaluated to justify sample sizes. Based on the safety and pharmacokinetic study done in humans 26, the sample size (n=6, at each dose) was deemed sufficient to evaluate safety and provide a pharmacokinetic profile of naringenin in serum.
The cohorts 1 and 2 were conducted in series. There was an interval of two weeks between the two cohorts during which and interim safety analysis was conducted. A summary of adverse events and blood safety markers for Cohort 1 was compiled and reviewed by the investigators, without breaking the blind. Dose safety was investigated by compiling by treatment (placebo, NAR 150, NAR 300, NAR 600, and NAR 900) a list of adverse events such as frequency of headaches and vomiting. The frequency of these events was counted and compared with the placebo. Changes in blood safety markers from pre-dose to 24 h post-dose were evaluated for significant differences among the doses. If there was a significant difference between the doses, the mean baseline value for each dose was added to the mean change from baseline for that dose and compared with the reference range for the marker. Pairwise comparisons of NAR 150, NAR 600 and placebo were tested with a linear mixed effect model for each of the parameters across all time points. Significance was set at p<0.05. Individual minimum and maximum values were evaluated by the medical investigator to determine clinical significance.
Pharmacokinetic profile was analyzed using a non-compartmental model 27. The Cmax and Tmax were taken directly from the individual serum data. The elimination rate constant (k) was obtained by means of linear regression analysis of the semi-logarithmic serum concentration-time curve using the time points that best fit the terminal elimination phase of the curve. The T½ was calculated by dividing In 2 by k. The AUC0 −24h, was estimated using the linear trapezoidal method. The Clearance(oral) was calculated as: Dose/AUC0 – 24h. The Cmax, Tmax, T½, AUC0 – 24h, and Clearance(oral) for each subject were determined and the means for subjects given NAR 150 and NAR 600 were calculated. All analyses were performed using SAS Version 9.4 software (SAS Institute Inc., Cary NC), by the study biostatistician.
Results
The chromatogram for the quantitative analysis of naringenin in the extract is presented in the Supplementary Appendix (Figure 2). The extract consisting of the plant materials found in the fruit and peel of Citrus Sinensis contained 28% naringenin and 8.5% naringin. Prunin was observed at a different ion (m/z = 578.1883, [-ESI]) and quantified as 2.5%. Eighteen subjects enrolled in and completed the study. Data related to 18 subjects were included in the analysis. Baseline characteristics of subjects are presented in Table 2. There were significant differences between the doses in the alanine aminotransferase, creatinine phosphokinase, and potassium concentrations, and eosinophil, and white blood cell counts. However, the mean change from baseline added to the mean baseline value was within the respective reference range. Individual values below and above the reference ranges pre- or post-dose were not clinically significant. The minimum and maximum values that were outside of the reference range are provided in the Supplementary Appendix.
Table 2.
Subject characteristics at baseline including age, BMI, gender, and race
n = 18 | |
---|---|
Mean ± Standard Deviation | |
Age | 38 ± 15.1 |
BMI (kg/m2) | 24.8 ± 4.1 |
n (%) | |
Gender | |
Female | 12 (67) |
Male | 6 (33) |
Race | |
White | 12 (67) |
Black | 4 (22) |
Asian | 2 (11) |
The AEs reported during the study and their intensities are presented in Table 3. There were 14 total AEs reported. Three AEs (placebo, NAR 300, and NAR 900) were reported at Visit 3 and were therefore ongoing on completion of the study. There was a statistically significant overall effect of dose on the intensity of the AEs (p=0.014). However, the worst effect was observed following ingestion of the placebo, and there was no statistically significant effect between the placebo and any of the doses.
Table 3.
Adverse events reported during the study. Each check mark represents an event
Event | Placebo | NAR 150 | NAR 300 | NAR 600 | NAR 900 | Mild | Moderate |
---|---|---|---|---|---|---|---|
Itching | √†√‡ | √ | √ | ||||
Rash | √‡ | √ | |||||
Acne | √ | √ | |||||
Cyst on Foot | √† | √ | |||||
Vomiting | √§ | √ | |||||
Diarrhea | √§ | √ | |||||
Abdominal Pain | √¶ | √ | |||||
Headache | √¶ | √ | √√ | ||||
Drowsiness | √ | √ | |||||
Joint pain | √† | √ | |||||
Sinus Congestion | √ | √ | |||||
Visual Disturbance | √ | √ |
Reported at Visit 3 and unresolved at the end of the study.
Events reported by same subjects
Events reported by same subjects
Events reported by same subjects.
The mean total serum concentrations after ingestion of NAR 150 or NAR 600, or placebo over 24 hours, and at four and 24 hours after ingestion of NAR 300 and NAR 900 are presented in Figure 1AB. Pharmacokinetic parameters of naringenin in serum are presented in Table 4. The Cmax of naringenin increased by 307.5% in the NAR 600 dose compared to NAR 150 (p=0.01). There was no difference in the Tmax (p=0.49) between NAR 150 and NAR 600, although the Tmax increased by 31.5%, following ingestion of NAR 600 compared to NAR 150. The four-hour serum concentration increased by four times (404%) after ingestion of NAR 900 compared to NAR 300 (p=0.001, Range NAR 300: 4.93 – 20.43 μM [4h] and 0.02 −1.30 μM [24h], Range NAR 900: 13.53 – 83.43 μM [4h] and 0.03 – 0.89 μM [24h]).
Figure 1.
(A) Serum kinetics of naringenin after ingestion of Extract of whole oranges containing 150 mg (NAR 150) and 600 mg (NAR 600) of naringenin. (B) Four- and 24-hour serum concentration of naringenin after ingestion of extract of whole oranges containing 300 mg (NAR 300) and 900 mg (NAR 900) of naringenin. Concentration of aglycones was determined by LC-MS after hydrolysis of β-glucuronidase-sulphatase. Values are means ± SEM.
Table 4.
Pharmacokinetic parameters of serum naringenin over 24 hours after an oral dose of 150 mg and 600 mg of naringenin in an extract from whole oranges (NAR) administered in capsule form. Values are mean ±SEM.
Dose | AUC0–24h† (μM×h) |
Cmax (μM) |
Tmax (h) |
T½‡ (h) |
AOC§ (L/h) |
---|---|---|---|---|---|
NAR 150 | 67.61 ± 24.26 | 15.76 ± 7.88 | 3.17 ± 0.74 | 3.0 | 10.21 ± 2.34 |
Range | 35.56 – 120.34 | 6.43 – 36.85 | 2 – 4 | 4.58 – 15.49 | |
NAR 600 | 199.05 ± 24.36 | 48.45 ± 7.88 | 2.41 ± 0.74 | 2.65 | 13.70 ± 2.34 |
Range | 97.63 – 316.74 | 22.01 – 94.94 | 2 – 3.5 | 6.96 – 22.57 | |
Dose | AUC0–24h† (μg/ml)×h |
Cmax (μg/ml) |
|||
NAR 150 | 18.41 ± 6.61 | 4.29 ± 2.15 | |||
Range | 9.68 – 32.77 | 1.75 – 10.03 | |||
NAR 600 | 54.19 ± 6.63 | 13.19 ± 2.15 | |||
Range | 26.58 – 86.24 | 5.99 – 25.85 | |||
p-value | 0.001 | 0.01 | 0.49 | 0.39 | 0.31 |
Area under the 24-hour serum concentration-time curve
Based on log transformed data using 3.5 to 24 hour time-points for best fit of terminal phase elimination curve
Apparent Oral Clearance (Dose/AUC0–24h (μg/ml)×h
Based on the best fit of the terminal elimination curve, the 3.5, 4, 4.5, 6, 12, and 24 hour time-points were used in the determination of k (r2=0.63 [NAR 150] and r2=0.90 [NAR 600]), for calculation of T½. The T½ was approximately 12% lower after ingestion of NAR 600 compared to NAR 150, but the difference was not statistically significant (p=0.39). Serum concentrations of naringenin were approximately proportional to the dose administered, whether in the fasted or fed state. The apparent oral clearance increased by 34% following ingestion of NAR 600 compared to NAR 150, but the difference was not significant (p=0.31).
Discussion
This is the first study to establish the safety and tolerability of four single ascending doses (150, 300, 600, 900 mg) of naringenin in the aglycone form in humans, and to determine a complete pharmacokinetic profile including Cmax, Tmax, T½, AUC0 −24h, and Clearance(oral) at 150 mg and 600 mg doses. The kinetic curves of naringenin concentrations measured in serum over 24 hours after ingestion of NAR 150 and NAR 600 indicate that naringenin is present in circulation and has a half-life of approximately three hours. Naringenin is absorbed into circulation approximately proportional to the dose administered in both the fasted (NAR 150 and NAR 600) and fed states (NAR 300 and NAR 900) and is cleared from circulation within 24 hours.
Citrus flavonoids such as naringenin have diverse and powerful biological properties 31. Scientific research on the physiologic relevance of naringenin has been limited to in vitro and animal models. Studies seeking to understand the therapeutic potential of naringenin in humans, have been hindered by ingestion of citrus juices or fruits whereby, the circulating concentration of naringenin is low. In cell culture and animal models, the range of naringenin determined to elicit a desired response ranges from 1–200 μM 31,32. In humans, the maximum plasma concentrations achieved from food sources of naringenin has only reached <1 μM 15,16,19,20.
In the only other clinical trial of naringenin in the aglycone form 26, ingestion of a single 135 mg dose of the pure compound delivered in a solid dispersion capsule produced a Cmax of 7.39 μM in 3.67 hours. In comparison, ingestion of 150 mg naringenin delivered in an extract from whole oranges produced a Cmax that was two-fold higher in less time (Tmax=3.17 h). Following ingestion of 600 mg naringenin, the Cmax was more than three-fold higher than the 150 mg dose, and the peak concentration was reached in 2.41 hours. Following ingestion of 900 mg naringenin, the serum naringenin concentration was more than four-fold higher than the 300 mg dose. The serum concentrations of naringenin following ascending doses show that naringenin absorption is approximately proportional to the dose administered. The half-life of naringenin following the 150 mg (3 h) and 600 mg doses (2.65 h) was not significantly different between the doses, and is consistent with the 2.31 h reported previously 26.
In addition to naringenin, the extract from Citrus Sinensis also contained 8.5% naringin and 2.5% prunin. It is unlikely that naringin contributed to the naringenin Cmax since naringin must reach the colon and be hydrolyzed by the gut bacteria for absorption. Previous studies have shown that the Cmax values for flavanone metabolites occurred approximately five hours after ingestion of citrus fruits 16,19,20 containing naringin. Although prunin can be hydrolyzed in the human small intestine 33,34, little is known of its bioavailability.
The evidence in the literature supports a role for naringenin in the treatment of dyslipidemia, insulin resistance, hepatic steatosis, obesity, and atherosclerosis 31. Several methods of drug design for delivery of naringenin are in the process of development such as liposomes, naringenin-encapsulated nanoparticles, self-nanoemulsifying drug delivery systems (SNEDDS), and nanosuspension 35. However, the safety and efficacy of these delivery routes have never been tested in humans. The present study using an extract from whole sweet oranges demonstrates that serum naringenin concentrations are higher when naringenin is delivered along with its native plant materials compared to the isolated form delivered in solid dispersion capsules 26. Synergistic effects can be produced if the constituents of a plant extract interact with one another in order to improve the solubility and thereby enhance the bioavailability of a particular component of an extract, compared to the isolated constituent 36. This possibility is not uncommon in phyto-pharmacology, especially with polyphenols 37, and enhanced absorption of naringenin following ingestion of the extract is likely due to a synergy among its various plant constituents.
Our study is limited by the lack of information on urinary excretion of naringenin metabolites. Flavonoid bioavailability studies have used urinary excretion as a measure of absorption. However, urinary excretion does not account for the possibility of metabolites being sequestered in tissues 38. Moreover, in addition to urinary excretion, a significant proportion of bioavailable flavonoid metabolites may be excreted via bile into the feces 39. In the study by Kanaze et al, a 135 mg dose of naringenin produced a urinary excretion of 5.81% and the relative urine excretion of naringenin reported in the literature has a wide range from 4% to 30% of the dose administered 19,20. Although it has been suggested that the naringenin metabolites appearing in circulation are treated by the body as xenobiotics and are removed from the system, analysis of the serum concentrations provides valuable information on the pharmacokinetic profile of naringenin 38. Further, we measured serum naringenin concentrations following hydrolysis of the conjugated form; however, the method provides an estimation of serum naringenin concentrations 15,16,20,26,40.
Little is known about tissue uptake and disposition of flavonoid metabolites in humans which is important for the effects of naringenin on cell and tissue function 41. In rodent studies, dosing with naringenin or naringin produced inconsistent results. Following a single gastric gavage of naringenin (50mg/kg) in rats, naringenin aglycone was detected at higher levels in tissues than in plasma after 18 h 42. However, following gastric gavage of naringin at 210 mg/kg twice daily (17 doses) in rats, free forms of naringin and naringenin were not detected in plasma or major organs.43 Future studies should evaluate the safety and physiologic effects of multiple dosing of naringenin on energy expenditure and glucose metabolism in humans, which will provide information on tissue uptake.
In conclusion, the purpose of this study was to determine the safety and tolerability of naringenin and evaluate its pharmacokinetic profile. We show that single doses of 150, 300, 600, and 900 mg of naringenin are safe and well tolerated in humans. At these doses, naringenin metabolites are present in circulation, and are cleared within 24 hours. Since naringenin at 8 μM concentrations is effective in primary human adipocytes and primary human adipose tissue 9, 300 mg ingested twice daily would be sufficient to elicit a physiologic response.
Supplementary Material
Acknowledgments
Supported by U54 GM104940 from the National Institute of General Medical Sciences of the National Institutes of Health (NIH), which funds the Louisiana Clinical and Translational Science Center and in part by the National Center For Complementary & Integrative Health and the Office of Dietary Supplements of the NIH under Award Number P50AT002776 which funds the Botanical Dietary Supplements Research Center of PBRC, the NIH under Award Number T32A T004094, and the NORC Center Grant #P30DK072476 entitled “Nutrition and Metabolic Health Through the Lifespan” sponsored by NIDDK.
Contributor Information
Candida J. Rebello, Pharmacology Clinical Trials, Pennington Biomedical Research Center (PBRC), Baton Rouge, Louisiana
Robbie A. Beyl, Biostatistics, PBRC, Baton Rouge, Louisiana
Juan J. L. Lertora, Clinical Pharmacology, PBRC, Baton Rouge, Louisiana
Frank L. Greenway, Pharmacology Clinical Trials, Pennington Biomedical Research Center (PBRC), Baton Rouge, Louisiana
Eric Ravussin, Human Translational Physiology, PBRC, Baton Rouge, Louisiana.
David M. Ribnicky, Rutgers University Botanical Center, New Jersey
Alexander Poulev, Rutgers University Botanical Center, New Jersey.
Brandon J. Kennedy, Department of Chemistry, University of Tennessee, Knoxville, Tennessee
Hector F. Castro, Department of Chemistry, University of Tennessee, Knoxville, Tennessee Biological and Small Molecule Chemistry Core, University of Tennessee, Knoxville, Tennessee.
Shawn R. Campagna, Department of Chemistry, University of Tennessee, Knoxville, Tennessee Biological and Small Molecule Chemistry Core, University of Tennessee, Knoxville, Tennessee.
Ann A. Coulter, Pharmacology Clinical Trials, Pennington Biomedical Research Center (PBRC), Baton Rouge, Louisiana
Leanne M. Redman, Reproductive Endocrinology, PBRC, Baton Rouge, Louisiana Rutgers University Botanical Center, New Jersey.
References
- 1.Erdman JW Jr., Balentine D, Arab L, et al. Flavonoids and heart health: proceedings of the ILSI North America Flavonoids Workshop, May 31-June 1, 2005, Washington, DC. J Nutr. 2007;137(3 Suppl 1):718S–737S. [DOI] [PubMed] [Google Scholar]
- 2.Kawaii S, Tomono Y, Katase E, Ogawa K, Yano M. Quantitation of flavonoid constituents in citrus fruits. J Agric Food Chem. 1999;47(9):3565–3571. [DOI] [PubMed] [Google Scholar]
- 3.Davies JN, Hobson GE. The constituents of tomato fruit--the influence of environment, nutrition, and genotype. Crit Rev Food Sci Nutr. 1981;15(3):205–280. [DOI] [PubMed] [Google Scholar]
- 4.Zygmunt K, Faubert B, MacNeil J, Tsiani E. Naringenin, a citrus flavonoid, increases muscle cell glucose uptake via AMPK. Biochemical and biophysical research communications. 2010;398(2):178–183. [DOI] [PubMed] [Google Scholar]
- 5.Kannappan S, Anuradha CV. Naringenin enhances insulin-stimulated tyrosine phosphorylation and improves the cellular actions of insulin in a dietary model of metabolic syndrome. Eur J Nutr. 2010;49(2):101–109. [DOI] [PubMed] [Google Scholar]
- 6.Goldwasser J, Cohen PY, Yang E, Balaguer P, Yarmush ML, Nahmias Y. Transcriptional regulation of human and rat hepatic lipid metabolism by the grapefruit flavonoid naringenin: role of PPARalpha, PPARgamma and LXRalpha. PLoS One. 2010;5(8):e12399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Bhattacharya S, Oksbjerg N, Young JF, Jeppesen PB. Caffeic acid, naringenin and quercetin enhance glucose-stimulated insulin secretion and glucose sensitivity in INS-1E cells. Diabetes Obes Metab. 2014;16(7):602–612. [DOI] [PubMed] [Google Scholar]
- 8.Thaiss CA, Itav S, Rothschild D, et al. Persistent microbiome alterations modulate the rate of post-dieting weight regain. Nature. 2016. [DOI] [PubMed] [Google Scholar]
- 9.Rebello CJ, Greenway FL, Lau FH, et al. Naringenin Promotes Thermogenic Gene Expression in Human White Adipose Tissue. Obesity (Silver Spring). 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Herman MA, Peroni OD, Villoria J, et al. A novel ChREBP isoform in adipose tissue regulates systemic glucose metabolism. Nature. 2012;484(7394):333–338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Shepherd PR, Kahn BB. Glucose transporters and insulin action--implications for insulin resistance and diabetes mellitus. N Engl J Med. 1999;341(4):248–257. [DOI] [PubMed] [Google Scholar]
- 12.Joshipura KJ, Ascherio A, Manson JE, et al. Fruit and vegetable intake in relation to risk of ischemic stroke. JAMA. 1999;282(13):1233–1239. [DOI] [PubMed] [Google Scholar]
- 13.Cassidy A, Rimm EB, O’Reilly EJ, et al. Dietary flavonoids and risk of stroke in women. Stroke. 2012;43(4):946–951. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Yamada T, Hayasaka S, Shibata Y, et al. Frequency of citrus fruit intake is associated with the incidence of cardiovascular disease: the Jichi Medical School cohort study. J Epidemiol. 2011;21(3):169–175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Manach C, Morand C, Gil-Izquierdo A, Bouteloup-Demange C, Remesy C. Bioavailability in humans of the flavanones hesperidin and narirutin after the ingestion of two doses of orange juice. Eur J Clin Nutr. 2003;57(2):235–242. [DOI] [PubMed] [Google Scholar]
- 16.Erlund I, Meririnne E, Alfthan G, Aro A. Plasma kinetics and urinary excretion of the flavanones naringenin and hesperetin in humans after ingestion of orange juice and grapefruit juice. J Nutr. 2001;131(2):235–241. [DOI] [PubMed] [Google Scholar]
- 17.Nielsen IL, Chee WS, Poulsen L, et al. Bioavailability is improved by enzymatic modification of the citrus flavonoid hesperidin in humans: a randomized, double-blind, crossover trial. J Nutr. 2006;136(2):404–408. [DOI] [PubMed] [Google Scholar]
- 18.Zeng X, Su W, Bai Y, et al. Urinary metabolite profiling of flavonoids in Chinese volunteers after consumption of orange juice by UFLC-Q-TOF-MS/MS. J Chromatogr B Analyt Technol Biomed Life Sci. 2017;1061–1062:79–88. [DOI] [PubMed] [Google Scholar]
- 19.Manach C, Williamson G, Morand C, Scalbert A, Remesy C. Bioavailability and bioefficacy of polyphenols in humans. I. Review of 97 bioavailability studies. Am J Clin Nutr. 2005;81(1 Suppl):230S–242S. [DOI] [PubMed] [Google Scholar]
- 20.Brett GM, Hollands W, Needs PW, et al. Absorption, metabolism and excretion of flavanones from single portions of orange fruit and juice and effects of anthropometric variables and contraceptive pill use on flavanone excretion. Br J Nutr. 2009;101(5):664–675. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Bailey DG, Malcolm J, Arnold O, Spence JD. Grapefruit juice-drug interactions. British journal of clinical pharmacology. 1998;46(2):101–110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Schmiedlin-Ren P, Edwards DJ, Fitzsimmons ME, et al. Mechanisms of enhanced oral availability of CYP3A4 substrates by grapefruit constituents. Decreased enterocyte CYP3A4 concentration and mechanism-based inactivation by furanocoumarins. Drug Metab Dispos. 1997;25(11):1228–1233. [PubMed] [Google Scholar]
- 23.Guo LQ, Fukuda K, Ohta T, Yamazoe Y. Role of furanocoumarin derivatives on grapefruit juice-mediated inhibition of human CYP3A activity. Drug Metab Dispos. 2000;28(7):766–771. [PubMed] [Google Scholar]
- 24.Edwards DJ, Bernier SM. Naringin and naringenin are not the primary CYP3A inhibitors in grapefruit juice. Life Sci. 1996;59(13):1025–1030. [DOI] [PubMed] [Google Scholar]
- 25.Saita T, Fujito H, Mori M. Screening of furanocoumarin derivatives in citrus fruits by enzyme-linked immunosorbent assay. Biol Pharm Bull. 2004;27(7):974–977. [DOI] [PubMed] [Google Scholar]
- 26.Kanaze FI, Bounartzi MI, Georgarakis M, Niopas I. Pharmacokinetics of the citrus flavanone aglycones hesperetin and naringenin after single oral administration in human subjects. Eur J Clin Nutr. 2007;61(4):472–477. [DOI] [PubMed] [Google Scholar]
- 27.Foster DM and Vicini P. Non-compartmental and compartmental approaches to pharmacokinetic data analysis In Principles of Clinical Pharmacology (Third Edition), Atkinson AJ Jr., Huang S-M, Lertora JJL, Markey SP, editors. Academic Press, 2012, p. 97–116.23. [Google Scholar]
- 28.Kanaze FI, Kokkalou E, Georgarakis M, Niopas I. Validated high-performance liquid chromatographic method utilizing solid-phase extraction for the simultaneous determination of naringenin and hesperetin in human plasma. J Chromatogr B Analyt Technol Biomed Life Sci. 2004;801(2):363–367. [DOI] [PubMed] [Google Scholar]
- 29.Clasquin MF, Melamud E, Rabinowitz JD. LC-MS data processing with MAVEN: a metabolomic analysis and visualization engine. Curr Protoc Bioinformatics. 2012;Chapter 14:Unit14 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Melamud E, Vastag L, Rabinowitz JD. Metabolomic analysis and visualization engine for LC-MS data. Anal Chem. 2010;82(23):9818–9826. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Mulvihill EE, Burke AC, Huff MW. Citrus Flavonoids as Regulators of Lipoprotein Metabolism and Atherosclerosis. Annu Rev Nutr. 2016;36:275–299. [DOI] [PubMed] [Google Scholar]
- 32.Rani N, Bharti S, Krishnamurthy B, et al. Pharmacological Properties and Therapeutic Potential of Naringenin: A Citrus Flavonoid of Pharmaceutical Promise. Curr Pharm Des. 2016;22(28):4341–4359. [DOI] [PubMed] [Google Scholar]
- 33.Berrin JG, McLauchlan WR, Needs P, et al. Functional expression of human liver cytosolic beta-glucosidase in Pichia pastoris. Insights into its role in the metabolism of dietary glucosides. Eur J Biochem. 2002;269(1):249–258. [DOI] [PubMed] [Google Scholar]
- 34.Day AJ, Canada FJ, Diaz JC, et al. Dietary flavonoid and isoflavone glycosides are hydrolysed by the lactase site of lactase phlorizin hydrolase. FEBS Lett. 2000;468(2–3):166–170. [DOI] [PubMed] [Google Scholar]
- 35.Joshi R, Kulkarni YA, Wairkar S. Pharmacokinetic, pharmacodynamic and formulations aspects of Naringenin: An update. Life Sci. 2018;215:43–56. [DOI] [PubMed] [Google Scholar]
- 36.Wagner H, Ulrich-Merzenich G. Synergy research: approaching a new generation of phytopharmaceuticals. Phytomedicine. 2009;16(2–3):97–110. [DOI] [PubMed] [Google Scholar]
- 37.Butterweck V, Wall A, Lieflander-Wulf U, Winterhoff H, Nahrstedt A. Effects of the total extract and fractions of Hypericum perforatum in animal assays for antidepressant activity. Pharmacopsychiatry. 1997;30 Suppl 2:117–124. [DOI] [PubMed] [Google Scholar]
- 38.Crozier A, Jaganath IB, Clifford MN. Dietary phenolics: chemistry, bioavailability and effects on health. Nat Prod Rep. 2009;26(8):1001–1043. [DOI] [PubMed] [Google Scholar]
- 39.Ma LY, Liu RH, Xu XD, Yu MQ, Zhang Q, Liu HL. The pharmacokinetics of C-glycosyl flavones of Hawthorn leaf flavonoids in rat after single dose oral administration. Phytomedicine. 2010;17(8–9):640–645. [DOI] [PubMed] [Google Scholar]
- 40.Zeng X, Su W, Zheng Y, et al. Pharmacokinetics, Tissue Distribution, Metabolism, and Excretion of Naringin in Aged Rats. Front Pharmacol. 2019;10:34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Cassidy A, Minihane AM. The role of metabolism (and the microbiome) in defining the clinical efficacy of dietary flavonoids. Am J Clin Nutr. 2017;105(1):10–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.El Mohsen MA, Marks J, Kuhnle G, et al. The differential tissue distribution of the citrus flavanone naringenin following gastric instillation. Free Radic Res. 2004;38(12):1329–1340. [DOI] [PubMed] [Google Scholar]
- 43.Lin SP, Hou YC, Tsai SY, Wang MJ, Chao PD. Tissue distribution of naringenin conjugated metabolites following repeated dosing of naringin to rats. Biomedicine (Taipei). 2014;4:16. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.