Next Article in Journal
Assessment of Heavy Metals and Trace Elements in the Human Milk of Women Living in Latvia and an Evaluation of Influencing Factors
Previous Article in Journal
Breastfeeding Perceptions and Decisions among Hispanic Participants in the Special Supplemental Nutrition Program for Women, Infants, and Children: A Qualitative Study
Previous Article in Special Issue
Pattern of Consumption of Sports Supplements of Spanish Handball Players: Differences According to Gender and Competitive Level
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

In Vitro Activation of Human Adrenergic Receptors and Trace Amine-Associated Receptor 1 by Phenethylamine Analogues Present in Food Supplements

by
Nicole E. T. Pinckaers
1,2,*,
W. Matthijs Blankesteijn
1,3,
Anastasiya Mircheva
1,
Xiao Shi
4,5,
Antoon Opperhuizen
1,6,
Frederik-Jan van Schooten
1,2 and
Misha F. Vrolijk
1,2
1
Department of Pharmacology and Toxicology, Maastricht University, 6200 MD Maastricht, The Netherlands
2
Research Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, 6200 MD Maastricht, The Netherlands
3
School for Cardiovascular Diseases (CARIM), Maastricht University, 6200 MD Maastricht, The Netherlands
4
Research Service, Veterans Affairs Portland Health Care System, Portland, OR 97239, USA
5
Department of Psychiatry, Oregon Health and Science University, Portland, OR 97239, USA
6
Office for Risk Assessment and Research, Netherlands Food and Consumer Product Safety Authority, 3540 AA Utrecht, The Netherlands
*
Author to whom correspondence should be addressed.
Nutrients 2024, 16(11), 1567; https://doi.org/10.3390/nu16111567
Submission received: 12 April 2024 / Revised: 15 May 2024 / Accepted: 16 May 2024 / Published: 22 May 2024
(This article belongs to the Special Issue Sport Supplementation for Performance and Health (Volume II))

Abstract

:
Pre-workout supplements are popular among sport athletes and overweight individuals. Phenethylamines (PEAs) and alkylamines (AA) are widely present in these supplements. Although the health effects of these analogues are not well understood yet, they are hypothesised to be agonists of adrenergic (ADR) and trace amine-associated receptors (TAARs). Therefore, we aimed to pharmacologically characterise these compounds by investigating their activating properties of ADRs and TAAR1 in vitro. The potency and efficacy of the selected PEAs and AAs was studied by using cell lines overexpressing human ADRα1A1B1D2a2B12 or TAAR1. Concentration–response relationships are expressed as percentages of the maximal signal obtained by the full ADR agonist adrenaline or the full TAAR1 agonist phenethylamine. Multiple PEAs activated ADRs (EC50 = 34 nM–690 µM; Emax = 8–105%). Almost all PEAs activated TAAR1 (EC50 = 1.8–92 µM; Emax = 40–104%). Our results reveal the pharmacological profile of PEAs and AAs that are often used in food supplements. Several PEAs have strong agonistic properties on multiple receptors and resemble potencies of the endogenous ligands, indicating that they might further stimulate the already activated sympathetic nervous system in exercising athletes via multiple mechanisms. The use of supplements containing one, or a combination of, PEA(s) may pose a health risk for their consumers.

1. Introduction

In recent years, the availability of food supplements with varying purposes on the market and their use are rising. In particular, pre-workout supplements and fat burners are becoming a more and more popular category of food supplements among sport athletes and overweight individuals [1] due to their advertised promises of improved sports performance and weight loss. Whereas a study on the use of such products in the Dutch society in 2012 showed that only 5.8% of regularly exercising participants were using sports enhancement supplements [2], a more recent study showed that 52.6% of the (amateur) sport athletes are using or have ever used such products [3]. This shows the substantial increase in the popularity of these nutritional supplements in the past few years.
Despite the large-scale use of these supplements, their adverse health effects are poorly investigated. Over the years, several cases of serious adverse health effects, like palpitations, myocardial infarction, cardiac arrest and brain haemorrhages, have been linked to the use of pre-workout and fat burner supplements [3,4,5,6,7,8,9]. Most of these adverse health effects are specifically associated with the cardiovascular system [10]. Since pharmacological and toxicological data about the ingredients of these supplements are often lacking, causally linking the adverse health effects to the use of the supplements and mechanistically explaining these effects remain challenging.
Most likely, exposure to the pharmacologically active substances (PASs) present in these supplements explains the evoked adverse health events. Various studies have identified the presence of known PASs and structurally related compounds in pre-workout and fat burner supplements, of which phenethylamine and alkylamine analogues constitute a large group [8,11,12,13,14,15,16,17,18,19,20,21,22,23]. Phenethylamines and alkylamines are two classes of molecules that have been shown to have psychoactive and stimulant effects [24,25]. These compounds are structurally similar to endogenous catecholamines, such as adrenaline, noradrenaline and dopamine (Figure 1a), and to exogenous stimulants like amphetamine. Catecholamines and amphetamines are both known to induce sympathetic activation via the stimulation of adrenergic receptors (ADRs) and the human trace amine-associated receptor 1 (TAAR1) located in the central nervous system (CNS) and the periphery [26,27,28]. Ultimately, these will both lead to increased activation of the cardiovascular system, which is physiologically shown by increased heart rate and blood pressure [29,30].
Remarkably, for most of the detected phenethylamines and alkylamines, no pharmacological profile is available. It is therefore not known if and to what extent these compounds interact with the TAAR1 and ADR and thus how they might affect the cardiovascular system. Despite this lack of knowledge, supplements containing these compounds are used on a large scale and often in high doses [3,11,19,31]. For example, intake levels of methylsynephrine were found to be two times higher than the maximum prescribed dosage of the former pharmaceutical Carnigen, which was formulated with methylsynephrine [19,32]. Besides the high dosages, this compound was present in a sport supplement that also contained other PASs (e.g., β-methylphenethylamine and dimethylphenethylamine), which might both lead to adverse health effects. It is therefore essential to gain more knowledge about the pharmacological profile of phenethylamine and alkylamine compounds that are often used as nutritional supplements. Therefore, this study aims to pharmacologically characterise a selection of these phenethylamine and alkylamine analogues in vitro by assessing their potency and efficacy to activate human ADRs, α1, α2, β1 and β2, and TAAR1.

2. Materials and Methods

2.1. Selection of Phenethylamine and Alkylamine Analogues for Receptor Activation Assays

Search engines PubMed and Scopus were used to collect research papers on the detection of phenethylamine and alkylamine analogues in nutritional supplements. The key words/phrases used for the search were as follows: “pharmacologically active compounds supplements”, “bioactive compounds supplements”, “weight loss supplements”, “pre-workout supplements”, “fatburner”, “sport supplements”, “phenethylamine supplements”, “alkylamine supplements” and “amine supplements”. The most frequently reported analogues were selected to include in the in vitro pharmacological screening. The selected compounds are presented in Figure 1b,c [8,11,12,13,14,15,16,17,18,19,20,21,22,23].

2.2. Chemicals

Probenecid (purity ≥ 98%), adrenaline bitartrate, beta-methylphenethylamine (99%), phenethylamine (99.7%), tyramine hydrochloride (≥98%), halostachine (99%), hordenine (≥97.5%), p-octopamine (≥95%), dimethylbutylamine (98%), dimethylhexylamine (99%), dimethylaminoethanol (99.8%), puromycin dihydrochloride (10 mg/mL solution in mQ) and L-ascorbic acid (99%) were purchased from Sigma-Aldrich (St. Louis, MO, USA). p-synephrine hydrochloride (99.6%) and higenamine hydrochloride (98.98%) were purchased from MedChemExpress (Monmouth Junction, NJ, USA). Methyltyramine (97%) and 1,3-dimethylamylamine hydrochloride (95%) were purchased from Apollo Scientific (Stockport, UK). Sodium bicarbonate and isobutylmethylxanthine (IBMX) were purchased from Merck (Darmstadt, Germany). Fluo-8 AM (97%) was purchased from AAT Bioquest (Sunnyvale, CA, USA). Methylsynephrine hydrochloride (99.8%) was purchased from Mikromol (Luckenwalde, Germany), isopropyloctopamine acetate (99.9%) from Honeywell Fluka (Seetze, Germany), isopropyloctopamine hydrochloride (98%) from Toronto Research Chemicals (Toronto, ON, Canada) and N,N-dimethylphenethylamine hydrochloride (>99%) from GLPBio (Montclair, NJ, USA).

2.3. Adrenergic Receptor (ADR) Activation

Cells overexpressing human ADRs α1A, α1B, α1D, α2A, α2B, β1 and β2 (Eurofins Discovery Services, St. Charles, MO, USA, product numbers: HTS087RTA, HTS158RTA, HTS216RTA, HTS096RTA, HTS157LRTA, HTS104RTA and HTS073RTA, respectively) were used to study the potency (EC50) and efficacy (Emax) of the selected compounds for these receptors by quantifying Ca2+ influx upon receptor activation. ADRα1A, α1B, α1D, α2A, β1 and β2 were overexpressed in rat chem-1 cells. ADRα2B was overexpressed in HEK293 cells. The cells were seeded (10,000–50,000 cells/well) in a black 96-well plate (VWR International, Leuven, Belgium) and incubated at 37 °C and 5% CO2 for 24 h. After 24 h, the cells were washed with Hank’s balanced salt solution (HBSS) (Gibco, Paisley, UK) containing 0.35 g/L sodium bicarbonate and incubated with 5 µg/mL Fluo-8 AM in HBSS (pH 7.4) supplemented with 2.5 mM probenecid for 1 h. Baseline fluorescence (excitation: 490 nm; emission: 525 nm) was measured for 15 s with a read interval of 1.3 s, whereafter the selected test compounds were injected at a dispense rate of 78 µL/s to the cells using FlexStation 3 (Molecular Devices, Sunnyvale, CA, USA). After the addition of the test compounds, fluorescence was measured for another 165 s using the same settings. Peak tops of the obtained fluorescence response–time curves were corrected for baseline fluorescence and used to construct concentration–response relationships.

2.4. Trace Amine-Associated Receptor 1 (TAAR1) Activation

HEK293T cells stably expressing human TAAR1 (Appendix A), kindly donated by Aaron Janowsky, PhD from VA Portland Health Care System, were used to study the potency (EC50) and efficacy (Emax) of the selected compounds for this receptor by quantifying intracellular cyclic adenosine monophosphate (cAMP) levels upon receptor activation. The cells were grown in Dulbecco’s Modified Eagle’s Medium with 4.5 g/L glucose and 584 mg/L L-glutamine (Sigma-Aldrich, St. Louis, MO, USA), supplemented with 10% foetal bovine serum (Gibco, Grand Island, NY, USA), 100 units/mL penicillin, 100 µg/mL streptomycin (Gibco, Grand Island, NY, USA), 26 mM sodium bicarbonate and 2 µg/mL puromycin, and were maintained in a humidified incubator with 5% CO2. The cells were seeded (90,000–250,000 cells/well) in a 48-well plate two days before the assay. Two hours before the assay, the cells were incubated with the culture medium containing 10% charcoal-stripped foetal bovine serum (Gibco, Grand Island, NY, USA). The assay was performed in Earle’s balanced salt solution (EBSS) (Gibco, Paisley, UK), containing 15 mM HEPES (Gibco, Paisley, UK), 1 mM IBMX and 0.02% ascorbic acid. First, the cells were pre-incubated for 20 min in the supplemented buffer. Subsequently, the cells were exposed to the test compounds for 1 h in an incubator at 37 °C with 5% CO2. After 1 h, the buffer was discarded and the cells were lysed with 3% trichloroacetic acid and incubated at room temperature on a plate shaker for 2 h. Cell lysates were further diluted and cAMP was measured using a cAMP ELISA kit (Cayman Chemical, Ann Arbor, MI, USA), according to the manufacturer’s protocol.

2.5. Data Analysis

Concentration–response relationships are shown as mean ± SD. For ADR activation, concentration–response relationships are presented as percentages of the maximal intracellular Ca2+ signal obtained by the full ADR agonist adrenaline and corrected for baseline measurements. Concentration–response relationships for TAAR1 activation are presented as percentages of the maximal intracellular cAMP level induced by full the TAAR1 agonist phenethylamine and corrected for baseline cAMP accumulation. Concentration–response curves were fitted to non-linear regression curves with a standard Hill slope of 1 (GraphPad Software, version 10.1.2, San Diego, CA, USA). Experiments were run in duplicate in 7–9 independent experiments for ADR and were run in 3–5 independent experiments for TAAR1. The EC50 and Emax values are presented as an average of the independent experiments, with a 95% confidence interval (95% CI).

3. Results

3.1. Adrenergic Receptor α1A/B/D Activation

The concentration–response curves of the compounds that activated the three ADRα1 subtypes α1A, α1B and α1D are presented in Figure 2a,b,c, respectively. In addition, ADR-activating potency (EC50) and efficacy (Emax) values of the selected compounds are presented in Table 1. Adrenaline activated ADRα1A, α1B and α1D with EC50 values of 6.5 nM, 6.1 nM and 1.7 nM, respectively. Four of the selected phenethylamines were found to activate ADRα1A. p-octopamine and p-synephrine activated ADRα1A with EC50 values of 11 µM and 2.4 µM, respectively, and Emax values of 87% and 82%. Furthermore, halostachine and methylsynephrine activated ADRα1A with EC50 values of 8.7 µM and 44 µM and Emax values of 59% and 16%, respectively. Similarly, five compounds activated ADRα1B and ADRα1D. p-Octopamine and p-synephrine activated ADRα1B and ADRα1D with EC50 values of 3.9 µM and 0.66 µM, respectivelyfor ADRα1B (Emax = 94% and 91%); the EC50 values for ADRα1D were 1.2 µM and 1.7 µM, respectively (Emax = 100% and 80%). Additionally, halostachine activated ADRα1B and ADRα1D with EC50 values of 1.1 µM and 2.1 µM, respectively, and acted as a partial agonist (Emax = 77% for ADRα1B and Emax = 82% for ADRα1D). Both hordenine and dimethylphenethylamine acted as weak agonists for ADRα1B and ADRα1D, with EC50 values of 5.7 µM and 6.1 µM, respectively; for ADRα1B (Emax = 37% and 19%), the values were 34 µM and 8.4 µM, respectively, and for ADRα1D, Emax = 23% and 7.9%. The other phenethylamine analogues and all of the alkylamines did not activate these ADRs at concentrations up to 300 µM.

3.2. Adrenergic Receptor α2A/B Activation

The concentration–response relationships of the compounds that activated ADRα2A are presented in Figure 3. The concentration–response relationship of adrenaline at ADRα2B is shown in Figure A2 in Appendix B. In addition, the activating potency (EC50) and efficacy (Emax) values of the selected compounds are presented in Table 1. Adrenaline activated ADRα2A with an EC50 value of 8.4 nM and ADRα2B with an EC50 value of 23 nM. ADRα2A was only activated by the compounds p-synephrine (EC50 = 100 µM and Emax = 89%) and hordenine (EC50 = 690 µM and Emax = 12%) (Figure 3). The other phenethylamine analogues and all of the alkylamines did not activate these ADRs at concentrations up to 300 µM.

3.3. Adrenergic Receptor β1/2 Activation

The concentration–response curves of the studied compounds that activated ADRβ1 and ADRβ2 are presented in Figure 4a,b. In addition, the activating potency (EC50) and efficacy (Emax) values of the selected compounds are presented in Table 1. Adrenaline activated ADRβ1 with an EC50 of 9.9 nM and ADRβ2 with an EC50 of 67 nM. Five of the studied compounds were found to activate ADRβ1 and only one compound activated ADRβ2. Isopropyloctopamine and higenamine exhibited strong agonistic properties at ADRβ1, with EC50 values of 117 nM and 34 nM, respectively (Emax = 105% for both). In comparison, p-octopamine, p-synephrine and methylsynephrine were less potent, with EC50 values of 5.5 µM, 28 µM and 25 µM, respectively (Emax = 88%, 64% and 75%). ADRβ2 was only partially activated by higenamine, with an EC50 value of 0.47 µM (Emax = 31%). The other phenethylamine analogues and all of the alkylamines did not activate these ADRs at concentrations up to 300 µM.

3.4. Trace Amine-Associated Receptor 1 (TAAR1) Activation

The concentration–response curves of the studied compounds that activated TAAR1 are presented in Figure 5. In addition, the activating potency (EC50) and efficacy (Emax) values of the selected compounds are presented in Table 1. Phenethylamine activated TAAR1 with an EC50 value of 8.8 µM and an Emax value of 97%. Ten of the selected phenethylamine analogues were found to activate TAAR1. Higenamine, β-methylphenethylamine, tyramine and methyltyramine were found to be the most potent agonists, with EC50 values of 0.98 µM (Emax = 93%), 2.1 µM (Emax = 77%), 9.5 µM (Emax = 77%) and 23 µM (Emax = 83%), respectively. Isopropyloctopamine also had an EC50 value in this lower range (EC50 = 1.8 µM) but was least effective (Emax = 40%). In addition, dimethylphenethylamine, p-octopamine, hordenine, halostachine and p-synephrine activated TAAR1 with EC50 values of 21 µM (Emax = 64%), 46 µM (Emax = 85%), 47 µM (Emax = 82%), 74 µM (Emax = 104%) and 92 µM (Emax = 85%), respectively. Methylsynephrine did not activate TAAR1 at concentrations up to 300 µM. Alkylamine analogues 1,3-dimethylamylamine, dimethylbutylamine and dimethylhexylamine activated TAAR1 only at a concentration of 300 µM, with an effect of 52%, 20% and 36%, respectively, of the maximal effect obtained by phenethylamine (see Figure A3 in Appendix C). Dimethylaminoethanol activated the receptor with an effect of approximately 5% of the maximal effect obtained by phenethylamine.
Table 1. Human adrenergic receptor (ADR)- and trace amine-associated receptor 1 (TAAR1)-activating potency (EC50) and efficacy (Emax) of adrenaline and the selected phenethylamine and alkylamine analogues. Emax is the maximal signal obtained by the full ADR agonist adrenaline or the full TAAR1 agonist phenethylamine. Compounds with a dash (–) showed no effects at concentrations up to 300 µM (n = 3). Compounds with an * showed only an effect at 300 µM and their effects at 300 µM are presented in the Emax column as % of the maximal effect obtained by phenethylamine. Values are shown as an average (95% confidence interval) of 7–9 independent experiments with 2 technical replicates for ADR and 3–5 independent experiments with 2 technical replicates for TAAR1.
Table 1. Human adrenergic receptor (ADR)- and trace amine-associated receptor 1 (TAAR1)-activating potency (EC50) and efficacy (Emax) of adrenaline and the selected phenethylamine and alkylamine analogues. Emax is the maximal signal obtained by the full ADR agonist adrenaline or the full TAAR1 agonist phenethylamine. Compounds with a dash (–) showed no effects at concentrations up to 300 µM (n = 3). Compounds with an * showed only an effect at 300 µM and their effects at 300 µM are presented in the Emax column as % of the maximal effect obtained by phenethylamine. Values are shown as an average (95% confidence interval) of 7–9 independent experiments with 2 technical replicates for ADR and 3–5 independent experiments with 2 technical replicates for TAAR1.
ADRα1A ADRα1B ADRα1D ADRα2A ADRβ1 ADRβ2 TAAR1
EC50 (M)Emax (%)EC50 (M)Emax (%)EC50 (M)Emax (%)EC50 (M)Emax (%)EC50 (M)Emax (%)EC50 (M)Emax (%)EC50 (M)Emax (%)
Adrenaline6.5 × 10−9
(6.0–7.0 × 10−9)
98
(98–98)
6.1 × 10−10
(5.6–6.7 × 10−10)
99
(98–99)
1.7 × 10−9
(1.5–1.9 × 10−9)
99
(97–100)
8.4 × 10−9
(7.4–9.4 × 10−9)
105
(104–106)
9.9 × 10−9
(3.6–5.9 × 10−9)
99
(97–100)
6.7 × 10−8
(5.4–7.9 × 10−8)
104
(103–105)
Phenethylamines
Phenethylamine------------8.8 × 10−6
(6.8–11 × 10−6)
97
(96–99)
β-Methylphenethyl-amine------------2.1 × 10−6
(1.9–2.3 × 10−6)
77
(74–80)
Tyramine------------9.5 × 10−6
(5.3–14 × 10−6)
77
(75–80)
Methyltyramine---- -------2.3 × 10−5
(1.8–2.9 × 10−5)
83
(78–87)
p-Synephrine2.4 × 10−6
(2.1–2.7 × 10−6)
82
(79–84)
6.6 × 10−7
(5.8–7.4 × 10−7)
91
(89–92)
1.7 × 10−6
(1.4–1.9 × 10−6)
80
(78–81)
1.0 × 10−4
(0.87–1.2 × 10−4)
89
(83–94)
2.8 × 10−5
(2.6–3.0 × 10−6)
64
(59–69)
--9.2 × 10−5
(7.8–11 × 10−5)
85
(82–87)
Methylsynephrine4.4 × 10−5
(3.5–5.3 × 10−5)
16
(14–18)
------2.5 × 10−5
(2.3–2.8 × 10−5)
75
(70–80)
----
Higenamine--------3.4 × 10−8
(2.8–4.0 × 10−8)
105
(102–108)
4.7 × 10−7
(4.2–5.3 × 10−7)
31
(29–33)
9.8 × 10−7
(8.4–11 × 10−7)
93
(80–106)
Halostachine8.7 × 10−6
(7.1–10 × 10−6)
59
(57–60)
1.1 × 10−6
(0.98–1.2 × 10−6)
77
(75–80)
2.1 × 10−6
(1.9–2.3 × 10−6)
82
(79–85)
------7.4 × 10−5
(6.4–8.4 × 10−5)
100
(94–114)
Hordenine--5.7 × 10−6
(5.3–6.0 × 10−6)
37
(34–39)
3.4 × 10−5
(2.2–4.5 × 10−5)
23
(20–25)
6.9 × 10−4
(3.7–10 × 10−4)
28
(22–33)
----4.7 × 10−5
(3.5–5.8 × 10−5)
82
(78–86)
p-Octopamine1.1 × 10−5
(0.93–1.2 × 10−5)
87
(86–89)
3.9 × 10−6
(3.5–4.3 × 10−6)
94
(92–96)
1.2 × 10−6
(1.0–1.3 × 10−6)
100
(99–102)
--5.5 × 10−6
(5.0–6.0 × 10−6)
88
(87–90)
--4.6 × 10−5
(3.8–5.3 × 10−5)
85
(77–92)
Isopropyloctopamine--------1.2 × 10−7
(0.99–1.4 × 10−7)
105
(100–109)
--1.8 × 10−6
(0.95–2.6 × 10−6)
40
(35–45)
Dimethylphenethylamine--6.1 × 10−6
(5.4–6.9 × 10−6)
19
(17–21)
8.4 × 10−6
(7.3–9.6 × 10−6)
7.9
(7.3–8.5)
------2.1 × 10−5
(1.2–2.9 × 10−5)
64
(59–70)
Alkylamines
1,3-Dimethylamylamine------------*55
(50–61)
Dimethylbutylamine------------*20
(17–23)
Dimethylhexylamine------------*5.3
(3.4–7.2)
Dimethylaminoethanol------------*36
(34–38)

4. Discussion

In this study, the pharmacological profile of a group phenethylamine and alkylamine analogues, which are frequently detected in sport and weight loss supplements [8,11,12,13,14,15,16,17,18,19,20,21,22,23], was determined by assessing the receptor-activating potencies and efficacies at human ADRα1A, α1B, α1D, α2a, α2B, β1 and β2 and TAAR1. These receptors play a key role in the homeostatic regulation of the (cardio)vascular system, mainly by means of (in)direct stimulation of the sympathetic nervous system (SNS), which regulates heart rate and blood pressure [28,30]. During rest, the SNS and the parasympathetic nervous system (PNS) are tightly kept in balance by performing the opposite physiological functions [33]. In a state of exercise, this balance shifts toward the SNS, which becomes more activated in order to increase the oxygen supply in the exercising muscles. To achieve this increased oxygen delivery, the respiratory and cardiovascular system are stimulated directly by ADRs and indirectly by TAAR1. Our results show that multiple phenethylamine analogues are pharmacologically active at these receptors, which suggests that they can activate the SNS in addition to exercise.
ADRs selectively bind the catecholamines noradrenaline and adrenaline after their release from sympathetic nerve endings or the adrenal gland. This interaction leads to an intracellular signalling cascade that ultimately results in a physiological effect, such as vascular smooth muscle contraction and increased heart rate, both being critically involved in cardiac function and blood pressure homeostasis. ADRs can generally be classified into α1, α2, β1 and β2 receptors, each having different roles in the regulation of cardiovascular homeostasis [34].
The α ADRs modulate sympathetic activity in the central and peripheral nervous system and thereby control cardiovascular function. ADRα1 is expressed in vascular smooth muscle cells and plays a major role in blood pressure regulation by mediating vascular smooth muscle contraction [35]. The analogues that activated ADRα1 in the present study were mainly partial agonists and were more than hundred times less potent than the endogenous agonist adrenaline. p-Synephrine and p-octopamine were the most effective and potent phenethylamines on the ADRα1 subtypes in this study, with EC50 values more than 1000 times higher than reported plasma concentrations in humans after single oral doses of 20–50 mg p-synephrine [36,37]. Haller et al. (2005) showed an increase in both systolic and diastolic blood pressure in humans after consumption of a weight loss supplement containing a mixture of PASs, including p-synephrine [36]. Remarkably, increasing the dose of p-synephrine in this mixture did not alter the blood pressure, which may imply that p-synephrine alone was not responsible for the increase in blood pressure. Another explanation could be that p-synephrine is not a selective ADR α agonist and that its ADRα1 effects may be counteracted by the activation of central ADRα2A. However, it is difficult to causally link any of the observed effects to one of the ingredients, as the nutritional supplement also contained other PASs that were known for their cardiovascular side effects [38]. This emphasises the need for research approaches that study single-ingredient effects before studying mixture pharmacology. So far, no studies have been reported on the effects of p-octopamine on blood pressure in humans [39,40]. Pharmacodynamic and pharmacokinetic research of most of the studied phenethylamines is scarce. A few pharmacokinetic studies on the partial ADRα1 agonist hordenine after oral consumption in humans, with limited subject numbers (n = 3), are published. Human plasma levels of 2.7 nM and 15 nM after a 100 mg and 5.5 mg oral dose, respectively, were previously reported [41,42]. These oral doses did not result in plasma concentrations in the same range as the EC50 values in our study, which are three orders of magnitude higher. However, Biesterbos et al. (2019) reported a maximal daily dose of 669 mg hordenine in nutritional supplements, which indicates that the hordenine intake via nutritional supplements could be higher [11]. To date, no studies on the effects of hordenine on the cardiovascular system in humans have been published. In horses, hordenine was found to activate the sympathetic nervous system (SNS), as it increased heart rate and respiratory rate [43]. Hence, our study suggests that hordenine could have effects on blood pressure in humans, shown by its activating properties on ADRα1B and ADRα1D.
Our study also pharmacologically characterised the selected phenethylamines and alkylamines on two ADRα2 subtypes: ADRα2A and ADRα2B. The α2 ADR is involved in the regulation of peripheral vascular resistance. None of the studied phenethylamines and alkylamines activated ADRα2B. Only p-synephrine and hordenine were able to partially activate ADRα2A, with potencies that were >12000 times lower compared to the endogenous agonist adrenaline. Hordenine showed an efficacy smaller than 30% compared to adrenaline and therefore is unlikely to exhibit major effects on blood pressure control via ADR α2A. p-synephrine had an efficacy of 89% and is therefore more likely to cause physiological effects. ADRα2 activation is shown to result in divergent physiological effects, depending on the site of action. The activation of ADRα2A in the CNS decreases blood pressure by inhibiting synaptic noradrenaline release. On the other hand, ADRα2A in vascular smooth muscle cells causes smooth muscle cell contraction, leading to vasoconstriction and, in this way, increases blood pressure [44,45].
Besides ADRα1 and ADRα2, the activating effects of the phenethylamine analogues were also screened for β1 ADRs, which play a major role in regulating cardiac contractility and heart rate. Similarly to α1 and α2, p-synephrine also acted as an ADRβ1 agonist. The addition of a methyl group to the carbon chain of p-synephrine did not alter its efficacy and potency on ADRβ1, as methylsynephrine showed similar results. Both compounds were approximately 2500 times less potent than the endogenous agonist adrenaline. p-Octopamine was slightly more effective and >500 times less potent than adrenaline on this receptor subtype. Both isopropyloctopamine and higenamine acted as full agonists and were found to have potency values in the same order of magnitude as adrenaline (approximately 10 and 3 times less potent than adrenaline). As ADRβ1 is predominantly located on cardiomyocytes in the heart, these findings suggest that these compounds may increase cardiac contraction force and rate [46]. This is also shown by Feng et al. (2012) who established a significant increased heart rate from 68 bpm at baseline to 126 bpm during a maximal plasma concentration of 8.4 nM higenamine in humans [47]. Case reports have revealed sympathetic overstimulation (leading to adverse effects such as palpitations, chest pain, tachycardia and even cardiac arrest) after consumption of multi-ingredient weight loss supplements containing isopropyloctopamine [8,17]. An increased heart rate was also observed after intravenous administration of isopropyloctopamine in subjects pre-treated with the non-selective ADRβ antagonist propranolol [48], which suggests that isopropyloctopamine competes with this ADRβ antagonist for receptor binding. These reports clearly highlight the cardiovascular risks of using sympathomimetic compounds like isopropyloctopamine in relatively high doses.
The β2 ADR plays an important in role bronchodilation and the regulation of vascular homeostasis by mediating vascular smooth muscle relaxation and it contributes to an increased heart contraction force and rate after activation in the heart [49,50]. Compared to ADRβ1, it less intensively contributes to this latter effect as the ratio of ADRβ1 and ADRβ2 is 77% to 23%, respectively [51]. Higenamine was the only analogue found to act as a partial ADRβ2 agonist in the present study and was found to be seven times less potent than the full agonist adrenaline. Feng et al. (2012) observed that higenamine administration led to a slight decrease in diastolic blood pressure in humans, which could be explained by ADRβ2-mediated vasodilatation [47].
TAAR1 is expressed in both brain and peripheral tissues (e.g., blood vessels and the heart), where it is suggested to play a role in both direct and indirect activation of the SNS [27,28,52,53]. In the brain, TAAR1 is involved in stimulating synaptic neurotransmitter release and in synaptic neurotransmitter uptake inhibition [54], which both indirectly activate the SNS. Peripheral TAAR1 is found to indirectly activate the SNS by stimulating noradrenaline release in nerve endings and by a direct effect on vasoconstriction [55]. Broadley and Mehta (2023) recently proposed that phenethylamine-induced vasoconstriction in human, pig and rat blood vessels was a direct effect of TAAR1, as an ADR α1 antagonist did not inhibit vasoconstriction [28,56,57,58]. In the present study, ten of the studied phenethylamine analogues acted as TAAR1 agonists, with their efficacy ranging from 40 to 100%. Strikingly, β-methylphenethylamine, isopropyloctopamine and higenamine were more potent than the endogenous agonist phenethylamine. The other studied phenethylamine analogues were less potent than phenethylamine, but the EC50 values were still within a 10-fold change. Phenethylamine, tyramine, p-octopamine and p-synephrine are endogenous agonists of TAAR1 that are present in the CNS at concentrations more than hundred times lower than their traditional analogues (e.g., dopamine, serotonin and noradrenaline) and are therefore called “trace amines” [59]. Although the exact (patho)physiological role of TAAR1 is not known yet, the use of TAAR1 agonists is linked to severe cardiovascular health effects. Supplemental use of β-methylphenethylamine is previously linked to haemorrhagic strokes in exercising individuals [4,5]. Additionally, β-methylphenethylamine also elevated blood pressure in rats through indirect activation of the SNS with a similar efficacy to its isomer amphetamine, which is a well-known TAAR1-agonist [60,61]. These amphetamine-like effects caused by higher concentrations of trace amines (reached, for instance, by means of supplemental intake) are different from the effects caused by the endogenous trace amines at concentrations a hundred to a thousand times lower. At high concentrations, the activation of TAAR1 in the CNS will affect neurotransmission by increasing neurotransmitter release and inhibiting the reuptake of dopamine, noradrenaline and serotonin. This will consequently lead to overactivation of the SNS and the consequential overstimulation of the cardiovascular system [54,59,62]. These data show that the use of phenethylamines in high doses via supplements may stimulate the cardiovascular system and thereby pose a significant risk to human health.
In order to exert effects via TAAR1 localised in the brain, agonists need to cross the blood–brain barrier and pass through the cell membrane, as TAAR1—unlike other G protein-coupled receptors—is localised in the cytoplasm [61]. Tyramine and phenethylamines have previously been shown to pass through lipid bilayers by means of passive diffusion [63]. Additionally, many trace amines have a high affinity for Organic Cation Transporter 2 (OCT2) and dopamine, noradrenaline and serotonin transporters [27,64] and are therefore expected to reach intracellular TAAR1.
The alkylamine analogue 1,3-dimethylamylamine exclusively and partially activated TAAR1, but only at a concentration of 300 µM, which is too high to be physiologically relevant. The other alkylamine analogues also activated TAAR1 only at 300 µM but were even less effective. Neither of the alkylamine analogues activated ADRs at concentrations up to 300 µM. However, multiple case studies describe a link between adverse cardiovascular events and the consumption of pre-workout supplements containing these substances [65,66,67]. These studies suggest that also alkylamines might have sympathomimetic effects after consumption. Our current study shows that these effects cannot be mechanistically explained by their interaction with ADRs and probably not either by TAAR1. Recent studies have indeed proposed different pharmacological mechanisms for these types of substances, namely, their interaction with noradrenaline and dopamine transporters in the brain [25,68].
The links between cardiovascular toxicity and the use of weight loss and pre-workout supplements are in line with the historical evolution of the development of obesity medication [69]. Sympathomimetics are particularly of interest for weight loss medication as part of obesity treatment [70]. The application of sympathomimetics in weight loss management has extensively been studied in the past. Nevertheless, only a few drugs are currently approved (e.g., phentermine/topiramate, liraglutide, semaglutide, naltrexone/bupropion and orlistat), which are all indirectly acting sympathomimetics [71]. Cardiovascular side effects have been the main reason for the withdrawal of most of these drugs [69,71,72]. In particular, sympathomimetics (e.g., sibutramine, phentermine, methamphetamine and fenfluramine) that inhibit neurotransmitter reuptake or stimulate neurotransmitter release have failed to fulfil cardiovascular safety standards [69,72]. Despite the fact that they have failed to be successful for safe weight loss, a lot of these withdrawn sympathomimetics are nowadays drugs of abuse (e.g., in weight loss and pre-workout supplements) [73]. In this study, we showed that phenethylamine analogues are another potential group of compounds of abuse, as they could potentially act as sympathomimetics by activating ADRs or TAAR1.

5. Conclusions

Taken together, our results reveal the pharmacological activity of a selection of phenethylamine analogues regarding ADRs (α1, α2, β1 and β2) and TAAR1. Supplements containing these compounds are highly popular among (amateur and professional sports) individuals to improve their performance and/or to lose weight. Many of the phenethylamine analogues were shown to be (potent) activators of ADRs and TAAR1. Remarkably, some of the compounds (isopropyloctopamine and higenamine) have a potency comparable to the endogenous agonists for the receptor. Moreover, several phenethylamines have agonistic activity on multiple of these receptors (Table 1), indicating that they might further stimulate the already activated SNS in exercising individuals through multiple ways. For instance, higenamine is a potent agonist for both TAAR1 and ADR β1 and p-synephrine is a potent agonist for α1, α2, β1 and TAAR1, meaning that these compounds could affect the cardiovascular system directly via the ADRs and TAAR1 in the heart and vasculature and indirectly via TAAR1 in the brain. These individual effects may both contribute to an amplified combined effect on the cardiovascular system and may thereby adversely affect cardiovascular parameters like blood pressure and heart rate. Besides this, users may not always be aware of the presence of phenethylamine or its analogues, since for many of these supplements, the names of the plant-derived extracts are listed on the label, like Citrus aurantium (bitter orange), Acacia rigidula, Hordeum vulgare (barley) or Ariocarpus retusus (cactus), which are natural sources of phenethylamine and many of its studied analogues [22,74,75,76,77]. Consequently, the use of supplements containing one, or a combination of, phenethylamine(s) may pose a serious health risk to their consumers, especially when used by obese individuals with an already increased risk of cardiovascular diseases [78]. The collection of case reports on people using these supplements containing PASs indeed confirms this health risk, which is in line with the proven history of the development of obesity medication and its corresponding cardiotoxicity. However, since pharmacodynamic and pharmacokinetic information about the ingredients of these supplements is often lacking, more research needs to focus on the physiological effects of these compounds in humans.

Author Contributions

M.F.V., A.O. and F.-J.v.S. conceived the project. N.E.T.P., W.M.B., F.-J.v.S. and M.F.V. designed the project. N.E.T.P. and A.M. performed data acquisition. X.S. developed the HEK293T-hTAAR1 stable cell line. N.E.T.P. created the figures and tables. N.E.T.P. and M.F.V. wrote the manuscript. W.M.B., A.M. and X.S. reviewed the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This study was funded by the Dutch Ministry of Agriculture, Nature and Food Quality.

Data Availability Statement

The original contributions presented in this study are included in the article; further inquiries can be directed to the corresponding author.

Acknowledgments

We would like to thank Aaron Janowsky (VA Senior Research Career Scientist Award 5IK6BX005754) for kindly giving us the HEK293T-hTAAR1 stable cell line.

Conflicts of Interest

The authors declare no conflicts of interest.

Appendix A

Figure A1. (A1) Gene expression of human trace amine-associated receptor 1 (hTAAR1) as fold change in GAPDH in untransfected HEK293T cells and in hTAAR1-transfected HEK293T cells. Data are presented as mean (± SEM) of 2–3 independent experiments which were run in duplicate. (A2) cAMP production in cells as percentage of maximal signal obtained by phenethylamine (PEA) in hTAAR1-transfected HEK293T cells exposed to PEA (triangles) or RO5166017 (squares) and in untransfected HEK293T cells exposed to PEA (diamonds). Data are presented as mean (±SEM) of 2–3 independent experiments which were run in duplicate.
Figure A1. (A1) Gene expression of human trace amine-associated receptor 1 (hTAAR1) as fold change in GAPDH in untransfected HEK293T cells and in hTAAR1-transfected HEK293T cells. Data are presented as mean (± SEM) of 2–3 independent experiments which were run in duplicate. (A2) cAMP production in cells as percentage of maximal signal obtained by phenethylamine (PEA) in hTAAR1-transfected HEK293T cells exposed to PEA (triangles) or RO5166017 (squares) and in untransfected HEK293T cells exposed to PEA (diamonds). Data are presented as mean (±SEM) of 2–3 independent experiments which were run in duplicate.
Nutrients 16 01567 g0a1

Appendix B

Figure A2. Concentration–response relationship (mean ± SD) of adrenaline (closed circles) that activated adrenergic receptor (ADR) α2B in chem-1 cells. Data are presented as percentages of maximal fluorescent Ca2+ response obtained by adrenaline (n = 8).
Figure A2. Concentration–response relationship (mean ± SD) of adrenaline (closed circles) that activated adrenergic receptor (ADR) α2B in chem-1 cells. Data are presented as percentages of maximal fluorescent Ca2+ response obtained by adrenaline (n = 8).
Nutrients 16 01567 g0a2

Appendix C

Figure A3. Concentration–response relationships (mean ± SD) of alkylamine analogues 1,3-dimethylamylamine (closed bar), dimethylbutylamine (vertical striped bar), dimethylaminoethanol (open bar) and dimethylhexylamine (checkered bar) that activated human trace amine-associated receptor 1 (TAAR1) in HEK293T cells transfected with this receptor at a concentration of 300 µM. The data are presented as percentages of the maximal intracellular cAMP level obtained by phenethylamine (n = 3).
Figure A3. Concentration–response relationships (mean ± SD) of alkylamine analogues 1,3-dimethylamylamine (closed bar), dimethylbutylamine (vertical striped bar), dimethylaminoethanol (open bar) and dimethylhexylamine (checkered bar) that activated human trace amine-associated receptor 1 (TAAR1) in HEK293T cells transfected with this receptor at a concentration of 300 µM. The data are presented as percentages of the maximal intracellular cAMP level obtained by phenethylamine (n = 3).
Nutrients 16 01567 g0a3

References

  1. Alonso, M.R.; Fernández-García, B. Evolution of the use of sports supplements. PharmaNutrition 2020, 14, 100239. [Google Scholar] [CrossRef]
  2. Wardenaar, F.C.; Ceelen, I.J.M.; Van den Dool, R.; Witkamp, R.; Mensink, M. Het gebruik van voedingssupplementen en sportvoeding door meer en minder actieve Nederlanders—Een inventarisatie op basis van het Nationaal Sport Onderzoek 2012 onder de Nederlandse bevolking. Ned. Tijdschr. Voor Voeding Dietiek 2014, 69, 9. [Google Scholar]
  3. Razenberg, L.; Notenboom, S.; De Wit-Bos, L. Use of Workout Supplements by Sport Participants in The Netherlands; National Institute for Public Health and the Environment: Bilthoven, The Netherlands, 2021; Volume 0116. [CrossRef]
  4. Cohen, P.A.; Zeijlon, R.; Nardin, R.; Keizers, P.H.J.; Venhuis, B. Hemorrhagic Stroke Probably Caused by Exercise Combined With a Sports Supplement Containing β-Methylphenyl-ethylamine (BMPEA): A Case Report. Ann. Intern. Med. 2015, 162, 879–880. [Google Scholar] [CrossRef] [PubMed]
  5. Harris, B.F.; Winn, C.; Ableman, T.B. Hemorrhagic Stroke in a Young Healthy Male Following Use of Pre-Workout Supplement Animal Rage XL. Mil. Med. 2017, 182, e2030–e2033. [Google Scholar] [CrossRef] [PubMed]
  6. Smith, T.B.; Staub, B.A.; Natarajan, G.M.; Lasorda, D.M.; Poornima, I.G. Acute myocardial infarction associated with dietary supplements containing 1,3-dimethylamylamine and Citrus aurantium. Tex. Heart Inst. J. 2014, 41, 70–72. [Google Scholar] [CrossRef] [PubMed]
  7. Zeijlon, R.; Hantelius, V.; Wallerstedt, S.M.; Holmqvist, L. Sports nutrition supplements and adverse events—A meta-epidemiological study of case reports specifically addressing causality assessment. Eur. J. Clin. Pharmacol. 2022, 78, 1–9. [Google Scholar] [CrossRef]
  8. Bovee, T.F.; Mol, H.G.; Bienenmann-Ploum, M.E.; Heskamp, H.H.; Van Bruchem, G.D.; Van Ginkel, L.A.; Kooijman, M.; Lasaroms, J.J.; Van Dam, R.; Hoogenboom, R.L. Dietary supplement for energy and reduced appetite containing the β-agonist isopropyloctopamine leads to heart problems and hospitalisations. Food Addit. Contam. Part A Chem. Anal. Control Expo. Risk Assess. 2016, 33, 749–759. [Google Scholar] [CrossRef] [PubMed]
  9. De Jonge, M.L.L.; Kieviet, L.C.; Sierts, M.; Egberink, L.B.; Van der Heyden, M.A.G. Review of Case Reports on Adverse Events Related to Pre-workout Supplements Containing Synephrine. Cardiovasc. Toxicol. 2023, 23, 1–9. [Google Scholar] [CrossRef] [PubMed]
  10. Adami, P.E.; Koutlianos, N.; Baggish, A.; Bermon, S.; Cavarretta, E.; Deligiannis, A.; Furlanello, F.; Kouidi, E.; Marques-Vidal, P.; Niebauer, J.; et al. Cardiovascular effects of doping substances, commonly prescribed medications and ergogenic aids in relation to sports: A position statement of the sport cardiology and exercise nucleus of the European Association of Preventive Cardiology. Eur. J. Prev. Cardiol. 2022, 29, 559–575. [Google Scholar] [CrossRef]
  11. Biesterbos, J.W.H.; Sijm, D.; Van Dam, R.; Mol, H.G.J. A health risk for consumers: The presence of adulterated food supplements in the Netherlands. Food Addit. Contam. Part A Chem. Anal. Control Expo. Risk Assess. 2019, 36, 1273–1288. [Google Scholar] [CrossRef]
  12. Cohen, P.A.; Travis, J.C.; Venhuis, B.J. A synthetic stimulant never tested in humans, 1,3-dimethylbutylamine (DMBA), is identified in multiple dietary supplements. Drug Test. Anal. 2015, 7, 83–87. [Google Scholar] [CrossRef] [PubMed]
  13. Cohen, P.A.; Bloszies, C.; Yee, C.; Gerona, R. An amphetamine isomer whose efficacy and safety in humans has never been studied, β-methylphenylethylamine (BMPEA), is found in multiple dietary supplements. Drug Test. Anal. 2016, 8, 328–333. [Google Scholar] [CrossRef] [PubMed]
  14. Cohen, P.A.; Travis, J.C.; Keizers, P.H.J.; Deuster, P.; Venhuis, B.J. Four experimental stimulants found in sports and weight loss supplements: 2-amino-6-methylheptane (octodrine), 1,4-dimethylamylamine (1,4-DMAA), 1,3-dimethylamylamine (1,3-DMAA) and 1,3-dimethylbutylamine (1,3-DMBA). Clin. Toxicol. 2018, 56, 421–426. [Google Scholar] [CrossRef]
  15. Cohen, P.; Travis, J.C.; Keizers, P.H.J.; Boyer, F.E.; Venhuis, B.J. The stimulant higenamine in weight loss and sports supplements. Clin. Toxicol. 2019, 57, 125–130. [Google Scholar] [CrossRef]
  16. Cohen, P.A.; Travis, J.C.; Vanhee, C.; Ohana, D.; Venhuis, B.J. Nine prohibited stimulants found in sports and weight loss supplements: Deterenol, phenpromethamine (Vonedrine), oxilofrine, octodrine, beta-methylphenylethylamine (BMPEA), 1,3-dimethylamylamine (1,3-DMAA), 1,4-dimethylamylamine (1,4-DMAA), 1,3-dimethylbutylamine (1,3-DMBA) and higenamine. Clin. Toxicol. 2021, 59, 975–981. [Google Scholar] [CrossRef] [PubMed]
  17. Venhuis, B.; Keizers, P.; van Riel, A.; De Kaste, D. A cocktail of synthetic stimulants found in a dietary supplement associated with serious adverse events. Drug Test. Anal. 2014, 6, 578–581. [Google Scholar] [CrossRef] [PubMed]
  18. Avula, B.; Bae, J.Y.; Chittiboyina, A.G.; Wang, Y.H.; Wang, M.; Khan, I.A. Liquid chromatography-quadrupole time of flight mass spectrometric method for targeted analysis of 111 nitrogen-based compounds in weight loss and ergogenic supplements. J. Pharm. Biomed. Anal. 2019, 174, 305–323. [Google Scholar] [CrossRef] [PubMed]
  19. Duiven, E.; Van Loon, L.J.C.; Spruijt, L.; Koert, W.; De Hon, O.M. Undeclared Doping Substances are Highly Prevalent in Commercial Sports Nutrition Supplements. J. Sports Sci. Med. 2021, 20, 328–338. [Google Scholar] [CrossRef] [PubMed]
  20. Zhao, J.; Wang, M.; Avula, B.; Khan, I.A. Detection and quantification of phenethylamines in sports dietary supplements by NMR approach. J. Pharm. Biomed. Anal. 2018, 151, 347–355. [Google Scholar] [CrossRef]
  21. Yun, J.; Kwon, K.; Choi, J.; Jo, C.H. Monitoring of the amphetamine-like substances in dietary supplements by LC-PDA and LC-MS/MS. Food Sci. Biotechnol. 2017, 26, 1185–1190. [Google Scholar] [CrossRef]
  22. Pawar, R.S.; Sagi, S.; Leontyev, D. Analysis of bitter orange dietary supplements for natural and synthetic phenethylamines by LC-MS/MS. Drug Test. Anal. 2020, 12, 1241–1251. [Google Scholar] [CrossRef]
  23. ElSohly, M.A.; Gul, W. LC-MS-MS analysis of dietary supplements for N-ethyl-α-ethyl-phenethylamine (ETH), N, N-diethylphenethylamine and phenethylamine. J. Anal. Toxicol. 2014, 38, 63–72. [Google Scholar] [CrossRef]
  24. Hondebrink, L.; Zwartsen, A.; Westerink, R.H.S. Effect fingerprinting of new psychoactive substances (NPS): What can we learn from in vitro data? Pharmacol. Ther. 2018, 182, 193–224. [Google Scholar] [CrossRef]
  25. Rickli, A.; Hoener, M.C.; Liechti, M.E. Pharmacological profiles of compounds in preworkout supplements (“boosters”). Eur. J. Pharmacol. 2019, 859, 172515. [Google Scholar] [CrossRef]
  26. Rebec, G.V. Cocaine and Amphetamines. In Encyclopedia of Life Sciences; John Wiley & Sons: Hoboken, NJ, USA, 2012. [Google Scholar] [CrossRef]
  27. Rutigliano, G.; Accorroni, A.; Zucchi, R. The Case for TAAR1 as a Modulator of Central Nervous System Function. Front. Pharmacol. 2017, 8, 987. [Google Scholar] [CrossRef]
  28. Fehler, M.; Broadley, K.J.; Ford, W.R.; Kidd, E.J. Identification of trace-amine-associated receptors (TAAR) in the rat aorta and their role in vasoconstriction by β-phenylethylamine. Naunyn Schmiedeberg’s Arch. Pharmacol. 2010, 382, 385–398. [Google Scholar] [CrossRef]
  29. Broadley, K.J. The vascular effects of trace amines and amphetamines. Pharmacol. Ther. 2010, 125, 363–375. [Google Scholar] [CrossRef]
  30. Graham, R.M. Adrenergic receptors: Structure and function. Clevel. Clin. J. Med. 1990, 57, 481–491. [Google Scholar] [CrossRef]
  31. Wardenaar, F.; Van den Dool, R.; Ceelen, I.; Witkamp, R.; Mensink, M. Self-Reported Use and Reasons among the General Population for Using Sports Nutrition Products and Dietary Supplements. Sports 2016, 4, 33. [Google Scholar] [CrossRef]
  32. Cohen, P.A.; Avula, B.; Venhuis, B.; Travis, J.C.; Wang, Y.H.; Khan, I.A. Pharmaceutical doses of the banned stimulant oxilofrine found in dietary supplements sold in the USA. Drug Test. Anal. 2017, 9, 135–142. [Google Scholar] [CrossRef]
  33. Waxenbaum, J.A.; Reddy, V.; Varacallo, M. Anatomy, Autonomic Nervous System. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2024. Available online: https://www.ncbi.nlm.nih.gov/books/NBK539845/ (accessed on 1 December 2023).
  34. Bylund, D.B.; Eikenberg, D.C.; Hieble, J.P.; Langer, S.Z.; Lefkowitz, R.J.; Minneman, K.P.; Molinoff, P.B.; Ruffolo, R.R., Jr. Trendelenburg, U. International Union of Pharmacology nomenclature of adrenoceptors. Pharmacol. Rev. 1994, 46, 121–136. [Google Scholar] [PubMed]
  35. Docherty, J.R. Subtypes of functional α1-adrenoceptor. Cell. Mol. Life Sci. 2010, 67, 405–417. [Google Scholar] [CrossRef] [PubMed]
  36. Haller, C.A.; Benowitz, N.L.; Jacob, P. Hemodynamic effects of ephedra-free weight-loss supplements in humans. Am. J. Med. 2005, 118, 998–1003. [Google Scholar] [CrossRef] [PubMed]
  37. Haller, C.A.; Duan, M.; Jacob, P., 3rd; Benowitz, N. Human pharmacology of a performance-enhancing dietary supplement under resting and exercise conditions. Br. J. Clin. Pharmacol. 2008, 65, 833–840. [Google Scholar] [CrossRef] [PubMed]
  38. Aprotosoaie, A.C.; Miron, A.; Trifan, A.; Luca, V.S.; Costache, I.I. The Cardiovascular Effects of Cocoa Polyphenols-An Overview. Diseases 2016, 4, 39. [Google Scholar] [CrossRef] [PubMed]
  39. Stohs, S.J.; Shara, M.; Ray, S.D. p-Synephrine, ephedrine, p-octopamine and m-synephrine: Comparative mechanistic, physiological and pharmacological properties. Phytother. Res. 2020, 34, 1838–1846. [Google Scholar] [CrossRef] [PubMed]
  40. Stohs, S.J. Physiological functions and pharmacological and toxicological effects of p-octopamine. Drug Chem. Toxicol. 2015, 38, 106–112. [Google Scholar] [CrossRef] [PubMed]
  41. Sobiech, M.; Giebułtowicz, J.; Luliński, P. Application of Magnetic Core–Shell Imprinted Nanoconjugates for the Analysis of Hordenine in Human Plasma-Preliminary Data on Pharmacokinetic Study after Oral Administration. J. Agric. Food Chem. 2020, 68, 14502–14512. [Google Scholar] [CrossRef] [PubMed]
  42. Sommer, T.; Göen, T.; Budnik, N.; Pischetsrieder, M. Absorption, Biokinetics, and Metabolism of the Dopamine D2 Receptor Agonist Hordenine (N,N-Dimethyltyramine) after Beer Consumption in Humans. J. Agric. Food Chem. 2020, 68, 1998–2006. [Google Scholar] [CrossRef]
  43. Frank, M.; Weckman, T.J.; Wood, T.; Woods, W.E.; Tai, C.L.; Chang, S.L.; Ewing, A.; Blake, J.W.; Tobin, T. Hordenine: Pharmacology, pharmacokinetics and behavioural effects in the horse. Equine Vet. J. 1990, 22, 437–441. [Google Scholar] [CrossRef]
  44. Kanagy, N.L. α2-Adrenergic receptor signalling in hypertension. Clin. Sci. 2005, 109, 431–437. [Google Scholar] [CrossRef] [PubMed]
  45. Gavras, I.; Manolis, A.J.; Gavras, H. The alpha2—Adrenergic receptors in hypertension and heart failure: Experimental and clinical studies. J. Hypertens. 2001, 19, 2115–2124. [Google Scholar] [CrossRef] [PubMed]
  46. Alhayek, S.; Preuss, C.V. Beta 1 Receptors. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2024. Available online: https://www.ncbi.nlm.nih.gov/books/NBK532904/ (accessed on 14 August 2023).
  47. Feng, S.; Jiang, J.; Hu, P.; Zhang, J.Y.; Liu, T.; Zhao, Q.; Li, B.L. A phase I study on pharmacokinetics and pharmacodynamics of higenamine in healthy Chinese subjects. Acta Pharmacol. Sin. 2012, 33, 1353–1358. [Google Scholar] [CrossRef]
  48. Pilkington, T.R.E.; Lowe, R.D.; Foster, R.; Robinson, B.F.; Antonis, A. Effect of sympathomimetic compounds with Β-adrenergic effects on plasma free fatty acids in man. J. Lipid Res. 1966, 7, 73–76. [Google Scholar] [CrossRef] [PubMed]
  49. Hudzik, T.J.; Patel, M.; Brown, A. β(2)—Adrenoceptor agonist activity of higenamine. Drug Test. Anal. 2021, 13, 261–267. [Google Scholar] [CrossRef] [PubMed]
  50. Ferro, A.; Kaumann, A.J.; Brown, M.J. β-Adrenoceptor Subtypes in Human Coronary Artery: Desensitization of β2-Adrenergic Vasorelaxation by Chronic β1-Adrenergic Stimulation In Vitro. J. Cardiovasc. Pharmacol. 1995, 25, 134–141. [Google Scholar] [CrossRef] [PubMed]
  51. Bristow, M.R.; Ginsburg, R.; Umans, V.; Fowler, M.; Minobe, W.; Rasmussen, R.; Zera, P.; Menlove, R.; Shah, P.; Jamieson, S. Beta 1- and beta 2-adrenergic-receptor subpopulations in nonfailing and failing human ventricular myocardium: Coupling of both receptor subtypes to muscle contraction and selective beta 1-receptor down-regulation in heart failure. Circ. Res. 1986, 59, 297–309. [Google Scholar] [CrossRef] [PubMed]
  52. Barnes, D.A.; Galloway, D.A.; Hoener, M.C.; Berry, M.D.; Moore, C.S. TAAR1 Expression in Human Macrophages and Brain Tissue: A Potential Novel Facet of MS Neuroinflammation. Int. J. Mol. Sci. 2021, 22, 11576. [Google Scholar] [CrossRef] [PubMed]
  53. Lindemann, L.; Ebeling, M.; Kratochwil, N.A.; Bunzow, J.R.; Grandy, D.K.; Hoener, M.C. Trace amine-associated receptors form structurally and functionally distinct subfamilies of novel G protein-coupled receptors. Genomics 2005, 85, 372–385. [Google Scholar] [CrossRef]
  54. Xie, Z.; Miller, G.M. Beta-phenylethylamine alters monoamine transporter function via trace amine-associated receptor 1: Implication for modulatory roles of trace amines in brain. J. Pharmacol. Exp. Ther. 2008, 325, 617–628. [Google Scholar] [CrossRef]
  55. Koh, A.H.W.; Chess-Williams, R.; Lohning, A.E. Differential mechanisms of action of the trace amines octopamine, synephrine and tyramine on the porcine coronary and mesenteric artery. Sci. Rep. 2019, 9, 10925. [Google Scholar] [CrossRef] [PubMed]
  56. Broadley, K.J.; Mehta, D. Trace amine-induced vasoconstriction of human mammary artery and saphenous vein. Vasc. Pharmacol. 2023, 151, 107191. [Google Scholar] [CrossRef] [PubMed]
  57. Broadley, K.J.; Broadley, H.D. Non-adrenergic vasoconstriction and vasodilatation of guinea-pig aorta by β-phenylethylamine and amphetamine—Role of nitric oxide determined with L-NAME and NO scavengers. Eur. J. Pharmacol. 2018, 818, 198–205. [Google Scholar] [CrossRef] [PubMed]
  58. Herbert, A.A.; Kidd, E.J.; Broadley, K.J. Dietary trace amine-dependent vasoconstriction in porcine coronary artery. Br. J. Pharmacol. 2008, 155, 525–534. [Google Scholar] [CrossRef] [PubMed]
  59. Berry, M.D. Mammalian central nervous system trace amines. Pharmacologic amphetamines, physiologic neuromodulators. J. Neurochem. 2004, 90, 257–271. [Google Scholar] [CrossRef] [PubMed]
  60. Schindler, C.W.; Thorndike, E.B.; Rice, K.C.; Partilla, J.S.; Baumann, M.H. The Supplement Adulterant β-Methylphenethylamine Increases Blood Pressure by Acting at Peripheral Norepinephrine Transporters. J. Pharmacol. Exp. Ther. 2019, 369, 328–336. [Google Scholar] [CrossRef]
  61. Underhill, S.M.; Hullihen, P.D.; Chen, J.; Fenollar-Ferrer, C.; Rizzo, M.A.; Ingram, S.L.; Amara, S.G. Amphetamines signal through intracellular TAAR1 receptors coupled to Gα13 and GαS in discrete subcellular domains. Mol. Psychiatry 2021, 26, 1208–1223. [Google Scholar] [CrossRef]
  62. Lindemann, L.; Hoener, M.C. A renaissance in trace amines inspired by a novel GPCR family. Trends Pharmacol. Sci. 2005, 26, 274–281. [Google Scholar] [CrossRef]
  63. Berry, M.D.; Shitut, M.R.; Almousa, A.; Alcorn, J.; Tomberli, B. Membrane permeability of trace amines: Evidence for a regulated, activity-dependent, nonexocytotic, synaptic release. Synapse 2013, 67, 656–667. [Google Scholar] [CrossRef]
  64. Berry, M.D.; Hart, S.; Pryor, A.R.; Hunter, S.; Gardiner, D. Pharmacological characterization of a high-affinity p-tyramine transporter in rat brain synaptosomes. Sci. Rep. 2016, 6, 38006. [Google Scholar] [CrossRef]
  65. Eliason, M.J.; Eichner, A.; Cancio, A.; Bestervelt, L.; Adams, B.D.; Deuster, P.A. Case Reports: Death of Active Duty Soldiers Following Ingestion of Dietary Supplements Containing 1,3-Dimethylamylamine (DMAA). Mil. Med. 2012, 177, 1455–1459. [Google Scholar] [CrossRef] [PubMed]
  66. Gee, P.; Tallon, C.; Long, N.; Moore, G.; Boet, R.; Jackson, S. Use of recreational drug 1,3 Dimethylamylamine (DMAA) [corrected] associated with cerebral hemorrhage. Ann. Emerg. Med. 2012, 60, 431–434. [Google Scholar] [CrossRef] [PubMed]
  67. Lieberman, H.R.; Austin, K.G.; Farina, E.K. Surveillance of the armed forces as a sentinel system for detecting adverse effects of dietary supplements in the general population. Public Health Nutr. 2018, 21, 882–887. [Google Scholar] [CrossRef]
  68. Small, C.; Cheng, M.H.; Belay, S.S.; Bulloch, S.L.; Zimmerman, B.; Sorkin, A.; Block, E.R. The Alkylamine Stimulant 1,3-Dimethylamylamine Exhibits Substrate-Like Regulation of Dopamine Transporter Function and Localization. J. Pharmacol. Exp. Ther. 2023, 386, 266–273. [Google Scholar] [CrossRef] [PubMed]
  69. Onakpoya, I.J.; Heneghan, C.J.; Aronson, J.K. Post-marketing withdrawal of anti-obesity medicinal products because of adverse drug reactions: A systematic review. BMC Med. 2016, 14, 191. [Google Scholar] [CrossRef] [PubMed]
  70. Astrup, A.; Lundsgaard, C. What do pharmacological approaches to obesity management offer? Linking pharmacological mechanisms of obesity management agents to clinical practice. Exp. Clin. Endocrinol. Diabetes 1998, 106 (Suppl. 2), 29–34. [Google Scholar] [CrossRef] [PubMed]
  71. Chakhtoura, M.; Haber, R.; Ghezzawi, M.; Rhayem, C.; Tcheroyan, R.; Mantzoros, C.S. Pharmacotherapy of obesity: An update on the available medications and drugs under investigation. EClinicalMedicine 2023, 58, 101882. [Google Scholar] [CrossRef] [PubMed]
  72. Müller, T.D.; Blüher, M.; Tschöp, M.H.; DiMarchi, R.D. Anti-obesity drug discovery: Advances and challenges. Nat. Rev. Drug Discov. 2022, 21, 201–223. [Google Scholar] [CrossRef]
  73. White, C.M. Continued Risk of Dietary Supplements Adulterated with Approved and Unapproved Drugs: Assessment of the US Food and Drug Administration’s Tainted Supplements Database 2007 through 2021. J. Clin. Pharmacol. 2022, 62, 928–934. [Google Scholar] [CrossRef]
  74. Braga, D.; McLaughlin, J. Cactus Alkaloids, V. Isolation of Hordenine and N-Methyltyramine from Ariocarpus retusus. Planta Medica 1969, 17, 87–94. [Google Scholar] [CrossRef]
  75. Clement, B.A.; Goff, C.M.; Forbes, T.D.A. Toxic amines and alkaloids from Acacia rigidula. Phytochemistry 1998, 49, 1377–1380. [Google Scholar] [CrossRef]
  76. Sander, L.C.; Putzbach, K.; Nelson, B.C.; Rimmer, C.A.; Bedner, M.; Thomas, J.B.; Porter, B.J.; Wood, L.J.; Schantz, M.M.; Murphy, K.E.; et al. Certification of standard reference materials containing bitter orange. Anal. Bioanal. Chem. 2008, 391, 2023–2034. [Google Scholar] [CrossRef] [PubMed]
  77. Pawar, R.S.; Grundel, E.; Fardin-Kia, A.R.; Rader, J.I. Determination of selected biogenic amines in Acacia rigidula plant materials and dietary supplements using LC–MS/MS methods. J. Pharm. Biomed. Anal. 2014, 88, 457–466. [Google Scholar] [CrossRef]
  78. Guh, D.P.; Zhang, W.; Bansback, N.; Amarsi, Z.; Birmingham, C.L.; Anis, A.H. The incidence of co-morbidities related to obesity and overweight: A systematic review and meta-analysis. BMC Public Health 2009, 9, 88. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Molecular structures of (a) well-known activators of sympathetic nervous system: adrenaline, noradrenaline and dopamine, (b) phenethylamines and (c) alkylamines present in nutritional supplements.
Figure 1. Molecular structures of (a) well-known activators of sympathetic nervous system: adrenaline, noradrenaline and dopamine, (b) phenethylamines and (c) alkylamines present in nutritional supplements.
Nutrients 16 01567 g001
Figure 2. Concentration–response relationships (mean ± SD) of compounds that activated human adrenergic receptor (ADR) α1A (a), α1B (b) and α1D (c) in chem-1 cells overexpressing these receptors. Data are presented as percentages of maximal fluorescent Ca2+ response obtained by adrenaline (n = 7–9).
Figure 2. Concentration–response relationships (mean ± SD) of compounds that activated human adrenergic receptor (ADR) α1A (a), α1B (b) and α1D (c) in chem-1 cells overexpressing these receptors. Data are presented as percentages of maximal fluorescent Ca2+ response obtained by adrenaline (n = 7–9).
Nutrients 16 01567 g002
Figure 3. Concentration–response relationships (mean ± SD) of compounds that activated human adrenergic receptor (ADR) α2A in chem-1 cells. Data are presented as percentages of maximal fluorescent Ca2+ response obtained by adrenaline (n = 8).
Figure 3. Concentration–response relationships (mean ± SD) of compounds that activated human adrenergic receptor (ADR) α2A in chem-1 cells. Data are presented as percentages of maximal fluorescent Ca2+ response obtained by adrenaline (n = 8).
Nutrients 16 01567 g003
Figure 4. Concentration–response relationships (mean ± SD) of compounds that activated human adrenergic receptor (ADR) β1 (a) and β2 (b) in chem-1 cells overexpressing these receptors. Data are presented as percentages of maximal fluorescent Ca2+ response obtained by adrenaline (n = 8).
Figure 4. Concentration–response relationships (mean ± SD) of compounds that activated human adrenergic receptor (ADR) β1 (a) and β2 (b) in chem-1 cells overexpressing these receptors. Data are presented as percentages of maximal fluorescent Ca2+ response obtained by adrenaline (n = 8).
Nutrients 16 01567 g004
Figure 5. Concentration–response relationships (mean ± SD) of (a) phenethylamine, tyramine, methyltyramine and higenamine, (b) phenethylamine, p-synephrine, hordenine, dimethylphenethylamine and halostachine, and (c) phenethylamine, β-methylphenethylamine, p-octopamine and isopropyloctopamine, which activated human trace amine-associated receptor 1 (TAAR1) in HEK293T cells stably transfected with this receptor. Data are presented as percentages of maximal intracellular cAMP level obtained by phenethylamine (n = 3–5).
Figure 5. Concentration–response relationships (mean ± SD) of (a) phenethylamine, tyramine, methyltyramine and higenamine, (b) phenethylamine, p-synephrine, hordenine, dimethylphenethylamine and halostachine, and (c) phenethylamine, β-methylphenethylamine, p-octopamine and isopropyloctopamine, which activated human trace amine-associated receptor 1 (TAAR1) in HEK293T cells stably transfected with this receptor. Data are presented as percentages of maximal intracellular cAMP level obtained by phenethylamine (n = 3–5).
Nutrients 16 01567 g005
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Pinckaers, N.E.T.; Blankesteijn, W.M.; Mircheva, A.; Shi, X.; Opperhuizen, A.; Schooten, F.-J.v.; Vrolijk, M.F. In Vitro Activation of Human Adrenergic Receptors and Trace Amine-Associated Receptor 1 by Phenethylamine Analogues Present in Food Supplements. Nutrients 2024, 16, 1567. https://doi.org/10.3390/nu16111567

AMA Style

Pinckaers NET, Blankesteijn WM, Mircheva A, Shi X, Opperhuizen A, Schooten F-Jv, Vrolijk MF. In Vitro Activation of Human Adrenergic Receptors and Trace Amine-Associated Receptor 1 by Phenethylamine Analogues Present in Food Supplements. Nutrients. 2024; 16(11):1567. https://doi.org/10.3390/nu16111567

Chicago/Turabian Style

Pinckaers, Nicole E. T., W. Matthijs Blankesteijn, Anastasiya Mircheva, Xiao Shi, Antoon Opperhuizen, Frederik-Jan van Schooten, and Misha F. Vrolijk. 2024. "In Vitro Activation of Human Adrenergic Receptors and Trace Amine-Associated Receptor 1 by Phenethylamine Analogues Present in Food Supplements" Nutrients 16, no. 11: 1567. https://doi.org/10.3390/nu16111567

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop