• No results found

Changes in adipose tissue mRNA expression due to perinatal exposure to bisphenol A in rats

N/A
N/A
Protected

Academic year: 2022

Share "Changes in adipose tissue mRNA expression due to perinatal exposure to bisphenol A in rats"

Copied!
11
0
0

Loading.... (view fulltext now)

Full text

(1)

1

Department of Women’s and Children’s health

Biomedical Laboratory Science Programme

Degree project 15 credits spring 2014

Changes in adipose tissue mRNA expression due to perinatal exposure to bisphenol A in rats

Gunilla Chen

Supervisors:

Margareta Halin Lejonklou, Department of Medical Sciences; Occupational and Environmental Medicine, Uppsala University

Tomas Waldén, Department of Medical Cell Biology, Uppsala University

(2)

2 ABSTRACT

Bisphenol A (BPA) is an estrogen receptor binding chemical, widely used in the plastics industry, and as such commonly encountered from plastic containers etc.

Even at very low doses, BPA is believed to induce obesity and to have various endocrine disruptive effects. The purpose of this study was to determine possible gene expression changes in gonadal and inguinal adipose tissue from rats perinatally exposed to BPA. The method used was quantitative real-time PCR, and genes found to be up-regulated were PLZF, adiponectin, RXRa and Tcf21, while down-regulated genes were PPARγ, Tmem26, EsR1, Resistin, LPL, Chemerin, Serpina6, TFAM and Ahr. This is so far largely unsupported by other studies, and more research is needed.

Keywords

Obesity ・ endocrine disruptor ・ metabolism ・ PPARγ ・ estrogen receptors

(3)

3 INTRODUCTION

In recent years there have been rising concerns that our exposure to chemicals may have greater detrimental impact on our health than previously thought, even with the low doses we receive every day, especially during early childhood [1].

Studies have shown that obesity rates in young children and infants have increased. There is growing evidence that this is partly due to endocrine- disrupting chemicals (EDCs) acting as obesity agents by increasing adipocyte numbers, altering energy balance into the storing of energy, as well as changing the appetite/satiety balance [2].

One such EDC, Bisphenol A (BPA), binds the estrogen receptor and has been of particular interest because of its wide use in the plastics industry with a yearly production of 4.5 million tons

1

. BPA is a somewhat water-soluble compound which has been found to leach from plastics. Our most common ingestion sources are via digestion or oral mucosal absorption from leaching food containers made from polycarbonate plastics or from the leaching epoxy lining of water pipes [3].

In the European Union, BPA has been banned from the production of baby bottles since March 2011. However, studies show that even plastic items reportedly free of BPA also leach estrogen-like substances. Although each chemical occurs in low concentration, together they amount to a much greater hormonal effect [4]. Other sources of BPA include inhalation of dust, the leaching from dental sealants into the mouth, and from contact with thermal paper such as receipts either skin-to- mouth [3] or even directly through the skin [5].

Even though the substance is not accumulated in tissue due to its rapid metabolism in the gastrointestinal tract and liver [6], our daily exposure to it maintains low concentrations in our blood at all times [3]. Also, exposure via anything but digestion bypasses the first-

pass metabolism of liver and gut, increasing bioavailability [7].

A problem with all studies measuring the effects of BPA is the establishment of negative controls. Due to the ubiquitousness of BPA, one has to work on the premise that even the negative control has not escaped exposure to BPA. This is a problem even in animal studies, since housing cages and water bottles are made from plastics, and common feed types contain varying levels of phytoestrogens, plant-derived estrogen-like substances [8].

Studies have estimated that the average BPA exposure for young children lies around 0.04 μg/kg/day, with the average daily human intake being somewhere between 0.48 and 1.6 μg/kg body weight/day from food sources. Concentrations of unconjugated, biologically active BPA measured in human serum range between 0.2 and 20 ng/mL [3]. The No-Adverse-effect level (NOAEL) of BPA has been set to 50 mg/kg body weight/day by the US Environmental Protection Agency (EPA), which is far above the normal daily intake. The maximum acceptable daily exposure reference dose (RfD) was set to one thousandth of this, 50 μg/kg body weight/day. There are, however, several problems with establishing “safe” levels of BPA exposure. Since BPA is acting like a hormone, ____________________

1 www.kemi.se/Innehall/Fragor-i-fokus/Bisfenol-A-BPA/ 20140515

(4)

4

and hormones have effects even at extremely low doses, BPA is also likely to have effect at very low doses. Studies have even shown higher effect at low doses, resulting in nonmonotonic dose-response curves [9]. Nonmonotonic dose- response curves are U-shaped, with higher response at very high and very low doses, probably due to high doses inhibiting the low-dose response. Also, since many different EDCs apart from BPA add their effects, the total effect will be much higher.

Numerous studies have shown effects at doses below the NOAEL and the RfD, with impact on brain development, behaviour, reproduction, growth and metabolism [9]. Furthermore, the effects of hormones depend on the time of exposure, with greater effects in developing individuals, meaning that lower levels of accepted doses are needed for children and pregnant women [10].

This experiment was designed to be a pilot study. The purpose was to single out genes of interest for analysis by examining differences in expression using Real-Time quantitative PCR (RT-qPCR). Material for the study was gonadal and inguinal adipose tissue from rats exposed to BPA both during prenatal development and from their mother’s milk until weaning. The genes are important for fat metabolism, and include PPARγ, RXRa, adiponectin, PLZF and LPL.

Peroxisome proliferator-activated receptor gamma (PPARγ), also known as NR1C3, is a nuclear receptor that forms a heterodimer with the retinoid X receptor (RXRs). It is considered to be a very important regulator of adipogenesis, increasing weight and adipocyte cell numbers, and promoting differentiation of multipotent stromal cells into adipose tissue at the expense of bone tissue [2]. Adiponectin (ADIPOQ) is a hormone secreted from adipose tissue specifically, and is insulin sensitizing and anti-inflammatory. It is a possible candidate for diabetes treatment [11]. Promyelocytic leukemia zinc finger (PLZF) is a transcription factor specific to invariant natural killer T-cells, and is shown to represses adipogenesis when overexpressed [12]. Lipoprotein lipase (LPL) plays an important role in lipid metabolism as well as increasing cellular uptake of lipoproteins [13].

MATERIALS AND METHODS Study objects

Nine Fisher 344 rats were kept in cages made from polysulfone plastic, with glass water bottles. They were given unlimited access to water and food (standard pellets) and BPA was added to the drinking water (tap water with 1 % EtOH vehicle) at day ten of gestation. BPA concentrations in the drinking water were determined as to reach a final concentration per kilogram bodyweight of 0.5 (Low dose), 50 (High dose) and 500 (Very High dose) μg, with actual concentrations being 0.4, 30–38 and 333–422 μg/kg/day (measured in plasma).

The control group received plain water. Their offspring were killed at five weeks

of age at which time tissue samples were collected.

(5)

5

Since the low dosing of BPA is not acutely toxic, animals subjected to it are not expected to suffer. The most distressing procedure was the sedation prior to termination. Ethical permission was granted on the 16th of April 2013 by the Swedish Board of Agriculture.

Samples

Samples were taken from nine litters with ten females and twelve males: 22 retroperitonal adipose tissue samples (BP1–22), 22 interscapular white adipose tissue samples (BW1–22), 22 interscapular brown adipose tissue sample (BB1–

22) and 14 liver samples (BL1–14). All adipose tissue samples were preserved in AllProtect (Qiagen) and subsequently frozen in −70ºC. The liver samples were homogenized using the Ultra-Turrax (Ika) and preserved in RNAlater (Qiagen) before freezing in −70ºC, except for sample BL1 and BL2 which had been snap frozen in liquid nitrogen prior to storing in −70ºC. Also analyzed were 22 cDNA samples from inguinal adipose tissue (BIn1–22) and 21 cDNA samples from gonadal adipose tissue (BG1–15, BG18, BG20–26; of these BG18 and BG23 were from the same individual with the difference that BG18 was homogenized with Ultra-Turrax and BG23 not). See table 1 for a full list of inguinal and gonadal samples.

Table 1.The number of males and females in each dose group for inguinal and gonadal white adipose tissue.

group n, males n, females

iWAT control 3 3

low dose 2 3

high dose 4 2

very high dose 2 2

gWAT control 4 3

low dose 2 2

high dose 4 2

very high dose 2 2

TRIzol RNA extraction

The tissue samples were run in the Bullet Blender (Next Advance) with 0.5 ml

Lysing Matrix D (MP-Biomedicals) och 1 ml TRIzol (Life Technologies) at speed 6

for one minute, or until fully homogenized, after which they were put on ice for 5

minutes. Chloroform (200 μl) was added to each sample which was then

vortexed for 15 seconds and put on ice for at least 3 minutes. The samples were

centrifuged (4°C, 12000 × g, 15 min) and 500 μl isopropanol was added to the

aquatic phase. Samples were put on ice for one hour and centrifuged again (4°C,

12000 × g, 10 min). The supernatant was removed and 1 ml 75 % EtOH was

added to the pellet, which was then centrifuged (4°C, 7500 × g, 5 min). The

alcohol was removed, 30 μl Diethylpyrocarbonate (DEPC) treated water was

added, and the samples were vortexed for 30 seconds.

(6)

6

Purity and concentration of the samples was determined using Nanodrop (Thermo Scientific), with following dilution using DEPC-treated water to reach a maximum concentration of 600 ng/μl when needed. Samples were frozen and stored at −70°C.

cDNA Synthesis

From each sample, 500 ng RNA was used, diluted to a total volume of 13.2 μl. The master mix for each sample contained 2 μl 10 × RT-buffer, 0.8 μl 25 × dNTPs, 2 μl random primers, 1 μl Multiscribe RT och 1 μl RNase inhibitor (all from Life Technologies). Samples were run on GeneAmp 9700 (Applied Biosystems. Step 1:

25°C for 10 min, step 2: 37°C for 120 min, step 3: 85°C for 5 min, and last cooling to 4°C). The synthesised cDNA was diluted to 200 μl using DEPC-treated water and frozen in −20°C.

qPCR procedure and verifications of method reliability

Only samples from gonadal and inguinal adipose tissue were fully analyzed in this pilot study.

Four μl cDNA in duplicates was used for each analysis. The master mix for each sample consisted of 5.6 μl DEPC-treaded water, 10 μl SsoFast EvaGreen (Bio- Rad), 0.2 μl Forward Primer (30 μM) och 0.2 μl Reverse Primer (30 μM). qPCR was run on a CFX96 (Bio-Rad. 95°C for 30 sec, (95°C for 5 sec, 58°C for 5 sec) × 40 cycles with a melting curve from 65–95°C). See table 2 for a list of primers used.

Table 2. Primers (Invitrogen)

Gene Forward Primer Reverse Primer

TFIIB CCTGGCAGGAGTCCTATCTCT ACCAGCAATATCCCCGATTT PPARγ TCCTCCTGTTGACCCAGAGCAT CCACCAACTTCGGAATCAGCT

Tmem26 GTCTCGGGAATCCTTGTGG CGGGGCATACCAGTTTCA

EsR1 GATGGGCTTATTGACCAACC TGGAGATTCAAGTCCCCAAA

Resistin ATCAAGACTTCAGCTCCCTACTG GTGACGGTTGTGCCTTCTG

LPL CAGAGAAGGGGCTTGGAGAT TTCATTCAGCAGGGAGTCAA

Chemerin TGCCTGAAGAAGGACTGGA TGCCTGAAGAAGGACTGGA Serpina6 AGCAAAGCACAACCAGAAGG GGCGAGTGACATTGTTCAGTT

TFAM AGCTAAACACCCAGATGCAAA TCAGCTTTAAAATCCGCTTCA

ADIPOQ TGGTCACAATGGGATACCG CCCTTAGGACCAAGAACACCT

RXRa ACATGCAGATGGACAAGACG GGGTTTGAGAGCCCCTTAGA

Ahr CTTCAGATGCCGGCTGAG CCTCCCTTGGAAATTCATTG

Tcf21 CATTCACCCAGTCAACCTGA CCACTTCCTTTAGGTCACTCTCA Leptin CCAGGATCAATGACATTTCACA AATGAAGTCCAAACCGGTGA

Scd1 GAAGCGAGCAACCGACAG GGTGGTCGTGTAGGAACTGG

AdipoR1 AGCACCGGCAGACAAGAG GGTGGGTACAACACCACTCA

Nrip1 GAGCTGTTCTCAGGACCCATT CCTCAGTCTACTGCGCACCT

Primer efficiency was tested for TFIIB and PPARγ using cDNA diluted 1:1, 1:4,

1:16, 1:64, 1:256 and 1:1024.

(7)

7

A 2 % (w/v) agarose gel electrophoresis was performed with samples from qPCR runs with TFIIB, PPARγ and leptin, as well as samples from a run with Scd1 which had shown double peaks in its melting curve. GeneRuler 100 bp (ThermoScientific) was used as ladder.

qPCR data treatment

Ct-value duplicates with a difference exceeding 0.4 were excluded, as were any Ct-values exhibiting double peaks in their melting curves. BG3 and BG6 were litter adjusted and thus not included. Ct-values were calculated as the means for every pair of duplicates and were normalized against the endogenous control (TFIIB), after which they were linearized for easier comparison.

GraphPad Prism was used for Dunn᾿s Multiple Comparison Test and Microsoft Excel for calculating unpaired Student᾿s t-tests for the different groups.

RESULTS qPCR results

Results from the qPCR were divided into groups according to their BPA exposure levels. The statistical significance of any gene expression changes were determined using unpaired Student᾿s t-test (p ≤ 0.05 deemed significant). Gene expression changes in adipose tissue are shown in table 3.

Table 3. Relative gene expression changes deemed significant according to Student᾿s t-test (p ≤ 0.05) in adipose tissue in groups exposed to BPA compared to control groups (Ld = Low dose, Hd

= High dose, VHd = Very High dose). Gonadal adipose tissue in the left table and inguinal adipose tissue in the right table. Increases in gene expression marked as upward pointing arrows and decreases as downward pointing arrows (m = male group, f = female group).

Ld Hd VHd Hd+VH

d Ld Hd VHd Hd+VH

d PPARγ

↓ m ↓ m ↓ m ↓ m f

PPARγ

↓ m ↓ m ↓ m ↓ m

Tmem26

- - ↓ f ↓ f

Tmem26

- - - ↓ m

PLZF

- ↑ f - ↑ f

Adiponectin

- ↑ f - ↑ f

EsR1

- ↓ f ↓ f ↓ f

RXRa

- - ↑ f -

Resistin

- ↓ f ↓ f ↓f

Ahr

- - ↓ f -

LPL

- ↓ f ↓ f ↓ f

Tcf21

- - ↑ f -

Chemerin

- - - ↓ f

Serpina6

- - - ↓ f

TFAM

- ↓ f ↓ f ↓ f

(8)

8

Down-regulated genes were PPARγ, Tmem26, EsR1, Resistin, LPL, Chemerin, Serpina6, TFAM and Ahr, and up-regulated genes PLZF, adiponectin, RXRa and Tcf21.

Agarose gel electrophoresis results

The agarose gel showed multiple bands from Scd1 samples displaying multiple peaks in their melting curves from the qPCR analysis, but single bands of correct sizes for TFIIB (102 nt), PPARγ (119 nt) and leptin (71 nt) (see figure 1).

Figure 1. Two percent agarose gel. Lane 1: ladder (GeneRuler 100 bp ThermoScientific, fragment lengths 100–1000 bp). Lane 2: Scd1 product not exhibiting double melting curve peaks. Lanes 3–4:

Scd1 products exhibiting double peaks. Lane 5:

TFIIB product with single peak. Lane 6: PPARγ product with single peak. Lanes 7–10: Leptin products from different samples, all with single peaks. Lane 11: ladder again.

DISCUSSION

The purpose of this study was to examine possible gene expression differences in adipose tissue due to BPA exposure, using qPCR. Values were normalized using the endogenous control gene TFIIB, encoding part of the RNA polymerase preinitiation complex. A second and possibly third endogenous control would ideally have been used [14], but other endogenous controls tested (LRP10, EIF3K and PGK1) showed various problems including low expression (data not included).

Several of the qPCR results showed double peaks in their melting curves, signifying either genomic DNA contamination, inadequate primer specificity or primer-dimer formation, necessitating the exclusion of the results from five genes, Scd1, FoxP3, AdipoR1, Nrip1 and F4/80. A temperature gradient to determine the optimal primer temperature was run for several of these primers, and the results need to be used for further analyses. Three samples in particular repeatedly exhibited double peaks with many primers and were excluded whenever double peaks occurred. These were likely to be either contaminated with genomic DNA or otherwise degraded.

The agarose gel electrophoresis showed no unwanted product from samples lacking melting curve double peaks. All products exhibited correct product length. Efficiency tests for TFIIB and PPARγ showed excellent linearity in the concentration range tested.

No significant results were obtained when data was analysed using Dunn᾿s multiple comparison test. Due to this being a pilot study, and the subject number

1 2 3 4 5 6 7 8 9 10 11

(9)

9

small, this was to be expected. The purpose of this work was to find tendencies for gene expression changes, later to be fully explored in the larger study.

The results from the qPCR showed significant changes in expression in many of the genes, but of different weight. Tmem26, PLZF, adiponectin, RXRa, Ahr and Tcf21 only showed significance in one dose/sex group, whereas PPARγ, EsR1, resistin, LPL and TFAM showed significance in at least two dose/sex groups.

Most significant changes were found in female dose groups, which might be explained by the fact that BPA binds to the estrogen receptor.

The most interesting result was for PPARγ, which showed signs of decrease in all male dose groups when compared to the control group. It was also the only gene showing significant change in the low dose group. These results are the opposite of a previous study by Somm et al. [15], in which exposure to BPA caused an increase of PPARγ expression. This overexpression was, however, only observed in females, while the present study noted the largest decrease in males.

Numerous studies have also shown weight-increases from BPA-exposure both in rats [16] and humans [17]: further reason to believe that the expression of PPARγ should increase rather than decrease with BPA exposure. One possible explanation is that the primers used for PPARγ in the current study only amplified one splice variant, PPARγ1. PPARγ1 is expressed at high levels in most tissues, whereas PPARγ2, the second splice variant, is specifically expressed in adipocytes and seems to be of greater importance in adipocyte differentiation [18].

The results from adiponectin also showed opposite results than expected, with levels increasing rather than decreasing, as in a study by Hugo et al. where adiponectin expression in cell cultures from human breast tissue and two abdominal fat depots decreased with exposure to BPA [19]. Also not in accordance to other studies was the decrease of resistin expression, which has been found to increase on exposure to octylphenol, an environmental estrogen [20].

LPL expression decreased in both female high-dose groups, and other studies have shown similar results in human female adult stem cells [21]. The expression of TFAM was decreased in the same groups, and has been shown to decrease in rat insulinoma cells on BPA exposure [22].

Also decreased in female high-dose groups was the expression of EsR1, which previous studies have found increased in fetal mouse prostate mesenchyme cells [23] and decreased in certain female rat brain cells [24] due to BPA exposure, leaving matters inconclusive.

Certain is that BPA is a chemical to be wary of at both low and high doses of

exposure, and hopefully the main study will answer more of our questions.

(10)

10 REFERENCES

[1] Vandenberg, L. N. (2014) Low-Dose Effects of Hormones and Endocrine Disruptors. Vitam. Horm. 94, 129-165

[2] Boekelheide, K. et al. (2012) Predicting Later-Life Outcomes of Early-Life Exposures. Environ. Health Perspect. 120(10), 1353-1361

[3] Vandenberg, L. N. et al. (2007) Human exposure to BPA. Reprod. Toxicol. 24, 139-177

[4] Yang, C. Z. et al. (2011) Most plastic products release estrogenic chemicals: a potential health problem that can be solved. Environ. Health Perspect. 119(7), 989-996

[5] Geens, T., Goeyens. L. and Covaci, A. (2011) Are potential sources for human exposure to bisphenol-A overlooked? Int. J. Hyg. Environ. Health 214(5), 339- 347

[6] Doerge, D. R. and Fisher, J W. (2010) Background Paper on Metabolism and Toxicokinetics of Bisphenol A. WHO 2010

[7] Gayrard, V. et al. (2013) High Bioavailability of Bisphenol A from Sublingual Exposure. Environ. Health Perspect. 121(8), 951-956

[8] Thigpen, J. E. et al. (2013) The Estrogenic Content of Rodent Diets, Bedding, Cages, and Water Bottles and Its Effect on Bisphenol A Studies. J. Am. Assoc. Lab.

Anim. Sci. 52(2), 130–141

[9] vom Saal, F. S. and Peterson Myers, J. (2008) Bisphenol A and risk of Metabolic Disorders. JAMA 300(11), 1353-1355

[10] Vandenberg, L. N. et al. (2013) Low dose effects of bisphenol A: An integrated review of in vitro, laboratory animal, and epidemiology studies.

Endocr. Disruptors e25078, 1-13

[11] Blüher, M. (2014) Adipokines - removing road blocks to obesity and diabetes therapy. Mol. Metab. 3(3), 230-240

[12] Mikkelsen, T. S. et al. (2010) Comparative epigenomic analysis of murine and human adipogenesis. Cell. 1;143(1), 156-169

[13] Mead, J. R., Irvine, S. A. and Ramji, D. P. (2002) Lipoprotein lipase: structure, function, regulation, and role in disease. J. Mol. Med. (Berl). 80(12), 753-769 [14] Vandesompele, J. et al. (2002) Accurate normalization of real-time quantitative RT-PCR data by geometric averaging of multiple internal control genes. Genome Biol. 3(7), research0034.1–0034.11

[15] Somm, E. et al. (2009) Perinatal Exposure to Bisphenol A Alters Early Adipogenesis in the Rat. Environ. Health Perspect. 117(10), 1549-1555

[16] Rubin, B. S. et al. (2001) Perinatal exposure to low doses of bisphenol A affects body weight, patterns of estrous cyclicity, and plasma LH levels. Environ.

Health Perspect. 109(7), 675-80

[17] Ko, A. et al. (2014) Association between Urinary Bisphenol A and Waist Circum-ference in Korean Adults. Toxicol. Res. 30(1), 39-44

[18] Aprile, M. et al. (2014) PPARG in Human Adipogenesis: Differential

Contribution of Canonical Transcripts and Dominant Negative Isoforms. PPAR

Res. 537865

(11)

11

[19] Hugo, E. R. et al. (2008) Bisphenol A at environmentally relevant doses inhibits adiponectin release from human adipose tissue explants and adipocytes.

Environ. Health Perspect. 116(12), 1642-1647

[20] Lee, M. J. et al. (2008) Octylphenol stimulates resistin gene expression in 3T3-L1 adipocytes via the estrogen receptor and extracellular signal-regulated kinase pathways. Am. J. Physiol. Cell Physiol. 294(6), C1542-51

[21] Linehan, C. et al. (2012) Bisphenol A-mediated suppression of LPL gene expression inhibits triglyceride accumulation during adipogenic differentiation of human adult stem cells. PLoS One 7(5), e36109

[22] Lin, Y. et al. (2013) Exposure to bisphenol A induces dysfunction of insulin secretion and apoptosis through the damage of mitochondria in rat insulinoma (INS-1) cells. Cell Death Dis. 17;4, e460

[23] Richter, C. A. et al. (2007) Estradiol and Bisphenol A Stimulate Androgen Receptor and Estrogen Receptor Gene Expression in Fetal Mouse Prostate Mesenchyme Cells. Environ. Health Perspect. 115(6), 902–908

[24] Rebuli, M. E. et al. (2014) Investigation of the Effects of Subchronic Low Dose Oral Exposure to Bisphenol A (BPA) and Ethinyl Estradiol (EE) on Estrogen Receptor Expression in the Juvenile and Adult Female Rat Hypothalamus.

Toxicol. Sci. [Epub ahead of print]

References

Related documents

Levels of KRT36 go from very high in normal control tongue tissue to extremely low in tumors, and tumor free tissue showed a big variation in expression with patients having levels

In summary, gene expression profiling of human adipocytes and adipose tissue during different conditions suggest that SAA, NQO1, CIDE-A and ZAG may be implicated in human

In both the Karnataka and Hyderabad data we have rich borrowing information, and we can measure the impacts of microfinance on several different types of household borrowing

Vi har inte funnit någon evidens i litteraturen om att det skulle finnas några specifika skillnader vad gäller prestation på spannivå mellan pojkar och flickor med

The results show that a material model with varying Young’s modulus through the thickness, based on a stiffness and porosity relationship, can produce similar densification

Male right:left testes area ratio was statistically significant, affected by treatment dose 70 μg/g egg, as well as liver weight and liver:body weight ratio, hepatosomatic index..

In extension to these findings we propose that estrogens, emanating from the systemic circulation or from local synthesis, by way of slow genomic and more rapid non-genomic

Distributionen av östrogenreceptor-innehållande nervceller är här snarlik den som ses för nervceller som producerar den smärthämmande kroppsegna opioiden enkefalin, vilket är av