<<

Digital Comprehensive Summaries of Uppsala Dissertations from the Faculty of Medicine 1616

Melatonin in the

FANNY SÖDERQUIST

ACTA UNIVERSITATIS UPSALIENSIS ISSN 1651-6206 ISBN 978-91-513-0814-2 UPPSALA urn:nbn:se:uu:diva-396347 2019 Dissertation presented at Uppsala University to be publicly examined in Humanistiska teatern, Thunbergsvägen 3, Uppsala, Friday, 17 January 2020 at 09:00 for the degree of Doctor of Philosophy (Faculty of Medicine). The examination will be conducted in Swedish. Faculty examiner: Professor Bodil Ohlsson (Lunds Universitet).

Abstract Söderquist, F. 2019. in the gastrointestinal tract. Digital Comprehensive Summaries of Uppsala Dissertations from the Faculty of Medicine 1616. 63 pp. Uppsala: Acta Universitatis Upsaliensis. ISBN 978-91-513-0814-2.

Melatonin is recognised as the pineal hormone regulating sleep and circadian rhythm. It has also been identified in peripheral tissues (mainly in animals) and thought to display a variety of actions, including anti-inflammatory properties, regulation of gastrointestinal (GI) functions, glucose homeostasis and beneficial effects in different tumour types. Patients with irritable bowel disorder commonly exhibit psychiatric co-morbidity and disturbances of the gut-brain axis have been proposed to play a role in these disorders. The focus of this thesis was to study melatonin and melatonin receptors in the normal human GI tract, the pancreas and small intestinal neuroendocrine tumours. The thesis also explores the complex relationship between GI symptoms and underlying psychiatric traits in the context of elevated levels of peripheral melatonin during waking hours. In paper I-II, tissue samples from the normal human GI tract and pancreas and tumour tissue from small intestinal neuroendocrine tumours were analysed for expression of melatonin and melatonin receptors using immunohistochemistry. For tumour patients, melatonin was also analysed in plasma and set in relation to symptoms and outcome. In paper III-IV, a cohort of young adults (18-25 years) seeking psychiatric care was examined for GI symptoms, melatonin levels in saliva, depressive symptoms and anxiety traits. Psychiatric assessments were performed using structured or semi structured interviews. Depressive symptoms were measured using the self-rating version of the Montgomery-Åsberg Depression Rating Scale; GI symptoms were measured using the Gastrointestinal Symptoms Rating Scale for ; and personality traits were evaluated using the Swedish Universities Scales of Personality. Melatonin and melatonin receptors were widely expressed in the normal human gut and pancreas (paper I) but even in small intestinal neuroendocrine tumours known to produce (paper II). The intensity of the melatonin immunoreactivity in tumour tissue was found to correlate with lower proliferation index. After treatment, plasma levels of melatonin were reduced in tumour patients. Young adult patients seeking psychiatric care reported more GI symptoms than healthy controls, regardless of the currently active psychotropic . The level of GI symptoms was associated with severity of depressive symptoms and trait anxiety (paper III). Higher postprandial levels of melatonin were associated with the GI symptoms of bloating and pain (paper IV). In summary, these findings demonstrate the widespread presence of melatonin in the human gut and confirm a link between melatonin, psychiatric health and GI symptoms.

Keywords: melatonin, MT1, MT2, gastointestinal tract, small intestinal neuroendocrine tumours, IBS, depression, anxiety, personality and affective disorder

Fanny Söderquist, Department of Neuroscience, Psychiatry, University Hospital, Akademiska sjukhuset, Uppsala University, SE-751 85 Uppsala, Sweden.

© Fanny Söderquist 2019

ISSN 1651-6206 ISBN 978-91-513-0814-2 urn:nbn:se:uu:diva-396347 (http://urn.kb.se/resolve?urn=urn:nbn:se:uu:diva-396347)

Lindrig sömnlöshet ökar genialiteten.

Brokiga iakttagelser Edith Södergran, 1919

To my family

List of Papers

This thesis is based on the following papers, which are referred to in the text by their Roman numerals.

I Söderquist F, Hellström PM, Cunningham JL. Human gastroentero- pancreatic expression of melatonin and its receptors MT1 and MT2. PLoS One. 2015 Mar 30;10(3):e0120195.

II Söderquist F, Janson ET, Rasmusson AJ, Ali A, Stridsberg M, Cunning- ham JL. Melatonin immunoreactivity in malignant small intestinal neu- roendocrine tumours. PLoS One. 2016 Oct 13;11(10):e0164354.

III Söderquist F, Syk M, Just D, Kubalija Novicic Z, Jacobsson A, Hell- ström PM, Ramklint M, Cunningham JL. Gastrointestinal symptoms, depression and trait anxiety in young adults seeking psychiatric care. Manuscript

IV Söderquist F, Sundberg I, Ramklint M, Widerström R, Hellström PM, Cunningham JL. The Relationship Between Daytime Sali- vary Melatonin and Gastrointestinal Symptoms in Young Adults Seek- ing Psychiatric Care. Psychosom Med. 2019 Jan;81(1):51-56.

Reprints were made with permission from the respective publishers.

Contents

Introduction ...... 11 What is a hormone? ...... 11 Melatonin, general aspects ...... 12 Melatonin receptors and interaction sites ...... 14 Melatonin and the brain ...... 14 The gut-brain axis ...... 15 Melatonin signalling in peripheral tissue ...... 16 Consequences of peripheral melatonin ...... 17 Melatonin in the gastro-pancreatic system ...... 17 Immunomodulatory functions of melatonin ...... 18 Melatonin and cancer ...... 19 Small intestinal neuroendocrine tumours ...... 20 Functional gastrointestinal disorders ...... 21 Psychiatric co-morbidity and functional gastrointestinal disorders ..... 22 Aims ...... 23 Materials and methods ...... 24 Design ...... 24 Setting ...... 24 Paper I ...... 24 Paper II ...... 24 Paper III and IV ...... 25 Study population and patient samples ...... 25 Paper I ...... 25 Paper II ...... 25 Paper III ...... 26 Paper IV ...... 26 Microarray expression analysis ...... 27 Immunohistochemistry ...... 29 Histological assessment – Paper I ...... 29 Histological assessment – Paper II ...... 29 Hormone collection and analysis ...... 29 Analysis of melatonin in plasma – Paper II ...... 29 Analysis of melatonin in saliva – Paper IV ...... 29

Psychiatric assessment and questionnaires – Paper III and IV ...... 30 Self-assessment – Paper III and IV ...... 30 Statistics ...... 31 Paper II ...... 31 Paper III ...... 31 Paper IV ...... 32 Summary of results ...... 33 Melatonin and expression (Paper I) ...... 33 Normal human GI tract ...... 33 Normal human pancreas ...... 33 Melatonin and melatonin receptors in SI-NETs (Paper II) ...... 33 Tissue expression ...... 33 Plasma levels ...... 34 Relationship between melatonin tissue intensity, plasma levels and clinical parameters ...... 34 GI symptoms and depressive symptoms (Paper III) ...... 34 Melatonin levels and GI symptoms (Paper IV) ...... 35 Discussion ...... 37 Melatonin and melatonin receptors in normal human gut and pancreas .. 37 Melatonin and melatonin receptors in SI-NETs ...... 39 GI symptoms and psychiatric comorbidity ...... 40 Daytime melatonin and GI symptoms ...... 41 Methodological considerations and limitations ...... 42 Clinical implications and future perspectives ...... 43 Ethics ...... 45 Conclusions ...... 46 Sammanfattning på svenska ...... 47 Bakgrund ...... 47 Syfte ...... 47 Metod ...... 48 Resultat och slutsats ...... 48 Acknowledgements ...... 50 References ...... 52

Abbreviations

AANAT Arylalkylamine N-acetyltransferase ANS Autonomic nervous system ATP Adenosine triphosphate AUDIT Use Disorders Identification Test BMI Body mass index CgA Chromogranin A CRH Corticotropin-releasing hormone CRHR1 Corticotropin-releasing hormone receptor 1 CYP Cytochrome P DSM Statistical Manual of Mental Disorders DUDIT Drug Use Disorders Identification Test EC Enterochromaffin ELISA Enzyme-linked immunosorbent assay ENS Enteric nervous system FGID Functional gastrointestinal disorder GAD Generalised anxiety disorder GDP Guanosine diphosphate GEO Gene Expression Omnibus GI Gastrointestinal GSRS-IBS Gastrointestinal Symptom Rating Scale-IBS GTP Guanosine triphosphate HIOMT Hydroxyindole-O-methyltransferase IBS Irritable bowel syndrome IDO Indolamine 2,3 dioxygenase IF Immunofluorescence IFN Interferon IHC Immunohistochemistry IL Interleukin IPANs Intrinsic primary afferents IR Immunoreactivity KYNA Kynurenic acid MADRS Montgomery-Åsberg Depression Rating Scale MDD Major depressive disorder min minutes M.I.N.I. Mini-International Neuropsychiatric Interview mRNA Messenger Ribonucleic acid

MT1 Melatonin receptor type 1 MT2 Melatonin receptor type 2 PCA Principal component analysis PCR Polymerase chain reaction PsTAT Psychic trait anxiety T-score PTSD Post-traumatic stress disorder QoL Quality of life ROS Reactive oxygen ROR/RZR Retinoid orphan receptors/retinoid Z receptors SCID-1 Structural Clinical Interview for DSM IV axis I SCN Suprachiasmatic nucleus SI-NET Small intestinal neuroendocrine tumour SSP Swedish Universities Scale of Personality SST Stress susceptibility T-score SSRI Selective serotonin reuptake inhibitors STAT Somatic trait anxiety T-score TDO Tryptophan 2,3 dioxygenase TNF Tumour necrosis factor UC

Introduction

What is a hormone?1 Hormones are chemical messengers that help the organs and cells to com- municate. There are many types of hormones: , peptides, amino ac- ids, steroids, fatty acid derivatives, ions and gases. All nucleated cells have the genetic information required to produce hormones (1). The target receptor, the type of cell that expresses it and its location all determine the effect of the hormone and the same hormone can exert a varie- ty of effects in different tissues. Ligand binding activates the receptor, which, in turn, sets off a cascade of intracellular signalling pathways result- ing in changed behaviour of the cell. The terms autocrine, paracrine and endocrine signalling describe three levels of signalling, based on where the target cell is located. Autocrine sig- nalling implies that the hormone sends its message to the same cell from which it was secreted (e.g., cancer cells producing growth factors that stimu- late their survival and proliferation) (2). In paracrine signalling the receiver of the hormonal message is neighbouring cells from where the hormone was secreted (e.g., neurotransmitters such as acetylcholine). Endocrine signalling refers to systemic effects of hormones; for example, insulin is produced in the pancreatic islets, secreted to the bloodstream and the target cells are adi- pocytes, hepatocytes and myocytes (3). Historically, descriptions of different hormonal effects have been simpli- fied explanations in keeping with the idea that one hormone has one action. This long-standing belief, however, is an oversimplification given the in- creasing knowledge of the growing number of diverse effects exerted by known hormones. For instance, it has recently been shown that insulin has other target cells in the brain and the effects extend far beyond glucose ho- meostasis and may even influence cognitive function (for a review, see (4)).

1 The paragraph ”What is a hormone?” has previously been published in the licentiate thesis Söderquist F. Melatonin and its receptors in the normal human gastrointestinal tract, pancreas and in small intestinal neuroendocrine tumours. 2017.

11 Melatonin, general aspects Melatonin is a small amphiphilic indoleamine, commonly known as the hormone involved in sleep and circadian rhythm in vertebrates. Its precursor is tryptophan via the synthesis of serotonin (Figure 1). Melatonin was first discovered when Aaron B. Lerner and his colleagues isolated it from the bovine pineal gland in 1958. It was named melatonin for its ability to lighten the skin of frogs (5). In the pineal gland production of melatonin follows a circadian rhythm. Signals from the suprachiasmatic nucleus stimulate the release of noradrena- line, which then activates adrenoreceptors on pinealocytes causing a rise in enzyme activity of arylalkylamine N-acetyltransferase (AANAT), the rate- limiting step in melatonin synthesis (1). This pineal system is effectively inhibited by light and melatonin release from the pineal gland is therefore a nocturnal event, with plasma levels steeply increasing soon after sunset to peak about 02:00, remain elevated throughout the night and then decline to lower levels by morning. During the daytime, plasma levels of melatonin are low and believed to be of a peripheral origin (6). Light exposure during the night results in lower plasma levels of melatonin (7).

12

Figure 1. Synthesis of melatonin from tryptophan (left) and kynurenine pathways (right). Enzymatic reactions catalysed by tryptophan hydroxylase (TPH1, TPH2), amino acid decarboxylase (AAD), arylalkylamine N-acetyltransferase (AANAT) and 5-hydroxyindole O-methyltransferase (HIOMT). Enzymatic reactions driving the kynurenine pathway are indoleamine dioxygenase (IDO), tryptophan-2,3- dioxygenase (TDO) and formamidase. Adapted from licentiate thesis (8).

Besides the pineal gland, melatonin is present in various peripheral tissues and organs. The two enzymes needed for melatonin synthesis from serotonin have been identified in retina, stomach, gut, spleen, liver, heart, skeletal muscle, bone marrow, testes, ovaries, placenta, skin and immune cells (9, 10). However, most studies are conducted in animals and studies in humans are needed to confirm the animal findings. Recent studies have proposed that melatonin may be produced within mitochondria as a protection against the high concentrations of free radicals produced in mitochondria. Acting as a powerful antioxidant is also the most basic function of melatonin (11). Hence, melatonin could be synthesised in all mitochondria-containing cells.

13 Circadian synthesis of melatonin begins at 3-5 months in a newborn infant and before endogenous production, melatonin is provided via breast (12). is associated with lower nocturnal melatonin amplitude and this decline appears to occur at approximately 40 years of age (13). Owing to its size and solubility, melatonin can easily pass cell mem- branes, including the blood-brain barrier and thus is also present in essential- ly all biological fluids: cerebrospinal fluid, saliva, bile, amniotic fluid, syno- vial fluid and (9). When administered, melatonin is rapidly ab- sorbed and distributed. Varying results for half-life elimination time have been reported, but ranges from 30-60 minutes (min) regardless of oral or intravenous administration (14, 15). The main route of degradation is via the liver and the CYP P450 enzymes, converting melatonin to 6- hydroxymelatonin, which is then conjugated with sulphate or glucuronide before secreted in the urine (16, 17).

Melatonin receptors and interaction sites There are two known membrane bound receptor subtypes for melatonin in humans and other mammals: receptor type 1 (MT1) and type 2 (MT2). They are both guanine nucleotide-binding regulatory (G-protein)-coupled receptors. The binding of melatonin results in intracellular signalling via second messengers (18). Both MT1 and MT2 receptors display a wide variety of signal transduction pathways in different tissues and cell types (19). A third receptor for melatonin named MT3 has been identified as the quinone reductase 2, which at least in part appears to be membrane-associated (20). Studies investigating the effect of melatonin administration on intestinal motility have also suggested that melatonin may act through the serotonin receptor 5-HT3, delaying gastric emptying and counteracting the actions of serotonin in the gut (21, 22). Melatonin can also bind to and interact with other intracellular proteins, including calmodulin, calreticulin and tubulin (23-25). Finally, melatonin can bind to nuclear receptors of the retinoid- related orphan nuclear hormone receptor (ROR/RZR) subfamily, which acts as transcription factors upon ligand binding (26, 27). Moreover, some of the actions of melatonin are receptor independent, including its free radical scavenging properties (11).

Melatonin and the brain Melatonin’s sleep-promoting effects have been extensively studied. In psy- chiatry melatonin and agomelatin, a melatonin receptor , are com- monly used to treat mood disorders and sleep disturbances. Alterations of the normal circadian secretory pattern of melatonin have been demonstrated in

14 several psychiatric disorders. However, the direction of this association is unclear and it is not fully understood whether circadian disturbances are a cause or a consequence of the psychiatric condition (28). Altered circadian rhythms and lower night-time melatonin levels have been demonstrated in neurodegenerative disorders, including Alzheimer’s, Huntington’s and Parkinson’s disease. Moreover, in patients with depression evening melatonin levels in saliva are decreased (29-31). Of note, in the context of melatonin’s anti-inflammatory properties, neuroinflammation has been proposed as a contributor to the pathogenesis in all of these conditions (32). Melatonin may also be involved in cognitive functions and behaviour. Middle-aged patients with cognitive impairment had a lower median noctur- nal melatonin response compared with healthy controls. Moreover, in pa- tients with affective disorders lower melatonin levels in the evening correlat- ed with poorer performance in verbal memory tasks (33, 34). A study using knock-out mouse models found that MT1 deletion increased anxiety-like behaviour, whereas deletion of the MT2 receptor resulted in anhedonia and generalised social avoidance, signifying depressive-like behaviour (35).

The gut-brain axis The brain and the gut share common signalling pathways that can be roughly divided into neural, endocrine and immunological routes of communication. This complex system is commonly referred to as the gut-brain axis. The system has implications for the function of the whole body in that it is linked to metabolism and the . The endocrine routes of gut-brain interactions involve the hypothalamic- pituitary-adrenal (HPA) axis and the release of gut peptides from enteroen- docrine cells in the intestinal wall. The activation of the HPA axis, resulting in elevated levels of cortisol, affects several aspects of gastrointestinal (GI) function. For example, the corticotropin-releasing hormone (CRH) is an important modulator of the gut-brain axis and mediates the stress response in both the brain and the gut (36) Activation of the CRH receptor 1 (CRHR1) appears to be involved in the colonic response to stress, with increased stress-related defecation in mice (37). The CRH-associated stimulation of colonic transit involves parasympathetic activation of colonic serotonin se- cretion and the binding to 5-HT3 receptors (38). In addition to the centrally active CRH, peripheral administration of the CRH stimulates colonic motili- ty and secretion, which appears to be CRHR1 dependent (39). Enteroendocrine cells of the GI mucosa produce various substances that regulate motility and secretion, as well as different gut peptides that affect hunger and satiety and regulate food intake and insulin release. The most common enteroendocrine cells are the enterochromaffin (EC) cells, which

15 produce and secrete serotonin and stain positive for chromogranin A (CgA) (40). The intestinal is also highly interactive with the gut-brain axis, influencing both neural and endocrine signalling, but also modulating the immune response. Short chain fatty acids (SCFAs) produced by intestinal can, for instance, stimulate EC cells to increase serotonin production (41). In addition, many of the common neurotransmitters in our nervous system can be synthesised by commensal bacteria (42). Moreover, intestinal macrophages located in the muscularis externa can, by the influence of commensal microbiota, cause changes in smooth muscle contractions of the colon (43).The influence of the on the gut-brain axis, howev- er, is beyond the scope of this thesis.

Melatonin signalling in peripheral tissue

MT1 and MT2 melatonin receptors have been identified in several organs and tissues primarily in animal studies (44-46). In humans, MT1 and MT2 have been identified in different parts of the brain, with notable differences in distribution between the two receptors (47-50). The MT1 receptor, identified in immune cells, has been more extensively investigated (51). Few studies, however, have investigated the receptor expression in the normal GI tract of humans. In one human study binding sites in the colon, caecum and appen- dix were identified using iodinated melatonin. Another human study found expression of MT1 in colon adenocarcinoma but also identified the MT1 re- ceptor in normal adjacent tissue of the colon using immunohistochemistry (52, 53). Table 1 summarises the present knowledge of MT1 and MT2 recep- tor distribution in normal human tissue.

Table 1. Summary of melatonin receptor expression in normal human tissue. Organ/tissue Receptor Method Reference

Brain MT1, MT2 PCR, IHC (47, 49, 50, 54, 55) Retina MT1, MT2 PCR, IHC (MT1) (56) Parotid glands MT1, MT2 (weakly) IHC (57) Cardiovascular MT1, MT2 PCR (58, 59) system Immune system MT1MT2 PCR (51, 60) Brown adipose MT1, MT2 PCR (61) tissue Breast MT1 IHC (55) Pancreas MT1 (alpha-cells), MT2 PCR (62, 63) (beta-cells) Gallbladder MT1 PCR, western blot, IF (64) GI tract 2[125I]iodomelatonin Autoradiography, PCR, (52, 53) binding sites, MT1 (colon) IHC, western blot Kidney MT1, MT2 PCR (65)

16 Ovary MT1, MT2 PCR (66) Uterus MT1, MT2 PCR, in situ hybridisation (67) Placenta MT1, MT2 IHC (68) Prostate MT1, MT2 IHC (69) Skin MT1, MT2 PCR (70) Abbreviations: GI = gastrointestinal, MT1/MT2 = melatonin receptor type 1/type 2, PCR = polymerase chain reaction, IHC = immunohistochemistry, IF = immunofluorescence.

Consequences of peripheral melatonin Melatonin in the gastro-pancreatic system As early as 1975, Kvetnoy and colleagues reported the presence of melato- nin in the human GI tract (71). From studies in animals, the cells responsible for the synthesis of gut melatonin are the EC cells (72, 73). The EC cells are neuroendocrine cells that line the intestinal mucosa and are the main source of gut-derived serotonin. They function as sensory cells, responding to vari- ous stimuli from the lumen. For example, changes in pH, glucose and amino acids cause release of serotonin, which, can activate neural reflexes that re- sult in changes in mucosal blood flow, secretion and gut motility (74). The amount of melatonin in the GI tract easily surpasses the amount in the pineal gland. It has also been demonstrated that melatonin produced in the gut most likely attributes to daytime circulating levels, as oral admin- istration of melatonin or its precursor tryptophan increases circulating levels of melatonin. An increase in melatonin in the portal blood was seen before that in the systemic circulation and the effect was almost completely elimi- nated by ligature of the portal vein, indicating that the GI tract was the source of this rise in circulating levels (6, 75). Pinealectomy reduces night- time levels of circulating melatonin, but it does not affect levels in the GI tract, suggesting that gut melatonin is independent of pineal production (76). Variations in GI melatonin levels appear to be related to fasting and food intake, with both resulting in increased formation of melatonin in the GI tract in response to food restriction, as well as postprandial elevations of melato- nin in human saliva (77, 78). Moreover, receptor expression appears to be highly variable with nutritional status. In rats up-regulation of MT1 mRNA transcripts increased after short-term fasting (79). Melatonin has been demonstrated to increase intracellular (Ca2+) in human and rat enterocytes, possibly through the MT2 receptor, as the ef- fect was eliminated when adding a predominantly MT2-selective antagonist (80). Another study has identified MT1 immunoreactivity (IR) in the human colon (53). The actions of gut melatonin include regulation of intestinal mo- tility, protection of intestinal mucosa by reducing the paracellular permeabil- ity and glucose homeostasis. Understanding the complex systems that con- trol intestinal motility is challenging, largely because many substances are

17 involved. Nevertheless, it has been proposed that melatonin and serotonin co-operate through a feedback system, in which melatonin appears to act as a physiological antagonist of serotonin, mainly dampening motility (81, 82). Melatonin also appears to influence glucose homeostasis and melatonin receptors have been identified in the human pancreas, with MT1 mRNA lo- cated largely in the glucagon-secreting cells of the pancreas, whereas MT2 mRNA seems to be primarily located in the insulin-producing cells (62, 63). For example, melatonin can affect transcription factors involved in insulin secretion (83) and mutations in the gene encoding the MT2 receptor, result- ing in impaired melatonin signalling increases the risk of type 2 diabetes (63). In summary, both melatonin and its receptors are widely expressed in dif- ferent organs and melatonin in the gut appears to play an important role in many regulatory functions. However, studies demonstrating the expression of melatonin and melatonin receptors in the human GI tract and pancreas are scarce.

Immunomodulatory functions of melatonin Perhaps the first and most important function of melatonin during evolution is as a powerful antioxidant, acting as a scavenger of free radicals (11, 84). In contrast to other well-known antioxidants, which generally neutralise one free radical per molecule, melatonin is far more powerful. The reason for this expansive anti-oxidative effect is the cascade of metabolites generated when melatonin interacts with reactive oxygen or nitrogen species. Many of these metabolites also have radical scavenging capacities, some even more powerful than melatonin (85). In addition, melatonin can reduce oxidative stress indirectly by influencing the expression and activity of other antioxi- dant enzymes (86). Melatonin also acts as a modulator of immune functions and human lym- phocytes produce large amounts of melatonin (87). According to a recent review, melatonin may act as a stimulatory factor on the immune system in immunosuppressive conditions and, in contrast, augment anti-inflammatory effects in acute inflammation (88). Melatonin and its precursor, serotonin, are abundant in the GI tract and both are important modulators of immune responses. For example, endocrine cells lining the gut can modulate the ac- tivity of immune cells. By interaction with EC cells, CD4+ T cells can in- crease serotonin production, and conversely, serotonin can regulate inflam- mation, activating immune cells to produce and secrete inflammatory media- tors (89). Pro-inflammatory cytokines (such as TNF and IFN-γ) increase intestinal paracellular permeability, which appears to play a role in the path- ophysiology of irritable bowel syndrome (IBS) and linked to more severe GI symptoms (90-92).

18 The immunomodulatory properties of serotonin seem to be chiefly stimu- latory, where administration of serotonin receptor antagonists can attenuate intestinal inflammation (93). In contrast, significant protective effects against stress-induced damage to the intestinal mucosa have been attributed to melatonin. Luminal melatonin strengthens tight junctions and reduces mucosal paracellular permeability in the duodenum (94), which prevents bacterial translocation and leakage of inflammatory substances. Moreover, melatonin binding to MT2 stimulates bicarbonate secretion in the duodenum, thereby protecting the mucosa from the acidic luminal contents propagated from the stomach (95). Finally, in rats melatonin administration reduced the damages seen in small intestinal microvasculature after exposure to systemic inflammation, possibly by limiting local immune cell recruitment (96). The authors proposed a therapeutic benefit of melatonin in mild systemic in- flammation. The synthesis of melatonin is dependent on the availability of its precur- sors, tryptophan and serotonin. Immune activation induces indolamine 2,3- dioxygenase (IDO), a key enzyme in the tryptophan metabolism, driving tryptophan towards the kynurenine pathway, away from serotonin and mela- tonin production (97). Both activation of IDO and the tryptophan 2,3 dioxy- genase (TDO) decrease serotonin availability, and consequently, less mela- tonin can be synthesised (98). The metabolism of tryptophan via the kynurenine pathway results in kynurenine and other metabolites involved in immunomodulatory processes linked to depression and suicidal behaviour (99). Metabolites of the kynurenine pathway include quinolinic acid (QUIN), which is associated with neurodegenerative changes and such disorders as multiple sclerosis, Alzheimer’s disease and major psychiatric disorders. On the other hand, the metabolite kynurenic acid (KYNA) is neuroprotective, counteracting the negative effects of QUIN (100). The enzyme converting kynurenine to KYNA is up-regulated by physical exercise, which can help explain the mechanisms underlying the positive effects of physical exercise in depression (101). In summary, low levels of melatonin, either due to a shift in tryptophan metabolism or disturbed melatonin signalling due to impaired receptor func- tion, may be linked to symptoms of depression and anxiety.

Melatonin and cancer When it was concluded that the risk of developing cancer is higher in shift workers exposed to light at night, melatonin and its involvement in cancer development became a subject of discussion. The increased risk is thought to be at least partly due to the suppression of nocturnal production of melatonin in the pineal gland (102). The anti-tumorigenic effects of melatonin have been investigated in several types of cancer. The overall findings demon-

19 strate that melatonin exhibits strong oncostatic effects, including anti- proliferative (103, 104) and pro-apoptotic actions (105). Many studies have investigated the influence of melatonin in mammary cancer and the oncostatic effect seen appears to be related to the regulation of different hormone receptors. However, melatonin also looks to inhibit cancer growth by affecting several signalling pathways. For instance, in prostate cancer melatonin causes up-regulation of the cell cycle regulatory protein p27kip1, which could inhibit tumour proliferation (106). In human MCF-7 breast cancer cells melatonin decreases cell proliferation by increas- ing the expression of p53 and p21WAF1 proteins, which induce cell cycle arrest (107). One recent study demonstrated that melatonin could inhibit cytochrome C release from mitochondria, an initiator signal for apoptosis, via MT1 receptors on the mitochondrial membrane (108). The opposite is found in haematological malignancies, where melatonin appears to induce apoptosis by activation of the intrinsic and extrinsic pathway (109).

Small intestinal neuroendocrine tumours EC cells are hormone-producing cells located throughout the GI tract. When these cells become malignant, they form neuroendocrine tumours, which also produce hormones. Small intestinal neuroendocrine tumours (SI-NETs) are characterised by their low proliferation rate and long survival expectancy and yet high propensity to metastasise to the mesentery and liver. The tu- mours are rare, with an incidence of 1.12 per 100 000 inhabitants in Sweden (110). However, a recent study using autopsy records discovered that the number might be higher, with subclinical disease and multiple primary tu- mours present before metastatic progression (111). SI-NETs produce and secrete serotonin and other bioactive agents such as tachykinins (112), causing the carcinoid syndrome, a set of symptoms that include diarrhoea, vomiting, cutaneous flush, carcinoid heart disease and bronchial constriction. At the time of diagnosis, metastases have generally already formed. The diagnosis of SI-NETs is based on the analysis of CgA and urinary 5- hydroxyindoleacetic acid (U-5-HIAA), radiology and histopathological analysis (113). Surgery is the primary treatment for SI-NETs, which delay the recurrence of the disease and reduce symptoms. When the tumour has spread, surgery is still beneficial to reduce hormone levels and prevent com- plications of fibrosis (e.g., intestinal obstruction and bowel ischemia) (114). Medical treatment includes somatostatin analogues, interferon-alpha, or both and aims to ameliorate symptoms caused by hormone production and delay progression of tumour growth (115-117). SI-NETs constitute a form of hormone-producing tumours resulting in major effects on gut function and symptoms. What is noteworthy is that alt- hough melatonin is only two enzymatic reactions from serotonin, the produc-

20 tion of melatonin in these tumours is yet to be investigated. If present, this could be an important disease model for understanding the effects of melato- nin in malignancy and on GI function.

Functional gastrointestinal disorders Functional GI disorders (FGIDs) are a group of disorders without clear path- ogenesis. They are disorders that are common and costly for society (and the individual), often causing considerable impact on health-related quality of life (QoL) (118). IBS is the most common FGID, with a pooled global prevalence of 11.2%, with most studies reporting a higher prevalence in women than in men (119). Symptoms of IBS include recurrent abdominal pain and bloating in combination with altered stool consistency and frequen- cy. The diagnosis of IBS is based on the Rome Criteria, with recurrent ab- dominal pain, at least one day per week for the past 3 months and associated with two or more of the following: 1) related to defecation, 2) associated with a change in frequency of stool and 3) associated with a change in the form of stool (120). Currently, there are no clinically accepted biomarkers for IBS, although several molecules have been investigated. In the context of visceral hyper- sensitivity, Chromogranin A, faecal calprotectin, some SCFAs and β- defensin 2 have been reported to be altered in IBS patients compared with controls (121). Changes in bile acid balance and altered colon transit have also been proposed as possible future biomarkers for significant gut dysfunc- tion in IBS (122). Peripheral factors reported to be involved in the pathophysiology of IBS are increased gut permeability, changes in enteroendocrine activity, dysregu- lation of the immune system and altered gut microbiota composition, all affecting the bi-directional communication between the gut and the brain commonly referred to as the gut-brain axis (the biochemical signalling that occurs between the GI tract and the central nervous system) (90, 123). Of potential relevance for melatonin is that higher levels of postprandial serotonin levels in plasma have been reported in patients with diarrhoea- predominant IBS and in patients with post-infectious IBS following gastro- enteritis. Patients with -predominant IBS, however, had lower levels of serotonin compared with controls (124). Melatonin has been studied in the treatment of IBS. Although the dosage of melatonin administered and the methodology of the studies vary widely, some studies have primarily found amelioration of abdominal pain and QoL in patients with IBS (125, 126).

21 Psychiatric co-morbidity and functional gastrointestinal disorders Psychiatric co-morbidity is high in patients with FGIDs and up to 60% of patients who seek care for FGIDs also suffer from psychiatric disorders, predominantly depression and anxiety (127). In a Hong Kong study based on telephone surveys with questionnaires, generalised anxiety disorder (GAD) was five times more common in respondents with IBS and respondents with both IBS and GAD had higher functional impairment than those suffering from only one of the disorders (128). Disturbances of the HPA axis are common in people with FGIDs, who typically present with high anxiety traits and psychiatric disorders. (127, 129). IBS patients have a higher increase in ACTH than controls after CRH administration and variations in the CRHR1 gene have been linked to high anxiety traits and an increased risk of developing stress-induced psycho- pathology (130, 131). Another proposed mechanism is via the CRH, mast cell activation and the release of proteases (132). In addition, stress can cause alterations in gut permeability, which further supports the link between the brain and gut in FGIDs. Most studies have investigated the prevalence of psychiatric disorders in patients with IBS, but less research has been done from a psychiatric per- spective, especially in younger patients in which the somatic disorder pano- rama is different from an older population. In this respect it is of interest to separate different types of GI symptoms in relation to anxiety traits but also current psychiatric diagnosis and depressive symptoms. Finally, as described above, previous work indicates that melatonin sig- nalling may be disturbed in some patients with depression and anxiety. The next question is whether different patterns of melatonin production during the day are related to specific symptoms of GI dysfunction in young adult patients with psychiatric morbidity.

22 Aims

The overall aim of this thesis was to study the relationship between melato- nin and its receptors in the human GI tract and gut function and psychiatric disorders. The specific aims of each paper were:

I To characterise the tissue expression of melatonin and its receptors in the normal human GI tract and pancreas.

II To characterise the expression of melatonin and its receptors in small intestinal neuroendocrine tumours and further investigate possible links between circulating levels of melatonin, GI symptoms, prognostic fac- tors and treatment response.

III To investigate the frequency and severity of IBS symptoms in young adults with mood or anxiety disorders.

IV To examine the possible associations between daytime levels of melato- nin and IBS symptoms in young adults with mood or anxiety disorders.

23 Materials and methods

Design The four studies of this thesis were cross-sectional observational studies.

Setting Paper I Human tissue, representing different parts of the GI tract, was acquired from resection margins from surgical material removed due to various malignan- cies in adult patients. Normal macro- and microscopic appearance of the tissue was a prerequisite for inclusion in the study. Gene expression data for key enzymes in tryptophan metabolism, seroto- nin and melatonin receptors from small intestine and pancreas were extract- ed from the Gene Expression Omnibus (GEO).

Paper II Tumour tissue, representing serial sections of primary tumours and metasta- ses from patients with SI-NETs was acquired from the Laboratory of Pathol- ogy and Cytology, Uppsala University Hospital in Sweden. Patients were treated at the Department of Endocrine Oncology, Uppsala University Hos- pital in Sweden. Plasma samples from patients with SI-NETs were collected before and af- ter medical or surgical treatment and promptly stored at -80° C until assayed. Medical records for patients with SI-NETs were examined and clinical data on age at time of diagnosis, body mass index (BMI), smoking history, diabetes, psychiatric medication, Ki67 and levels of U-5HIAA and CgA at the time of surgery or plasma sampling collected. Treatment response was classified as disease regression, stabilisation or progression, measured by biochemical or radiological examination at follow-up.

24 Paper III and IV Data used in paper III and IV were collected as part of the Uppsala Psychiat- ric Patient Samples (UPP) cohort, a project designed to collect biological material from patients seeking psychiatric care at the Department of General Psychiatry, Uppsala University Hospital. For study III, data was collected between 2013 and 2017 from patients 18-25 years old with primarily mood and anxiety disorders; for paper IV, data were collected from the same co- hort but between 2012 and 2014.

Study population and patient samples Paper I Tissue material from the GI tract from 39 individuals and pancreatic tissue from 3 individuals were analysed (17 purchased from Asterand, Detroit, MI, USA and 25 from the Department of Pathology, Uppsala University Hospi- tal, Uppsala, Sweden). Biopsies represented stomach (n=12), small intestine (n=11), appendix (n=3) and (n=13). No additional clinical data for the patients were known and thus not included in the analyses.

Paper II Fifty-two patients were included in the study. For histology assessment, only cases with paired sections of primary tumour and metastasis were included (n=26). For plasma analyses, patients who completed plasma collection at both time points (before and after treatment) were included (n=43). Seventeen patients were included in both tissue and plasma analyses (Figure 2).

25

Figure 2. Flowchart of study patients in paper II.

Paper III From the UPP cohort, which comprised patients, age 18-25 seeking psychi- atric outpatient care, 682 patients were eligible for this study. Patients with established inflammatory bowel disorder, endometriosis and Anorexia ner- vosa were excluded. In addition, patients were excluded who did not under- go a structured psychiatric assessment or when questionnaires were incom- plete or improperly completed. In total, 491 (72%) patients were included in the study. Healthy controls under the age of 30 years (n=139) were recruited from the staff of Uppsala University Hospital and students at Uppsala University. Of these, 85 (61%) were included in the statistical analyses (Figure 3).

Paper IV For paper IV, the study population originated from the same UPP cohort as for paper III. From 621 eligible patients, 264 (43%) agreed to participate in this study and 102 (39%) of these patients completed both saliva sampling and validated questionnaires for GI symptoms. Six patients were excluded from the analyses because they did not fulfil criteria for any DSM-IV axis I diagnosis.

26

Figure 3. Flowchart of study participants in paper III.

Microarray expression analysis

For paper I, raw data from human small intestinal epithelium, of which it is estimated that 25% of the captured cells are endocrine, and from human normal pancreas tissue were extracted from GEO (133). The gene expression series is summarised in Table 2. Data were imported into Expression Console software provided by Affy- metrix (http://www.affymetrix.com) and normalised with the robust multi- array average method (134). Log2 expression signals were then extracted. Using the statistical computing language R (http://www.r-project.org), gene expression data for melatonin and serotonin receptors as well as key en- zymes in melatonin synthesis were analysed in the data from small intestinal epithelium, whole pancreas and pancreatic islets. Table 4 presents a sum- mary of the genes analysed. Expression signals for parathyroid hormone were used as a negative control. Differences in gene expression between samples from colon biopsies in patients with ulcerative colitis (UC) and healthy controls were analysed us- ing the Linear Models for Microarray Analysis (Limma) R package on GEO2R (http://www.ncbi.nlm.nih.gov/geo/info/geo2r.html).

27 Table 2. Summary of the gene expression series from the Gene Expression Omnibus data archive used in the microarray analyses. Adapted from licentiate thesis (8).

Table 3. Summary of genes analysed using microarray analysis. Adapted from licen- tiate thesis (8).

28 Immunohistochemistry Tissue specimens representing the normal human GI tract and tumour tissue were stained for melatonin, MT1 and MT2 receptors and serotonin. Tissue sections from human skin served as positive controls for MT1 and MT2 and antibody specificity tests were performed for melatonin, MT1 and MT2. For double immunofluorescence staining, tissue sections were incubated with a cocktail of two primary antibodies. Detailed information on immunohisto- chemical methods, antibodies and positive/negative controls is provided in paper I.

Histological assessment – Paper I Two independent observers examined each tissue specimen microscopically and assessed immunoreactivity (IR) for melatonin, MT1 and MT2 receptors. Intensity was classified into four categories: negative, weak, medium or strong. IR localisation in different cell types was documented. Co-localisation studies of sections with double immunofluorescence staining were performed using a Zeiss 510 confocal microscope.

Histological assessment – Paper II Tumour tissue specimens were examined microscopically by three inde- pendent observers and IR intensity for melatonin, MT1 and MT2 was classi- fied after study I. The intraclass correlation coefficient between observers was 0.955, indicating high reliability. Tissue specimens were also analysed using Image J software and the intraclass correlation coefficient between computerised and manual scoring was high (0.934).

Hormone collection and analysis Analysis of melatonin in plasma – Paper II Plasma samples were collected at two time points (before and after treat- ment). Sampling was performed in the morning from fasting patients. Plas- ma levels of melatonin were measured using competitive radioimmunoassay (Melatonin direct RIA, LDN, Norhorn, Germany).

Analysis of melatonin in saliva – Paper IV Patients collected saliva samples at home at six time points for one day: when waking up, 30 min after waking up, at 11:00, 30 min after lunch, at 22:00 and bedtime. Salivary melatonin levels were measured at the Depart-

29 ment of Clinical Chemistry at Uppsala University Hospital, using the com- petitive enzyme-linked immunosorbent assay technique (ELISA) (Direct Salivary Melatonin Elisa EK-DSM. Bühlmann Laboratories AG.Schönenbuch. Switzerland). The three time points representing daytime melatonin measurements (30 min after waking up, at 11:00, 30 min after lunch) were selected for statistical analysis.

Figure 3. Illustration of saliva samples and ELISA with the three daytime measure- ment time points selected for analyses in paper IV.

Psychiatric assessment and questionnaires – Paper III and IV A trained doctor in psychiatry or a clinical psychologist performed the psy- chiatric assessment by structured or semi structured interviews, using the Structural Clinical Interview for DSM-IV Axis I Disorders – Clinical Ver- sion (SCID-I) or the Mini-International Neuropsychiatric Interview (M.I.N.I.). In conjunction with the psychiatric assessment, a physical exami- nation was performed and BMI recorded.

Self-assessment – Paper III and IV Montgomery-Åsberg Depression Rating Scale The self-rating version of the Montgomery-Åsberg Depression Rating Scale (MADRS-S) was used to rate depressive symptoms. The MADRS-S is a nine-item diagnostic questionnaire with items rated on a six-point Likert- scale. The items include mood, feeling of unease, sleep, appetite, ability to concentrate, initiative, emotional involvement, pessimism and zest for life. The overall score ranges from 0-54 points.

Gastrointestinal Symptoms Rating Scale for IBS The Gastrointestinal Symptoms Rating Scale for IBS (GSRS-IBS) was used to measure GI symptoms. The GSRS-IBS is a validated self-assessment in-

30 strument for the evaluation of IBS symptoms (140). The instrument contains 13 questions on a seven-point Likert-scale (1-7), spanning from no symp- toms to very severe symptoms during the past week. The overall score rang- es from 13-91. The questions are grouped into five symptom clusters depict- ing pain, bloating, constipation, diarrhoea and satiety.

Swedish Universities Scales of Personality The Swedish Universities Scales of Personality (SSP) instrument was used to estimate personality traits at the baseline assessment. The SSP is a revised version of the Karolinska Scales of Personality (KSP), a self-rating ques- tionnaire with improved psychometric properties. The instrument contains 91 items divided into 13 scales (141). Normative T-scores for the extracted scales of somatic trait anxiety (STAT), psychic trait anxiety (PsTAT) and stress susceptibility (SST) were used for data analysis in paper III.

Statistics Paper II Data were analysed using the Statistical Program Package for the Social Sciences (SPSS Inc., Chicago, IL, USA). Statistical analysis of the two co- horts, where 1) tumour tissue and 2) plasma samples were available, was conducted separately and they were not compared with each other. Survival was calculated from time of diagnosis to death (or, if still alive, to 25 April 2015) and analysis was performed using the Mantel-Cox log- rank test. The Wilcoxon signed-ranks test was applied for comparison of IR intensity between primary tumour and metastasis and for change in bi- omarker response between samplings. The Spearman’s test (rho) was used to calculate pairwise correlations be- tween IR intensity or plasma levels of melatonin and clinical parameters. Sex differences in plasma levels of melatonin were analysed using the Mann-Whitney U test.

Paper III The Mann-Whitney U test was performed to compare GSRS-IBS scores between patients and healthy control, followed by the Kruskal-Wallis one- way analysis of variance for comparison between three groups (patients with psychotropic medication, psychotropic medication-free patients and con- trols). A principal component analysis (PCA) was performed using normalised z-scores of the individual items of the MADRS-S and GSRS-IBS but also the scales of STA, PsTA, SS and BMI to identify principal components,

31 which accounted for most of the variance in the dataset. A two-step cluster analysis method was then performed to look for subgroups in the patient population. Spearman’s rank test (rho) was carried out to assess pairwise correlations between GSRS-IBS scores, MADRS-S scores and the STAT, PsTAT and SST SSP subscales. To control for possible confounding factors a general- ised linear model was constructed using the covariates sex, BMI and ongo- ing bulimia nervosa. A second model was then performed that also included the three scales representing trait anxiety.

Paper IV Generalised linear model analyses were conducted for salivary melatonin at the three time points in relation to GSRS-IBS scores. Sex, BMI, anti- depressive medication and oral contraception were included as potential confounders. The Bonferroni correction method was applied to adjust for multiple comparisons. Post-hoc explorations of correlations between the GSRS-IBS subscales and salivary melatonin were performed using the Spearman’s rank test.

32 Summary of results

Melatonin and melatonin receptor expression (Paper I) Normal human GI tract Positive IR for melatonin was found in EC cells throughout the GI tract. Co- localisation studies showed cellular localisation of melatonin in the cyto- plasm that was only partially overlapping with serotonin. In addition, diffuse melatonin IR was seen in the cytoplasm of enteric epithelial cells and mono- nuclear immune cells of the lamina propria. MT1 IR was negative in EC cells throughout the gut, whereas positive IR for MT2 was found in EC cells in 5 of 12 of the gastric sections and all of the sections from the small intestine and large intestine and appendix. In large intestinal epithelial cells both MT1 and MT2 IR were strong, with cellular localisation in the cytoplasm. Positive IR for both receptors was also seen in the myenteric plexus and vascular structures, with generally more positive sections and higher intensity for MT2 compared with MT1. In the submuco- sal plexus only positive IR for MT2 IR was seen. The gene expression analysis supported these results in that mRNA levels of MT2 were higher than MT1 in small intestinal epithelium enriched for EC cells.

Normal human pancreas Melatonin IR was identified in endocrine cells of pancreatic islets, where also MT2 IR was positive in a subset of endocrine cells. MT1 IR varied be- tween sections, with medium IR in endocrine cells in one section, whereas the other two tissue sections were negative for MT1. In the gene expression analysis mRNA for the gene encoding MT2 was higher than for MT1 in pancreatic tissue, which supports the IHC results.

Melatonin and melatonin receptors in SI-NETs (Paper II) Tissue expression Positive melatonin IR was seen in all tumour tissue sections, with varying IR intensity (weak to strong) in primary tumours and in metastases. MT1 IR was

33 weak or negative in tumour tissue, whereas MT2 IR was positive in all sec- tions representing primary tumours and in 92% of sections representing me- tastases. IR intensity of MT2 was lower in metastases compared with primary tumours (r=0.53; p=0.007).

Plasma levels The median level of melatonin in morning plasma from patients with SI- NETs was 26.0 pg/L [4.5–220.0] before treatment and 23.0 pg/L [8.9–90.7] after treatment. Patients with disease regression or stabilisation according to biochemical or radiological response had lower levels of plasma melatonin at the second sampling (r=0.356; p=0.038).

Relationship between melatonin tissue intensity, plasma levels and clinical parameters Higher melatonin IR intensity in primary tumours correlated with lower Ki67 (r=-0.446; p=0.022) and to less reported symptoms of diarrhoea (r=- 0.484; p=0.012). A positive correlation between higher levels of melatonin in plasma and increased nausea or vomiting was noted at both sampling time points (rho=0.337; p=0.027 and (rho=0.413; p=0.006). High melatonin levels at the second sampling (i.e. post-treatment) also correlated with the symptom of flush (rho=0.353; p=0.020).

GI symptoms and depressive symptoms (Paper III) In this study 491 patients (mainly with mood or anxiety disorders) and 85 healthy controls were included. Patients had higher MADRS-S scores (me- dian 23 vs. 4; p<0.001), and GSRS-IBS scores (median 30 vs. 22; p<0.001) than controls. There were no differences in total GSRS-IBS scores between patients receiving psychotropic medication and psychotropic medication-free patients. Using PCA, six factors were identified, which accounted for 63,9% of the variation in the dataset. They were characterised according to the items that loaded the highest on each construct and were labelled as follows: slow bowel, fast bowel, depressive symptoms, trait anxiety, disturbed appetite and BMI. A cluster analysis resulted in six clusters and the characteristics of the six cluster groups are shown in Figure 5. Higher MADRS-S total scores correlated with higher GSRS-IBS total scores and the association remained significant after controlling for sex, BMI and ongoing bulimia in a generalised linear model. The GSRS-IBS total

34 scores also correlated with all three SSP subscales analysed: STAT (rho=0.313; p<0.001), PsTAT (rho=0.147; p=0.001) and SST (rho=0.233; p<0.001). The correlation with STAT showed the largest effect size of the three subscales. In a second generalised linear model we discovered that all studied factors were independent predictors of higher GSRS-IBS total score.

Figure 4. Chord diagram of the different cluster groups for the six factors extracted from the principal component analysis. Abbreviations: MADRS-S = Montgomery- Åsberg Depression Rating Scale self-rating version, GSRS-IBS = Gastrointestinal Symptoms Rating Scale for Irritable Bowel Syndrome, STAT = Somatic trait anxie- ty T-score, PsTAT = Psychic trait anxiety T-score, SST = Stress susceptibility T- score, BMI = body mass index.

Melatonin levels and GI symptoms (Paper IV) Daytime melatonin levels in saliva were analysed and compared with report- ed GI symptoms in 96 young adult patients seeking psychiatric care for

35 mainly mood or anxiety disorders. The median levels of salivary melatonin were 6.9 (2.7–15) 30 min after waking up, 3.3 (1.8–5.8) at 11:00 hours and 2.8 (1.8–4.7) 30 min after lunch. The median total GSRS-IBS score was 31 (range=13–78). Higher levels of melatonin in saliva after lunch correlated with higher to- tal GSRS-IBS scores, a relationship that remained significant after control- ling for possible confounding factors (p=0.015, q=0.045) and bloating and pain contributed most to this association.

36 Discussion

The present studies have demonstrated the widespread presence of melatonin and melatonin receptors in the normal human gut, pancreas, and for the first time, in SI-NETs. In these tumour patients melatonin levels in plasma and IR in the tumours correlated with clinicopathological features. Moreover, young adult patients seeking psychiatric care commonly report GI symptoms, which are independently associated with sex, depressive symptom severity, trait anxiety and BMI. In agreement with our hypothesis of a role for mela- tonin in GI symptoms from paper II a link between daytime melatonin levels and GI symptoms was identified in young adults with psychiatric morbidity.

Melatonin and melatonin receptors in normal human gut and pancreas In paper I we demonstrated positive IR for melatonin in EC cells, a finding concurring with previous knowledge from animal studies in which the en- zymes needed for melatonin synthesis have been identified in EC cells (71, 142, 143). Microarray gene analyses also supported this finding and together these results strongly suggest the production of melatonin in EC cells in humans. In addition, a diffuse positive IR for melatonin was seen throughout the gut, with localisation in the cytoplasm of epithelial cells and the mucus layer lining the epithelium. A tendency towards higher intensity was ob- served in the aboral direction, indicating an accumulation of melatonin. Mel- atonin is amphiphilic and, unlike serotonin, not stored in secretory vesicles. It can easily cross cell membranes, which could explain the scattered distri- bution reported in IHC analyses. In addition to synthesis in EC cells, mela- tonin is produced by and the intestinal microbiota may fur- ther contribute to melatonin accumulation in the large intestine (144). Final- ly, knowing that vegetables and meat contain melatonin, part of the melato- nin noted in the mucosal lining may originate from ingested food. (6, 145). The distribution of MT1 and MT2 melatonin receptors was somewhat dif- ferent. MT2 IR was seen in EC cells, whereas MT1 IR was negative. In the epithelium IR for both receptors was positive, with a generally higher inten- sity for MT2. MT1 IR localisation in epithelial cells of the colon is in agree- ment with a study of human colonic mucosa (53). Beyond this, human stud-

37 ies are limited and receptor expression has mainly been investigated in ani- mals. A study in rats demonstrated MT1 mRNA in the sub-epithelial layers of the GI tract, with generally low levels of mRNA in the epithelium (79). The reason for this discrepancy may be related to differences in feeding be- haviour and diet, as melatonin levels vary with fasting and food intake, pos- sibly regulating receptor expression through a feedback mechanism. IHC studies of rat intestine found intense expression of MT2 in colonic smooth muscle, but not in the villi or the crypts of the mucosa (146). The finding of MT2 in the human colonic epithelium may, in addition to species-specific differences, be caused by the use of different antibodies. However, we show that the machinery for melatonin synthesis is actively expressed in the hu- man gut and thus likely not only absorbed from food containing melatonin. Some of the central functions of gut melatonin include protecting the mu- cosal barrier (e.g., by reducing paracellular permeability and increasing bi- carbonate secretion) (94, 95). These effects appear to be receptor-mediated given that the effect was abolished when adding a melatonin receptor antag- onist. In vascular structures IR for both receptors was positive and melatonin has recently been shown to reduce inflammatory damage to the microvascu- lature in the small intestine (96). In response to inflammation induced by lipopolysaccharide administration to rodents an increase in the AANAT enzyme has previously been demon- strated in macrophages in mice resulting in melatonin synthesis (147). The radical scavenging properties of melatonin, protecting lipids, proteins and nuclear DNA from oxidative stress, are receptor-independent, merely requir- ing the close presence of melatonin where the radical is formed (148). More- over, in patients with UC, both an increased number of EC cells and higher levels of HIOMT in colonic mucosa have been reported (149). In paper I, however, there were no significant differences in gene expression of key enzymes in melatonin synthesis or melatonin receptors in tissues from pa- tients with UC compared with controls. Melatonin appears to be involved in glucose homeostasis. In paper I pan- creatic endocrine cells demonstrated positive IR for melatonin and MT2. Gene expression analysis supported the finding of a dominating MT2 recep- tor expression, with higher levels of MTNR1B (encoding MT2) compared with MTNR1A (encoding MT1), which, however, is not consistent with pre- vious studies in which the relationship was found to be reversed (62). A re- cent study investigating the expression of MT1 and MT2 melatonin receptors in human pancreas found receptor expression in alpha, beta and delta cells, with the highest receptor density in alpha cells (150). Mutations in the MT2 receptor that cause impaired melatonin signalling, and decreased melatonin secretion increase the risk of type 2 diabetes (151, 152). One study in rats demonstrated that the administration of a MT1/MT2 agonist ameliorated im- paired glucose metabolism induced by chronic stress and high-fat diet. The authors concluded that the mechanism for this was through normalisation of

38 the HPA axis function, evidencing important regulatory receptor-mediated actions of melatonin in glucose homeostasis (153).

Melatonin and melatonin receptors in SI-NETs In paper II the presence of melatonin in SI-NETs has been described for the first time. Our analysis also revealed that all tumour sections (primary tu- mour and metastasis), showed positive IR for melatonin. Higher IR intensity was significantly associated with a lower Ki67. Ki67 is a widely used marker for proliferation and the discovered association could indicate anti- proliferative effects of melatonin in these tumours. The anti-tumorigenic effects of melatonin in other non-endocrine cancers are more extensively studied and include several mechanisms and signalling pathways that result in proliferation suppression, induction of apoptosis, anti-metastatic actions and a drive towards cell differentiation. The oncostatic effects of melatonin have recently been reviewed (106, 154). Most studies, however, have inves- tigated the effect of melatonin administration in different cancers, rather than the endogenous melatonin effect. In prostate cancer, for instance, melatonin administration stopped cell cycle progression, reduced cell growth and stim- ulated cellular differentiation (155). Melatonin receptor expression in different types of solid tumour varies and its clinical implications may be of varying importance. In GI neuroendo- crine tumours the influence of melatonin has, to our knowledge, not been investigated. In ovarian cancer, MT1 receptor expression was identified but there was no correlation between receptor expression and prognostic factors (156). In mammary and prostate cancer, however, a growth inhibitory effect of melatonin has been demonstrated and linked to the MT1 receptor (106, 157). Therefore, a more dominant expression of MT1 could have been ex- pected. The MT2 receptor has not been as extensively investigated in the context of tumour progression, although one study in mice found oncostatic effects in colon cancer, probably mediated via the MT2 receptor but also via nuclear receptors of the ROR/RZR family (158). In addition, the growth- inhibiting effects of melatonin in melanoma are presumably also mediated through the MT2 receptor (159). In paper II we investigated SI-NETs and found that MT2 IR was rich in primary tumours and generally lower in me- tastases. Down regulation of receptors may indicate a reduced sensitivity to the anti-proliferative effects of melatonin with increasing malignant progres- sion. Daytime levels of melatonin are primarily of peripheral origin and the gut constitutes a major source of extrapineal melatonin (78). With possible mela- tonin production in SI-NETs, higher circulating levels of melatonin could have been expected. However, as mentioned earlier, melatonin is not con- fined to the bloodstream but passes freely across membranes. Moreover,

39 clearance through the liver is high, which could explain why levels in plasma remain modest (75). In tumour patients with disease regression or stabilisa- tion plasma a reduction in levels of melatonin was noted at the second sam- pling (after treatment), possibly because of a reduction in tumour burden and less tumour-derived melatonin. The most prominent symptom of SI-NETs is diarrhoea, with watery stools up to 10 times per day. This symptom is mainly caused by the overwhelming release of serotonin. Melatonin affects motility and its actions appear to be largely inhibitory, with a longer colonic transit time (160). In rats melatonin administration attenuated the stress-induced increase of faecal output and decreased the dry weight of stool (161). In paper II patients with high mela- tonin IR in tumour sections reported less difficulty with diarrhoea, specula- tively related to a larger inhibitory effect of locally produced melatonin. In addition, circulating levels of melatonin correlated positively with more nau- sea or vomiting. Theoretically, slower intestinal transit can cause nausea because blocking of 5-HT3 receptors prolongs colonic transit time and 5-HT3 antagonists are used as an anti-emetic, relieving symptoms of nausea (162, 163). Other possible explanations for the symptom of nausea are high sero- tonin levels. Yet, in this study there was no correlation between the levels of U-5-HIAA at the time of sampling and nausea. Circulating levels of melato- nin also correlated with the symptom of flush. Other active substances, such as tachykinins, are known to be involved in flush (112). Because it has been shown to have vasodilatory effects by binding to the MT2 receptor in arterial smooth muscle, melatonin may also influence circulatory changes (164). Melatonin is often used in psychiatric care, to treat depression and sleep disturbances. The hypothesis that high circulating levels of melatonin could have an influence on mental health and sleep in these patients was also test- ed. The use of medication for depression, anxiety or sleep disturbances was explored in patient medical records, but no associations were identified. Fur- thermore, the study design did not allow for reliable evaluation of more complex psychiatric symptoms and information concerning mental health was not routinely requested or recorded.

GI symptoms and psychiatric comorbidity In paper III we investigated self-reported GI symptoms in young adult pa- tients seeking psychiatric care, predominantly for mood and anxiety disor- ders. Patients more commonly reported GI symptoms than healthy controls and higher GI symptom burden correlated with depressive symptom severi- ty, which is in accord with previous studies (129, 165-167). Patients with IBS often have psychiatric co-morbidity and implications of disturbed signalling pathways between the brain and the gut, which largely affects the HPA axis, have been proposed as a common pathogenesis. Pa-

40 tients with IBS appear to have an exaggerated stress response compared with controls, with both elevated levels of basal cortisol and stress-induced corti- sol levels (168). Early life stress, as modelled by maternal separation in ro- dents shows long-term alterations in gut functions, including a compromised intestinal mucosal barrier and bacterial translocation (169). Activation of the HPA axis also results in increased CRH production, which is implicated in stress-induced gut dysfunction. Surprisingly, there was no difference in GI symptom burden between pa- tients receiving psychotropic medication and those without. Both SSRI and other have been proposed to treat IBS. A recent review found that treatment with antidepressants, particularly tricyclic antidepres- sants, improved IBS symptoms, especially in patients with diarrhoea- predominant IBS (170). However, SSRI and other antidepressants have well- known GI side effects. Thus any positive effects may have been cancelled out in analyses of the total patient group. In a PCA and cluster analysis six cluster groups were identified demon- strating a heterogeneity in both psychiatric and GI symptoms. One of the cluster groups did not appear to have high GI symptoms, whereas all the others scored high on either symptoms of fast or slow bowel or disturbed appetite.

Daytime melatonin and GI symptoms In paper IV a link between daytime levels of salivary melatonin and GI symptoms was identified. Melatonin levels after lunch correlated with higher GSRS-IBS scores and more specifically to the symptoms of bloating and pain. A possible explanation for this link may be through the motility regu- lating actions of melatonin that can be both stimulatory and inhibitory de- pending on the concentration of melatonin (81). Daytime levels of melatonin are believed to be of peripheral origin, and because the gut constitutes a ma- jor source, circulating levels likely reflect the levels of the GI tract. The reli- ability of salivary melatonin as a reflection of plasma levels has previously been confirmed (171). An increased number of EC cells have been reported in patients with IBS compared with controls (172). This increased EC cell level could theoretically result in higher levels of melatonin during the day. Earlier studies have reported that melatonin levels in the GI tract are related to fasting and food intake, with both causing an elevation in melatonin levels (77, 78). However, studies in humans and in different animal species have reported ambiguous results, which may be a consequence of differences in feeding behaviour, the extent of food restriction and measuring methods. The correlation between melatonin and pain was somewhat surprising seeing that previous studies have indicated a beneficial effect of melatonin administration and abdominal pain, rectal pain sensitivity, and extra colonic

41 symptoms in patients with IBS (126, 173). In these studies, however, mela- tonin administration was at night, when food processing is less active. Because both depression and functional GI disorders (e.g., IBS) are relat- ed to increased inflammation, melatonin elevation may be secondary to im- mune activation and low-grade inflammation in this cohort.

Methodological considerations and limitations The work included in this thesis has several limitations. In paper I and II most of the results are based on immunohistochemical analyses A recog- nised limitation in immunohistochemical methods is the risk of cross- reactivity in which the antibody, instead of binding to the intended antigen, binds to something similar. Melatonin is a small molecule, similar to seroto- nin, and therefore cross-reactivity is challenging. To ensure reliability of the method we performed antibody neutralisation tests in which the antibody for melatonin was preincubated with both melatonin and serotonin. The anti- body did show partial IR for serotonin, indicating cross-reactivity. To further minimise this limitation, co-localisation studies were performed. Only partial overlap in melatonin and serotonin IR was seen, indicating that the antibody indeed binds to melatonin. In addition, there was no correlation between IR for serotonin and melatonin in serial sections, denoting separate antibody targets. Paper I aimed to characterise the expression of melatonin and melatonin receptors in normal human GI tract. However, the biopsies from normal tissue may have been influenced by local immunological processes. This possibility may have arisen because tissue specimens were obtained from patients undergoing surgery for different types of cancer. Given melatonin’s important functions in both cancer and modulation of inflammation, this might constitute an important confounding factor. Pancreatic tissue was only available from three individuals and the expression showed substantial varia- tion. The small sample size prevents any firm conclusions on receptor ex- pression. In paper II correlation analysis between melatonin IR or plasma levels of melatonin and clinical parameters (e.g., symptoms) were performed. The study design, however, was not optimal for this analysis because information about symptoms was extracted from medical records and not standardised in rating scales. The validity of this information is therefore questionable and should be considered exploratory. The GSRS-IBS in paper III and IV was used to evaluate GI symptoms in psychiatric patients, which is somewhat outside the intended scope The GSRS-IBS is originally designed to identify the most bothersome symptoms and evaluate response to treatment in patients with IBS. Still, in these studies the scale was chosen considering that it covers a variety of symptoms and

42 the possibility to classify GI symptoms into symptom clusters in patients not primarily seeking care for GI symptoms, but with an anticipated high preva- lence of GI symptoms. In paper II and IV melatonin levels, local in tissue, plasma and saliva were analysed. From these results, the origin of the melatonin measured is not possible to determine and several alternative or contributing sources of melatonin have to be considered. As an illustration, both plants and animals contain melatonin and intake of melatonin- or tryptophan-rich foods could contribute to higher levels (174). In paper II plasma samples were collected after overnight fasting, which minimises the interaction of dietary melatonin. In paper IV this interaction is probably higher, and a limitation is the lack of information for the term “lunch”. Patients collected saliva after lunch, but no information of what they ate or how much was available. The cross-sectional design (paper II and IV) used in these studies restricts in establishing cause- effect relationships between melatonin and GI symptoms.

Clinical implications and future perspectives Communication between the gut and brain is a rapidly growing field of re- search. It is a complex system that is difficult to grasp and possibly even more difficult to study. Many of the studies on melatonin in the GI tract have been conducted in animals. While animal models are necessary for practical reasons, to be able to study aetiological factors and mechanisms, the direct translation of animal results to humans cannot be assumed i.e. animal trial results may not always translate to humans. Of special importance in this context are differences in circadian rhythm, diet, feeding behaviour and microbiota composition (175, 176). Paper I in this thesis constitutes basic research, but is an essential piece of the puzzle. Indeed, some variations in melatonin receptor expression from previous animal studies were found (79). Further exploration of the melato- nin system in other clinical patient groups may add nuances to our under- standing of its regulation and function. In paper II, melatonin was identified in SI-NETs, which are known to produce serotonin. Based on previous research on EC cells, it is likely that the tumour cells also produce melatonin (72, 73). These tumours could there- fore be used as a model to study the effects of increased local melatonin production and accumulation. Moreover, contradictory results have been demonstrated on the role of melatonin in regulation of apoptosis in different studies. In various types of cancer, melatonin can induce apoptosis, whereas in normal tissues (such as neuronal mitochondria and umbilical endothelium) melatonin reduces apoptosis (105, 108, 177, 178). In this respect it is of in- terest that small intestinal neuroendocrine tumours generally display very low levels of apoptosis (179). In addition, a lower intensity of MT2 receptor

43 expression was seen in metastases compared with primary tumours, which may also account for changes related to malignant progression and impaired melatonin signalling. This indicates that the melatonin system may be a pos- sible treatment target worth further investigation. In paper I and II membrane-bound receptors were studied. Nuclear recep- tors of the ROR/RZR family may play a role in mediating the anti- proliferative effects of melatonin (180). Their function and impact in healthy tissue and melatonin-containing tumours are yet to be investigated. Behaviour and mood are influenced by numerous factors. It is central to identify subgroups of patients that may benefit from more targeted treatment, and for this purpose, novel biomarkers are needed. One of the challenges when studying IBS and mood disorders is that both are wide concepts with large differences within the patient groups. Therefore it is essential to identi- fy the underlying causes of both disorders and to find subgroups that are more alike. In paper III cluster analysis revealed different cluster groups characterised by varying levels of psychiatric and GI symptoms. The link between depression, anxiety and somatic symptoms requires fur- ther investigation and if any conclusions are to be stated regarding causality, long-term observation time and prospective studies are needed. In addition, repeated measurements of possible biomarkers (in this case melatonin) fol- lowing treatment or intervention are desirable. Considering the pronounced anti-inflammatory actions of melatonin, measurements of inflammation with respect to melatonin levels and GI symptoms may further identify subgroups of patients with possibly different underlying pathology. Some of the common pathogenic mechanisms of functional GI disorders and depression include inflammation and changes in microbiota composi- tion. Some bacteria can produce melatonin but also other factors that may have secondary effects on melatonin pathways (181). The administration of probiotics has shown hopeful results in improving not only symptoms of IBS but also in reducing depressive symptoms (182). These results, however, are still in the very early stages and remote from stable evidence-based treat- ments. Mutations in genes affecting melatonin synthesis and signalling have been linked to autism spectrum disorders (183, 184). In future studies vulnerabil- ity factors, such as genetic variants affecting melatonin synthesis, gut dys- function and metabolic alterations, should be studied in relation to clinical symptom subgroups that extend beyond autism. Possible future biomarkers need to be evaluated concerning not only gut function and GI symptoms but also psychiatric symptoms in order to develop strategies for screening and treatment.

44 Ethics

The projects included in this thesis all obtained ethical approval by the Re- gional Ethics Committee in Uppsala (Dnr 2007/143/1 with decisions 2007- 06-13 and 2012-09-14, Dnr 2012/81, Dnr 2012/81/1, Dnr 2013/219). All study patients signed a written informed consent. For study I, written in- formed consent was obtained for material collection, but not for this specific study on normal tissue in that the data were analysed anonymously and sam- ples cannot be traced back to individuals. The Regional Ethics committee waived the need for consent in this case in accordance with Swedish law.

45 Conclusions

Melatonin and its receptors are widely expressed in the human gut and pan- creas: in EC cells, epithelial cells, in immune cells, vascular structures and muscular cells. This widespread expression is consistent with the multiple proposed roles of gut melatonin, including regulation of motility, gut perme- ability, immunomodulation and influence of glucose homeostasis and meta- bolic control. Neuroendocrine tumours derived from EC cells display positive melato- nin IR and higher melatonin IR intensity is associated with lower prolifera- tion index, supporting the previous findings of melatonin as an oncostatic agent. Melatonin IR intensity and circulating levels of melatonin in patients with SI-NETs are associated with GI symptoms and treatment response. Young adult patients seeking psychiatric care report more GI symptoms than controls, regardless of ongoing psychotropic medication. Moreover, GI symptom burden is associated with depressive symptom severity and anxiety traits. In the same cohort salivary melatonin levels after lunch are associated with increased GI symptoms, especially bloating and pain. Taken together, the present results confirm a link between endogenous melatonin regulation and IBS symptoms in patients with psychiatric disor- ders.

46 Sammanfattning på svenska

Bakgrund Melatonin är ett hormon som produceras i tallkottkörteln och som är in- volverat i reglering av sömn och dygnsrytm. Melatonin finns också i många perifera organ och vävnader och har betydligt fler funktioner utöver sömnreglering. Förekomsten av melatonin och melatoninreceptorer har framför allt studerats hos djur och mag-tarmkanalen har föreslagits vara den största källan till perifert producerat melatonin och mer specifikt enterokro- maffincellerna, som också producerar serotonin. När enterokromaffinceller blir maligna, bildas hormonproducerande tumörer som visserligen är ovan- liga, men som ger stora bekymmer med symptom i form av diarré på grund av kraftig frisättning av serotonin. En av de viktigaste egenskaperna hos melatonin är dess antiinflammato- riska effekt och förmågan att binda fria syreradikaler, något som har varit avgörande evolutionärt, till exempel för energiproduktion i mitokondrier, där stora mängder fria radikaler bildas. Melatonin är också viktig för reglering av glukosmetabolismen och har visat sig ha positiva effekter vid olika typer av cancer. I mag-tarmkanalen reglerar melatonin motorik och sekretion, samt skyddar tarmslemhinnan, som utgör en viktig barriär för att motverka uppkomsten av låggradig inflammation. Olika processer i mage och tarm kan genom signalering till hjärnan ge förändringar i psykiskt mående och beteende, något som på engelska brukar kallas ”gut-brain axis” och avser kommunikationen mellan tarmen och hjärnan. Kommunikationen består av neurologiska, endokrina och immunol- ogiska signaler. Uppkomsten av funktionella tarmsjukdomar som irritabel tarm eller IBS tros åtminstone delvis bero på störningar i detta kommu- nikationssystem och patienter med IBS lider oftare av depression och ångest än kontroller.

Syfte Syftet med de olika delarbetena i den här avhandlingen har varit följande:

I. Att undersöka uttrycket av melatonin och melatoninreceptorer i normal tarm och bukspottkörtel från människa.

47 II. Att undersöka uttrycket av melatonin och melatoninreceptorer i tumörvävnad från tunntarmskarcinoider, hormonproducerande tumörer, som utgår från enterocromaffinceller, samt att studera sambandet mellan melatoninnivåer, mag-tarmbesvär och prog- nostiska faktorer. III. Att undersöka förekomsten av olika typer av mag-tarmbesvär hos unga patienter med psykiatriska sjukdomar, framför allt de- pression och ångest jämfört med kontroller och vidare att un- dersöka om det är någon skillnad mellan patienter som har psyk- iatrisk medicinering och de som inte har det. IV. Att undersöka sambandet mellan melatoninnivåer i saliv på dag- tid och besvär från mag-tarmkanalen.

Metod I studie I och II, användes immunohistokemi för att identifiera och påvisa uttryck av melatonin och melatoninreceptorer i vävnadsbiopsier från män- niska. Biopsierna i studie I utgjordes av normal tarm och bukspottkörtel från patienter som opererats av annan anledning. I studie II analyserades biopsier från tunntarmskarcinoidtumörer. För tumörpatienter mättes även nivåer av melatonin i plasma med en metod som kallas RIA vid två tillfällen. Melato- ninnivåer jämfördes med kliniska parametrar så som besvär från mag- tarmkanalen och sjukdomsutfall. I studie III och IV undersöktes patienter, 18-25 år som sökte vård på psykiatrimottagningen för unga vuxna, avseende förekomsten av mag- tarmbesvär, melatoninnivåer i saliv, depressiva symptom och ångestbenä- genhet. Melatonin i saliv mättes med ELISA och patienterna fyllde i skatt- ningsskalor för mag-tarmbesvär, depressiva symptom och olika person- lighetsdrag som ångestbenägenhet.

Resultat och slutsats I. Uttryck av melatonin och melatoninreceptorer identifierades i stora delar av mag-tarmkanalen och i flera olika typer av celler, bland annat enterokromaffinceller, epitelceller, immunceller, muskelceller och celler som omger blodkärl. Uttryck av melato- nin och dess receptorer påvisades också i bukspottkörteln, med varierande intensitet. II. Uttryck av melatonin och åtminstone en av receptorerna för melatonin identifierades i tumörvävnad från hormonproduc- erande tunntarmskarcinoider. Det fanns ett samband mellan högre intensitet av melatonin i tumörvävnad och lägre prolifera-

48 tion. Det fanns också samband mellan högre nivåer av melatonin i plasma och mindre diarrésymptom. Melatoninnivåer var lägre efter behandling hos patienter med stabil eller minskad tumörbörda. III. Unga patienter med psykiatriska sjukdomar, framför allt depres- sion och ångest hade mer mag-tarmbesvär än friska kontroller, oavsett om de hade pågående behandling med psykiatriska läkemedel eller inte.

Sammantaget visar dessa studier att melatonin och dess receptorer finns i stora delar av tarmen hos människa och vi har, för första gången, visat att melatonin också finns i tunntarmskarcinoider. Det finns sannolikt en kop- pling mellan melatonin producerat i tarmen och symptom från mag- tarmkanalen, även om ytterligare studier behövs för att undersöka ver- kningsmekanismerna bakom detta samband. Patienter med depression och ångest har en ökad förekomst av mag-tarmbesvär jämfört med kontroller och detta bör efterfrågas vid behandling och uppföljning av psykiatriska sjuk- domar. Ökad kunskap om kommunikationen mellan mage och tarm är nödvändig för att bättre förstå och kunna styra behandlingen av såväl depres- sion och ångest som IBS. Den här avhandlingen är en pusselbit i arbetet att försöka hitta biologiska markörer som kan användas för att skilja mellan olika subgrupper inom den heterogena gruppen unga med depression och ångest.

49 Acknowledgements

This work was carried out at the Department of Neuroscience, Psychiatry, Uppsala University. I would like to express my deepest gratitude to everyone involved in this thesis project.

Janet Cunningham, my brilliant, energetic, inspiring supervisor. Thank you for believing in me and understanding that life is complex and multifaceted. Without you, I would never have taken on this challenge. Thank you for always looking after (all of) me.

Annica Rasmusson, my co-supervisor and general fixer. Your ability to iden- tify the gaps and find the solutions to bridge them has been instrumental for this work. Thank you for your practical compassion.

Lisa Ekselius, originally my associate supervisor, for making this work pos- sible and supporting my supervisor so she could support me.

Mikaela Syk, thank you for your brilliant mind, eternal patience and accura- cy.

Isak Sundberg, for outstanding collaboration and picking up the slack when sleep was non-existing.

Mia Ramklint, for sound scientific discussions and understanding of psy- chometric data.

Per Hellström, for bringing your expertise in gastroenterology in general and IBS in particular.

David Just, for exceptional and immediate help with statistics and visualisa- tions.

Zorana Kurbalija Novicic, for patiently teaching me about PCA and cluster analysis and for great scientific discussions.

Hans Arinell, for your patience and sound scepticism and always willing to answer my many statistical questions.

50 A-C Fält, our university administrator, who knows everything and everyone. Without you, this PhD process would be an administrative nightmare.

Åsa Forsberg, laboratory technician for excellent technical assistance with immunohistochemical staining.

Leslie Shaps, for brilliant and thorough proofreading of the thesis.

Thank you to all my co-authors that include Eva Tiensuu-Janson, Mats Stridsberg, Abir Ali and Rebecka Widerström, for all your hard work, wise comments and excellent collaboration.

Emelie Ahnfelt, for pep talks throughout the process of this thesis, both on scientific and personal matters.

Thanks to all my colleagues in my clinical everyday life at the department of Gynaecology and Obstetrics, for teaching, inspiring and supporting me in my clinical work. You truly do an amazing work!

Tahmineh Badiei, for your compassion, distraction and support.

My Dala family, for very little research but a lot of everything else!

The entire Brink family and especially Maria and Hans for all the love, help and support with our children. Without you, we would never manage.

My dearest “klicken”, Hanna Minna, Amanda and Agnes, “klacken” and all of your lovely children for a lifetime of friendship and support, completely unrelated to research.

My family, my mother Carina and my father Lennart for unconditional love, support and inspiration. Bibi and my brothers John and Linus with families. Thank you all for being a part of my life.

Most of all, thank you John, my friend and partner in life and love for every- thing that you do and for being the brilliant, fun, anxiety-free person that you are. Also thank you for our wonderful children, the other two overwhelming projects during this highly full-packed period in our lives. Olle and Mikkel, thank you for all the love and distraction.

51 References

1. Werner S. Endokrinologi. 3 ed: Liber; 2015. 2. Alberts BJ, A; Lewis, J; Morgan, D; Raff, M; Roberts, K; Walter, P. Molecular biology of the Cell. sixth ed. New York: Garland Science; 2015. 3. Erlandsson-Albertsson CG, U Cellbiologi: Studentlitteratur; 2007. 4. Biessels GJ, Reagan LP. Hippocampal insulin resistance and cognitive dysfunction. Nature reviews Neuroscience. 2015;16(11):660-71. 5. Lerner AB, Case JD, Takahashi Y, Lee TH, Mori W. Isolation of Melatonin, the Pineal Gland Factor That Lightens Melanocytes. J Am Chem Soc. 1958;80(10):2587-. 6. Huether G. The contribution of extrapineal sites of melatonin synthesis to circulating melatonin levels in higher vertebrates. Experientia. 1993;49(8):665-70. 7. Claustrat B, Brun J, Chazot G. The basic and pathophysiology of melatonin. Sleep medicine reviews. 2005;9(1):11-24. 8. Söderquist F. Melatonin and its receptors in the normal human gastrointestinal tract, pancreas and in small intestinal neuroendocrine tumours. 2017. 9. Acuna-Castroviejo D, Escames G, Venegas C, Diaz-Casado ME, Lima- Cabello E, Lopez LC, et al. Extrapineal melatonin: sources, regulation, and potential functions. Cellular and molecular life sciences : CMLS. 2014;71(16):2997-3025. 10. Carrillo-Vico A, Guerrero JM, Lardone PJ, Reiter RJ. A review of the multiple actions of melatonin on the immune system. Endocrine. 2005;27(2):189-200. 11. Tan DX, Manchester LC, Liu X, Rosales-Corral SA, Acuna-Castroviejo D, Reiter RJ. Mitochondria and chloroplasts as the original sites of melatonin synthesis: a hypothesis related to melatonin's primary function and evolution in eukaryotes. J Pineal Res. 2013;54(2):127-38. 12. Anderson G, Vaillancourt C, Maes M, Reiter RJ. and the gut- brain axis: is there a role for melatonin? Biomolecular concepts. 2017;8(3- 4):185-95. 13. Zhou JN, Liu RY, van Heerikhuize J, Hofman MA, Swaab DF. Alterations in the circadian rhythm of salivary melatonin begin during middle-age. J Pineal Res. 2003;34(1):11-6. 14. Andersen LP, Werner MU, Rosenkilde MM, Harpsoe NG, Fuglsang H, Rosenberg J, et al. Pharmacokinetics of oral and intravenous melatonin in healthy volunteers. BMC pharmacology & toxicology. 2016;17:8. 15. Markantonis SL, Tsakalozou E, Paraskeva A, Staikou C, Fassoulaki A. Melatonin pharmacokinetics in premenopausal and postmenopausal healthy female volunteers. J Clin Pharmacol. 2008;48(2):240-5. 16. Ma X, Idle JR, Krausz KW, Gonzalez FJ. Metabolism of melatonin by human cytochromes p450. Drug Metab Dispos. 2005;33(4):489-94.

52 17. Arendt J. Melatonin. Clin Endocrinol (Oxf). 1988;29(2):205-29. 18. Masana MI, Dubocovich ML. Melatonin receptor signaling: finding the path through the dark. Sci STKE. 2001;2001(107):pe39. 19. Pandi-Perumal SR, Trakht I, Srinivasan V, Spence DW, Maestroni GJ, Zisapel N, et al. Physiological effects of melatonin: role of melatonin receptors and signal transduction pathways. Prog Neurobiol. 2008;85(3):335- 53. 20. Boutin JA, Ferry G. Is There Sufficient Evidence that the Melatonin Binding Site MT3 Is Quinone Reductase 2? J Pharmacol Exp Ther. 2019;368(1):59- 65. 21. Kasimay O, Cakir B, Devseren E, Yegen BC. Exogenous melatonin delays gastric emptying rate in rats: role of CCK2 and 5-HT3 receptors. J Physiol Pharmacol. 2005;56(4):543-53. 22. Esteban-Zubero E, Lopez-Pingarron L, Alatorre-Jimenez MA, Ochoa-Moneo P, Buisac-Ramon C, Rivas-Jimenez M, et al. Melatonin's role as a co- adjuvant treatment in colonic diseases: A review. Life Sci. 2017;170:72-81. 23. Benitez-King G, Anton-Tay F. Calmodulin mediates melatonin cytoskeletal effects. Experientia. 1993;49(8):635-41. 24. Macias M, Escames G, Leon J, Coto A, Sbihi Y, Osuna A, et al. Calreticulin- melatonin. An unexpected relationship. European journal of biochemistry. 2003;270(5):832-40. 25. Melendez J, Maldonado V, Ortega A. Effect of melatonin on beta-tubulin and MAP2 expression in NIE-115 cells. Neurochemical research. 1996;21(6):653-8. 26. Smirnov AN. Nuclear melatonin receptors. Biochemistry (Mosc). 2001;66(1):19-26. 27. Wiesenberg I, Missbach M, Carlberg C. The potential role of the transcription factor RZR/ROR as a mediator of nuclear melatonin signaling. Restorative neurology and neuroscience. 1998;12(2-3):143-50. 28. Pacchierotti C, Iapichino S, Bossini L, Pieraccini F, Castrogiovanni P. Melatonin in psychiatric disorders: a review on the melatonin involvement in psychiatry. Frontiers in neuroendocrinology. 2001;22(1):18-32. 29. Wu YH, Swaab DF. The human pineal gland and melatonin in aging and Alzheimer's disease. J Pineal Res. 2005;38(3):145-52. 30. Videnovic A, Noble C, Reid KJ, Peng J, Turek FW, Marconi A, et al. Circadian melatonin rhythm and excessive daytime sleepiness in Parkinson disease. JAMA neurology. 2014;71(4):463-9. 31. Sundberg I, Ramklint M, Stridsberg M, Papadopoulos FC, Ekselius L, Cunningham JL. Salivary Melatonin in Relation to Depressive Symptom Severity in Young Adults. PLoS One. 2016;11(4):e0152814. 32. Brites D, Fernandes A. Neuroinflammation and Depression: Microglia Activation, Extracellular Microvesicles and microRNA Dysregulation. Frontiers in cellular neuroscience. 2015;9:476. 33. Waller KL, Mortensen EL, Avlund K, Fagerlund B, Lauritzen M, Gammeltoft S, et al. Melatonin and cortisol profiles in late midlife and their association with age-related changes in cognition. Nature and science of sleep. 2016;8:47-53. 34. Naismith SL, Hermens DF, Ip TK, Bolitho S, Scott E, Rogers NL, et al. Circadian profiles in young people during the early stages of affective disorder. Translational psychiatry. 2012;2:e123.

53 35. Liu J, Clough SJ, Dubocovich ML. Role of the MT1 and MT2 melatonin receptors in mediating depressive- and anxiety-like behaviors in C3H/HeN mice. Genes, brain, and behavior. 2017;16(5):546-53. 36. Fukudo S. Role of corticotropin-releasing hormone in irritable bowel syndrome and intestinal inflammation. Journal of gastroenterology. 2007;42 Suppl 17:48-51. 37. Martinez V, Wang L, Rivier J, Grigoriadis D, Tache Y. Central CRF, urocortins and stress increase colonic transit via CRF1 receptors while activation of CRF2 receptors delays gastric transit in mice. The Journal of physiology. 2004;556(Pt 1):221-34. 38. Nakade Y, Fukuda H, Iwa M, Tsukamoto K, Yanagi H, Yamamura T, et al. Restraint stress stimulates colonic motility via central corticotropin-releasing factor and peripheral 5-HT3 receptors in conscious rats. Am J Physiol Gastrointest Liver Physiol. 2007;292(4):G1037-44. 39. Tache Y, Million M. Role of Corticotropin-releasing Factor Signaling in Stress-related Alterations of Colonic Motility and Hyperalgesia. Journal of neurogastroenterology and motility. 2015;21(1):8-24. 40. Gunawardene AR, Corfe BM, Staton CA. Classification and functions of enteroendocrine cells of the lower gastrointestinal tract. International journal of experimental pathology. 2011;92(4):219-31. 41. Reigstad CS, Salmonson CE, Rainey JF, 3rd, Szurszewski JH, Linden DR, Sonnenburg JL, et al. Gut microbes promote colonic serotonin production through an effect of short-chain fatty acids on enterochromaffin cells. FASEB J. 2015;29(4):1395-403. 42. Galland L. The gut microbiome and the brain. Journal of medicinal food. 2014;17(12):1261-72. 43. Muller PA, Koscso B, Rajani GM, Stevanovic K, Berres ML, Hashimoto D, et al. Crosstalk between Muscularis Macrophages and Enteric Neurons Regulates Gastrointestinal Motility. Cell. 2014;158(5):1210. 44. Slominski RM, Reiter RJ, Schlabritz-Loutsevitch N, Ostrom RS, Slominski AT. Melatonin membrane receptors in peripheral tissues: distribution and functions. Mol Cell Endocrinol. 2012;351(2):152-66. 45. Dubocovich ML, Markowska M. Functional MT1 and MT2 melatonin receptors in mammals. Endocrine. 2005;27(2):101-10. 46. Ekmekcioglu C. Melatonin receptors in humans: biological role and clinical relevance. Biomed Pharmacother. 2006;60(3):97-108. 47. Klosen P, Lapmanee S, Schuster C, Guardiola B, Hicks D, Pevet P, et al. MT1 and MT2 melatonin receptors are expressed in nonoverlapping neuronal populations. J Pineal Res. 2019;67(1):e12575. 48. Weaver DR, Stehle JH, Stopa EG, Reppert SM. Melatonin receptors in human hypothalamus and pituitary: implications for circadian and reproductive responses to melatonin. J Clin Endocrinol Metab. 1993;76(2):295-301. 49. Reppert SM, Godson C, Mahle CD, Weaver DR, Slaugenhaupt SA, Gusella JF. Molecular characterization of a second melatonin receptor expressed in human retina and brain: the Mel1b melatonin receptor. Proc Natl Acad Sci U S A. 1995;92(19):8734-8. 50. Mazzucchelli C, Pannacci M, Nonno R, Lucini V, Fraschini F, Stankov BM. The melatonin receptor in the human brain: cloning experiments and distribution studies. Brain research Molecular brain research. 1996;39(1- 2):117-26.

54 51. Pozo D, Garcia-Maurino S, Guerrero JM, Calvo JR. mRNA expression of nuclear receptor RZR/RORalpha, melatonin membrane receptor MT, and hydroxindole-O-methyltransferase in different populations of human immune cells. J Pineal Res. 2004;37(1):48-54. 52. Poon AM, Chow PH, Mak AS, Pang SF. Autoradiographic localization of 2[125I]iodomelatonin binding sites in the gastrointestinal tract of mammals including humans and birds. J Pineal Res. 1997;23(1):5-14. 53. Nemeth C, Humpeler S, Kallay E, Mesteri I, Svoboda M, Rogelsperger O, et al. Decreased expression of the melatonin receptor 1 in human colorectal adenocarcinomas. J Biol Regul Homeost Agents. 2011;25(4):531-42. 54. Weaver DR, Reppert SM. The Mel1a melatonin receptor gene is expressed in human suprachiasmatic nuclei. Neuroreport. 1996;8(1):109-12. 55. Dillon DC, Easley SE, Asch BB, Cheney RT, Brydon L, Jockers R, et al. Differential expression of high-affinity melatonin receptors (MT1) in normal and malignant human breast tissue. Am J Clin Pathol. 2002;118(3):451-8. 56. Scher J, Wankiewicz E, Brown GM, Fujieda H. MT(1) melatonin receptor in the human retina: expression and localization. Invest Ophthalmol Vis Sci. 2002;43(3):889-97. 57. Isola M, Ekstrom J, Diana M, Solinas P, Cossu M, Lilliu MA, et al. Subcellular distribution of melatonin receptors in human parotid glands. Journal of anatomy. 2013;223(5):519-24. 58. Ekmekcioglu C, Haslmayer P, Philipp C, Mehrabi MR, Glogar HD, Grimm M, et al. Expression of the MT1 melatonin receptor subtype in human coronary arteries. J Recept Signal Transduct Res. 2001;21(1):85-91. 59. Ekmekcioglu C, Thalhammer T, Humpeler S, Mehrabi MR, Glogar HD, Holzenbein T, et al. The melatonin receptor subtype MT2 is present in the human cardiovascular system. J Pineal Res. 2003;35(1):40-4. 60. Lardone PJ, Carrillo-Vico A, Naranjo MC, De Felipe B, Vallejo A, Karasek M, et al. Melatonin synthesized by Jurkat human leukemic line is implicated in IL-2 production. J Cell Physiol. 2006;206(1):273-9. 61. Brydon L, Petit L, Delagrange P, Strosberg AD, Jockers R. Functional expression of MT2 (Mel1b) melatonin receptors in human PAZ6 adipocytes. Endocrinology. 2001;142(10):4264-71. 62. Ramracheya RD, Muller DS, Squires PE, Brereton H, Sugden D, Huang GC, et al. Function and expression of melatonin receptors on human pancreatic islets. J Pineal Res. 2008;44(3):273-9. 63. Lyssenko V, Nagorny CL, Erdos MR, Wierup N, Jonsson A, Spegel P, et al. Common variant in MTNR1B associated with increased risk of type 2 diabetes and impaired early insulin secretion. Nat Genet. 2009;41(1):82-8. 64. Aust S, Thalhammer T, Humpeler S, Jager W, Klimpfinger M, Tucek G, et al. The melatonin receptor subtype MT1 is expressed in human gallbladder epithelia. J Pineal Res. 2004;36(1):43-8. 65. Drew JE, Williams LM, Hannah LT, Barrett P, Abramovich DR. Melatonin receptors in the human fetal kidney: 2-[125I]iodomelatonin binding sites correlated with expression of Mel1a and Mel1b receptor genes. J Endocrinol. 1998;156(2):261-7. 66. Niles LP, Wang J, Shen L, Lobb DK, Younglai EV. Melatonin receptor mRNA expression in human granulosa cells. Mol Cell Endocrinol. 1999;156(1-2):107-10. 67. Schlabritz-Loutsevitch N, Hellner N, Middendorf R, Muller D, Olcese J. The human myometrium as a target for melatonin. J Clin Endocrinol Metab. 2003;88(2):908-13.

55 68. Lanoix D, Beghdadi H, Lafond J, Vaillancourt C. Human placental trophoblasts synthesize melatonin and express its receptors. J Pineal Res. 2008;45(1):50-60. 69. Tam CW, Mo CW, Yao KM, Shiu SY. Signaling mechanisms of melatonin in antiproliferation of hormone-refractory 22Rv1 human prostate cancer cells: implications for prostate cancer chemoprevention. J Pineal Res. 2007;42(2):191-202. 70. Slominski A, Pisarchik A, Zbytek B, Tobin DJ, Kauser S, Wortsman J. Functional activity of serotoninergic and melatoninergic systems expressed in the skin. J Cell Physiol. 2003;196(1):144-53. 71. Raikhlin NT, Kvetnoy IM, Tolkachev VN. Melatonin may be synthesised in enterochromaffin cells. Nature. 1975;255(5506):344-5. 72. Raikhlin NT, Kvetnoy, I. M, Kadagidze, Z. G., Sokolov, A. V. Immunomorphological studies on synthesis of melaotnin in enterochromaffine cells. Acta Histochem Cytochem. 1978;11(1):75-7. 73. Bubenik GA, Brown GM, Grota LJ. Immunohistological localization of melatonin in the rat digestive system. Experientia. 1977;33(5):662-3. 74. Ahlman H, Nilsson. The gut as the largest endocrine organ in the body. Ann Oncol. 2001;12 Suppl 2:S63-8. 75. Huether G, Poeggeler B, Reimer A, George A. Effect of tryptophan administration on circulating melatonin levels in chicks and rats: evidence for stimulation of melatonin synthesis and release in the gastrointestinal tract. Life Sci. 1992;51(12):945-53. 76. Bubenik GA, Brown GM. Pinealectomy reduces melatonin levels in the serum but not in the gastrointestinal tract of rats. Biological signals. 1997;6(1):40-4. 77. Huether G. Melatonin synthesis in the gastrointestinal tract and the impact of nutritional factors on circulating melatonin. Ann N Y Acad Sci. 1994;719:146-58. 78. Bubenik GA. Gastrointestinal melatonin: localization, function, and clinical relevance. Dig Dis Sci. 2002;47(10):2336-48. 79. Sotak M, Mrnka L, Pacha J. Heterogeneous expression of melatonin receptor MT1 mRNA in the rat intestine under control and fasting conditions. J Pineal Res. 2006;41(2):183-8. 80. Sjoblom M, Safsten B, Flemstrom G. Melatonin-induced calcium signaling in clusters of human and rat duodenal enterocytes. Am J Physiol Gastrointest Liver Physiol. 2003;284(6):G1034-44. 81. Thor PJ, Krolczyk G, Gil K, Zurowski D, Nowak L. Melatonin and serotonin effects on gastrointestinal motility. J Physiol Pharmacol. 2007;58 Suppl 6:97- 103. 82. Bubenik GA. The effect of serotonin, N-acetylserotonin, and melatonin on spontaneous contractions of isolated rat intestine. J Pineal Res. 1986;3(1):41- 54. 83. Bazwinsky-Wutschke I, Wolgast S, Muhlbauer E, Albrecht E, Peschke E. Phosphorylation of cyclic AMP-response element-binding protein (CREB) is influenced by melatonin treatment in pancreatic rat insulinoma beta-cells (INS-1). J Pineal Res. 2012;53(4):344-57. 84. Galano A, Tan DX, Reiter RJ. Melatonin as a natural ally against oxidative stress: a physicochemical examination. J Pineal Res. 2011;51(1):1-16. 85. Galano A, Tan DX, Reiter RJ. On the free radical scavenging activities of melatonin's metabolites, AFMK and AMK. J Pineal Res. 2013;54(3):245-57.

56 86. Rodriguez C, Mayo JC, Sainz RM, Antolin I, Herrera F, Martin V, et al. Regulation of antioxidant enzymes: a significant role for melatonin. J Pineal Res. 2004;36(1):1-9. 87. Carrillo-Vico A, Calvo JR, Abreu P, Lardone PJ, Garcia-Maurino S, Reiter RJ, et al. Evidence of melatonin synthesis by human lymphocytes and its physiological significance: possible role as intracrine, autocrine, and/or paracrine substance. FASEB J. 2004;18(3):537-9. 88. Carrillo-Vico A, Lardone PJ, Alvarez-Sanchez N, Rodriguez-Rodriguez A, Guerrero JM. Melatonin: buffering the immune system. Int J Mol Sci. 2013;14(4):8638-83. 89. Khan WI, Ghia JE. Gut hormones: emerging role in immune activation and inflammation. Clinical and experimental immunology. 2010;161(1):19-27. 90. Piche T, Barbara G, Aubert P, Bruley des Varannes S, Dainese R, Nano JL, et al. Impaired intestinal barrier integrity in the colon of patients with irritable bowel syndrome: involvement of soluble mediators. Gut. 2009;58(2):196- 201. 91. Zhou Q, Zhang B, Verne GN. Intestinal membrane permeability and hypersensitivity in the irritable bowel syndrome. Pain. 2009;146(1-2):41-6. 92. Hu YJ, Wang YD, Tan FQ, Yang WX. Regulation of paracellular permeability: factors and mechanisms. Molecular biology reports. 2013;40(11):6123-42. 93. Shajib MS, Khan WI. The role of serotonin and its receptors in activation of immune responses and inflammation. Acta physiologica. 2015;213(3):561- 74. 94. Sommansson A, Nylander O, Sjoblom M. Melatonin decreases duodenal epithelial paracellular permeability via a nicotinic receptor-dependent pathway in rats in vivo. J Pineal Res. 2012. 95. Sjoblom M, Flemstrom G. Melatonin in the duodenal lumen is a potent stimulant of mucosal bicarbonate secretion. J Pineal Res. 2003;34(4):288-93. 96. Lansink MO, Patyk V, de Groot H, Effenberger-Neidnicht K. Melatonin reduces changes to small intestinal microvasculature during systemic inflammation. Journal of Surgical Research.211:114-25. 97. Morris G, Carvalho AF, Anderson G, Galecki P, Maes M. The Many Neuroprogressive Actions of Tryptophan Catabolites (TRYCATs) that may be Associated with the Pathophysiology of Neuro-Immune Disorders. Current pharmaceutical design. 2016;22(8):963-77. 98. Maes M, Anderson G. Overlapping the Tryptophan Catabolite (TRYCAT) and Melatoninergic Pathways in Alzheimer's Disease. Current pharmaceutical design. 2016;22(8):1074-85. 99. Brundin L, Sellgren CM, Lim CK, Grit J, Palsson E, Landen M, et al. An enzyme in the kynurenine pathway that governs vulnerability to suicidal behavior by regulating excitotoxicity and neuroinflammation. Translational psychiatry. 2016;6(8):e865. 100. Myint AM. Kynurenines: from the perspective of major psychiatric disorders. The FEBS journal. 2012;279(8):1375-85. 101. Agudelo LZ, Femenia T, Orhan F, Porsmyr-Palmertz M, Goiny M, Martinez- Redondo V, et al. Skeletal muscle PGC-1alpha1 modulates kynurenine metabolism and mediates resilience to stress-induced depression. Cell. 2014;159(1):33-45. 102. Haus EL, Smolensky MH. Shift work and cancer risk: potential mechanistic roles of circadian disruption, light at night, and sleep deprivation. Sleep medicine reviews. 2013;17(4):273-84.

57 103. Shiu SY. Towards rational and evidence-based use of melatonin in prostate cancer prevention and treatment. J Pineal Res. 2007;43(1):1-9. 104. Cos S, Sanchez-Barcelo EJ. Melatonin and mammary pathological growth. Frontiers in neuroendocrinology. 2000;21(2):133-70. 105. Sainz RM, Mayo JC, Rodriguez C, Tan DX, Lopez-Burillo S, Reiter RJ. Melatonin and cell death: differential actions on apoptosis in normal and cancer cells. Cellular and molecular life sciences : CMLS. 2003;60(7):1407- 26. 106. Shiu SY, Leung WY, Tam CW, Liu VW, Yao KM. Melatonin MT(1) receptor-induced transcriptional up-regulation of p27(Kip1) in prostate cancer antiproliferation is mediated via inhibition of constitutively active nuclear factor kappa B (NF-kappaB): potential implications on prostate cancer chemoprevention and therapy. J Pineal Res. 2012. 107. Mediavilla MD, Cos S, Sanchez-Barcelo EJ. Melatonin increases p53 and p21WAF1 expression in MCF-7 human breast cancer cells in vitro. Life Sci. 1999;65(4):415-20. 108. Suofu Y, Li W, Jean-Alphonse FG, Jia J, Khattar NK, Li J, et al. Dual role of mitochondria in producing melatonin and driving GPCR signaling to block cytochrome c release. Proc Natl Acad Sci U S A. 2017;114(38):E7997- E8006. 109. Casado-Zapico S, Martin V, Garcia-Santos G, Rodriguez-Blanco J, Sanchez- Sanchez AM, Luno E, et al. Regulation of the expression of death receptors and their ligands by melatonin in haematological cancer cell lines and in leukaemia cells from patients. J Pineal Res. 2011;50(3):345-55. 110. Landerholm K, Falkmer S, Jarhult J. Epidemiology of small bowel carcinoids in a defined population. World journal of surgery. 2010;34(7):1500-5. 111. Eriksson J, Norlen O, Ogren M, Garmo H, Ihre-Lundgren C, Hellman P. Primary small intestinal neuroendocrine tumors are highly prevalent and often multiple before metastatic disease develops. Scandinavian journal of surgery : SJS : official organ for the Finnish Surgical Society and the Scandinavian Surgical Society. 2019:1457496919874484. 112. Cunningham JL, Janson ET, Agarwal S, Grimelius L, Stridsberg M. Tachykinins in endocrine tumors and the carcinoid syndrome. European journal of endocrinology / European Federation of Endocrine Societies. 2008;159(3):275-82. 113. Oberg K, Akerstrom G, Rindi G, Jelic S, Group EGW. Neuroendocrine gastroenteropancreatic tumours: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2010;21 Suppl 5:v223-7. 114. Norlen O, Stalberg P, Oberg K, Eriksson J, Hedberg J, Hessman O, et al. Long-term results of surgery for small intestinal neuroendocrine tumors at a tertiary referral center. World journal of surgery. 2012;36(6):1419-31. 115. Strosberg J, Kvols L. Antiproliferative effect of somatostatin analogs in gastroenteropancreatic neuroendocrine tumors. World journal of gastroenterology. 2010;16(24):2963-70. 116. Kvols LK, Moertel CG, O'Connell MJ, Schutt AJ, Rubin J, Hahn RG. Treatment of the malignant carcinoid syndrome. Evaluation of a long-acting somatostatin analogue. N Engl J Med. 1986;315(11):663-6. 117. Kolby L, Persson G, Franzen S, Ahren B. Randomized clinical trial of the effect of interferon alpha on survival in patients with disseminated midgut carcinoid tumours. Br J Surg. 2003;90(6):687-93.

58 118. Creed F, Ratcliffe J, Fernandez L, Tomenson B, Palmer S, Rigby C, et al. Health-related quality of life and health care costs in severe, refractory irritable bowel syndrome. Annals of internal medicine. 2001;134(9 Pt 2):860- 8. 119. Lovell RM, Ford AC. Global prevalence of and risk factors for irritable bowel syndrome: a meta-analysis. Clinical gastroenterology and hepatology : the official clinical practice journal of the American Gastroenterological Association. 2012;10(7):712-21 e4. 120. Mearin F, Lacy BE, Chang L, Chey WD, Lembo AJ, Simren M, et al. Bowel Disorders. Gastroenterology. 2016. 121. Mujagic Z, Jonkers D, Ludidi S, Keszthelyi D, Hesselink MA, Weerts Z, et al. Biomarkers for visceral hypersensitivity in patients with irritable bowel syndrome. Neurogastroenterology and motility : the official journal of the European Gastrointestinal Motility Society. 2017;29(12). 122. Vijayvargiya P, Camilleri M, Burton D, Busciglio I, Lueke A, Donato LJ. Bile and fat excretion are biomarkers of clinically significant diarrhoea and constipation in irritable bowel syndrome. Alimentary pharmacology & therapeutics. 2019;49(6):744-58. 123. Ohman L, Tornblom H, Simren M. Crosstalk at the mucosal border: importance of the gut microenvironment in IBS. Nature reviews Gastroenterology & hepatology. 2015;12(1):36-49. 124. Dunlop SP, Coleman NS, Blackshaw E, Perkins AC, Singh G, Marsden CA, et al. Abnormalities of 5-hydroxytryptamine metabolism in irritable bowel syndrome. Clinical gastroenterology and hepatology : the official clinical practice journal of the American Gastroenterological Association. 2005;3(4):349-57. 125. Siah KT, Wong RK, Ho KY. Melatonin for the treatment of irritable bowel syndrome. World journal of gastroenterology. 2014;20(10):2492-8. 126. Song GH, Leng PH, Gwee KA, Moochhala SM, Ho KY. Melatonin improves abdominal pain in irritable bowel syndrome patients who have sleep disturbances: a randomised, double blind, placebo controlled study. Gut. 2005;54(10):1402-7. 127. Levy RL, Olden KW, Naliboff BD, Bradley LA, Francisconi C, Drossman DA, et al. Psychosocial aspects of the functional gastrointestinal disorders. Gastroenterology. 2006;130(5):1447-58. 128. Lee S, Wu J, Ma YL, Tsang A, Guo WJ, Sung J. Irritable bowel syndrome is strongly associated with generalized anxiety disorder: a community study. Alimentary pharmacology & therapeutics. 2009;30(6):643-51. 129. Sibelli A, Chalder T, Everitt H, Workman P, Windgassen S, Moss-Morris R. A systematic review with meta-analysis of the role of anxiety and depression in irritable bowel syndrome onset. Psychological medicine. 2016;46(15):3065-80. 130. Weger M, Sandi C. High anxiety trait: A vulnerable phenotype for stress- induced depression. Neuroscience and biobehavioral reviews. 2018;87:27-37. 131. Kano M, Muratsubaki T, Van Oudenhove L, Morishita J, Yoshizawa M, Kohno K, et al. Altered brain and gut responses to corticotropin-releasing hormone (CRH) in patients with irritable bowel syndrome. Scientific reports. 2017;7(1):12425. 132. Overman EL, Rivier JE, Moeser AJ. CRF induces intestinal epithelial barrier injury via the release of mast cell proteases and TNF-alpha. PLoS One. 2012;7(6):e39935.

59 133. Barrett T, Wilhite SE, Ledoux P, Evangelista C, Kim IF, Tomashevsky M, et al. NCBI GEO: archive for functional genomics data sets--update. Nucleic Acids Res. 2013;41(Database issue):D991-5. 134. Li C, Wong WH. Model-based analysis of oligonucleotide arrays: expression index computation and outlier detection. Proc Natl Acad Sci U S A. 2001;98(1):31-6. 135. Leja J, Essaghir A, Essand M, Wester K, Oberg K, Totterman TH, et al. Novel markers for enterochromaffin cells and gastrointestinal neuroendocrine carcinomas. Modern pathology : an official journal of the United States and Canadian Academy of Pathology, Inc. 2009;22(2):261-72. 136. Pei H, Li L, Fridley BL, Jenkins GD, Kalari KR, Lingle W, et al. FKBP51 affects cancer cell response to chemotherapy by negatively regulating Akt. Cancer cell. 2009;16(3):259-66. 137. Badea L, Herlea V, Dima SO, Dumitrascu T, Popescu I. Combined gene expression analysis of whole-tissue and microdissected pancreatic ductal adenocarcinoma identifies genes specifically overexpressed in tumor epithelia. Hepato-gastroenterology. 2008;55(88):2016-27. 138. Taneera J, Lang S, Sharma A, Fadista J, Zhou Y, Ahlqvist E, et al. A systems genetics approach identifies genes and pathways for type 2 diabetes in human islets. Cell metabolism. 2012;16(1):122-34. 139. Noble CL, Abbas AR, Cornelius J, Lees CW, Ho GT, Toy K, et al. Regional variation in gene expression in the healthy colon is dysregulated in ulcerative colitis. Gut. 2008;57(10):1398-405. 140. Wiklund IK, Fullerton S, Hawkey CJ, Jones RH, Longstreth GF, Mayer EA, et al. An irritable bowel syndrome-specific symptom questionnaire: development and validation. Scandinavian journal of gastroenterology. 2003;38(9):947-54. 141. Gustavsson JP, Bergman H, Edman G, Ekselius L, von Knorring L, Linder J. Swedish universities Scales of Personality (SSP): construction, internal consistency and normative data. Acta psychiatrica Scandinavica. 2000;102(3):217-25. 142. Bubenik GA. Localization of melatonin in the digestive tract of the rat. Effect of maturation, diurnal variation, melatonin treatment and pinealectomy. Hormone research. 1980;12(6):313-23. 143. Holloway WR, Grota LJ, Brown GM. Determination of immunoreactive melatonin in the colon of the rat by immunocytochemistry. J Histochem Cytochem. 1980;28(3):255-62. 144. Manchester LC, Poeggeler B, Alvares FL, Ogden GB, Reiter RJ. Melatonin immunoreactivity in the photosynthetic prokaryote Rhodospirillum rubrum: implications for an ancient antioxidant system. Cellular & molecular biology research. 1995;41(5):391-5. 145. Reiter RJ, Tan DX, Burkhardt S, Manchester LC. Melatonin in plants. reviews. 2001;59(9):286-90. 146. Stebelova K, Anttila K, Manttari S, Saarela S, Zeman M. Immunohistochemical definition of MT(2) receptors and melatonin in the gastrointestinal tissues of rat. Acta Histochem. 2010;112(1):26-33. 147. Muxel SM, Pires-Lapa MA, Monteiro AW, Cecon E, Tamura EK, Floeter- Winter LM, et al. NF-kappaB drives the synthesis of melatonin in RAW 264.7 macrophages by inducing the transcription of the arylalkylamine-N- acetyltransferase (AA-NAT) gene. PLoS One. 2012;7(12):e52010. 148. Reiter RJ, Tan DX, Fuentes-Broto L. Melatonin: a multitasking molecule. Prog Brain Res. 2010;181:127-51.

60 149. Chojnacki C, Wisniewska-Jarosinska M, Kulig G, Majsterek I, Reiter RJ, Chojnacki J. Evaluation of enterochromaffin cells and melatonin secretion exponents in ulcerative colitis. World journal of gastroenterology. 2013;19(23):3602-7. 150. Zibolka J, Bazwinsky-Wutschke I, Muhlbauer E, Peschke E. Distribution and density of melatonin receptors in human main pancreatic islet cell types. J Pineal Res. 2018;65(1):e12480. 151. McMullan CJ, Schernhammer ES, Rimm EB, Hu FB, Forman JP. Melatonin secretion and the incidence of type 2 diabetes. JAMA. 2013;309(13):1388- 96. 152. Bonnefond A, Clement N, Fawcett K, Yengo L, Vaillant E, Guillaume JL, et al. Rare MTNR1B variants impairing function contribute to type 2 diabetes. Nat Genet. 2012;44(3):297-301. 153. Zhou J, Wang D, Luo X, Jia X, Li M, Laudon M, et al. Melatonin Receptor Agonist Piromelatine Ameliorates Impaired Glucose Metabolism in Chronically Stressed Rats Fed a High-Fat Diet. J Pharmacol Exp Ther. 2018;364(1):55-69. 154. Bhattacharya S, Patel KK, Dehari D, Agrawal AK, Singh S. Melatonin and its ubiquitous anticancer effects. Molecular and cellular biochemistry. 2019;462(1-2):133-55. 155. Sainz RM, Mayo JC, Tan DX, Leon J, Manchester L, Reiter RJ. Melatonin reduces prostate cancer cell growth leading to neuroendocrine differentiation via a receptor and PKA independent mechanism. The Prostate. 2005;63(1):29-43. 156. Jablonska K, Pula B, Zemla A, Kobierzycki C, Kedzia W, Nowak-Markwitz E, et al. Expression of the MT1 melatonin receptor in ovarian cancer cells. Int J Mol Sci. 2014;15(12):23074-89. 157. Srinivasan V, Spence DW, Pandi-Perumal SR, Trakht I, Cardinali DP. Therapeutic actions of melatonin in cancer: possible mechanisms. Integr Cancer Ther. 2008;7(3):189-203. 158. Winczyk K, Pawlikowski M, Lawnicka H, Kunert-Radek J, Spadoni G, Tarzia G, et al. Effects of melatonin and melatonin receptors ligand N-[(4- methoxy-1H-indol-2-yl)methyl]propanamide on murine Colon 38 cancer growth in vitro and in vivo. Neuro Endocrinol Lett. 2002;23 Suppl 1:50-4. 159. Roberts JE, Wiechmann AF, Hu DN. Melatonin receptors in human uveal melanocytes and melanoma cells. J Pineal Res. 2000;28(3):165-71. 160. Lu WZ, Song GH, Gwee KA, Ho KY. The effects of melatonin on colonic transit time in normal controls and IBS patients. Dig Dis Sci. 2009;54(5):1087-93. 161. Tan W, Zhou W, Luo HS, Liang CB, Xia H. The inhibitory effect of melatonin on colonic motility disorders induced by water avoidance stress in rats. European review for medical and pharmacological sciences. 2013;17(22):3060-7. 162. Talley NJ, Phillips SF, Haddad A, Miller LJ, Twomey C, Zinsmeister AR, et al. GR 38032F (), a selective 5HT3 receptor antagonist, slows colonic transit in healthy man. Dig Dis Sci. 1990;35(4):477-80. 163. Gregory RE, Ettinger DS. 5-HT3 receptor antagonists for the prevention of chemotherapy-induced nausea and vomiting. A comparison of their pharmacology and clinical efficacy. Drugs. 1998;55(2):173-89. 164. Masana MI, Doolen S, Ersahin C, Al-Ghoul WM, Duckles SP, Dubocovich ML, et al. MT(2) melatonin receptors are present and functional in rat caudal artery. J Pharmacol Exp Ther. 2002;302(3):1295-302.

61 165. Karling P, Danielsson A, Adolfsson R, Norrback KF. No difference in symptoms of irritable bowel syndrome between healthy subjects and patients with recurrent depression in remission. Neurogastroenterology and motility : the official journal of the European Gastrointestinal Motility Society. 2007;19(11):896-904. 166. Karling P, Maripuu M, Wikgren M, Adolfsson R, Norrback KF. Association between gastrointestinal symptoms and affectivity in patients with bipolar disorder. World journal of gastroenterology. 2016;22(38):8540-8. 167. Gros DF, Antony MM, McCabe RE, Swinson RP. Frequency and severity of the symptoms of irritable bowel syndrome across the anxiety disorders and depression. Journal of anxiety disorders. 2009;23(2):290-6. 168. Chang L, Sundaresh S, Elliott J, Anton PA, Baldi P, Licudine A, et al. Dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis in irritable bowel syndrome. Neurogastroenterology and motility : the official journal of the European Gastrointestinal Motility Society. 2009;21(2):149-59. 169. Barreau F, Ferrier L, Fioramonti J, Bueno L. Neonatal maternal deprivation triggers long term alterations in colonic epithelial barrier and mucosal immunity in rats. Gut. 2004;53(4):501-6. 170. Kulak-Bejda A, Bejda G, Waszkiewicz N. Antidepressants for irritable bowel syndrome-A systematic review. Pharmacological reports : PR. 2017;69(6):1366-79. 171. Voultsios A, Kennaway DJ, Dawson D. Salivary melatonin as a circadian phase marker: validation and comparison to plasma melatonin. Journal of biological rhythms. 1997;12(5):457-66. 172. Spiller RC, Jenkins D, Thornley JP, Hebden JM, Wright T, Skinner M, et al. Increased rectal mucosal enteroendocrine cells, T lymphocytes, and increased gut permeability following acute Campylobacter enteritis and in post- dysenteric irritable bowel syndrome. Gut. 2000;47(6):804-11. 173. Saha L, Malhotra S, Rana S, Bhasin D, Pandhi P. A preliminary study of melatonin in irritable bowel syndrome. Journal of clinical gastroenterology. 2007;41(1):29-32. 174. Reiter RJ, Manchester LC, Tan DX. Melatonin in walnuts: influence on levels of melatonin and total antioxidant capacity of blood. Nutrition. 2005;21(9):920-4. 175. Ziegler A, Gonzalez L, Blikslager A. Large Animal Models: The Key to Translational Discovery in Digestive Disease Research. Cellular and molecular gastroenterology and hepatology. 2016;2(6):716-24. 176. Turner PV. The role of the gut microbiota on animal model reproducibility. Animal models and experimental medicine. 2018;1(2):109-15. 177. Cui J, Li Z, Zhuang S, Qi S, Li L, Zhou J, et al. Melatonin alleviates inflammation-induced apoptosis in human umbilical vein endothelial cells via suppression of Ca(2+)-XO-ROS-Drp1-mitochondrial fission axis by activation of AMPK/SERCA2a pathway. Cell stress & chaperones. 2018;23(2):281-93. 178. Yun CW, Kim S, Lee JH, Lee SH. Melatonin Promotes Apoptosis of Colorectal Cancer Cells via Superoxide-mediated ER Stress by Inhibiting Cellular Prion Protein Expression. Anticancer Res. 2018;38(7):3951-60. 179. Cunningham JL, Grimelius L, Sundin A, Agarwal S, Janson ET. Malignant ileocaecal serotonin-producing carcinoid tumours: the presence of a solid growth pattern and/or Ki67 index above 1% identifies patients with a poorer prognosis. Acta Oncol. 2007;46(6):747-56.

62 180. Garcia-Navarro A, Gonzalez-Puga C, Escames G, Lopez LC, Lopez A, Lopez-Cantarero M, et al. Cellular mechanisms involved in the melatonin inhibition of HT-29 human colon cancer cell proliferation in culture. J Pineal Res. 2007;43(2):195-205. 181. Forsythe P, Sudo N, Dinan T, Taylor VH, Bienenstock J. Mood and gut feelings. Brain, behavior, and immunity. 2010;24(1):9-16. 182. Pinto-Sanchez MI, Hall GB, Ghajar K, Nardelli A, Bolino C, Lau JT, et al. Probiotic longum NCC3001 Reduces Depression Scores and Alters Brain Activity: A Pilot Study in Patients With Irritable Bowel Syndrome. Gastroenterology. 2017;153(2):448-59 e8. 183. Pagan C, Goubran-Botros H, Delorme R, Benabou M, Lemiere N, Murray K, et al. Disruption of melatonin synthesis is associated with impaired 14-3-3 and miR-451 levels in patients with autism spectrum disorders. Scientific reports. 2017;7(1):2096. 184. Benabou M, Rolland T, Leblond CS, Millot GA, Huguet G, Delorme R, et al. Heritability of the melatonin synthesis variability in autism spectrum disorders. Scientific reports. 2017;7(1):17746.

63 Acta Universitatis Upsaliensis Digital Comprehensive Summaries of Uppsala Dissertations from the Faculty of Medicine 1616 Editor: The Dean of the Faculty of Medicine

A doctoral dissertation from the Faculty of Medicine, Uppsala University, is usually a summary of a number of papers. A few copies of the complete dissertation are kept at major Swedish research libraries, while the summary alone is distributed internationally through the series Digital Comprehensive Summaries of Uppsala Dissertations from the Faculty of Medicine. (Prior to January, 2005, the series was published under the title “Comprehensive Summaries of Uppsala Dissertations from the Faculty of Medicine”.)

ACTA UNIVERSITATIS UPSALIENSIS Distribution: publications.uu.se UPPSALA urn:nbn:se:uu:diva-396347 2019