Ayşe Şiva Acar1, Oytun Erbaş1,2

1ERBAS Institute of Experimental Medicine, Illinois, USA & Gebze, Turkey
2Department of Physiology, Medical Faculty of Demiroğlu Bilim University, Istanbul, Turkey

Keywords: Bipolar spectrum disorder, GLP-1, liraglutide, mood disorders, neuroprotection, obesity, schizophrenia

Abstract

GLP-1 (Glucagon-like peptide-1) receptor agonists have incretin, cardioprotective and neuroprotective effects. Liraglutide, a GLP-1 receptor agonist, is used to treat comorbid diseases such as obesity and diabetes through its incretin and cardioprotective effects. The therapeutic targets of Exenatide on neurodegenerative diseases have recently been concentrated, particularly after the discovery of GLP-1's neuroprotective effects. Mood disorders and metabolic comorbidities are the two groups of diseases that have the greatest global impact on mortality. There is also a significant correlation between patients with mood disorders and patients with obesity and T2DM (Type 2 diabetes mellitus), according to epidemiological data. GLP-1 receptor agonists are exceedingly being used to treat psychological disorders. According to the results of preclinical studies, Liraglutide can prevent cognitive function regression and develop normal functions. This condition suggests that GLP-1 receptor agonists could be useful in the treatment of psychiatric disorders in the future. This review examined the relationship between GLP-1 Receptor Agonists and Psychiatric diseases.

The glucagon-like peptide-1 (GLP-1) is an incretin hormone with a peptide structure that can be synthesized in a variety of locations all through the body and performs a variety of functions.[1] GLP-1 produced mainly by L cells in small intestine distal ileum[2] and colon, with a very small amount produced by alpha cells found in the pancreas.[1]

Glucagon-like peptide-1 receptor agonists (GLP1RA) activate the GLP-1 receptors (GLP-1Rs) in the body, which is attached to essential amino acids, fatty acids, or glucose that convert into post-nutrient intake basic components,[3] enabling multiple intracellular pathways to work, including cAMP/ PKA/CREB and PI3K/Akt.[4] GLP-1R activation occurs when nutrients are consumed orally, but not when nutrients are consumed intravenously (İV).[5]

Dipeptidyl Peptidase-4 (DPP-4) is a serine protease that separates dipeptides from polypeptides' amino-terminal. CD56 is another term for the same subject.[6] Due to proteolytic enzyme DPP-4's rapid inhibition, the half-life of GLP-1 in circulation is less than 2 minutes.[7] DPP-4 inhibitors (DPP4-I) are a new variant of oral hypoglycemic agent that increases GLP-1 levels in the bloodstream and are used to treatment of diabetes.[8]

GLUCAGON-LIKE PEPTIDE-1 RECEPTORS

In addition to the ability to produce GLP-1, the brain contains many GLP-1Rs. GLP-1Rs are produced predominantly in neurons, astrocytes and microglia in the brain.[9] Cardiovascular functions are controlled by GLP-1Rs found in Nucleus tractus solitarius (NTS) connected with area postrema (AP). In a study of dogs, it was observed that glp-1 infusion stimulates cardiac flow and improves hemodynamics of the left heart in dilate cardiomyopathy caused by fast pacing.[10] Increased cardiovascular function is independent of catecholamines during the iv infusion of GLP-1.[11]

Glucagon-like peptide-1 receptors are also located in reward areas of the brain, where flavor or drug consumption affects elevated dopamine levels and glutaminergic neuron transmission. GLP1-R agonists act by inhibiting pentagastrin, which has gastrin-like effect, and gastrin, which is stimulated by food and prevents stomach emptying.[12] So, food consumption and substance use gradually decline, and weight loss results.[13]

EFFECTS OF GLUCAGON-LIKE PEPTIDE-1

The incretin effect on pancreatic beta cells is one of the functions that makes GLP-1 so famous today. The release of insulin, which is stimulated following glucose intake into the bloodstream, causes the incretin effect in glucose metabolism.[14] GLP-1 is secreted by the Leptin, which is a hormone synthesized from L-cells in distal ileum.

Leptin tolerance and a reduction in GLP-1 levels are seen in people who eat a high-fat diet and become obese.[15] GLP-1RA and DPP-4 inhibitors are also successful in the treatment of type 2 diabetes mellitus (T2DM) though breaking down the leptin resistance.[3] In particular, Liraglutide and Exenatide are powerful T2DM receptor agonists in the treatment of GLP-1.[16]

CARDIOPROTECTIVE EFFECTS

Despite the fact that GLP-1RAs have cardioprotective effects, DPP-4 inhibitors are ineffective in patients with cardiovascular disease.[17] GLP-1 receptors reduce left ventricular functions in the isolated rat heart, thus reducing ischemia and infarction sizes.[18] In a retrospective study, it was reported with the LEADER study that the risk of cardiovascular disease in diabetic patients who had been given Liraglutide had decreased with the placebo group. But no obvious cardiovascular changes were reported compared to placebo if Lixisenatide which was also GLP-1RA in another study, was administered to diabetic patients with acute coronary syndrome (ACS).[19]

In the high-risk population for ACS, a comparable analysis was not performed in non-diabetic patients. As a result, it's unknown if GLP-1 RA has therapeutic efficacy in non-diabetic patients with ACS.[20] Furthermore, clinical trials in T2DM patients have shown that GLP-1RA[20,21] is more effective than DPP-4 inhibitors[22,23] in preventing the development of cardiovascular risks.

NEUROPROTECTIVE EFFECTS

There are many cases where GLP-1 and GLP1RAs active in the brain. GLP-1 and GLP-1RAs block apoptosis induced by hypoxia or methylglyoxal (MG), a product of chronic hyperglycemia, by regulating glucose levels in the brain.[24-26] It also shows a neuroprotective effect by minimizing amyloid precursor protein (APP) accumulation in the brain[27] and attempting to control neuronal damage that occurs after ischemia.[28,29]

The mouse brain has been observed to reduce chronic inflammation following radiation exposure with Liraglutide application.[30] Furthermore, it is also reported that GLP-1R agonists are increasing the progenitor cell proliferation in dentate gyrus and the antagonists are decreasing the progenitor cell proliferation and have a positive effect on synaptic plasticity.[31,32]

Liraglutide, a long-acting GLP-1RA, can penetrate circumventricular organs (CVOs) and hypothalamic neurons by crossing the blood brain barrier.[33,34] Likewise, DPP-4 inhibitors are also thought to be capable of crossing the intact blood brain barrier.[35]

In a study of T2DM mice, findings of a high degree of learning and memory impairment were observed through tests of mEPM (modified elevated plus maze) and PA (passive avoidance). Exenatide is thought to be a drug to reduce the effects of cognitive diseases in later periods due to improving cognitive ability and stimulating the molecular mechanisms of memory storage in mice.[36]

GLUCAGON-LIKE PEPTIDE-1 EFFECTS ON NEURODEGENERATIVE DISEASES

Many in vivo and in vitro studies have shown that GLP-1RAs have protective effects in neurodegenerative diseases such as Alzheimer's (AD) and Parkinson's disease (PD),[37] raising the possibility that GLP-1RAs may be used to treat these diseases.[38]

GLP-1 AND ALZHEIMER'S DISEASE (AD)

Alzheimer's disease is a deadly neurodegenerative disease characterized by dementia and decreased cognitive functions (such as language and thought disorders) that result in the progression of memory loss. Today, AD is the most common among neurodegenerative diseases. Although the etiology of AD has not been conclusively determined, the common pathological properties that patients have are characterized by the accumulation of non-neuron beta-amyloid (Aβ) plaques and neurofibrillary tangles of intra-neuron hyperphosphorylene tau proteins.[39] Today, no therapies that affect the progression of the disease have been developed, and patients are only given symptomatic care.[40]

The preclinical models studied showed reduced amyloid accumulation and neuroinflammation with Liraglutide application,[41,42] improved cognitive outcomes with increased brain glucose metabolism,[43] and increased proliferation of neuronal progenitor cells.[44]

GLP-1 AND PARKINSON'S DISEASE (PD)

Parkinson's disease is characterized by the death of dopaminergic neurons in the substantia nigra, central nervous system, which appears tremor and rigidity symptoms.[45] It is the most common second neurodegenerative disease after AD.[46]

The coexistence of dopaminergic neurons and insulin receptors in Substantia nigra makes the association between T2DM and Parkinson's even stronger. Dopamine depletion in the striatum and reduced insulin signaling in basal ganglia are also associated with the issue. Previous studies have confirmed that patients with insulin resistance and T2DM are in the high risk group for PD as a result of the high rate of abnormal glucose tolerance seen in Parkinson's patients.[47] It is therefore thought that antidiabetic agents such as peroxisome proliferator activated receptor-γ, GLP-1, and DPP-4 may be important therapeutic targets in the pathophysiological mechanism of PD.[47-50]

At the end of a 2020 meta-analysis, Exenatide use was reported to have prevented cognitive impairment and extended lifetime by improving quality of life by increasing in UPRDS (Unified Parkinson's Disease Rating Scale) scores with a cognitive test score in Parkinson's patients.[51-53] A study with Exenatide in patients with Parkinson's supported that Exenatide can be used for motor dysfunction diseases’ treatment[54] causing decreases non-motor symptoms and motor complications.[50]

GLP-1 AND ISCHEMIC STROKE

Ischemia is one of the major causes of death in the world.[55] An ischemic stroke can be performed 4-6 hours after a thrombolysis or thrombectomy when the patient's body is suitable for the procedure. However, only a small percentage of patients are eligible for the treatment.[56]

Although animal studies on the treatment of GLP-1 in stroke are extremely high, clinical studies on humans have recently begun to increase.[24] The immediate use of GLP-1RA following a stroke shows a good therapeutic window of up to 3 hours in mice, compared to approximately 36 hours in a human. This suggests that using GLP-1RA to treat ischemic stroke is a favorable treatment option.[57] According to tissue plasminogen activators (tPA) whose therapeutic window is 4.5 hours, GLP-1RAs have been shown to have a much longer effect and have a potential to be used in acute stroke.[17]

MOOD DISORDERS AND SCHIZOPHRENIA

Mood disorders and schizophrenia are the most common and severe observed chronic mental disorders.[58]

GLP-1R gene expression has a large difference between the healthy group and patients with mood disorders when factors such as age, sex, ethnicity are kept stable.[58] It has been reported through animal studies that declining numbers of GLP-1 positive cells in the brain is associated with obesity and metabolic syndrome.[59] In 2018, association between Body Mass Index (BMI) and GLP-1R expression over the brain tissues of death psychiatric patients was investigated. Excess GLP-1R and GLP-2R expression was detected in the postmortem brains of people with mental conditions as a result of this research.[58]

In a meta-analysis study, it was reported that GLP-1RAs may be effective at blocking the weight increase observed as a side effect in patients on Clozapine/Olanzapine, medications prescribed for treating schizophrenia and similar mood disorders.[60,61] Another study of patients using Clozapine found that after adding liraglutide to the treatment of schizophrenia patients, their HbA1c levels decreased from 10.0% to 6.5%, and as much as 8.7% of their body weight was lost.

As a result of epidemiological studies on humans, correlation is observed between mood disorders and metabolic comorbidities such as obesity, and T2DM.[62-65] While the increased prevalence of depression is observed in T2DM people, depression is also considered to be a risk factor for the development of diabetes.[66,67] A study reported that diseases such as obesity, T2DM and metabolic syndrome were observed 2 times more frequently in patients with mood disorder compared to the general population[68] One of the common factors underlying the link between obesity/diabetes and mood disorders is hypothalamic-pituitary-adrenal (HPA) axis dysfunction.[69] The co-observation of these diseases also reveals conditions that negatively affect the pathology of two diseases, such as obesity, the rapid rise of insulin resistance, increased suicide attempts, resistance to treatment.[70-72]

For non-diabetic individuals with mood disorders, GLP-1R agonists are safe and well tolerable agents. It is also thought that it may have beneficial effects that can alleviate metabolic disorders.[73]

Due to Preclinical studies, although the potential impact of Liraglutide on individuals with mood disorder was reported,[73] no noticeable effects have been found in trials with Exenatide in patients with schizophrenia.[74]

EATING DISORDERS

Eating disorders are serious psychiatric illnesses characterized by overeating and weight control, which can result in death by progressing if left untreated.[75] Eating behavior is inhibited by GLP-1 receptor activation found at the CA1 site in the ventral hippocampal area.[76] GLP-1 receptor agonists such as Liraglutide are thought to be effective in treating overeating by affecting this region.[77]

Emotional eating status has an activationreducing effect in the insula region of the brain in T2DM patients and in the amygdala in obese patients. Glucose intake reduces food consumption in the succeeding period, increasing the feeling of satiety, but GLP-1 receptor antagonists disappear these changes.[78] Levels of GLP-1 in obese people have not changed if the meal, which is not tasty or tasty, is eaten.[79]

Overeating is defined as consuming an abnormally large amount of food in a short period of time, with a subjective sense of loss of control.[80] In T2DM patients with an eating disorder, the GLP-1 effect has been observed to be low.[81,82]

A study involving normal weight and obese adults using Liraglutide examined the weight status of individuals with mood disorders. According to the findings of the study, Liraglutide improved metabolic parameters in non-diabetic individuals with mood disorders, and after treatment, subcortical structure and frontal gray matter volumes associated with weight loss increased. This increase has been associated with improvement in cognitive functions.[83]

A study of Bulimia nervosa patients found that reduced GLP-1 levels caused overeating episodes.[84] Although the relationship between bulimia and GLP-1 could not be explained clearly[85] GLP-1RA Liraglutide was suggested that there would be safe and effective treatments in Bulimia nervosa and overeating disorders.[86]

ANHEDONIA AND DEPRESSION

Depression is a psychiatric disease associated with extreme blood-brain barrier permeability, neuroinflammation and synaptic dysfunction,[87,88] adversely affecting the lives of the patient and his family.[89] In the severity and incidence of depression, the gene pool and environmental stressors that one and their family have are thought to be effective.[90]

Anhedonia is a reduction in the ability to motivate or enjoy and manifests in many psychiatric disorders.[91] Although many antidepressants are available for anhedonia and depression, 30% of diagnosed patients do not have the required therapeutic effect in these treatments.[92]

In the study among patients diagnosed with 100 control and 164 schizophrenia/bipolar spectrum disorders, GLP-1R polymorphisms’ Anhedonia's laboratory-based measurement method as a probabilistic reward learning task response bias was observed in Anhedonia and depression phenotypes.[93]

Another study in rats reported that depression and metabolic abnormalities that develop as a result of prolonged use of atypical antipsychotics with Liraglutide could be blocked.[94]

MAJOR DEPRESSIVE DISORDER

Major Depressive disorder is a neuropsychiatric disorder common in society, characterized by the observation of emotion condition changes such as irritability or state of sadness to the extent that it affects one’s work and family life, lasting at least 2 weeks. These mood changes can be cited as examples of anorexia, sexual reluctance, inability to enjoy, crying, suicidal ideation, slowing down in speech and actions.[95] Major depressive disorder (MDB) can be observed with comorbid conditions and obesity.[96]

BIPOLAR SPECTRUM DISORDER

Bipolar disorder (BD) is a chronic psychiatric disease that repeats itself manically and depressively in two extreme phases with certain periods.[97,98] Diagnosis is easier to make during the manic period when symptoms are more observable. Most bipolar patients may not get an accurate diagnosis during the early periods of the disease, as the cycle begins with a period of depression.[99]

To diagnose Bipolar Disorder type 1, at least one manic episode is needed.[100] These patients also experience depressive periods, but the presence of manic periods for diagnosis is distinctive.[101]

In bipolar disorder type 2, no manic periods are observed. It is replaced by one or more hypomanic periods and one or more major depressive periods that are not as severe as the manic period.[100]

A study of patients with BD and a control group found that the level of GLP-1 was significantly lower in patients with BD but it was also noted that GLP-1 levels were not enough to stage the disease.[102]

As a result of the study of 57 Bipolar patients, 24 of type 1, 33 of type 2, the low levels of GLP-1, glucagon and Ghrelin and high levels of the Gastric inhibitor polypeptide (GIP) are common qualifications in all patients and can have a role in the pathogenesis and diagnosis of BD, particularly in the depressive phase.[103]

Patients with BD have three times higher incidence of T2DM. At the same time, severe BD is extremely likely to occur in comorbid T2DM patients, and these patients are extremely resistant to treatment. T2DM and Bipolar patients have higher cardiovascular morbidity and mortality. These two disorders may be related from common pathophysiological origins, such as LHPA (limbichypothalamic-pituitary-adrenal) or mitochondrial dysfunction, or from common genetic connections or epigenetic interactions. It is thought that these drugs could be used to treat T2DM after studies have shown that the drugs used for bipolar spectrum disorder facilitate glycemic control.[104]

DEPRESSION AND ANXIETY

GLP-1RA can have significant effects on psychological stress responses as well as mood disorders.[105] Liraglutide has the mitigating effect of symptoms on depressive and anxiety behaviors caused by corticosteroids and regulates the endocrine system by normalizing Adrenocorticotropic hormone (ACTH) levels after stress treatment. As a consequence it is conceivable that it could be used as an antidepressant drug in the long term.[106]

In a pilot study with 19 volunteers, daily use of 1.8 mg of Liraglutide proved safe and well tolerable for non-diabetics with mood disorder.[83]

SUBSTANCE ABUSE

GLP-1 Rs are located in areas important for the brain's reward mechanism. GLP-1 R's are helping reduce delicious food intake and cocaine, amphetamine, opioid, nicotine and alcohol use by significantly altering dopamine levels and glutamatergic neurotransmission following food or drug intake.[13]

A study in those with cocaine addict people found that following cocaine injection, GLP-1 levels were significantly reduced, thus triggering more cocaine intake and subjective anxiety in the person.[107] Exenatide-4 supplementation decreased cocaine-induced preference for the conditioned location and dopamine release.[108]

Exenatide-4 (2.4 g/kg) decreased abnormal amphetamine mobility, conditional location preference, and accumulated accumbal dopamine release without disturbing the drug's spontaneous locomotor activity in amphetamine users.[108]

Exenatide reduced the locomotor behavior induced by nicotine in mice and blocked locomotor sensitivity. It also reduced the release of accumulated dopamine and the preference for nicotine-derived conditioned places.[108]

Ethanol intake and spontaneous locomotor activity decreased depending on dose with Exenatide-4 in rats.[109] Alcohol addicts received a significant decrease in alcohol intake and alcoholseeking behaviors as a result of receiving Exenatide-4 treatment for 8 months.[108]

A study in alcohol-dependent mice observed significantly reduced ethanol intake with AC3174, an Exenatide analogue.[110] Another agonist, Liraglutide, also eased the rise in alcohol-related dopamine and conditional location preference, preventing alcohol abstinence-related drinking.[111]

Another trial in monkeys showed that Liraglutide and Exenatide decreased alcohol consumption without causing adverse effects including dehydration or nausea/vomiting.[112]

SUICIDE

The inhibitory effect of Sitagliptin, a very powerful inhibitor of DPP-4, stops at plateau stage 93%. High levels of Sitagliptin causes levels of the increasing hormones and contributes to preventing hypoglycemia after overdose due to the fact that the activity of the incretin is dependent on glucose.[113]

Depression prevalence is high in patients with diabetes, and depression is a strong risk factor for suicide.[8] Sitagliptin is recommended for treating diabetes in the elderly and patients with suicidal psychiatric disorders.[8] Although nocturnal hypoglycemia could not be completely ruled out in the Japanese woman patient who attempted suicide by taking 1700 mg Sitagliptin aged 86, apparent hypoglycemia was observed later and as a result of this condition, Sitagliptin is defined as a low-risk oral hypoglycemic agent.

CONCLUSION-RESULT

When we examined the studies done on GLP-1, it was concluded that receptor agonists improve cognitive functions and inhibit reduction in functions due to their antidiabetic effects, as well as their neuroprotective effects. The number of clinical trials studying its use in treating neurodegenerative disorders such as AD and PD is continuously rising, with promising therapeutic findings in the near term. However, the number of studies on neuroprotection should be increased even further, and not only cognitive properties as well as their effects on social behaviors should be investigated.

Larger randomized controlled studies should be conducted on the topic, and studies should be translated and expanded into more languages so that studies to accelerate around the world and include more scientists. Psychiatric disorders that can be observed along with metabolic abnormalities such as diabetes mellitus should certainly not be overlooked, but follow-up and treatment should be done in consultation with psychiatry-endocrinology.

Likewise, reducing mortality in patients with bipolar spectrum disorder, reducing cardiovascular risk factors as much as possible and treating metabolic disorders that have the potential to develop to improving patients’ quality of life are also extremely critical for physicians.

It is also thought that GLP-1RAs can be used to treat substance abuse through a significant reduction in addiction and similar behaviors for cocaine, amphetamines, alcohol and nicotine.

GLP-1RAs are seen as an effective agent for potential mortality in patients with metabolic comorbidity and emotion condition disorder due to their beneficial impact on both weight control and cognitive functions.

Cite this article as: Şiva Acar A, Erbaş O. Glucagon-Like Peptide-1 and Psychiatric Disorder. JEB Med Sci 2021;2(2):106-115.

Conflict of Interest

The authors declared no conflicts of interest with respect to the authorship and/or publication of this article.

Financial Disclosure

The authors received no financial support for the research and/or authorship of this article.

References

  1. Calsolaro V, Edison P. Novel GLP-1 (glucagon-like peptide-1) analogues and insulin in the treatment for Alzheimer's disease and other neurodegenerative diseases. CNS Drugs 2015;29:1023-39.
  2. Feng X, Zhong S, Yang J, Wang Y, Liu J. Effects on glucagon-like peptide-1 secretion by distal ileal administration of nutrients. Obes Surg 2013;23:1774-82.
  3. Andersen ES, Deacon CF, Holst JJ. Do we know the true mechanism of action of the DPP-4 inhibitors? Diabetes Obes Metab 2018;20:34-41.
  4. Thorens B. Expression cloning of the pancreatic beta cell receptor for the gluco-incretin hormone glucagon-like peptide 1. Proc Natl Acad Sci U S A 1992;89:8641-5.
  5. Unger RH, Ohneda A, Valverde I, Eisentraut AM, Exton J. Characterization of the responses of circulating glucagon-like immunoreactivity to intraduodenal and intravenous administration of glucose. J Clin Invest 1968;47:48-65.
  6. Mentlein R, Gallwitz B, Schmidt WE. Dipeptidyl-peptidase IV hydrolyses gastric inhibitory polypeptide, glucagonlike peptide-1(7-36)amide, peptide histidine methionine and is responsible for their degradation in human serum. Eur J Biochem 1993;214:829-35.
  7. Deacon CF, Nauck MA, Toft-Nielsen M, Pridal L, Willms B, Holst JJ. Both subcutaneously and intravenously administered glucagon-like peptide I are rapidly degraded from the NH2-terminus in type II diabetic patients and in healthy subjects. Diabetes 1995;44:1126-31.
  8. Furukawa S, Kumagi T, Miyake T, Ueda T, Niiya T, Nishino K, et al. Suicide attempt by an overdose of sitagliptin, an oral hypoglycemic agent: A case report and a review of the literature. Endocr J 2012;59:329-33.
  9. Hamilton A, Hölscher C. Receptors for the incretin glucagon-like peptide-1 are expressed on neurons in the central nervous system. Neuroreport 2009 Aug;20:1161-6.
  10. Nikolaidis LA, Elahi D, Hentosz T, Doverspike A, Huerbin R, Zourelias L, et al. Recombinant glucagon-like peptide-1 increases myocardial glucose uptake and improves left ventricular performance in conscious dogs with pacing-induced dilated cardiomyopathy. Circulation 2004;110:955-61.
  11. Razeghi P, Young ME, Alcorn JL, Moravec CS, Frazier OH, Taegtmeyer H. Metabolic gene expression in fetal and failing human heart. Circulation 2001;104:2923-31.
  12. Willms B, Werner J, Holst JJ, Orskov C, Creutzfeldt W, Nauck MA. Gastric emptying, glucose responses, and insulin secretion after a liquid test meal: Effects of exogenous glucagon-like peptide-1 (GLP-1)-(7-36) amide in type 2 (noninsulin-dependent) diabetic patients. J Clin Endocrinol Metab 1996;81:327-32.
  13. Eren-Yazicioglu CY, Yigit A, Dogruoz RE, Yapici-Eser H. Can GLP-1 be a target for reward system related disorders? A qualitative synthesis and systematic review analysis of studies on palatable food, drugs of abuse, and alcohol. Front Behav Neurosci 2021;14:614884.
  14. Campbell JE, Drucker DJ. Pharmacology, physiology, and mechanisms of incretin hormone action. Cell Metab 2013;17:819-37.
  15. Anini Y, Brubaker PL. Role of leptin in the regulation of glucagon-like peptide-1 secretion. Diabetes 2003;52:252-9.
  16. Trujillo JM, Nuffer W, Ellis SL. GLP-1 receptor agonists: A review of head-to-head clinical studies. Ther Adv Endocrinol Metab 2015;6:19-28.
  17. Marlet IR, Ölmestig JNE, Vilsbøll T, Rungby J, Kruuse C. Neuroprotective mechanisms of glucagon-like peptide1-based therapies in ischaemic stroke: A systematic review based on pre-clinical studies. Basic Clin Pharmacol Toxicol 2018;122:559-69.
  18. Gros R, You X, Baggio LL, Kabir MG, Sadi AM, Mungrue IN, et al. Cardiac function in mice lacking the glucagon-like peptide-1 receptor. Endocrinology 2003;144:2242-52.
  19. Pfeffer MA, Claggett B, Diaz R, Dickstein K, Gerstein HC, Køber LV, et al. Lixisenatide in patients with type 2 diabetes and acute coronary syndrome. N Engl J Med 2015;373:2247-57.
  20. Marso SP, Daniels GH, Brown-Frandsen K, Kristensen P, Mann JF, Nauck MA, et al. Liraglutide and cardiovascular outcomes in type 2 diabetes. N Engl J Med 2016;375:311-22.
  21. Marso SP, Bain SC, Consoli A, Eliaschewitz FG, Jódar E, Leiter LA, et al. Semaglutide and cardiovascular outcomes in patients with type 2 diabetes. N Engl J Med 2016;375:1834-44.
  22. Green JB, Bethel MA, Armstrong PW, Buse JB, Engel SS, Garg J, et al. Effect of Sitagliptin on cardiovascular outcomes in type 2 diabetes. N Engl J Med 2015;373:232-42.
  23. White WB, Cannon CP, Heller SR, Nissen SE, Bergenstal RM, Bakris GL, et al. Alogliptin after acute coronary syndrome in patients with type 2 diabetes. N Engl J Med 2013;369:1327-35.
  24. Lerche S, Brock B, Rungby J, Bøtker HE, Møller N, Rodell A, et al. Glucagon-like peptide-1 inhibits blood-brain glucose transfer in humans. Diabetes 2008;57:325-31.
  25. Li Y, Perry T, Kindy MS, Harvey BK, Tweedie D, Holloway HW, et al. GLP-1 receptor stimulation preserves primary cortical and dopaminergic neurons in cellular and rodent models of stroke and Parkinsonism. Proc Natl Acad Sci U S A 2009;106:1285-90.
  26. Iwai T, Suzuki M, Kobayashi K, Mori K, Mogi Y, Oka J. The influences of juvenile diabetes on memory and hippocampal plasticity in rats: improving effects of glucagon-like peptide-1. Neurosci Res 2009;64:67-74.
  27. Perry T, Lahiri DK, Sambamurti K, Chen D, Mattson MP, Egan JM, et al. Glucagon-like peptide-1 decreases endogenous amyloid-beta peptide (Abeta) levels and protects hippocampal neurons from death induced by Abeta and iron. J Neurosci Res 2003;72:603-12.
  28. Zhang H, Liu Y, Guan S, Qu D, Wang L, Wang X, et al. An orally active allosteric GLP-1 receptor agonist is neuroprotective in cellular and rodent models of stroke. PLoS One 2016;11:e0148827.
  29. Zhang H, Meng J, Zhou S, Liu Y, Qu D, Wang L, et al. Intranasal delivery of exendin-4 confers neuroprotective effect against cerebral ischemia in mice. AAPS J 2016;18:385-94.
  30. Parthsarathy V, Hölscher C. The type 2 diabetes drug liraglutide reduces chronic inflammation induced by irradiation in the mouse brain. Eur J Pharmacol 2013;700:42-50.
  31. McClean PL, Gault VA, Harriott P, Hölscher C. Glucagonlike peptide-1 analogues enhance synaptic plasticity in the brain: a link between diabetes and Alzheimer's disease. Eur J Pharmacol 2010;630:158-62.
  32. Hamilton A, Patterson S, Porter D, Gault VA, Holscher C. Novel GLP-1 mimetics developed to treat type 2 diabetes promote progenitor cell proliferation in the brain. J Neurosci Res 2011;89:481-9.
  33. Secher A, Jelsing J, Baquero AF, Hecksher-Sørensen J, Cowley MA, Dalbøge LS, et al. The arcuate nucleus mediates GLP-1 receptor agonist liraglutide-dependent weight loss. J Clin Invest 2014;124:4473-88.
  34. Vella A, Shah P, Reed AS, Adkins AS, Basu R, Rizza RA. Lack of effect of exendin-4 and glucagon-like peptide1-(7,36)-amide on insulin action in non-diabetic humans. Diabetologia 2002;45:1410-5.
  35. Fuchs H, Binder R, Greischel A. Tissue distribution of the novel DPP-4 inhibitor BI 1356 is dominated by saturable binding to its target in rats. Biopharm Drug Dispos 2009;30:229-40.
  36. Gumuslu E, Mutlu O, Celikyurt IK, Ulak G, Akar F, Erden F, et al. Exenatide enhances cognitive performance and upregulates neurotrophic factor gene expression levels in diabetic mice. Fundam Clin Pharmacol 2016;30:376-84.
  37. Batista AF, Bodart-Santos V, De Felice FG, Ferreira ST. Neuroprotective actions of glucagon-like peptide-1 (GLP-1) analogues in Alzheimer's and Parkinson's diseases. CNS Drugs 2019;33:209-23.
  38. Hölscher C. Potential role of glucagon-like peptide-1 (GLP-1) in neuroprotection. CNS Drugs 2012;26:871-82.
  39. 2020 Alzheimer's disease facts and figures. Alzheimers Dement 2020.
  40. Cummings JL, Morstorf T, Zhong K. Alzheimer's disease drug-development pipeline: Few candidates, frequent failures. Alzheimers Res Ther 2014;6:37.
  41. Darsalia V, Mansouri S, Ortsäter H, Olverling A, Nozadze N, Kappe C, et al. Glucagon-like peptide-1 receptor activation reduces ischaemic brain damage following stroke in type 2 diabetic rats. Clin Sci (Lond) 2012;122:473-83.
  42. Teramoto S, Miyamoto N, Yatomi K, Tanaka Y, Oishi H, Arai H, et al. Exendin-4, a glucagon-like peptide-1 receptor agonist, provides neuroprotection in mice transient focal cerebral ischemia. J Cereb Blood Flow Metab 2011;31:1696-705.
  43. Klunk WE, Mathis CA, Price JC, Lopresti BJ, DeKosky ST. Two-year follow-up of amyloid deposition in patients with Alzheimer's disease. Brain 2006;129:2805-7.
  44. Femminella GD, Frangou E, Love SB, Busza G, Holmes C, Ritchie C, et al. Evaluating the effects of the novel GLP-1 analogue liraglutide in Alzheimer's disease: Study protocol for a randomised controlled trial (ELAD study). Trials 2019;20:191.
  45. Opara J, Małecki A, Małecka E, Socha T. Motor assessment in Parkinson`s disease. Ann Agric Environ Med 2017;24:411-5.
  46. Cabreira V, Massano J. Parkinson's disease: Clinical review and update. Acta Med Port 2019;32:661-70. Portuguese.
  47. Hu G, Jousilahti P, Bidel S, Antikainen R, Tuomilehto J. Type 2 diabetes and the risk of Parkinson's disease. Diabetes Care 2007;30:842-7.
  48. Lima MM, Targa AD, Noseda AC, Rodrigues LS, Delattre AM, dos Santos FV, et al. Does Parkinson's disease and type-2 diabetes mellitus present common pathophysiological mechanisms and treatments? CNS Neurol Disord Drug Targets 2014;13:418-28.
  49. Hogg E, Athreya K, Basile C, Tan EE, Kaminski J, Tagliati M. High prevalence of undiagnosed insulin resistance in non-diabetic subjects with Parkinson's disease. J Parkinsons Dis 2018;8:259-65.
  50. Wang SY, Wu SL, Chen TC, Chuang CS. Antidiabetic agents for treatment of Parkinson's disease: A metaanalysis. Int J Environ Res Public Health 2020;17:4805.
  51. Ramaker C, Marinus J, Stiggelbout AM, Van Hilten BJ. Systematic evaluation of rating scales for impairment and disability in Parkinson's disease. Mov Disord 2002;17:867-76.
  52. Yun SP, Kam TI, Panicker N, Kim S, Oh Y, Park JS, et al. Block of A1 astrocyte conversion by microglia is neuroprotective in models of Parkinson's disease. Nat Med 2018;24:931-8.
  53. Zhang L, Zhang L, Li L, Hölscher C. Neuroprotective effects of the novel GLP-1 long acting analogue semaglutide in the MPTP Parkinson's disease mouse model. Neuropeptides 2018;71:70-80.
  54. Mulvaney CA, Duarte GS, Handley J, Evans DJ, Menon S, Wyse R, et al. GLP-1 receptor agonists for Parkinson's disease. Cochrane Database Syst Rev 2020;7:CD012990.
  55. Feigin VL, Forouzanfar MH, Krishnamurthi R, Mensah GA, Connor M, Bennett DA, et al. Global and regional burden of stroke during 1990-2010: Findings from the Global Burden of Disease Study 2010. Lancet 2014;383:245-54.
  56. Wardlaw JM, Murray V, Berge E, del Zoppo G, Sandercock P, Lindley RL, et al. Recombinant tissue plasminogen activator for acute ischaemic stroke: An updated systematic review and meta-analysis. Lancet 2012;379:2364-72.
  57. Yang D, Nakajo Y, Iihara K, Kataoka H, Yanamoto H. Alogliptin, a dipeptidylpeptidase-4 inhibitor, for patients with diabetes mellitus type 2, induces tolerance to focal cerebral ischemia in non-diabetic, normal mice. Brain Res 2013;1517:104-13.
  58. Mansur RB, Fries GR, Trevizol AP, Subramaniapillai M, Lovshin J, Lin K, et al. The effect of body mass index on glucagon-like peptide receptor gene expression in the post mortem brain from individuals with mood and psychotic disorders. Eur Neuropsychopharmacol 2019;29:137-46.
  59. Kappe C, Tracy LM, Patrone C, Iverfeldt K, Sjöholm Å. GLP-1 secretion by microglial cells and decreased CNS expression in obesity. J Neuroinflammation 2012;9:276.
  60. Fulton B, Goa KL. Olanzapine. A review of its pharmacological properties and therapeutic efficacy in the management of schizophrenia and related psychoses. Drugs 1997;53:281-98.
  61. Siskind D, Hahn M, Correll CU, Fink-Jensen A, Russell AW, Bak N, et al. Glucagon-like peptide-1 receptor agonists for antipsychotic-associated cardio-metabolic risk factors: A systematic review and individual participant data meta-analysis. Diabetes Obes Metab 2019;21:293- 302.
  62. Crump C, Sundquist K, Winkleby MA, Sundquist J. Comorbidities and mortality in bipolar disorder: A Swedish national cohort study. JAMA Psychiatry 2013;70:931-9.
  63. Gomes FA, Almeida KM, Magalhães PV, Caetano SC, Kauer-Sant'Anna M, Lafer B, et al. Cardiovascular risk factors in outpatients with bipolar disorder: A report from the Brazilian Research Network in Bipolar Disorder. Braz J Psychiatry 2013;35:126-30.
  64. Perugi G, Quaranta G, Belletti S, Casalini F, Mosti N, Toni C, et al. General medical conditions in 347 bipolar disorder patients: Clinical correlates of metabolic and autoimmune-allergic diseases. J Affect Disord 2015;170:95-103.
  65. Mansur RB, Brietzke E, McIntyre RS. Is there a "metabolicmood syndrome"? A review of the relationship between obesity and mood disorders. Neurosci Biobehav Rev 2015;52:89-104.
  66. Anderson RJ, Freedland KE, Clouse RE, Lustman PJ. The prevalence of comorbid depression in adults with diabetes: A meta-analysis. Diabetes Care 2001;24:1069-78.
  67. Carnethon MR, Kinder LS, Fair JM, Stafford RS, Fortmann SP. Symptoms of depression as a risk factor for incident diabetes: Findings from the National Health and Nutrition Examination Epidemiologic Follow-up Study, 1971-1992. Am J Epidemiol 2003;158:416-23.
  68. Vancampfort D, Mitchell AJ, De Hert M, Sienaert P, Probst M, Buys R, et al. Prevalence and predictors of type 2 diabetes mellitus in people with bipolar disorder: A systematic review and meta-analysis. J Clin Psychiatry 2015;76:1490-9.
  69. Hendrick V, Altshuler L, Whybrow P. Psychoneuroendocrinology of mood disorders. The hypothalamic-pituitary-thyroid axis. Psychiatr Clin North Am 1998;21:277-92.
  70. Calkin CV, Ruzickova M, Uher R, Hajek T, Slaney CM, Garnham JS, et al. Insulin resistance and outcome in bipolar disorder. Br J Psychiatry 2015;206:52-7.
  71. Goldstein BI, Liu SM, Schaffer A, Sala R, Blanco C. Obesity and the three-year longitudinal course of bipolar disorder. Bipolar Disord 2013;15:284-93.
  72. Mansur RB, Rizzo LB, Santos CM, Asevedo E, Cunha GR, Noto MN, et al. Adipokines, metabolic dysfunction and illness course in bipolar disorder. J Psychiatr Res 2016;74:63-9.
  73. Mansur RB, Ahmed J, Cha DS, Woldeyohannes HO, Subramaniapillai M, Lovshin J, et al. Liraglutide promotes improvements in objective measures of cognitive dysfunction in individuals with mood disorders: A pilot, open-label study. J Affect Disord 2017;207:114-20.
  74. Ishøy PL, Fagerlund B, Broberg BV, Bak N, Knop FK, Glenthøj BY, et al. No cognitive-enhancing effect of GLP-1 receptor agonism in antipsychotic-treated, obese patients with schizophrenia. Acta Psychiatr Scand 2017;136:52-62.
  75. Marcus MD, Wildes JE. Obesity: Is it a mental disorder? Int J Eat Disord 2009;42:739-53.
  76. Hsu TM, Noble EE, Liu CM, Cortella AM, Konanur VR, Suarez AN, et al. A hippocampus to prefrontal cortex neural pathway inhibits food motivation through glucagon-like peptide-1 signaling. Mol Psychiatry 2018;23:1555-65.
  77. Robert SA, Rohana AG, Shah SA, Chinna K, Wan Mohamud WN, Kamaruddin NA. Improvement in binge eating in non-diabetic obese individuals after 3 months of treatment with liraglutide - A pilot study. Obes Res Clin Pract 2015;9:301-4.
  78. Meyer-Gerspach AC, Ly HG, Borgwardt S, Dupont P, Beglinger C, Van Oudenhove L, et al. Endogenous GLP-1 alters postprandial functional connectivity between homeostatic and reward-related brain regions involved in regulation of appetite in healthy lean males: A pilotstudy. Diabetes Obes Metab 2018;20:2330-8.
  79. Rigamonti AE, Piscitelli F, Aveta T, Agosti F, De Col A, Bini S, et al. Anticipatory and consummatory effects of (hedonic) chocolate intake are associated with increased circulating levels of the orexigenic peptide ghrelin and endocannabinoids in obese adults. Food Nutr Res 2015;59:29678.
  80. Battle DE. Diagnostic and Statistical Manual of Mental Disorders (DSM). Codas 2013;25:191-2.
  81. Da Porto A, Casarsa V, Colussi G, Catena C, Cavarape A, Sechi L. Dulaglutide reduces binge episodes in type 2 diabetic patients with binge eating disorder: A pilot study. Diabetes Metab Syndr 2020;14:289-92.
  82. van Bloemendaal L, Veltman DJ, ten Kulve JS, Drent ML, Barkhof F, Diamant M, et al. Emotional eating is associated with increased brain responses to food-cues and reduced sensitivity to GLP-1 receptor activation. Obesity (Silver Spring) 2015;23:2075-82.
  83. Mansur RB, Zugman A, Ahmed J, Cha DS, Subramaniapillai M, Lee Y, et al. Treatment with a GLP-1R agonist over four weeks promotes weight loss-moderated changes in frontal-striatal brain structures in individuals with mood disorders. Eur Neuropsychopharmacol 2017;27:1153-62.
  84. Dossat AM, Bodell LP, Williams DL, Eckel LA, Keel PK. Preliminary examination of glucagon-like peptide-1 levels in women with purging disorder and bulimia nervosa. Int J Eat Disord 2015;48:199-205.
  85. Naessén S, Carlström K, Holst JJ, Hellström PM, Hirschberg AL. Women with bulimia nervosa exhibit attenuated secretion of glucagon-like peptide 1, pancreatic polypeptide, and insulin in response to a meal. Am J Clin Nutr 2011;94:967-72.
  86. McElroy SL, Mori N, Guerdjikova AI, Keck PE Jr. Would glucagon-like peptide-1 receptor agonists have efficacy in binge eating disorder and bulimia nervosa? A review of the current literature. Med Hypotheses 2018;111:90-3.
  87. Najjar S, Pearlman DM, Devinsky O, Najjar A, Zagzag D. Neurovascular unit dysfunction with blood-brain barrier hyperpermeability contributes to major depressive disorder: A review of clinical and experimental evidence. J Neuroinflammation 2013;10:142.
  88. Felger JC, Treadway MT. Inflammation effects on motivation and motor activity: Role of dopamine. Neuropsychopharmacology 2017;42:216-41.
  89. Hamilton JP, Farmer M, Fogelman P, Gotlib IH. Depressive rumination, the default-mode network, and the dark matter of clinical neuroscience. Biol Psychiatry 2015;78:224-30.
  90. Belmaker RH, Agam G. Major depressive disorder. N Engl J Med 2008;358:55-68.
  91. Rizvi SJ, Pizzagalli DA, Sproule BA, Kennedy SH. Assessing anhedonia in depression: Potentials and pitfalls. Neurosci Biobehav Rev 2016;65:21-35.
  92. Al-Harbi KS. Treatment-resistant depression: Therapeutic trends, challenges, and future directions. Patient Prefer Adherence 2012;6:369-88.
  93. Yapici-Eser H, Appadurai V, Eren CY, Yazici D, Chen CY, Öngür D, et al. Association between GLP-1 receptor gene polymorphisms with reward learning, anhedonia and depression diagnosis. Acta Neuropsychiatr 2020;32:218-25.
  94. Sharma AN, Ligade SS, Sharma JN, Shukla P, Elased KM, Lucot JB. GLP-1 receptor agonist liraglutide reverses long-term atypical antipsychotic treatment associated behavioral depression and metabolic abnormalities in rats. Metab Brain Dis 2015;30:519-27.
  95. Kessler RC, Berglund P, Demler O, Jin R, Koretz D, Merikangas KR, et al. The epidemiology of major depressive disorder: Results from the National Comorbidity Survey Replication (NCS-R). JAMA 2003;289:3095-105.
  96. Rahman S, Alzarea S. Glial mechanisms underlying major depressive disorder: Potential therapeutic opportunities. Prog Mol Biol Transl Sci 2019;167:159-78.
  97. Tondo L, Vázquez GH, Baldessarini RJ. Depression and mania in bipolar disorder. Curr Neuropharmacol 2017;15:353-8.
  98. Merikangas KR, Jin R, He JP, Kessler RC, Lee S, Sampson NA, et al. Prevalence and correlates of bipolar spectrum disorder in the world mental health survey initiative. Arch Gen Psychiatry 2011;68:241-51.
  99. Ghaemi SN, Sachs GS, Chiou AM, Pandurangi AK, Goodwin K. Is bipolar disorder still underdiagnosed? Are antidepressants overutilized? J Affect Disord 1999;52:135-44.
  100. Renk K, White R, Lauer BA, McSwiggan M, Puff J, Lowell A. Bipolar disorder in children. Psychiatry J 2014;2014:928685.
  101. Barnett JH, Smoller JW. The genetics of bipolar disorder. Neuroscience 2009;164:331-43.
  102. Erşan S, Kurt A. Evaluation of glucagon-like peptide-1, adropin, and desnutrin levels and related factors in patients with bipolar disorder. Alpha Psychiatry 2021;22:1-6.
  103. Rosso G, Cattaneo A, Zanardini R, Gennarelli M, Maina G, Bocchio-Chiavetto L. Glucose metabolism alterations in patients with bipolar disorder. J Affect Disord 2015;184:293-8.
  104. Calkin CV, Gardner DM, Ransom T, Alda M. The relationship between bipolar disorder and type 2 diabetes: More than just co-morbid disorders. Ann Med 2013;45:171-81.
  105. Möller C, Sommer W, Thorsell A, Rimondini R, Heilig M. Anxiogenic-like action of centrally administered glucagon-like peptide-1 in a punished drinking test. Prog Neuropsychopharmacol Biol Psychiatry 2002;26:119-22.
  106. Weina H, Yuhu N, Christian H, Birong L, Feiyu S, Le W. Liraglutide attenuates the depressive- and anxiety-like behaviour in the corticosterone induced depression model via improving hippocampal neural plasticity. Brain Res 2018;1694:55-62.
  107. Bouhlal S, Ellefsen KN, Sheskier MB, Singley E, Pirard S, Gorelick DA, et al. Acute effects of intravenous cocaine administration on serum concentrations of ghrelin, amylin, glucagon-like peptide-1, insulin, leptin and peptide YY and relationships with cardiorespiratory and subjective responses. Drug Alcohol Depend 2017;180:68-75.
  108. Egecioglu E, Engel JA, Jerlhag E. The glucagon-like peptide 1 analogue, exendin-4, attenuates the rewarding properties of psychostimulant drugs in mice. PLoS One 2013;8:e69010.
  109. Bornebusch AB, Fink-Jensen A, Wörtwein G, Seeley RJ, Thomsen M. Glucagon-like peptide-1 receptor agonist treatment does not reduce abuse-related effects of opioid drugs. eNeuro 2019;6:ENEURO.0443-18.2019.
  110. Suchankova P, Yan J, Schwandt ML, Stangl BL, Caparelli EC, Momenan R, et al. The glucagon-like peptide-1 receptor as a potential treatment target in alcohol use disorder: Evidence from human genetic association studies and a mouse model of alcohol dependence. Transl Psychiatry 2015;5:e583.
  111. Vallöf D, Kalafateli AL, Jerlhag E. Brain region specific glucagon-like peptide-1 receptors regulate alcohol-induced behaviors in rodents. Psychoneuroendocrinology 2019;103:284-95.
  112. Thomsen M, Dencker D, Wörtwein G, Weikop P, Egecioglu E, Jerlhag E, et al. The glucagon-like peptide 1 receptor agonist Exendin-4 decreases relapse-like drinking in socially housed mice. Pharmacol Biochem Behav 2017;160:14-20.
  113. Bergman AJ, Stevens C, Zhou Y, Yi B, Laethem M, De Smet M, et al. Pharmacokinetic and pharmacodynamic properties of multiple oral doses of sitagliptin, a dipeptidyl peptidase-IV inhibitor: A double-blind, randomized, placebo-controlled study in healthy male volunteers. Clin Ther 2006;28:55-72.