Dopamine in the Regulation of Glucose Homeostasis, Pathogenesis of Type 2 Diabetes, and Chronic Conditions of Impaired Dopamine Activity/Metabolism: Implication for Pathophysiological and Therapeutic Purposes
Abstract
:1. Background
2. The Effects of Dopamine on Pancreatic Islets and Insulin and Glucagon Secretion
3. Dopamine in the Pathogenesis and Treatment of Traditional Chronic Diabetes-Related Complications
Diabetes-Related Traditional Chronic Complication | Role of Dopamine | Effect | Rationale for Treatment (Dopamine Agonists or Levodopa) |
---|---|---|---|
Retinopathy [45,46,47,48,49,50,51,52,53,54,55] | Impaired intraretinal metabolism (deficiency) | Defective photoreceptor adaptation to light | Yes |
Chronic renal disease [56,57,58,59,60] | Impaired renal metabolism (glomerular filtration-depended reduction) | Dysregulation in water and natrium resorption; promotion of glomerular hyperfiltration; micro- and macroalbuminuria | Scanty evidence or negative results |
Neuropathy [64,65,66] | Defective axonal transport; impaired metabolism (accumulation due to inadequate conversion to noradrenaline?) | Implication for painful neuropathy | No (dopamine antagonists) |
Stroke [72,73,74,75,76,77,78] | Impaired cerebral metabolism (deficiency) | Loss of motivation, motor impairment, and pathogenic role in post-stroke neuropsychiatric disorder | Scanty evidence or negative results |
Cardiovascular diseases [61,62,63,64,65,66,67,68,69,70] | Impaired cardiac metabolism (accumulation due to inadequate conversion to noradrenaline?); striatal deficiency | Increased risk of heart failure, impaired coronary vasodilatation, cardiac autonomic neuropathy | Scanty evidence or negative results |
4. The Pathophysiological Link between Type 2 Diabetes and Chronic Disorders Characterized by Impaired Dopamine Activity/Metabolism
4.1. Parkinson’s Disease
4.2. Huntington’s Disease
4.3. Attention-Deficit/Hyperactivity Disorder
4.4. Addictions
Diseases and Conditions | Pathophysiological Mechanisms |
---|---|
Diabetes and Parkinson’s disease [85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125] |
|
Diabetes and Huntington’s disease [135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156,157] |
|
Diabetes and ADHD [161,162,163,164,165,166,167,168,169,170,171,172,173,174,175,176,177,178] |
|
Diabetes and addictions [191,192,193,194,195,196,197,198,199,200,201,202,203,204,205,206,207,208,209,210,211,212,213,214,215,216,217,218,219,220] |
|
5. Therapeutic Implications
List of Medications | Therapeutic Area | Approved for | Mechanism of Action | Beneficial Effects * | Detrimental Effects * |
---|---|---|---|---|---|
Metformin | Diabetology | Diabetes mellitus | AMPK activator | Enhancement of cellular energy metabolism, improvement of autophagy and mitochondrial performance and redox homeostasis, anti-inflammatory effect, reduction in β-secretase 1 expression (AMPK activation) [244,245,246,247,248,249,250] | Dose-dependent adverse effects (abdominal pain or discomfort, nausea, diarrhea), impaired intestinal adsorption of Vitamin B12 [251,252,253] |
Acarbose | Diabetology | Diabetes mellitus | Intestine α-glucosidase inhibitor | Reduction in synthesis of biomarkers associated with adverse outcomes (proinflammatory cytokines, microRNA 10a-5p) [254,255] | Adverse intestinal effects |
Pioglitazone | Diabetology | Diabetes mellitus | PPAR-γ agonism | Anti-inflammatory and anti-atherosclerotic properties, insulin-sensitizing effect, attenuation of neuroinflammation [264,265,266,267,268,269,270,271,272,273,274] | Weight gain, water and sodium retention, intensive monitoring, or contraindication in case of heart failure, renal insufficiency, and macular edema |
Gliptins | Diabetology | Diabetes mellitus | DPP-IV inhibitors | Suppression of NFkB, reduction in the expression of RAGE, anti-inflammatory and anti-apoptotic properties, enhancement of brain-derived neurotrophic factors, reinforcement of anti-oxidative systems, increase in striatal dopamine synthesis [281,282,283,284,285,286,287,288,289,290,291,292] | - |
GLP-1RAs | Diabetology | Diabetes mellitus | GLP-1 agonism | Contrasting nigrostriatal injury and promoting neurogenesis, improvement of neuroinflammation and neuronal metabolic activity [118,119,120,121,122,123,124,125] | Relevant weight loss and reduction in appetite, potential for retinal injury |
Gliflozins | Diabetology | Diabetes mellitus | SGLT2 inhibitors | Improvement of energy utilization, reduction in oxidative stress and neuroinflammation, improvement of endoplasmic stress and autophagy, potential for exogen USP30 inhibitor [310,311,312,313,314,315,316,317] | Potential risk of genitourinary infections, hypotension, dehydration, and rapid decline of renal function, especially in older patients |
Secretagogues (Sulphonylureas, Glinides) | Diabetology | Diabetes mellitus | K inward channel inhibitors | Relevant improvement in short-term glucose control [318] | High risk of hypoglycemia, short durability, lack of evidence of extra-glycemic benefits, increased risk of dementia [319] |
Cabergoline, Bromocriptine, Apomorphine, Pramipexole, Rotigotine | Endocrinology/ Neurology | Diabetes mellitus Parkinson’s disease | Dopamine agonism | Improved glucose control; improved motor symptoms; reduced oxidative stress; possible CV benefits [226,227,228,229,230,231,232,233,234,235] | Mitral valve damage; impulse control disorders; short-term efficacy |
Entacapone, Tolcapone, Opicapone | Neurology | Parkinson’s disease | COMT inhibitors | Not well established | - |
Rivastigmine | Neurology | Parkinson’s disease | Acetylcholinesterase inhibitor | Not well established | - |
Amantadine | Neurology | Parkinson’s disease | Dopamine enhancer | Suppression of glucagon synthesis and stimulation of insulin release in response to oral glucose load [320] | Hypoglycemia (?) |
Istradefylline | Neurology | Parkinson’s disease | Adenosine (A2A) receptor antagonists | Potential for relevant impairment of intestinal glucose absorption (amelioration of non-fasting glycemia) [321] | - |
Pimavanserin | Neurology | Parkinson’s disease | Serotonin (5-HT2A) receptor inverse agonism | Mitigation of appetite, delaying gastric emptying, weight loss [322] | - |
Safinamide | Neurology | Parkinson’s disease | MAO-B reversible inhibitor | - | Potential affection of insulin secretion, apoptosis of β-cells (hyperglycemia and risk of new-onset T2D) [323] |
Deutetrabenazine, Tetrabenazine | Neurology | Huntington’s disease | VMAT2 reversible inhibitors | Neutral effect on glucose and metabolic parameters [324] | Slight weight gain [325] |
Methylphenidate, Lisdexamfetamine, Atomoxetine | Psychiatry | ADHD (adults) | Noradrenaline and dopamine reuptake inhibitors | Antidepressant effect, significant improvement of eating disorders, weight loss, improved glucose control [326] | Gastrointestinal discomfort, weight loss, or inability to gain weight |
Buprenorphine | Neurology | Addictions | Opiate (mu) receptor partial agonism | Restriction of sugar consumption, caloric intake, and weight loss [327] | Constipation, nausea, and vomiting |
Lofexidine | Psychiatry | Addictions | Central adrenergic (α2) receptor agonism | - | Enhancement of glucagon secretion, reduction in insulin secretion, lipolysis, gluconeogenesis, hyperglycemia [328] |
Naltrexone | Psychiatry | Addictions | Opiate (mu) receptor antagonism | Relevant attenuation of impulsive eating and purging behaviors, weight loss, improvement of glucose control in patients with diabetes [329] | Constipation, nausea |
Buprenorphine/Naloxone | Psychiatry | Addictions | Combining partial agonism and antagonism on opiate (mu) receptor | Restriction of sugar consumption, caloric intake, and weight loss [327] | Constipation, nausea, and vomiting |
6. Conclusions and Future Perspectives
Author Contributions
Funding
Conflicts of Interest
Abbreviations
References
- Available online: https://pubchem.ncbi.nlm.nih.gov/compound/Dopamine (accessed on 1 August 2023).
- Daubner, S.C.; Le, T.; Wang, S. Tyrosine hydroxylase and regulation of dopamine synthesis. Arch. Biochem. Biophys. 2011, 508, 1–12. [Google Scholar] [CrossRef] [PubMed]
- Yang, S.; Boudier-Revéret, M.; Choo, Y.J.; Chang, M.C. Association between Chronic Pain and Alterations in the Mesolimbic Dopaminergic System. Brain Sci. 2020, 10, 701. [Google Scholar] [CrossRef]
- Yadav, S.K.; Prakash, J.; Chouhan, S.; Westfall, S.; Verma, M.; Singh, T.D.; Singh, S.P. Comparison of the neuroprotective potential of Mucuna pruriens seed extract with estrogen in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD mice model. Neurochem. Int. 2014, 65, 1–13. [Google Scholar] [CrossRef]
- Guzmán, F. The Four Dopamine Pathways Relevant to Antipsychotics Pharmacology. Available online: http://psychopharmacologyinstitute.com/antipsychotics-videos/dopamine-pathways-antipsychotics-pharmacology/ (accessed on 15 November 2016).
- Quickel, K.E., Jr.; Feldman, J.M.; Lebovitz, H.E. Enhancement of insulin secretion in adult onset diabetics by methysergide maleate: Evidence for an endogenous biogenic monoamine mechanism as a factor in the impaired insulin secretion in diabetes mellitus. J. Clin. Endocrinol. Metab. 1971, 33, 877–881. [Google Scholar] [CrossRef] [PubMed]
- Lundquist, I. Insulin secretion. Its regulation by monoamines and acid amyloglucosidase. Acta Physiol. Scand. Suppl. 1971, 372, 1–47. [Google Scholar]
- Lebovitz, H.E.; Feldman, J.M. Pancreatic biogenic amines and insulin secretion in health and disease. Fed. Proc. 1973, 32, 1797–1802. [Google Scholar] [PubMed]
- Gagliardino, J.J.; Iturriza, F.C.; Hernandez, R.E.; Zieher, L.M. Effect of catecholamines precursors on insulin secretion. Endocrinology 1970, 87, 823–825. [Google Scholar] [CrossRef] [PubMed]
- Quickel, K.E., Jr.; Feldman, J.M.; Lebovitz, H.E. Inhibition of insulin secretion by serotonin and dopamine: Species variation. Endocrinology 1971, 89, 1295–1302. [Google Scholar] [CrossRef]
- Ericson, L.E.; Håkanson, R.; Lundquist, I. Accumulation of dopamine in mouse pancreatic B-cells following injection of L-DOPA. Localization to secretory granules and inhibition of insulin secretion. Diabetologia 1977, 13, 117–124. [Google Scholar] [CrossRef] [PubMed]
- Itoh, M.; Furman, B.L.; Gerich, J.E. Dopaminergic suppression of pancreatic somatostatin secretion. Acta Endocrinol. 1982, 101, 56–61. [Google Scholar] [CrossRef] [PubMed]
- Jetton, T.L.; Liang, Y.; Cincotta, A.H. Systemic treatment with sympatholytic dopamine agonists improves aberrant β-cell hyperplasia and GLUT2, glucokinase, and insulin immunoreactive levels in ob/ob mice. Metabolism 2001, 50, 1377–1384. [Google Scholar] [CrossRef] [PubMed]
- Ustione, A.; Piston, D.W. Dopamine synthesis and D3 receptor activation in pancreatic β-cells regulates insulin secretion and intracellular [Ca(2+)] oscillations. Mol. Endocrinol. 2012, 26, 1928–1940. [Google Scholar] [CrossRef]
- Barnett, A.H.; Chapman, C.; Gailer, K.; Hayter, C.J. Effect of bromocriptine on maturity onset diabetes. Postgrad. Med. J. 1980, 56, 11–14. [Google Scholar] [CrossRef] [PubMed]
- Garcia Barrado, M.J.; Iglesias Osma, M.C.; Blanco, E.J.; Carretero Hernández, M.; Sánchez Robledo, V.; Catalano Iniesta, L.; Carrero, S.; Carretero, J. Dopamine modulates insulin release and is involved in the survival of rat pancreatic beta cells. PLoS ONE 2015, 10, e0123197. [Google Scholar] [CrossRef] [PubMed]
- Tomaschitz, A.; Ritz, E.; Kienreich, K.; Pieske, B.; März, W.; Boehm, B.O.; Drechsler, C.; Meinitzer, A.; Pilz, S. Circulating dopamine and C-peptide levels in fasting nondiabetic hypertensive patients: The Graz Endocrine Causes of Hypertension study. Diabetes Care 2012, 35, 1771–1773. [Google Scholar] [CrossRef]
- Simpson, N.; Maffei, A.; Freeby, M.; Burroughs, S.; Freyberg, Z.; Javitch, J.; Leibel, R.L.; Harris, P.E. Dopamine-mediated autocrine inhibitory circuit regulating human insulin secretion in vitro. Mol. Endocrinol. 2012, 26, 1757–1772. [Google Scholar] [CrossRef] [PubMed]
- Kopf, D.; Gilles, M.; Paslakis, G.; Medlin, F.; Lederbogen, F.; Lehnert, H.; Deuschle, M. Insulin secretion and sensitivity after single-dose amisulpride, olanzapine or placebo in young male subjects: Double blind, cross-over glucose clamp study. Pharmacopsychiatry 2012, 45, 223–228. [Google Scholar] [CrossRef] [PubMed]
- Maffei, A.; Segal, A.M.; Alvarez-Perez, J.C.; Garcia-Ocaña, A.; Harris, P.E. Anti-incretin, Anti-proliferative Action of Dopamine on β-Cells. Mol. Endocrinol. 2015, 29, 542–557. [Google Scholar] [CrossRef]
- Tavares, G.; Rosendo-Silva, D.; Simões, F.; Eickhoff, H.; Marques, D.; Sacramento, J.F.; Capucho, A.M.; Seiça, R.; Conde, S.V.; Matafome, P. Circulating Dopamine Is Regulated by Dietary Glucose and Controls Glucagon-like 1 Peptide Action in White Adipose Tissue. Int. J. Mol. Sci. 2023, 24, 2464. [Google Scholar] [CrossRef]
- Chien, H.Y.; Chen, S.M.; Li, W.C. Dopamine receptor agonists mechanism of actions on glucose lowering and their connections with prolactin actions. Front. Clin. Diabetes Healthc. 2023, 4, 935872. [Google Scholar] [CrossRef] [PubMed]
- Ganic, E.; Johansson, J.K.; Bennet, H.; Fex, M.; Artner, I. Islet-specific monoamine oxidase A and B expression depends on MafA transcriptional activity and is compromised in type 2 diabetes. Biochem. Biophys. Res. Commun. 2015, 468, 629–635. [Google Scholar] [CrossRef] [PubMed]
- Available online: https://www.ncbi.nlm.nih.gov/gene/389692 (accessed on 31 August 2023).
- Oetjen, E.; Blume, R.; Cierny, I.; Schlag, C.; Kutschenko, A.; Krätzner, R.; Stein, R.; Knepel, W. Inhibition of MafA transcriptional activity and human insulin gene transcription by interleukin-1beta and mitogen-activated protein kinase kinase kinase in pancreatic islet beta cells. Diabetologia 2007, 50, 1678–1687. [Google Scholar] [CrossRef] [PubMed]
- Schmidt, R.E.; Geller, D.M.; Johnson, E.M., Jr. Characterization of increased plasma dopamine-β-hydroxylase activity in rats with experimental diabetes. Diabetes 1981, 30, 416–423. [Google Scholar] [CrossRef]
- Hurst, J.H.; Nisula, B.C.; Stolk, J.M. Circulating dopamine-β-hydroxylase in the rat: Importance of altered disposal pathways in experimental diabetes. J. Pharmacol. Exp. Ther. 1982, 220, 108–114. [Google Scholar] [PubMed]
- Lozovsky, D.; Saller, C.F.; Kopin, I.J. Dopamine receptor binding is increased in diabetic rats. Science 1981, 214, 1031–1033. [Google Scholar] [CrossRef]
- Kwok, R.P.; Juorio, A.V. Concentration of striatal tyramine and dopamine metabolism in diabetic rats and effect of insulin administration. Neuroendocrinology 1986, 43, 590–596. [Google Scholar] [CrossRef] [PubMed]
- Kwok, R.P.; Walls, E.K.; Juorio, A.V. The concentration of dopamine, 5-hydroxytryptamine, and some of their acid metabolites in the brain of genetically diabetic rats. Neurochem. Res. 1985, 10, 611–616. [Google Scholar] [CrossRef] [PubMed]
- Chaudhry, S.; Bernardes, M.; Harris, P.E.; Maffei, A. Gastrointestinal dopamine as an anti-incretin and its possible role in bypass surgery as therapy for type 2 diabetes with associated obesity. Minerva Endocrinol. 2016, 41, 43–56. [Google Scholar]
- Leblanc, H.; Lachelin, G.C.; Abu-Fadil, S.; Yen, S.S. The effect of dopamine infusion on insulin and glucagon secretion in man. J. Clin. Endocrinol. Metab. 1977, 44, 196–198. [Google Scholar] [CrossRef]
- Pernet, A.; Hammond, V.A.; Blesa-Malpica, G.; Burrin, J.; Orskov, H.; Alberti, K.G.; Johnston, D.G. The metabolic effects of dopamine in man. Eur. J. Clin. Pharmacol. 1984, 26, 23–28. [Google Scholar] [CrossRef]
- Keck, F.S.; Foldenauer, A.; Wolf, C.F.; Zeller, G.; Meyerhoff, C.; Dolderer, M.; Loos, U.; Pfeiffer, E.F. Differential effects of dopamine on glucoregulatory hormones in rats. Diabetes Res. Clin. Pract. 1990, 8, 155–159. [Google Scholar] [CrossRef] [PubMed]
- Deshpande, A.D.; Harris-Hayes, M.; Schootman, M. Epidemiology of diabetes and diabetes-related complications. Phys. Ther. 2008, 88, 1254–1264. [Google Scholar] [CrossRef]
- Gaster, B.; Hirsch, I.B. The effects of improved glycemic control on complications in type 2 diabetes. Arch. Intern. Med. 1998, 158, 134–140. [Google Scholar] [CrossRef]
- Nørgaard, C.H.; Mosslemi, M.; Lee, C.J.; Torp-Pedersen, C.; Wong, N.D. The Importance and Role of Multiple Risk Factor Control in Type 2 Diabetes. Curr. Cardiol. Rep. 2019, 21, 35. [Google Scholar] [CrossRef] [PubMed]
- Monnier, L.; Colette, C.; Schlienger, J.L.; Bauduceau, B.; ROwens, D. Glucocentric risk factors for macrovascular complications in diabetes: Glucose ‘legacy’ and ‘variability’-what we see, know and try to comprehend. Diabetes Metab. 2019, 45, 401–408. [Google Scholar] [CrossRef]
- Ceriello, A.; Prattichizzo, F. Variability of risk factors and diabetes complications. Cardiovasc. Diabetol. 2021, 20, 101. [Google Scholar] [CrossRef]
- ElSayed, N.A.; Aleppo, G.; Aroda, V.R.; Bannuru, R.R.; Brown, F.M.; Bruemmer, D.; Collins, B.S.; Hilliard, M.E.; Isaacs, D.; Johnson, E.L.; et al. Addendum. 3. Prevention or Delay of Type 2 Diabetes and Associated Comorbidities: Standards of Care in Diabetes–2023. Diabetes Care 2023, 46 (Suppl. S1), S41–S48, Erratum in Diabetes Care 2023, 46, 1716–1717. [Google Scholar] [CrossRef] [PubMed]
- Davies, M.J.; Drexel, H.; Jornayvaz, F.R.; Pataky, Z.; Seferović, P.M.; Wanner, C. Cardiovascular outcomes trials: A paradigm shift in the current management of type 2 diabetes. Cardiovasc. Diabetol. 2022, 21, 144. [Google Scholar] [CrossRef] [PubMed]
- Dardano, A.; Miccoli, R.; Bianchi, C.; Daniele, G.; Del Prato, S. Invited review. Series: Implications of the recent CVOTs in type 2 diabetes: Which patients for GLP-1RA or SGLT-2 inhibitor? Diabetes Res. Clin. Pract. 2020, 162, 108112. [Google Scholar] [CrossRef] [PubMed]
- Gasecka, A.; Siwik, D.; Gajewska, M.; Jaguszewski, M.J.; Mazurek, T.; Filipiak, K.J.; Postuła, M.; Eyileten, C. Early Biomarkers of Neurodegenerative and Neurovascular Disorders in Diabetes. J. Clin. Med. 2020, 9, 2807. [Google Scholar] [CrossRef] [PubMed]
- Popova, E. Role of Dopamine in Retinal Function. In Webvision: The Organization of the Retina and Visual System; Kolb, H., Fernandez, E., Nelson, R., Eds.; University of Utah Health Sciences Center: Salt Lake City, UT, USA, 1995. Available online: https://www.ncbi.nlm.nih.gov/books/NBK561740/ (accessed on 28 May 2020).
- Jain, V.; Liang, P.J.M.; Raja, S.; Mikhael, M.; Cameron, M.A. Light activation of the dopaminergic system occurs after eye-opening in the mouse retina. Front. Ophthalmol. 2023, 3, 1184627. [Google Scholar] [CrossRef]
- Wubben, T.J. Dopamine and Early Retinal Dysfunction in Diabetes: Insights From a Phase 1 Study. Diabetes 2020, 69, 1339–1340. [Google Scholar] [CrossRef] [PubMed]
- Allen, R.S.; Khayat, C.T.; Feola, A.J.; Win, A.S.; Grubman, A.R.; Chesler, K.C.; He, L.; Dixon, J.A.; Kern, T.S.; Iuvone, P.M.; et al. Diabetic rats with high levels of endogenous dopamine do not show retinal vascular pathology. Front. Neurosci. 2023, 17, 1125784. [Google Scholar] [CrossRef] [PubMed]
- Lee, Y.J.; Jeon, H.Y.; Lee, A.J.; Kim, M.; Ha, K.S. Dopamine ameliorates hyperglycemic memory-induced microvascular dysfunction in diabetic retinopathy. FASEB J. 2022, 36, e22643. [Google Scholar] [CrossRef] [PubMed]
- Upreti, S.; Sen, S.; Nag, T.C.; Ghosh, M.P. Insulin like growth factor-1 works synergistically with dopamine to attenuate diabetic retinopathy by downregulating vascular endothelial growth factor. Biomed. Pharmacother. 2022, 149, 112868. [Google Scholar] [CrossRef]
- Hendrick, A.; Smith, J.; Stelton, C.; Barb, S.; Yan, J.; Cribbs, B.; Jain, N.; Yeh, S.; Hubbard, G.B.; He, L.; et al. Dopamine metabolite levels in the vitreous of diabetic and non-diabetic humans. Exp. Eye Res. 2020, 195, 108040. [Google Scholar] [CrossRef]
- Motz, C.T.; Chesler, K.C.; Allen, R.S.; Bales, K.L.; Mees, L.M.; Feola, A.J.; Maa, A.Y.; Olson, D.E.; Thule, P.M.; Iuvone, P.M.; et al. Novel Detection and Restorative Levodopa Treatment for Preclinical Diabetic Retinopathy. Diabetes 2020, 69, 1518–1527. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Z.; Zhou, Y.; Zhao, H.; Xu, J.; Yang, X. Association Between Pathophysiological Mechanisms of Diabetic Retinopathy and Parkinson’s Disease. Cell. Mol. Neurobiol. 2022, 42, 665–675. [Google Scholar] [CrossRef]
- Chen, P.; Li, J.; Li, Z.; Yu, D.; Ma, N.; Xia, Z.; Meng, X.; Liu, X. 18F-FP-CIT dopamine transporter PET findings in the striatum and retina of type 1 diabetic rats. Ann. Nucl. Med. 2023, 37, 219–226. [Google Scholar] [CrossRef]
- Bethel, M.A.; Diaz, R.; Castellana, N.; Bhattacharya, I.; Gerstein, H.C.; Lakshmanan, M.C. HbA1c Change and Diabetic Retinopathy During GLP-1 Receptor Agonist Cardiovascular Outcome Trials: A Meta-analysis and Meta-regression. Diabetes Care 2021, 44, 290–296. [Google Scholar] [CrossRef]
- Kapoor, I.; Sarvepalli, S.M.; D’Alessio, D.; Grewal, D.S.; Hadziahmetovic, M. GLP-1 receptor agonists and diabetic retinopathy: A meta-analysis of randomized clinical trials. Surv. Ophthalmol. 2023, 68, 1071–1083. [Google Scholar] [CrossRef] [PubMed]
- Olivares-Hernández, A.; Figuero-Pérez, L.; Cruz-Hernandez, J.J.; González Sarmiento, R.; Usategui-Martin, R.; Miramontes-González, J.P. Dopamine Receptors and the Kidney: An Overview of Health- and Pharmacological-Targeted Implications. Biomolecules 2021, 11, 254. [Google Scholar] [CrossRef] [PubMed]
- Matsuyama, T.; Ohashi, N.; Ishigaki, S.; Isobe, S.; Tsuji, N.; Fujikura, T.; Tsuji, T.; Kato, A.; Miyajima, H.; Yasuda, H. The Relationship between the Intrarenal Dopamine System and Intrarenal Renin-angiotensin System Depending on the Renal Function. Intern. Med. 2018, 57, 3241–3247. [Google Scholar] [CrossRef] [PubMed]
- Hirose, M.; Tomoda, F.; Koike, T.; Yamazaki, H.; Ohara, M.; Liu, H.; Kagitani, S.; Inoue, H. Imbalance of renal production between 5-hydroxytryptamine and dopamine in patients with essential hypertension complicated by microalbuminuria. Am. J. Hypertens. 2013, 26, 227–233. [Google Scholar] [CrossRef] [PubMed]
- Zhang, M.Z.; Yao, B.; Yang, S.; Yang, H.; Wang, S.; Fan, X.; Yin, H.; Fogo, A.B.; Moeckel, G.W.; Harris, R.C. Intrarenal dopamine inhibits progression of diabetic nephropathy. Diabetes 2012, 61, 2575–2584. [Google Scholar] [CrossRef] [PubMed]
- Mejía-Rodríguez, O.; Herrera-Abarca, J.E.; Ceballos-Reyes, G.; Avila-Diaz, M.; Prado-Uribe, C.; Belio-Caro, F.; Salinas-González, A.; Vega-Gomez, H.; Alvarez-Aguilar, C.; Lindholm, B.; et al. Cardiovascular and renal effects of bromocriptine in diabetic patients with stage 4 chronic kidney disease. Biomed. Res. Int. 2013, 2013, 104059. [Google Scholar] [CrossRef] [PubMed]
- Neumann, J.; Hofmann, B.; Dhein, S.; Gergs, U. Role of Dopamine in the Heart in Health and Disease. Int. J. Mol. Sci. 2023, 24, 5042. [Google Scholar] [CrossRef] [PubMed]
- Lokhandwala, M.F.; Barrett, R.J. Cardiovascular dopamine receptors: Physiological, pharmacological and therapeutic implications. J. Auton. Pharmacol. 1982, 2, 189–215. [Google Scholar] [CrossRef]
- Neubauer, B.; Christensen, N.J. Norepinephrine, epinephrine, and dopamine contents of the cardiovascular system in long-term diabetics. Diabetes 1976, 25, 6–10. [Google Scholar] [CrossRef]
- Vinik, A.I.; Casellini, C.; Parson, H.K.; Colberg, S.R.; Nevoret, M.L. Cardiac Autonomic Neuropathy in Diabetes: A Predictor of Cardiometabolic Events. Front. Neurosci. 2018, 12, 591. [Google Scholar] [CrossRef]
- Vijayakumar, S.; Vaduganathan, M.; Butler, J. Glucose-Lowering Therapies and Heart Failure in Type 2 Diabetes Mellitus: Mechanistic Links, Clinical Data, and Future Directions. Circulation 2018, 137, 1060–1073. [Google Scholar] [CrossRef]
- Frishman, W.H.; Grewall, P. Serotonin and the heart. Ann. Med. 2000, 32, 195–209. [Google Scholar] [CrossRef]
- Mokhles, M.M.; Trifirò, G.; Dieleman, J.P.; Haag, M.D.; van Soest, E.M.; Verhamme, K.M.; Mazzaglia, G.; Herings, R.; de Luise, C.; Ross, D.; et al. The risk of new onset heart failure associated with dopamine agonist use in Parkinson’s disease. Pharmacol. Res. 2012, 65, 358–364. [Google Scholar] [CrossRef] [PubMed]
- Fouad Shalaby, M.A.; Abd El Latif, H.A.; El Yamani, M.; Galal, M.A.; Kamal, S.; Sindi, I.; Masaood, R. Therapeutic activity of sarpogrelate and dopamine D2 receptor agonists on cardiovascular and renal systems in rats with alloxan-induced diabetes. BMC Pharmacol. Toxicol. 2021, 22, 64. [Google Scholar] [CrossRef]
- Giamouzis, G.; Butler, J.; Starling, R.C.; Karayannis, G.; Nastas, J.; Parisis, C.; Rovithis, D.; Economou, D.; Savvatis, K.; Kirlidis, T.; et al. Impact of dopamine infusion on renal function in hospitalized heart failure patients: Results of the Dopamine in Acute Decompensated Heart Failure (DAD-HF) Trial. J. Card. Fail. 2010, 16, 922–930. [Google Scholar] [CrossRef] [PubMed]
- Triposkiadis, F.K.; Butler, J.; Karayannis, G.; Starling, R.C.; Filippatos, G.; Wolski, K.; Parissis, J.; Parisis, C.; Rovithis, D.; Koutrakis, K.; et al. Efficacy and safety of high dose versus low dose furosemide with or without dopamine infusion: The Dopamine in Acute Decompensated Heart Failure II (DAD-HF II) trial. Int. J. Cardiol. 2014, 172, 115–121. [Google Scholar] [CrossRef] [PubMed]
- Sharma, K.; Vaishnav, J.; Kalathiya, R.; Hu, J.R.; Miller, J.; Shah, N.; Hill, T.; Sharp, M.; Tsao, A.; Alexander, K.M.; et al. Randomized Evaluation of Heart Failure With Preserved Ejection Fraction Patients With Acute Heart Failure and Dopamine: The ROPA-DOP Trial. JACC Heart Fail. 2018, 6, 859–870. [Google Scholar] [CrossRef] [PubMed]
- Gower, A.; Tiberi, M. The Intersection of Central Dopamine System and Stroke: Potential Avenues Aiming at Enhancement of Motor Recovery. Front. Synaptic Neurosci. 2018, 10, 18. [Google Scholar] [CrossRef]
- Vitrac, C.; Nallet-Khosrofian, L.; Iijima, M.; Rioult-Pedotti, M.S.; Luft, A. Endogenous dopamine transmission is crucial for motor skill recovery after stroke. IBRO Neurosci. Rep. 2022, 13, 15–21. [Google Scholar] [CrossRef]
- Stinear, C.M. Dopamine for motor recovery after stroke: Where to from here? Lancet Neurol. 2019, 18, 514–515. [Google Scholar] [CrossRef] [PubMed]
- Sami, M.B.; Faruqui, R. The effectiveness of dopamine agonists for treatment of neuropsychiatric symptoms post brain injury and stroke. Acta Neuropsychiatr. 2015, 27, 317–326. [Google Scholar] [CrossRef] [PubMed]
- Villa, M.; Martínez-Vega, M.; Del Pozo, A.; Muneta-Arrate, I.; Gómez-Soria, A.; Muguruza, C.; de Hoz-Rivera, M.; Romero, A.; Silva, L.; Callado, L.F.; et al. The Role of the Dopamine System in Post-Stroke Mood Disorders in Newborn Rats. Int. J. Mol. Sci. 2023, 24, 3229. [Google Scholar] [CrossRef]
- Ford, G.A.; Bhakta, B.B.; Cozens, A.; Cundill, B.; Hartley, S.; Holloway, I.; Meads, D.; Pearn, J.; Ruddock, S.; Sackley, C.M.; et al. Dopamine Augmented Rehabilitation in Stroke (DARS): A multicentre double-blind, randomised controlled trial of co-careldopa compared with placebo, in addition to routine NHS occupational and physical therapy, delivered early after stroke on functional recovery. Effic. Mech. Eval. 2019, 6, 1–172. [Google Scholar]
- Scheidtmann, K.; Fries, W.; Müller, F.; Koenig, E. Effect of levodopa in combination with physiotherapy on functional motor recovery after stroke: A prospective, randomised, double-blind study. Lancet 2001, 358, 787–790. [Google Scholar] [CrossRef]
- Klein, M.O.; Battagello, D.S.; Cardoso, A.R.; Hauser, D.N.; Bittencourt, J.C.; Correa, R.G. Dopamine: Functions, Signaling, and Association with Neurological Diseases. Cell. Mol. Neurobiol. 2019, 39, 31–59. [Google Scholar] [CrossRef]
- Tysnes, O.B.; Storstein, A. Epidemiology of Parkinson’s disease. J. Neural Transm. 2017, 124, 901–905. [Google Scholar] [CrossRef]
- O’Reilly, S.; Loncin, M.; Cooksey, B. Dopamine and basal ganglia disorders. Neurology 1965, 15, 980–984. [Google Scholar] [CrossRef] [PubMed]
- Váradi, C. Clinical Features of Parkinson’s Disease: The Evolution of Critical Symptoms. Biology 2020, 9, 103. [Google Scholar] [CrossRef]
- Day, J.O.; Mullin, S. The Genetics of Parkinson’s Disease and Implications for Clinical Practice. Genes 2021, 12, 1006. [Google Scholar] [CrossRef] [PubMed]
- Simon, D.K.; Tanner, C.M.; Brundin, P. Parkinson Disease Epidemiology, Pathology, Genetics, and Pathophysiology. Clin. Geriatr. Med. 2020, 36, 1–12. [Google Scholar] [CrossRef]
- Chohan, H.; Senkevich, K.; Patel, R.K.; Bestwick, J.P.; Jacobs, B.M.; Bandres Ciga, S.; Gan-Or, Z.; Noyce, A.J. Type 2 Diabetes as a Determinant of Parkinson’s Disease Risk and Progression. Mov. Disord. 2021, 36, 1420–1429. [Google Scholar] [CrossRef] [PubMed]
- Komici, K.; Femminella, G.D.; Bencivenga, L.; Rengo, G.; Pagano, G. Diabetes Mellitus and Parkinson’s Disease: A Systematic Review and Meta-Analyses. J. Park. Dis. 2021, 11, 1585–1596. [Google Scholar] [CrossRef] [PubMed]
- Han, K.; Kim, B.; Lee, S.H.; Kim, M.K. A nationwide cohort study on diabetes severity and risk of Parkinson disease. NPJ Park. Dis. 2023, 9, 11. [Google Scholar] [CrossRef]
- Gallego, M.; Setién, R.; Izquierdo, M.J.; Casis, O.; Casis, E. Diabetes-induced biochemical changes in central and peripheral catecholaminergic systems. Physiol. Res. 2003, 52, 735–741. [Google Scholar] [CrossRef]
- Rocha, E.M.; De Miranda, B.; Sanders, L.H. Alpha-synuclein: Pathology, mitochondrial dysfunction and neuroinflammation in Parkinson’s disease. Neurobiol. Dis. 2018, 109, 249–257. [Google Scholar] [CrossRef] [PubMed]
- Dehay, B.; Bourdenx, M.; Gorry, P.; Przedborski, S.; Vila, M.; Hunot, S.; Singleton, A.; Olanow, C.W.; Merchant, K.M.; Bezard, E.; et al. Targeting α-synuclein for treatment of Parkinson’s disease: Mechanistic and therapeutic considerations. Lancet Neurol. 2015, 14, 855–866. [Google Scholar] [CrossRef]
- Meade, R.M.; Fairlie, D.P.; Mason, J.M. Alpha-synuclein structure and Parkinson’s disease—Lessons and emerging principles. Mol. Neurodegener. 2019, 14, 29. [Google Scholar] [CrossRef] [PubMed]
- Chen, R.; Gu, X.; Wang, X. α-Synuclein in Parkinson’s disease and advances in detection. Clin. Chim. Acta 2022, 529, 76–86. [Google Scholar] [CrossRef] [PubMed]
- Tofaris, G.K. Initiation and progression of α-synuclein pathology in Parkinson’s disease. Cell. Mol. Life Sci. 2022, 79, 210. [Google Scholar] [CrossRef]
- Marogianni, C.; Sokratous, M.; Dardiotis, E.; Hadjigeorgiou, G.M.; Bogdanos, D.; Xiromerisiou, G. Neurodegeneration and Inflammation-An Interesting Interplay in Parkinson’s Disease. Int. J. Mol. Sci. 2020, 21, 8421. [Google Scholar] [CrossRef] [PubMed]
- Asslih, S.; Damri, O.; Agam, G. Neuroinflammation as a Common Denominator of Complex Diseases (Cancer, Diabetes Type 2, and Neuropsychiatric Disorders). Int. J. Mol. Sci. 2021, 22, 6138. [Google Scholar] [CrossRef]
- Sarparanta, J.; García-Macia, M.; Singh, R. Autophagy and Mitochondria in Obesity and Type 2 Diabetes. Curr. Diabetes Rev. 2017, 13, 352–369. [Google Scholar] [CrossRef]
- Geng, X.; Lou, H.; Wang, J.; Li, L.; Swanson, A.L.; Sun, M.; Beers-Stolz, D.; Watkins, S.; Perez, R.G.; Drain, P. α-Synuclein binds the K(ATP) channel at insulin-secretory granules and inhibits insulin secretion. Am. J. Physiol. Endocrinol. Metab. 2011, 300, E276–E286. [Google Scholar] [CrossRef] [PubMed]
- Wijesekara, N.; Ahrens, R.; Wu, L.; Langman, T.; Tandon, A.; Fraser, P.E. α-Synuclein Regulates Peripheral Insulin Secretion and Glucose Transport. Front. Aging Neurosci. 2021, 13, 665348. [Google Scholar] [CrossRef]
- Vidal-Martinez, G.; Yang, B.; Vargas-Medrano, J.; Perez, R.G. Could α-Synuclein Modulation of Insulin and Dopamine Identify a Novel Link Between Parkinson’s Disease and Diabetes as Well as Potential Therapies? Front. Mol. Neurosci. 2018, 11, 465. [Google Scholar] [CrossRef]
- Yang, L.; Wang, H.; Liu, L.; Xie, A. The Role of Insulin/IGF-1/PI3K/Akt/GSK3β Signaling in Parkinson’s Disease Dementia. Front. Neurosci. 2018, 12, 73. [Google Scholar] [CrossRef] [PubMed]
- Tong, M.; Dong, M.; de la Monte, S.M. Brain insulin-like growth factor and neurotrophin resistance in Parkinson’s disease and dementia with Lewy bodies: Potential role of manganese neurotoxicity. J. Alzheimers Dis. 2009, 16, 585–599. [Google Scholar] [CrossRef]
- Bhattamisra, S.K.; Shin, L.Y.; Saad, H.I.B.M.; Rao, V.; Candasamy, M.; Pandey, M.; Choudhury, H. Interlink Between Insulin Resistance and Neurodegeneration with an Update on Current Therapeutic Approaches. CNS Neurol. Disord. Drug Targets 2020, 19, 174–183. [Google Scholar] [CrossRef] [PubMed]
- De Iuliis, A.; Montinaro, E.; Fatati, G.; Plebani, M.; Colosimo, C. Diabetes mellitus and Parkinson’s disease: Dangerous liaisons between insulin and dopamine. Neural Regen. Res. 2022, 17, 523–533. [Google Scholar] [CrossRef] [PubMed]
- Ramalingam, M.; Kim, S.J. Insulin on activation of autophagy with integrins and syndecans against MPP+-induced α-synuclein neurotoxicity. Neurosci. Lett. 2016, 633, 94–100. [Google Scholar] [CrossRef] [PubMed]
- Clark, I.A.; Vissel, B. Therapeutic implications of how TNF links apolipoprotein E, phosphorylated tau, α-synuclein, amyloid-β and insulin resistance in neurodegenerative diseases. Br. J. Pharmacol. 2018, 175, 3859–3875. [Google Scholar] [CrossRef] [PubMed]
- Ruiz-Pozo, V.A.; Tamayo-Trujillo, R.; Cadena-Ullauri, S.; Frias-Toral, E.; Guevara-Ramírez, P.; Paz-Cruz, E.; Chapela, S.; Montalván, M.; Morales-López, T.; Simancas-Racines, D.; et al. The Molecular Mechanisms of the Relationship between Insulin Resistance and Parkinson’s Disease Pathogenesis. Nutrients 2023, 15, 3585. [Google Scholar] [CrossRef] [PubMed]
- Greig, N.H.; Mattson, M.P.; Perry, T.; Chan, S.L.; Giordano, T.; Sambamurti, K.; Rogers, J.T.; Ovadia, H.; Lahiri, D.K. New therapeutic strategies and drug candidates for neurodegenerative diseases: p53 and TNF-alpha inhibitors, and GLP-1 receptor agonists. Ann. N. Y. Acad. Sci. 2004, 1035, 290–315. [Google Scholar] [CrossRef] [PubMed]
- Cabou, C.; Burcelin, R. GLP-1, the gut-brain, and brain-periphery axes. Rev. Diabet. Stud. 2011, 8, 418–431. [Google Scholar] [CrossRef] [PubMed]
- Katsurada, K.; Yada, T. Neural effects of gut- and brain-derived glucagon-like peptide-1 and its receptor agonist. J. Diabetes Investig. 2016, 7 (Suppl. S1), 64–69. [Google Scholar] [CrossRef]
- Chen, X.Y.; Chen, L.; Yang, W.; Xie, A.M. GLP-1 Suppresses Feeding Behaviors and Modulates Neuronal Electrophysiological Properties in Multiple Brain Regions. Front. Mol. Neurosci. 2021, 14, 793004. [Google Scholar] [CrossRef] [PubMed]
- Baggio, L.L.; Drucker, D.J. Glucagon-like peptide-1 receptors in the brain: Controlling food intake and body weight. J. Clin. Investig. 2014, 124, 4223–4226. [Google Scholar] [CrossRef] [PubMed]
- Harkavyi, A.; Abuirmeileh, A.; Lever, R.; Kingsbury, A.E.; Biggs, C.S.; Whitton, P.S. Glucagon-like peptide 1 receptor stimulation reverses key deficits in distinct rodent models of Parkinson’s disease. J. Neuroinflammation 2008, 5, 19. [Google Scholar] [CrossRef]
- Bertilsson, G.; Patrone, C.; Zachrisson, O.; Andersson, A.; Dannaeus, K.; Heidrich, J.; Kortesmaa, J.; Mercer, A.; Nielsen, E.; Rönnholm, H.; et al. Peptide hormone exendin-4 stimulates subventricular zone neurogenesis in the adult rodent brain and induces recovery in an animal model of Parkinson’s disease. J. Neurosci. Res. 2008, 86, 326–338. [Google Scholar] [CrossRef]
- Rachmany, L.; Tweedie, D.; Li, Y.; Rubovitch, V.; Holloway, H.W.; Miller, J.; Hoffer, B.J.; Greig, N.H.; Pick, C.G. Exendin-4 induced glucagon-like peptide-1 receptor activation reverses behavioral impairments of mild traumatic brain injury in mice. Age 2013, 35, 1621–1636. [Google Scholar] [CrossRef] [PubMed]
- 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. [Google Scholar] [CrossRef] [PubMed]
- Femminella, G.D.; Edison, P. Evaluation of neuroprotective effect of glucagon-like peptide 1 analogs using neuroimaging. Alzheimers Dement. 2014, 10 (Suppl. S1), S55–S61. [Google Scholar] [CrossRef] [PubMed]
- Simuni, T.; Brundin, P. Is exenatide the next big thing in Parkinson’s disease? J. Park. Dis. 2014, 4, 345–347. [Google Scholar] [CrossRef] [PubMed]
- Aviles-Olmos, I.; Dickson, J.; Kefalopoulou, Z.; Djamshidian, A.; Ell, P.; Soderlund, T.; Whitton, P.; Wyse, R.; Isaacs, T.; Lees, A.; et al. Exenatide and the treatment of patients with Parkinson’s disease. J. Clin. Investig. 2013, 123, 2730–2736. [Google Scholar] [CrossRef] [PubMed]
- Aviles-Olmos, I.; Dickson, J.; Kefalopoulou, Z.; Djamshidian, A.; Kahan, J.; Ell, P.; Whitton, P.; Wyse, R.; Isaacs, T.; Lees, A.; et al. Motor and cognitive advantages persist 12 months after exenatide exposure in Parkinson’s disease. J. Park. Dis. 2014, 4, 337–344. [Google Scholar] [CrossRef]
- Athauda, D.; Maclagan, K.; Skene, S.S.; Bajwa-Joseph, M.; Letchford, D.; Chowdhury, K.; Hibbert, S.; Budnik, N.; Zampedri, L.; Dickson, J.; et al. Exenatide once weekly versus placebo in Parkinson’s disease: A randomised, double-blind, placebo-controlled trial. Lancet 2017, 390, 1664–1675. [Google Scholar] [CrossRef]
- Hölscher, C. Novel dual GLP-1/GIP receptor agonists show neuroprotective effects in Alzheimer’s and Parkinson’s disease models. Neuropharmacology 2018, 136, 251–259. [Google Scholar] [CrossRef]
- Reich, N.; Hölscher, C. The neuroprotective effects of glucagon-like peptide 1 in Alzheimer’s and Parkinson’s disease: An in-depth review. Front. Neurosci. 2022, 16, 970925. [Google Scholar] [CrossRef]
- Cheng, D.; Yang, S.; Zhao, X.; Wang, G. The Role of Glucagon-Like Peptide-1 Receptor Agonists (GLP-1 RA) in Diabetes-Related Neurodegenerative Diseases. Drug Des. Dev. Ther. 2022, 16, 665–684. [Google Scholar] [CrossRef]
- Chen, S.D.; Chuang, Y.C.; Lin, T.K.; Yang, J.L. Alternative role of glucagon-like Peptide-1 receptor agonists in neurodegenerative diseases. Eur. J. Pharmacol. 2023, 938, 175439. [Google Scholar] [CrossRef] [PubMed]
- Nowell, J.; Blunt, E.; Gupta, D.; Edison, P. Antidiabetic agents as a novel treatment for Alzheimer’s and Parkinson’s disease. Ageing Res. Rev. 2023, 89, 101979. [Google Scholar] [CrossRef]
- Andhale, R.; Shrivastava, D. Huntington’s Disease: A Clinical Review. Cureus 2022, 14, e28484. [Google Scholar] [CrossRef] [PubMed]
- Schulte, J.; Littleton, J.T. The biological function of the Huntingtin protein and its relevance to Huntington’s Disease pathology. Curr. Trends Neurol. 2011, 5, 65–78. [Google Scholar]
- Roos, R.A. Huntington’s disease: A clinical review. Orphanet J. Rare Dis. 2010, 5, 40. [Google Scholar] [CrossRef]
- McColgan, P.; Tabrizi, S.J. Huntington’s disease: A clinical review. Eur. J. Neurol. 2018, 25, 24–34. [Google Scholar] [CrossRef] [PubMed]
- Albin, R.L.; Reiner, A.; Anderson, K.D.; Dure, L.S., 4th; Handelin, B.; Balfour, R.; Whetsell, W.O., Jr.; Penney, J.B.; Young, A.B. Preferential loss of striato-external pallidal projection neurons in presymptomatic Huntington’s disease. Ann. Neurol. 1992, 31, 425–430. [Google Scholar] [CrossRef] [PubMed]
- Bird, E.D. Chemical pathology of Huntington’s disease. Annu. Rev. Pharmacol. Toxicol. 1980, 20, 533–551. [Google Scholar] [CrossRef]
- Glass, M.; Dragunow, M.; Faull, R.L. The pattern of neurodegeneration in Huntington’s disease: A comparative study of cannabinoid, dopamine, adenosine and GABA(A) receptor alterations in the human basal ganglia in Huntington’s disease. Neuroscience 2000, 97, 505–519. [Google Scholar] [CrossRef] [PubMed]
- Bibb, J.A.; Yan, Z.; Svenningsson, P.; Snyder, G.L.; Pieribone, V.A.; Horiuchi, A.; Nairn, A.C.; Messer, A.; Greengard, P. Severe deficiencies in dopamine signaling in presymptomatic Huntington’s disease mice. Proc. Natl. Acad. Sci. USA 2000, 97, 6809–6814. [Google Scholar] [CrossRef]
- Jakel, R.J.; Maragos, W.F. Neuronal cell death in Huntington’s disease: A potential role for dopamine. Trends Neurosci. 2000, 23, 239–245. [Google Scholar] [CrossRef] [PubMed]
- Kumar, P.; Kalonia, H.; Kumar, A. Huntington’s disease: Pathogenesis to animal models. Pharmacol. Rep. 2010, 62, 1–14. [Google Scholar] [CrossRef] [PubMed]
- Illarioshkin, S.N.; Klyushnikov, S.A.; Vigont, V.A.; Seliverstov, Y.A.; Kaznacheyeva, E.V. Molecular Pathogenesis in Huntington’s Disease. Biochemistry 2018, 83, 1030–1039. [Google Scholar] [CrossRef] [PubMed]
- Wright, D.J.; Renoir, T.; Gray, L.J.; Hannan, A.J. Huntington’s Disease: Pathogenic Mechanisms and Therapeutic Targets. Adv. Neurobiol. 2017, 15, 93–128. [Google Scholar] [CrossRef]
- Farrer, L.A. Diabetes mellitus in Huntington disease. Clin. Genet. 1985, 27, 62–67. [Google Scholar] [CrossRef] [PubMed]
- Lee, C.Y.; Ro, J.S.; Jung, H.; Kim, M.; Jeon, B.; Lee, J.Y. Increased 10-Year Prevalence of Huntington’s Disease in South Korea: An Analysis of Medical Expenditure Through the National Healthcare System. J. Clin. Neurol. 2023, 19, 147–155. [Google Scholar] [CrossRef]
- Montojo, M.T.; Aganzo, M.; González, N. Huntington’s Disease and Diabetes: Chronological Sequence of its Association. J. Huntingt. Dis. 2017, 6, 179–188. [Google Scholar] [CrossRef] [PubMed]
- McDonald, T.S.; Lerskiatiphanich, T.; Woodruff, T.M.; McCombe, P.A.; Lee, J.D. Potential mechanisms to modify impaired glucose metabolism in neurodegenerative disorders. J. Cereb. Blood Flow Metab. 2023, 43, 26–43. [Google Scholar] [CrossRef]
- Podlacha, M.; Pierzynowska, K.; Gaffke, L.; Jerzemowska, G.; Piotrowska, E.; Węgrzyn, G. Behavioral- and blood-based biomarkers for Huntington’s disease: Studies on the R6/1 mouse model with prospects for early diagnosis and monitoring of the disease. Brain Behav. Immun. Health 2022, 23, 100482. [Google Scholar] [CrossRef] [PubMed]
- Li, L.; Sun, Y.; Zhang, Y.; Wang, W.; Ye, C. Mutant Huntingtin Impairs Pancreatic β-cells by Recruiting IRS-2 and Disturbing the PI3K/AKT/FoxO1 Signaling Pathway in Huntington’s Disease. J. Mol. Neurosci. 2021, 71, 2646–2658. [Google Scholar] [CrossRef] [PubMed]
- Ogilvie, A.C.; Gonzalez-Alegre, P.; Schultz, J.L. Diabetes Mellitus Is Associated With an Earlier Age of Onset of Huntington’s Disease. Mov. Disord. 2021, 36, 1033–1034. [Google Scholar] [CrossRef] [PubMed]
- Maiese, K. The Metabolic Basis for Nervous System Dysfunction in Alzheimer’s Disease, Parkinson’s Disease, and Huntington’s Disease. Curr. Neurovascular Res. 2023, 20, 314. [Google Scholar] [CrossRef] [PubMed]
- Dowling, R.J.; Topisirovic, I.; Fonseca, B.D.; Sonenberg, N. Dissecting the role of mTOR: Lessons from mTOR inhibitors. Biochim. Biophys. Acta 2010, 1804, 433–439. [Google Scholar] [CrossRef]
- Liu, Y.; Li, J.; Han, Y.; Chen, Y.; Liu, L.; Lang, J.; Yang, C.; Luo, H.; Ning, J. Advanced glycation end-products suppress autophagy by AMPK/mTOR signaling pathway to promote vascular calcification. Mol. Cell. Biochem. 2020, 471, 91–100. [Google Scholar] [CrossRef]
- Ramasubbu, K.; Devi Rajeswari, V. Impairment of insulin signaling pathway PI3K/Akt/mTOR and insulin resistance induced AGEs on diabetes mellitus and neurodegenerative diseases: A perspective review. Mol. Cell. Biochem. 2023, 478, 1307–1324. [Google Scholar] [CrossRef] [PubMed]
- Brás, I.C.; König, A.; Outeiro, T.F. Glycation in Huntington’s Disease: A Possible Modifier and Target for Intervention. J. Huntingt. Dis. 2019, 8, 245–256. [Google Scholar] [CrossRef] [PubMed]
- Vázquez-Manrique, R.P.; Farina, F.; Cambon, K.; Dolores Sequedo, M.; Parker, A.J.; Millán, J.M.; Weiss, A.; Déglon, N.; Neri, C. AMPK activation protects from neuronal dysfunction and vulnerability across nematode, cellular and mouse models of Huntington’s disease. Hum. Mol. Genet. 2016, 25, 1043–1058. [Google Scholar] [CrossRef] [PubMed]
- Hervás, D.; Fornés-Ferrer, V.; Gómez-Escribano, A.P.; Sequedo, M.D.; Peiró, C.; Millán, J.M.; Vázquez-Manrique, R.P. Metformin intake associates with better cognitive function in patients with Huntington’s disease. PLoS ONE 2017, 12, e0179283. [Google Scholar] [CrossRef] [PubMed]
- Sanchis, A.; García-Gimeno, M.A.; Cañada-Martínez, A.J.; Sequedo, M.D.; Millán, J.M.; Sanz, P.; Vázquez-Manrique, R.P. Metformin treatment reduces motor and neuropsychiatric phenotypes in the zQ175 mouse model of Huntington disease. Exp. Mol. Med. 2019, 51, 1–16. [Google Scholar] [CrossRef]
- Chang, C.C.; Lin, T.C.; Ho, H.L.; Kuo, C.Y.; Li, H.H.; Korolenko, T.A.; Chen, W.J.; Lai, T.J.; Ho, Y.J.; Lin, C.L. GLP-1 Analogue Liraglutide Attenuates Mutant Huntingtin-Induced Neurotoxicity by Restoration of Neuronal Insulin Signaling. Int. J. Mol. Sci. 2018, 19, 2505. [Google Scholar] [CrossRef]
- Shawki, S.M.; Saad, M.A.; Rahmo, R.M.; Wadie, W.; El-Abhar, H.S. Liraglutide Improves Cognitive and Neuronal Function in 3-NP Rat Model of Huntington’s Disease. Front. Pharmacol. 2021, 12, 731483. [Google Scholar] [CrossRef]
- Martin, B.; Golden, E.; Carlson, O.D.; Pistell, P.; Zhou, J.; Kim, W.; Frank, B.P.; Thomas, S.; Chadwick, W.A.; Greig, N.H.; et al. Exendin-4 improves glycemic control, ameliorates brain and pancreatic pathologies, and extends survival in a mouse model of Huntington’s disease. Diabetes 2009, 58, 318–328. [Google Scholar] [CrossRef] [PubMed]
- Sayed, N.H.; Fathy, N.; Kortam, M.A.; Rabie, M.A.; Mohamed, A.F.; Kamel, A.S. Correction to: Vildagliptin Attenuates Huntington’s Disease Through Activation of GLP-1 Receptor/PI3K/Akt/BDNF Pathway in 3-Nitropropionic Acid Rat Model. Neurotherapeutics 2022, 17, 252–268, Erratum in Neurotherapeutics 2022, 19, 686. [Google Scholar] [CrossRef] [PubMed]
- El-Sahar, A.E.; Rastanawi, A.A.; El-Yamany, M.F.; Saad, M.A. Dapagliflozin improves behavioral dysfunction of Huntington’s disease in rats via inhibiting apoptosis-related glycolysis. Life Sci. 2020, 257, 118076. [Google Scholar] [CrossRef] [PubMed]
- Koutsoklenis, A.; Honkasilta, J. ADHD in the DSM-5-TR: What has changed and what has not. Front. Psychiatry 2023, 13, 1064141. [Google Scholar] [CrossRef] [PubMed]
- Salari, N.; Ghasemi, H.; Abdoli, N.; Rahmani, A.; Shiri, M.H.; Hashemian, A.H.; Akbari, H.; Mohammadi, M. The global prevalence of ADHD in children and adolescents: A systematic review and meta-analysis. Ital. J. Pediatr. 2023, 49, 48. [Google Scholar] [CrossRef] [PubMed]
- Faraone, S.V.; Larsson, H. Genetics of attention deficit hyperactivity disorder. Mol. Psychiatry 2019, 24, 562–575. [Google Scholar] [CrossRef]
- Blum, K.; Chen, A.L.; Braverman, E.R.; Comings, D.E.; Chen, T.J.; Arcuri, V.; Blum, S.H.; Downs, B.W.; Waite, R.L.; Notaro, A.; et al. Attention-deficit-hyperactivity disorder and reward deficiency syndrome. Neuropsychiatr. Dis. Treat. 2008, 4, 893–918. [Google Scholar] [CrossRef] [PubMed]
- Cortese, S.; Moreira-Maia, C.R.; St Fleur, D.; Morcillo-Peñalver, C.; Rohde, L.A.; Faraone, S.V. Association Between ADHD and Obesity: A Systematic Review and Meta-Analysis. Am. J. Psychiatry 2016, 173, 34–43. [Google Scholar] [CrossRef] [PubMed]
- Mazor-Aronovitch, K.; Pinhas-Hamiel, O.; Pivko-Levy, D.; Modan-Moses, D.; Levek, N.; Miller, S.; Yackobovitch-Gavan, M.; Gruber, N.; Ben-Ami, M.; Stern, E.; et al. Dual diagnosis of type 1 diabetes mellitus and attention deficit hyperactivity disorder. Pediatr. Diabetes 2021, 22, 649–655. [Google Scholar] [CrossRef]
- Garcia-Argibay, M.; Li, L.; Du Rietz, E.; Zhang, L.; Yao, H.; Jendle, J.; Ramos-Quiroga, J.A.; Ribasés, M.; Chang, Z.; Brikell, I.; et al. The association between type 2 diabetes and attention- deficit/hyperactivity disorder: A systematic review, meta-analysis, and population-based sibling study. Neurosci. Biobehav. Rev. 2023, 147, 105076. [Google Scholar] [CrossRef]
- Kittel-Schneider, S.; Arteaga-Henriquez, G.; Vasquez, A.A.; Asherson, P.; Banaschewski, T.; Brikell, I.; Buitelaar, J.; Cormand, B.; Faraone, S.V.; Freitag, C.M.; et al. Non-mental diseases associated with ADHD across the lifespan: Fidgety Philipp and Pippi Longstocking at risk of multimorbidity? Neurosci. Biobehav. Rev. 2022, 132, 1157–1180. [Google Scholar] [CrossRef] [PubMed]
- Franz, A.P.; Bolat, G.U.; Bolat, H.; Matijasevich, A.; Santos, I.S.; Silveira, R.C.; Procianoy, R.S.; Rohde, L.A.; Moreira-Maia, C.R. Attention-Deficit/Hyperactivity Disorder and Very Preterm/Very Low Birth Weight: A Meta-analysis. Pediatrics 2018, 141, e20171645. [Google Scholar] [CrossRef] [PubMed]
- Garvey, W.T.; Mechanick, J.I.; Brett, E.M.; Garber, A.J.; Hurley, D.L.; Jastreboff, A.M.; Nadolsky, K.; Pessah-Pollack, R.; Plodkowski, R.; Reviewers of the AACE/ACE Obesity Clinical Practice Guidelines. American association of clinical endocrinologists and american college of endocrinology comprehensive clinical practice guidelines for Medical care of patients with obesity. Endocr. Pract. 2016, 22 (Suppl. S3), 1–203. [Google Scholar] [CrossRef]
- Villa, F.M.; Crippa, A.; Rosi, E.; Nobile, M.; Brambilla, P.; Delvecchio, G. ADHD and eating disorders in childhood and adolescence: An updated minireview. J. Affect. Disord. 2023, 321, 265–271. [Google Scholar] [CrossRef] [PubMed]
- Nazar, B.P.; Bernardes, C.; Peachey, G.; Sergeant, J.; Mattos, P.; Treasure, J. The risk of eating disorders comorbid with attention-deficit/hyperactivity disorder: A systematic review and meta-analysis. Int. J. Eat. Disord. 2016, 49, 1045–1057. [Google Scholar] [CrossRef]
- Catalá-López, F.; Hutton, B.; Núñez-Beltrán, A.; Page, M.J.; Ridao, M.; Macías Saint-Gerons, D.; Catalá, M.A.; Tabarés-Seisdedos, R.; Moher, D. The pharmacological and non-pharmacological treatment of attention deficit hyperactivity disorder in children and adolescents: A systematic review with network meta-analyses of randomised trials. PLoS ONE 2017, 12, e0180355. [Google Scholar] [CrossRef]
- Wang, G.J.; Volkow, N.D.; Fowler, J.S. The role of dopamine in motivation for food in humans: Implications for obesity. Expert. Opin. Ther. Targets 2002, 6, 601–609. [Google Scholar] [CrossRef]
- Danilovich, N.; Mastrandrea, L.D.; Cataldi, L.; Quattrin, T. Methylphenidate decreases fat and carbohydrate intake in obese teenagers. Obesity 2014, 22, 781–785. [Google Scholar] [CrossRef]
- Goldfield, G.S.; Lorello, C.; Doucet, E. Methylphenidate reduces energy intake and dietary fat intake in adults: A mechanism of reduced reinforcing value of food? Am. J. Clin. Nutr. 2007, 86, 308–315. [Google Scholar] [CrossRef]
- Liu, L.L.; Li, B.M.; Yang, J.; Wang, Y.W. Does dopaminergic reward system contribute to explaining comorbidity obesity and ADHD? Med. Hypotheses 2008, 70, 1118–1120. [Google Scholar] [CrossRef]
- Charach, G.; Karniel, E.; Grosskopf, I.; Rabinovich, A.; Charach, L. Methylphenidate has mild hyperglycemic and hypokalemia effects and increases leukocyte and neutrophil counts. Medicine 2020, 99, e20931. [Google Scholar] [CrossRef] [PubMed]
- Skibicka, K.P. The central GLP-1: Implications for food and drug reward. Front. Neurosci. 2013, 7, 181. [Google Scholar] [CrossRef] [PubMed]
- van Bloemendaal, L.; IJzerman, R.G.; Ten Kulve, J.S.; Barkhof, F.; Konrad, R.J.; Drent, M.L.; Veltman, D.J.; Diamant, M. GLP-1 receptor activation modulates appetite- and reward-related brain areas in humans. Diabetes 2014, 63, 4186–4196. [Google Scholar] [CrossRef] [PubMed]
- Anderberg, R.H.; Anefors, C.; Bergquist, F.; Nissbrandt, H.; Skibicka, K.P. Dopamine signaling in the amygdala, increased by food ingestion and GLP-1, regulates feeding behavior. Physiol. Behav. 2014, 136, 135–144. [Google Scholar] [CrossRef] [PubMed]
- Alavi, S.S.; Ferdosi, M.; Jannatifard, F.; Eslami, M.; Alaghemandan, H.; Setare, M. Behavioral Addiction versus Substance Addiction: Correspondence of Psychiatric and Psychological Views. Int. J. Prev. Med. 2012, 3, 290–294. [Google Scholar] [PubMed]
- Chen, C.Y.; Storr, C.L.; Anthony, J.C. Early-onset drug use and risk for drug dependence problems. Addict. Behav. 2009, 34, 319–322. [Google Scholar] [CrossRef]
- Haug, S.; Castro, R.P.; Kwon, M.; Filler, A.; Kowatsch, T.; Schaub, M.P. Smartphone use and smartphone addiction among young people in Switzerland. J. Behav. Addict. 2015, 4, 299–307. [Google Scholar] [CrossRef] [PubMed]
- Kramer, J.; Dick, D.M.; King, A.; Ray, L.A.; Sher, K.J.; Vena, A.; Vendruscolo, L.F.; Acion, L. Mechanisms of Alcohol Addiction: Bridging Human and Animal Studies. Alcohol Alcohol. 2020, 55, 603–607. [Google Scholar] [CrossRef]
- Caron, L.; Karkazis, K.; Raffin, T.A.; Swan, G.; Koenig, B.A. Nicotine addiction through a neurogenomic prism: Ethics, public health, and smoking. Nicotine Tob. Res. 2005, 7, 181–197. [Google Scholar] [CrossRef] [PubMed]
- Salsitz, E.A. Chronic Pain, Chronic Opioid Addiction: A Complex Nexus. J. Med. Toxicol. 2016, 12, 54–57. [Google Scholar] [CrossRef]
- Olsen, Y. What Is Addiction? History, Terminology, and Core Concepts. Med. Clin. N. Am. 2022, 106, 1–12. [Google Scholar] [CrossRef] [PubMed]
- Gardner, E.L. Addiction and brain reward and antireward pathways. Adv. Psychosom. Med. 2011, 30, 22–60. [Google Scholar] [CrossRef] [PubMed]
- Iovino, M.; Messana, T.; Lisco, G.; Mariano, F.; Giagulli, V.A.; Guastamacchia, E.; De Pergola, G.; Triggiani, V. Neuroendocrine Modulation of Food Intake and Eating Behavior. Endocr. Metab. Immune Disord. Drug Targets 2022, 22, 1252–1262. [Google Scholar] [CrossRef]
- Narayanan, N.S.; Guarnieri, D.J.; DiLeone, R.J. Metabolic hormones, dopamine circuits, and feeding. Front. Neuroendocrinol. 2010, 31, 104–112. [Google Scholar] [CrossRef] [PubMed]
- Hauck, C.; Cook, B.; Ellrott, T. Food addiction, eating addiction and eating disorders. Proc. Nutr. Soc. 2020, 79, 103–112. [Google Scholar] [CrossRef]
- Novelle, M.G.; Diéguez, C. Food Addiction and Binge Eating: Lessons Learned from Animal Models. Nutrients 2018, 10, 71. [Google Scholar] [CrossRef] [PubMed]
- Volkow, N.D.; Wise, R.A.; Baler, R. The dopamine motive system: Implications for drug and food addiction. Nat. Rev. Neurosci. 2017, 18, 741–752. [Google Scholar] [CrossRef] [PubMed]
- Nutt, D.J.; Lingford-Hughes, A.; Erritzoe, D.; Stokes, P.R. The dopamine theory of addiction: 40 years of highs and lows. Nat. Rev. Neurosci. 2015, 16, 305–312. [Google Scholar] [CrossRef] [PubMed]
- Constant, A.; Moirand, R.; Thibault, R.; Val-Laillet, D. Meeting of Minds around Food Addiction: Insights from Addiction Medicine, Nutrition, Psychology, and Neurosciences. Nutrients 2020, 12, 3564. [Google Scholar] [CrossRef]
- Römer, S.S.; Bliokas, V.; Teo, J.T.; Thomas, S.J. Food addiction, hormones and blood biomarkers in humans: A systematic literature review. Appetite 2023, 183, 106475. [Google Scholar] [CrossRef]
- Pak, K.; Seok, J.W.; Lee, M.J.; Kim, K.; Kim, I.J. Dopamine receptor and dopamine transporter in obesity: A meta-analysis. Synapse 2023, 77, e22254. [Google Scholar] [CrossRef] [PubMed]
- Volkow, N.D.; Wang, G.J.; Baler, R.D. Reward, dopamine and the control of food intake: Implications for obesity. Trends Cogn. Sci. 2011, 15, 37–46. [Google Scholar] [CrossRef]
- Morsali, M.; Poorolajal, J.; Shahbazi, F.; Vahidinia, A.; Doosti-Irani, A. Diet Therapeutics Interventions for Obesity: A Systematic Review and Network Meta-Analysis. J. Res. Health Sci. 2021, 21, e00521. [Google Scholar] [CrossRef] [PubMed]
- Muscogiuri, G.; El Ghoch, M.; Colao, A.; Hassapidou, M.; Yumuk, V.; Busetto, L.; Obesity Management Task Force (OMTF) of the European Association for the Study of Obesity (EASO). European Guidelines for Obesity Management in Adults with a Very Low-Calorie Ketogenic Diet: A Systematic Review and Meta-Analysis. Obes. Facts 2021, 14, 222–245. [Google Scholar] [CrossRef] [PubMed]
- Dominguez, L.J.; Veronese, N.; Di Bella, G.; Cusumano, C.; Parisi, A.; Tagliaferri, F.; Ciriminna, S.; Barbagallo, M. Mediterranean diet in the management and prevention of obesity. Exp. Gerontol. 2023, 174, 112121. [Google Scholar] [CrossRef]
- Franz, M.J.; VanWormer, J.J.; Crain, A.L.; Boucher, J.L.; Histon, T.; Caplan, W.; Bowman, J.D.; Pronk, N.P. Weight-loss outcomes: A systematic review and meta-analysis of weight-loss clinical trials with a minimum 1-year follow-up. J. Am. Diet. Assoc. 2007, 107, 1755–1767. [Google Scholar] [CrossRef] [PubMed]
- van Baak, M.A.; Mariman, E.C.M. Dietary Strategies for Weight Loss Maintenance. Nutrients 2019, 11, 1916. [Google Scholar] [CrossRef] [PubMed]
- Flore, G.; Preti, A.; Carta, M.G.; Deledda, A.; Fosci, M.; Nardi, A.E.; Loviselli, A.; Velluzzi, F. Weight Maintenance after Dietary Weight Loss: Systematic Review and Meta-Analysis on the Effectiveness of Behavioural Intensive Intervention. Nutrients 2022, 14, 1259. [Google Scholar] [CrossRef] [PubMed]
- Blomain, E.S.; Dirhan, D.A.; Valentino, M.A.; Kim, G.W.; Waldman, S.A. Mechanisms of Weight Regain following Weight Loss. ISRN Obes. 2013, 2013, 210524. [Google Scholar] [CrossRef] [PubMed]
- van Baak, M.A.; Mariman, E.C.M. Mechanisms of weight regain after weight loss—The role of adipose tissue. Nat. Rev. Endocrinol. 2019, 15, 274–287. [Google Scholar] [CrossRef]
- Busetto, L.; Bettini, S.; Makaronidis, J.; Roberts, C.A.; Halford, J.C.G.; Batterham, R.L. Mechanisms of weight regain. Eur. J. Intern. Med. 2021, 93, 3–7. [Google Scholar] [CrossRef] [PubMed]
- van Galen, K.A.; Schrantee, A.; Ter Horst, K.W.; la Fleur, S.E.; Booij, J.; Constable, R.T.; Schwartz, G.J.; DiLeone, R.J.; Serlie, M.J. Brain responses to nutrients are severely impaired and not reversed by weight loss in humans with obesity: A randomized crossover study. Nat. Metab. 2023, 5, 1059–1072. [Google Scholar] [CrossRef]
- Geisler, C.E.; Hayes, M.R. Metabolic hormone action in the VTA: Reward-directed behavior and mechanistic insights. Physiol. Behav. 2023, 268, 114236. [Google Scholar] [CrossRef] [PubMed]
- Rezitis, J.; Herzog, H.; Ip, C.K. Neuropeptide Y interaction with dopaminergic and serotonergic pathways: Interlinked neurocircuits modulating hedonic eating behaviours. Prog. Neuropsychopharmacol. Biol. Psychiatry 2022, 113, 110449. [Google Scholar] [CrossRef] [PubMed]
- Abizaid, A. Ghrelin and dopamine: New insights on the peripheral regulation of appetite. J. Neuroendocrinol. 2009, 21, 787–793. [Google Scholar] [CrossRef]
- Kawahara, Y.; Kawahara, H.; Kaneko, F.; Yamada, M.; Nishi, Y.; Tanaka, E.; Nishi, A. Peripherally administered ghrelin induces bimodal effects on the mesolimbic dopamine system depending on food-consumptive states. Neuroscience 2009, 161, 855–864. [Google Scholar] [CrossRef]
- Kawahara, Y.; Kaneko, F.; Yamada, M.; Kishikawa, Y.; Kawahara, H.; Nishi, A. Food reward-sensitive interaction of ghrelin and opioid receptor pathways in mesolimbic dopamine system. Neuropharmacology 2013, 67, 395–402. [Google Scholar] [CrossRef] [PubMed]
- Skibicka, K.P.; Shirazi, R.H.; Hansson, C.; Dickson, S.L. Ghrelin interacts with neuropeptide Y Y1 and opioid receptors to increase food reward. Endocrinology 2012, 153, 1194–1205. [Google Scholar] [CrossRef]
- Perez-Bonilla, P.; Santiago-Colon, K.; Leinninger, G.M. Lateral hypothalamic area neuropeptides modulate ventral tegmental area dopamine neurons and feeding. Physiol. Behav. 2020, 223, 112986. [Google Scholar] [CrossRef]
- Vallöf, D.; Kalafateli, A.L.; Jerlhag, E. Brain region specific glucagon-like peptide-1 receptors regulate alcohol-induced behaviors in rodents. Psychoneuroendocrinology 2019, 103, 284–295. [Google Scholar] [CrossRef]
- Sørensen, G.; Reddy, I.A.; Weikop, P.; Graham, D.L.; Stanwood, G.D.; Wortwein, G.; Galli, A.; Fink-Jensen, A. The glucagon-like peptide 1 (GLP-1) receptor agonist exendin-4 reduces cocaine self-administration in mice. Physiol. Behav. 2015, 149, 262–268. [Google Scholar] [CrossRef]
- Falk, S.; Petersen, J.; Svendsen, C.; Romero-Leguizamón, C.R.; Jørgensen, S.H.; Krauth, N.; Ludwig, M.Q.; Lundø, K.; Roostalu, U.; Skovbjerg, G.; et al. GLP-1 and nicotine combination therapy engages hypothalamic and mesolimbic pathways to reverse obesity. Cell Rep. 2023, 42, 112466. [Google Scholar] [CrossRef] [PubMed]
- Mietlicki-Baase, E.G.; Ortinski, P.I.; Reiner, D.J.; Sinon, C.G.; McCutcheon, J.E.; Pierce, R.C.; Roitman, M.F.; Hayes, M.R. Glucagon-like peptide-1 receptor activation in the nucleus accumbens core suppresses feeding by increasing glutamatergic AMPA/kainate signaling. J. Neurosci. 2014, 34, 6985–6992. [Google Scholar] [CrossRef] [PubMed]
- Kintscher, U. Reuptake inhibitors of dopamine, noradrenaline, and serotonin. In Appetite Control; Springer: Berlin/Heidelberg, Germany, 2012; pp. 339–347. [Google Scholar] [CrossRef]
- Billes, S.K.; Cowley, M.A. Inhibition of dopamine and norepinephrine reuptake produces additive effects on energy balance in lean and obese mice. Neuropsychopharmacology 2007, 32, 822–834. [Google Scholar] [CrossRef] [PubMed]
- Billes, S.K.; Cowley, M.A. Catecholamine reuptake inhibition causes weight loss by increasing locomotor activity and thermogenesis. Neuropsychopharmacology 2008, 33, 1287–1297. [Google Scholar] [CrossRef] [PubMed]
- Liu, M.; Ren, L.; Zhong, X.; Ding, Y.; Liu, T.; Liu, Z.; Yang, X.; Cui, L.; Yang, L.; Fan, Y.; et al. D2-Like Receptors Mediate Dopamine-Inhibited Insulin Secretion via Ion Channels in Rat Pancreatic β-Cells. Front. Endocrinol. 2020, 11, 152. [Google Scholar] [CrossRef]
- Uefune, F.; Aonishi, T.; Kitaguchi, T.; Takahashi, H.; Seino, S.; Sakano, D.; Kume, S. Dopamine Negatively Regulates Insulin Secretion Through Activation of D1-D2 Receptor Heteromer. Diabetes 2022, 71, 1946–1961. [Google Scholar] [CrossRef] [PubMed]
- Aslanoglou, D.; Bertera, S.; Sánchez-Soto, M.; Benjamin Free, R.; Lee, J.; Zong, W.; Xue, X.; Shrestha, S.; Brissova, M.; Logan, R.W.; et al. Dopamine regulates pancreatic glucagon and insulin secretion via adrenergic and dopaminergic receptors. Transl. Psychiatry 2021, 11, 59. [Google Scholar] [CrossRef]
- Farino, Z.J.; Morgenstern, T.J.; Maffei, A.; Quick, M.; De Solis, A.J.; Wiriyasermkul, P.; Freyberg, R.J.; Aslanoglou, D.; Sorisio, D.; Inbar, B.P.; et al. New roles for dopamine D2 and D3 receptors in pancreatic beta cell insulin secretion. Mol. Psychiatry 2020, 25, 2070–2085. [Google Scholar] [CrossRef]
- Mietlicki-Baase, E.G.; Ortinski, P.I.; Rupprecht, L.E.; Olivos, D.R.; Alhadeff, A.L.; Pierce, R.C.; Hayes, M.R. The food intake-suppressive effects of glucagon-like peptide-1 receptor signaling in the ventral tegmental area are mediated by AMPA/kainate receptors. Am. J. Physiol. Endocrinol. Metab. 2013, 305, E1367–E1374. [Google Scholar] [CrossRef] [PubMed]
- Available online: https://diabetesjournals.org/care/article/46/Supplement_1/S140/148057/9-Pharmacologic-Approaches-to-Glycemic-Treatment (accessed on 25 September 2023).
- Available online: https://diabetesjournals.org/care/article/34/4/789/38811/Bromocriptine-A-Sympatholytic-D2-Dopamine-Agonist (accessed on 25 September 2023).
- Aslanoglou, D.; Bertera, S.; Friggeri, L.; Sánchez-Soto, M.; Lee, J.; Xue, X.; Logan, R.W.; Lane, J.R.; Yechoor, V.K.; McCormick, P.J.; et al. Dual pancreatic adrenergic and dopaminergic signaling as a therapeutic target of bromocriptine. iScience 2022, 25, 104771. [Google Scholar] [CrossRef]
- Gaziano, J.M.; Cincotta, A.H.; O’Connor, C.M.; Ezrokhi, M.; Rutty, D.; Ma, Z.J.; Scranton, R.E. Randomized Clinical Trial of Quick-Release Bromocriptine Among Patients With Type 2 Diabetes on Overall Safety and Cardiovascular Outcomes. Diabetes Care 2016, 33, 1503–1508, Erratum in Diabetes Care 2016, 39, 1846. [Google Scholar] [CrossRef] [PubMed]
- Wang, R.; Shih, L.C. Parkinson’s disease—Current treatment. Curr. Opin. Neurol. 2023, 36, 302–308. [Google Scholar] [CrossRef]
- Dietrichs, E.; Alves, G.; Benjaminsen, E.; Johansen, K.K.; Tysnes, O.B. Treatment of motor symptoms in Parkinson’s disease. Tidsskr. Nor Laegeforen. 2023, 143. [Google Scholar] [CrossRef]
- Yamamoto, M. Do dopamine agonists provide neuroprotection? Neurology 1998, 51 (Suppl. S2), S10–S12. [Google Scholar] [CrossRef] [PubMed]
- Cacabelos, R. Parkinson’s Disease: From Pathogenesis to Pharmacogenomics. Int. J. Mol. Sci. 2017, 18, 551. [Google Scholar] [CrossRef] [PubMed]
- Woitalla, D.; Buhmann, C.; Hilker-Roggendorf, R.; Höglinger, G.; Koschel, J.; Müller, T.; Weise, D. Role of dopamine agonists in Parkinson’s disease therapy. J. Neural Transm. 2023, 130, 863–873. [Google Scholar] [CrossRef] [PubMed]
- Weiss, H.D.; Marsh, L. Impulse control disorders and compulsive behaviors associated with dopaminergic therapies in Parkinson disease. Neurol. Clin. Pract. 2012, 2, 267–274. [Google Scholar] [CrossRef] [PubMed]
- Masuzaki, H.; Kozuka, C.; Okamoto, S.; Yonamine, M.; Tanaka, H.; Shimabukuro, M. Brown rice-specific γ-oryzanol as a promising prophylactic avenue to protect against diabetes mellitus and obesity in humans. J. Diabetes Investig. 2019, 10, 18–25. [Google Scholar] [CrossRef] [PubMed]
- Kozuka, C.; Yabiku, K.; Sunagawa, S.; Ueda, R.; Taira, S.; Ohshiro, H.; Ikema, T.; Yamakawa, K.; Higa, M.; Tanaka, H.; et al. Brown rice and its component, γ-oryzanol, attenuate the preference for high-fat diet by decreasing hypothalamic endoplasmic reticulum stress in mice. Diabetes 2012, 61, 3084–3093. [Google Scholar] [CrossRef] [PubMed]
- Kozuka, C.; Sunagawa, S.; Ueda, R.; Higa, M.; Tanaka, H.; Shimizu-Okabe, C.; Ishiuchi, S.; Takayama, C.; Matsushita, M.; Tsutsui, M.; et al. γ-Oryzanol protects pancreatic β-cells against endoplasmic reticulum stress in male mice. Endocrinology 2015, 156, 1242–1250. [Google Scholar] [CrossRef] [PubMed]
- Kozuka, C.; Sunagawa, S.; Ueda, R.; Higa, M.; Ohshiro, Y.; Tanaka, H.; Shimizu-Okabe, C.; Takayama, C.; Matsushita, M.; Tsutsui, M.; et al. A novel insulinotropic mechanism of whole grain-derived γ-oryzanol via the suppression of local dopamine D2 receptor signalling in mouse islet. Br. J. Pharmacol. 2015, 172, 4519–4534. [Google Scholar] [CrossRef]
- Araujo, S.M.; de Paula, M.T.; Poetini, M.R.; Meichtry, L.; Bortolotto, V.C.; Zarzecki, M.S.; Jesse, C.R.; Prigol, M. Effectiveness of γ-oryzanol in reducing neuromotor deficits, dopamine depletion and oxidative stress in a Drosophila melanogaster model of Parkinson’s disease induced by rotenone. Neurotoxicology 2015, 51, 96–105. [Google Scholar] [CrossRef] [PubMed]
- Kumar, S.; Kumar, P. The Beneficial Effect of Rice Bran Extract Against Rotenone-Induced Experimental Parkinson’s Disease in Rats. Curr. Mol. Pharmacol. 2021, 14, 428–438. [Google Scholar] [CrossRef] [PubMed]
- Foretz, M.; Guigas, B.; Viollet, B. Metformin: Update on mechanisms of action and repurposing potential. Nat. Rev. Endocrinol. 2023, 19, 460–476. [Google Scholar] [CrossRef] [PubMed]
- Liu, Y.J.; Chern, Y. AMPK-mediated regulation of neuronal metabolism and function in brain diseases. J. Neurogenet. 2015, 29, 50–58. [Google Scholar] [CrossRef] [PubMed]
- Curry, D.W.; Stutz, B.; Andrews, Z.B.; Elsworth, J.D. Targeting AMPK Signaling as a Neuroprotective Strategy in Parkinson’s Disease. J. Park. Dis. 2018, 8, 161–181. [Google Scholar] [CrossRef] [PubMed]
- Rotermund, C.; Machetanz, G.; Fitzgerald, J.C. The Therapeutic Potential of Metformin in Neurodegenerative Diseases. Front. Endocrinol. 2018, 9, 400. [Google Scholar] [CrossRef] [PubMed]
- Ryu, Y.K.; Go, J.; Park, H.Y.; Choi, Y.K.; Seo, Y.J.; Choi, J.H.; Rhee, M.; Lee, T.G.; Lee, C.H.; Kim, K.S. Metformin regulates astrocyte reactivity in Parkinson’s disease and normal aging. Neuropharmacology 2020, 175, 108173. [Google Scholar] [CrossRef]
- Available online: https://www.ncbi.nlm.nih.gov/gene/586 (accessed on 5 September 2023).
- Mor, D.E.; Sohrabi, S.; Kaletsky, R.; Keyes, W.; Tartici, A.; Kalia, V.; Miller, G.W.; Murphy, C.T. Metformin rescues Parkinson’s disease phenotypes caused by hyperactive mitochondria. Proc. Natl. Acad. Sci. USA 2020, 117, 26438–26447. [Google Scholar] [CrossRef]
- Paudel, Y.N.; Angelopoulou, E.; Piperi, C.; Shaikh, M.F.; Othman, I. Emerging neuroprotective effect of metformin in Parkinson’s disease: A molecular crosstalk. Pharmacol. Res. 2020, 152, 104593. [Google Scholar] [CrossRef] [PubMed]
- Markowicz-Piasecka, M.; Sikora, J.; Szydłowska, A.; Skupień, A.; Mikiciuk-Olasik, E.; Huttunen, K.M. Metformin—A Future Therapy for Neurodegenerative Diseases: Theme: Drug Discovery, Development and Delivery in Alzheimer’s Disease Guest Editor: Davide Brambilla. Pharm. Res. 2017, 34, 2614–2627. [Google Scholar] [CrossRef] [PubMed]
- Huang, K.H.; Chang, Y.L.; Gau, S.Y.; Tsai, T.H.; Lee, C.Y. Dose-Response Association of Metformin with Parkinson’s Disease Odds in Type 2 Diabetes Mellitus. Pharmaceutics 2022, 14, 946. [Google Scholar] [CrossRef]
- Alrouji, M.; Al-Kuraishy, H.M.; Al-Gareeb, A.I.; Ashour, N.A.; Jabir, M.S.; Negm, W.A.; Batiha, G.E. Metformin role in Parkinson’s disease: A double-sword effect. Mol. Cell. Biochem. 2023. [Google Scholar] [CrossRef]
- Moore, E.; Mander, A.; Ames, D.; Carne, R.; Sanders, K.; Watters, D. Cognitive impairment and vitamin B12: A review. Int. Psychogeriatr. 2012, 24, 541–556. [Google Scholar] [CrossRef] [PubMed]
- Hanefeld, M. Cardiovascular benefits and safety profile of acarbose therapy in prediabetes and established type 2 diabetes. Cardiovasc. Diabetol. 2007, 6, 20. [Google Scholar] [CrossRef] [PubMed]
- Wang, H. MicroRNAs, Parkinson’s Disease, and Diabetes Mellitus. Int. J. Mol. Sci. 2021, 22, 2953. [Google Scholar] [CrossRef] [PubMed]
- Sundararajan, S.; Landreth, G.E. Antiinflammatory properties of PPARgamma agonists following ischemia. Drug News Perspect. 2004, 17, 229–236. [Google Scholar] [CrossRef] [PubMed]
- Tjokroprawiro, A. New approach in the treatment of T2DM and metabolic syndrome (focus on a novel insulin sensitizer). Acta Med. Indones. 2006, 38, 160–166. [Google Scholar]
- Kapadia, R.; Yi, J.H.; Vemuganti, R. Mechanisms of anti-inflammatory and neuroprotective actions of PPAR-gamma agonists. Front. Biosci. 2008, 13, 1813–1826. [Google Scholar] [CrossRef]
- Culman, J.; Nguyen-Ngoc, M.; Glatz, T.; Gohlke, P.; Herdegen, T.; Zhao, Y. Treatment of rats with pioglitazone in the reperfusion phase of focal cerebral ischemia: A preclinical stroke trial. Exp. Neurol. 2012, 238, 243–253. [Google Scholar] [CrossRef]
- White, A.T.; Murphy, A.N. Administration of thiazolidinediones for neuroprotection in ischemic stroke: A pre-clinical systematic review. J. Neurochem. 2010, 115, 845–853. [Google Scholar] [CrossRef]
- Tanaka, R.; Yamashiro, K.; Okuma, Y.; Shimura, H.; Nakamura, S.; Ueno, Y.; Tanaka, Y.; Miyamoto, N.; Tomizawa, Y.; Nakahara, T.; et al. Effects of Pioglitazone for Secondary Stroke Prevention in Patients with Impaired Glucose Tolerance and Newly Diagnosed Diabetes: The J-SPIRIT Study. J. Atheroscler. Thromb. 2015, 22, 1305–1316. [Google Scholar] [CrossRef]
- Kernan, W.N.; Viscoli, C.M.; Furie, K.L.; Young, L.H.; Inzucchi, S.E.; Gorman, M.; Guarino, P.D.; Lovejoy, A.M.; Peduzzi, P.N.; Conwit, R.; et al. Pioglitazone after Ischemic Stroke or Transient Ischemic Attack. N. Engl. J. Med. 2016, 374, 1321–1331. [Google Scholar] [CrossRef]
- de Jong, M.; van der Worp, H.B.; van der Graaf, Y.; Visseren, F.L.J.; Westerink, J. Pioglitazone and the secondary prevention of cardiovascular disease. A meta-analysis of randomized-controlled trials. Cardiovasc. Diabetol. 2017, 16, 134. [Google Scholar] [CrossRef] [PubMed]
- Jojo, G.M.; Kuppusamy, G. Scope of new formulation approaches in the repurposing of pioglitazone for the management of Alzheimer’s disease. J. Clin. Pharm. Ther. 2019, 44, 337–348. [Google Scholar] [CrossRef] [PubMed]
- Zamanian, M.Y.; Taheri, N.; Opulencia, M.J.C.; Bokov, D.O.; Abdullaev, S.Y.; Gholamrezapour, M.; Heidari, M.; Bazmandegan, G. Neuroprotective and Anti-inflammatory Effects of Pioglitazone on Traumatic Brain Injury. Mediat. Inflamm. 2022, 2022, 9860855. [Google Scholar] [CrossRef] [PubMed]
- Alhowail, A.; Alsikhan, R.; Alsaud, M.; Aldubayan, M.; Rabbani, S.I. Protective Effects of Pioglitazone on Cognitive Impairment and the Underlying Mechanisms: A Review of Literature. Drug Des. Dev. Ther. 2022, 16, 2919–2931. [Google Scholar] [CrossRef]
- Hirsch, E.C.; Breidert, T.; Rousselet, E.; Hunot, S.; Hartmann, A.; Michel, P.P. The role of glial reaction and inflammation in Parkinson’s disease. Ann. N. Y. Acad. Sci. 2003, 991, 214–228. [Google Scholar] [CrossRef] [PubMed]
- Dehmer, T.; Heneka, M.T.; Sastre, M.; Dichgans, J.; Schulz, J.B. Protection by pioglitazone in the MPTP model of Parkinson’s disease correlates with I kappa B alpha induction and block of NF kappa B and iNOS activation. J. Neurochem. 2004, 88, 494–501. [Google Scholar] [CrossRef]
- Randy, L.H.; Guoying, B. Agonism of Peroxisome Proliferator Receptor-Gamma may have Therapeutic Potential for Neuroinflammation and Parkinson’s Disease. Curr. Neuropharmacol. 2007, 5, 35–46. [Google Scholar] [CrossRef]
- Carta, A.R.; Pisanu, A. Modulating microglia activity with PPAR-γ agonists: A promising therapy for Parkinson’s disease? Neurotox. Res. 2013, 23, 112–123. [Google Scholar] [CrossRef] [PubMed]
- Burns, D.K.; Alexander, R.C.; Welsh-Bohmer, K.A.; Culp, M.; Chiang, C.; O’Neil, J.; Evans, R.M.; Harrigan, P.; Plassman, B.L.; Burke, J.R.; et al. Safety and efficacy of pioglitazone for the delay of cognitive impairment in people at risk of Alzheimer’s disease (TOMMORROW): A prognostic biomarker study and a phase 3, randomised, double-blind, placebo-controlled trial. Lancet Neurol. 2021, 20, 537–547. [Google Scholar] [CrossRef] [PubMed]
- Saunders, A.M.; Burns, D.K.; Gottschalk, W.K. Reassessment of Pioglitazone for Alzheimer’s Disease. Front. Neurosci. 2021, 15, 666958. [Google Scholar] [CrossRef] [PubMed]
- Simon, D.K.; Simuni, T.; Elm, J.; Clark-Matott, J.; Graebner, A.K.; Baker, L.; Dunlop, S.R.; Emborg, M.; Kamp, C.; Morgan, J.C.; et al. Peripheral Biomarkers of Parkinson’s Disease Progression and Pioglitazone Effects. J. Park. Dis. 2015, 5, 731–736. [Google Scholar] [CrossRef] [PubMed]
- Hussain, S.; Singh, A.; Baxi, H.; Taylor, B.; Burgess, J.; Antony, B. Thiazolidinedione use is associated with reduced risk of Parkinson’s disease in patients with diabetes: A meta-analysis of real-world evidence. Neurol. Sci. 2020, 41, 3697–3703. [Google Scholar] [CrossRef] [PubMed]
- Chen, L.; Tao, Y.; Li, J.; Kang, M. Pioglitazone use is associated with reduced risk of Parkinson’s disease in patients with diabetes: A systematic review and meta-analysis. J. Clin. Neurosci. 2022, 106, 154–158. [Google Scholar] [CrossRef]
- Sunnarborg, K.; Tiihonen, M.; Huovinen, M.; Koponen, M.; Hartikainen, S.; Tolppanen, A.M. Association between different diabetes medication classes and risk of Parkinson’s disease in people with diabetes. Pharmacoepidemiol. Drug Saf. 2022, 31, 875–882. [Google Scholar] [CrossRef] [PubMed]
- NINDS Exploratory Trials in Parkinson Disease (NET-PD) FS-ZONE Investigators. Pioglitazone in early Parkinson’s disease: A phase 2, multicentre, double-blind, randomised trial. Lancet Neurol. 2015, 14, 795–803, Erratum in Lancet Neurol. 2015, 14, 979. [Google Scholar] [CrossRef]
- Thornberry, N.A.; Gallwitz, B. Mechanism of action of inhibitors of dipeptidyl-peptidase-4 (DPP-4). Best Pract. Res. Clin. Endocrinol. Metab. 2009, 23, 479–486. [Google Scholar] [CrossRef]
- Nauck, M.A.; Meier, J.J. The incretin effect in healthy individuals and those with type 2 diabetes: Physiology, pathophysiology, and response to therapeutic interventions. Lancet Diabetes Endocrinol. 2016, 4, 525–536. [Google Scholar] [CrossRef]
- Shyamaladevi, B.; Dash, I.; Badrachalam, R.; Krishnan, M.; Panneerselvam, A.; Undru, S. An update on diagnosis and therapeutics for type-2 diabetes mellitus. Bioinformation 2023, 19, 295–298. [Google Scholar] [CrossRef]
- Abdelsalam, R.M.; Safar, M.M. Neuroprotective effects of vildagliptin in rat rotenone Parkinson’s disease model: Role of RAGE-NFκB and Nrf2-antioxidant signaling pathways. J. Neurochem. 2015, 133, 700–707. [Google Scholar] [CrossRef] [PubMed]
- Pariyar, R.; Bastola, T.; Lee, D.H.; Seo, J. Neuroprotective Effects of the DPP4 Inhibitor Vildagliptin in In Vivo and In Vitro Models of Parkinson’s Disease. Int. J. Mol. Sci. 2022, 23, 2388. [Google Scholar] [CrossRef] [PubMed]
- Badawi, G.A.; Abd El Fattah, M.A.; Zaki, H.F.; El Sayed, M.I. Sitagliptin and liraglutide reversed nigrostriatal degeneration of rodent brain in rotenone-induced Parkinson’s disease. Inflammopharmacology 2017, 25, 369–382. [Google Scholar] [CrossRef] [PubMed]
- Li, J.; Zhang, S.; Li, C.; Li, M.; Ma, L. Sitagliptin rescues memory deficits in Parkinsonian rats via upregulating BDNF to prevent neuron and dendritic spine loss. Neurol Res. 2018, 40, 736–743. [Google Scholar] [CrossRef]
- Abhangi, K.V.; Patel, J.I. Neuroprotective effects of linagliptin in a rotenone-induced rat model of Parkinson’s disease. Indian J. Pharmacol. 2022, 54, 46–50. [Google Scholar] [CrossRef]
- Yu, H.Y.; Sun, T.; Wang, Z.; Li, H.; Xu, D.; An, J.; Wen, L.L.; Li, J.Y.; Li, W.; Feng, J. Exendin-4 and linagliptin attenuate neuroinflammation in a mouse model of Parkinson’s disease. Neural Regen. Res. 2023, 18, 1818–1826. [Google Scholar] [CrossRef]
- Nassar, N.N.; Al-Shorbagy, M.Y.; Arab, H.H.; Abdallah, D.M. Saxagliptin: A novel antiparkinsonian approach. Neuropharmacology 2015, 89, 308–317. [Google Scholar] [CrossRef] [PubMed]
- Turnes, J.M.; Bassani, T.B.; Souza, L.C.; Vital, M.A.B.F. Ineffectiveness of saxagliptin as a neuroprotective drug in 6-OHDA-lesioned rats. J. Pharm. Pharmacol. 2018, 70, 1059–1068. [Google Scholar] [CrossRef] [PubMed]
- Svenningsson, P.; Wirdefeldt, K.; Yin, L.; Fang, F.; Markaki, I.; Efendic, S.; Ludvigsson, J.F. Reduced incidence of Parkinson’s disease after dipeptidyl peptidase-4 inhibitors-A nationwide case-control study. Mov. Disord. 2016, 31, 1422–1423. [Google Scholar] [CrossRef]
- Brauer, R.; Wei, L.; Ma, T.; Athauda, D.; Girges, C.; Vijiaratnam, N.; Auld, G.; Whittlesea, C.; Wong, I.; Foltynie, T. Diabetes medications and risk of Parkinson’s disease: A cohort study of patients with diabetes. Brain 2020, 143, 3067–3076. [Google Scholar] [CrossRef]
- Lietzau, G.; Magni, G.; Kehr, J.; Yoshitake, T.; Candeias, E.; Duarte, A.I.; Pettersson, H.; Skogsberg, J.; Abbracchio, M.P.; Klein, T.; et al. Dipeptidyl peptidase-4 inhibitors and sulfonylureas prevent the progressive impairment of the nigrostriatal dopaminergic system induced by diabetes during aging. Neurobiol. Aging 2020, 89, 12–23. [Google Scholar] [CrossRef] [PubMed]
- Jeong, S.H.; Chung, S.J.; Yoo, H.S.; Hong, N.; Jung, J.H.; Baik, K.; Lee, Y.H.; Sohn, Y.H.; Lee, P.H. Beneficial effects of dipeptidyl peptidase-4 inhibitors in diabetic Parkinson’s disease. Brain 2021, 144, 1127–1137. [Google Scholar] [CrossRef] [PubMed]
- Qin, X.; Zhang, X.; Li, P.; Wang, M.; Yan, L.; Bao, Z.; Liu, Q. Association Between Diabetes Medications and the Risk of Parkinson’s Disease: A Systematic Review and Meta-Analysis. Front. Neurol. 2021, 12, 678649. [Google Scholar] [CrossRef] [PubMed]
- Wang, S.Y.; Wu, S.L.; Chen, T.C.; Chuang, C.S. Antidiabetic Agents for Treatment of Parkinson’s Disease: A Meta-Analysis. Int. J. Environ. Res. Public Health 2020, 17, 4805. [Google Scholar] [CrossRef]
- Khalaf, M.M.; El-Sayed, M.M.; Kandeil, M.A.; Ahmed, S. A novel protective modality against rotenone-induced Parkinson’s disease: A pre-clinical study with dulaglutide. Int. Immunopharmacol. 2023, 119, 110170. [Google Scholar] [CrossRef]
- Karagiannis, T.; Avgerinos, I.; Liakos, A.; Del Prato, S.; Matthews, D.R.; Tsapas, A.; Bekiari, E. Management of type 2 diabetes with the dual GIP/GLP-1 receptor agonist tirzepatide: A systematic review and meta-analysis. Diabetologia 2022, 65, 1251–1261. [Google Scholar] [CrossRef]
- De Mesquita, Y.L.L.; Pera Calvi, I.; Reis Marques, I.; Almeida Cruz, S.; Padrao, E.M.H.; Carvalho, P.E.P.; da Silva, C.H.A.; Cardoso, R.; Moura, F.A.; Rafalskiy, V.V. Efficacy and safety of the dual GIP and GLP-1 receptor agonist tirzepatide for weight loss: A meta-analysis of randomized controlled trials. Int. J. Obes. 2023, 47, 883–892. [Google Scholar] [CrossRef]
- Lisco, G.; De Tullio, A.; Disoteo, O.; De Geronimo, V.; Piazzolla, G.; De Pergola, G.; Giagulli, V.A.; Jirillo, E.; Guastamacchia, E.; Sabbà, C.; et al. Basal insulin intensification with GLP-1RA and dual GIP and GLP-1RA in patients with uncontrolled type 2 diabetes mellitus: A rapid review of randomized controlled trials and meta-analysis. Front. Endocrinol. 2022, 13, 920541. [Google Scholar] [CrossRef]
- Lv, M.; Xue, G.; Cheng, H.; Meng, P.; Lian, X.; Hölscher, C.; Li, D. The GLP-1/GIP dual-receptor agonist DA5-CH inhibits the NF-κB inflammatory pathway in the MPTP mouse model of Parkinson’s disease more effectively than the GLP-1 single-receptor agonist NLY01. Brain Behav. 2021, 11, e2231. [Google Scholar] [CrossRef]
- Ji, C.; Xue, G.F.; Lijun, C.; Feng, P.; Li, D.; Li, L.; Li, G.; Hölscher, C. A novel dual GLP-1 and GIP receptor agonist is neuroprotective in the MPTP mouse model of Parkinson’s disease by increasing expression of BNDF. Brain Res. 2016, 1634, 1–11. [Google Scholar] [CrossRef] [PubMed]
- Cao, L.; Li, D.; Feng, P.; Li, L.; Xue, G.F.; Li, G.; Hölscher, C. A novel dual GLP-1 and GIP incretin receptor agonist is neuroprotective in a mouse model of Parkinson’s disease by reducing chronic inflammation in the brain. Neuroreport 2016, 27, 384–391. [Google Scholar] [CrossRef] [PubMed]
- Hölscher, C. Glucagon-like peptide 1 and glucose-dependent insulinotropic peptide hormones and novel receptor agonists protect synapses in Alzheimer’s and Parkinson’s diseases. Front. Synaptic Neurosci. 2022, 14, 955258. [Google Scholar] [CrossRef] [PubMed]
- Salameh, T.S.; Rhea, E.M.; Talbot, K.; Banks, W.A. Brain uptake pharmacokinetics of incretin receptor agonists showing promise as Alzheimer’s and Parkinson’s disease therapeutics. Biochem Pharmacol. 2023, 180, 114187, Erratum in Biochem. Pharmacol. 2023, 210, 115474. [Google Scholar] [CrossRef]
- Hsia, D.S.; Grove, O.; Cefalu, W.T. An update on sodium-glucose co-transporter-2 inhibitors for the treatment of diabetes mellitus. Curr. Opin. Endocrinol. Diabetes Obes. 2017, 24, 73–79. [Google Scholar] [CrossRef] [PubMed]
- De Fronzo, R.A.; Hompesch, M.; Kasichayanula, S.; Liu, X.; Hong, Y.; Pfister, M.; Morrow, L.A.; Leslie, B.R.; Boulton, D.W.; Ching, A.; et al. Characterization of renal glucose reabsorption in response to dapagliflozin in healthy subjects and subjects with type 2 diabetes. Diabetes Care 2013, 36, 3169–3176. [Google Scholar] [CrossRef]
- Vallon, V.; Thomson, S.C. Targeting renal glucose reabsorption to treat hyperglycaemia: The pleiotropic effects of SGLT2 inhibition. Diabetologia 2017, 60, 215–225. [Google Scholar] [CrossRef] [PubMed]
- List, J.F.; Whaley, J.M. Glucose dynamics and mechanistic implications of SGLT2 inhibitors in animals and humans. Kidney Int. Suppl. 2011, 79, S20–S27. [Google Scholar] [CrossRef] [PubMed]
- Sattar, N.; Lee, M.M.Y.; Kristensen, S.L.; Branch, K.R.H.; Del Prato, S.; Khurmi, N.S.; Lam, C.S.P.; Lopes, R.D.; McMurray, J.J.V.; Pratley, R.E.; et al. Cardiovascular, mortality, and kidney outcomes with GLP-1 receptor agonists in patients with type 2 diabetes: A systematic review and meta-analysis of randomised trials. Lancet Diabetes Endocrinol. 2021, 9, 653–662. [Google Scholar] [CrossRef]
- McGuire, D.K.; Shih, W.J.; Cosentino, F.; Charbonnel, B.; Cherney, D.Z.I.; Dagogo-Jack, S.; Pratley, R.; Greenberg, M.; Wang, S.; Huyck, S.; et al. Association of SGLT2 Inhibitors With Cardiovascular and Kidney Outcomes in Patients With Type 2 Diabetes: A Meta-analysis. JAMA Cardiol. 2021, 6, 148–158. [Google Scholar] [CrossRef]
- Lin, K.J.; Wang, T.J.; Chen, S.D.; Lin, K.L.; Liou, C.W.; Lan, M.Y.; Chuang, Y.C.; Chuang, J.H.; Wang, P.W.; Lee, J.J.; et al. Two Birds One Stone: The Neuroprotective Effect of Antidiabetic Agents on Parkinson Disease-Focus on Sodium-Glucose Cotransporter 2 (SGLT2) Inhibitors. Antioxidants 2021, 10, 1935. [Google Scholar] [CrossRef]
- Arab, H.H.; Safar, M.M.; Shahin, N.N. Targeting ROS-Dependent AKT/GSK-3β/NF-κB and DJ-1/Nrf2 Pathways by Dapagliflozin Attenuates Neuronal Injury and Motor Dysfunction in Rotenone-Induced Parkinson’s Disease Rat Model. ACS Chem. Neurosci. 2021, 12, 689–703. [Google Scholar] [CrossRef] [PubMed]
- Ahmed, S.; El-Sayed, M.M.; Kandeil, M.A.; Khalaf, M.M. Empagliflozin attenuates neurodegeneration through antioxidant, anti-inflammatory, and modulation of α-synuclein and Parkin levels in rotenone-induced Parkinson’s disease in rats. Saudi Pharm. J. 2022, 30, 863–873. [Google Scholar] [CrossRef] [PubMed]
- Motawi, T.K.; Al-Kady, R.H.; Abdelraouf, S.M.; Senousy, M.A. Empagliflozin alleviates endoplasmic reticulum stress and augments autophagy in rotenone-induced Parkinson’s disease in rats: Targeting the GRP78/PERK/eIF2α/CHOP pathway and miR-211-5p. Chem. Biol. Interact. 2022, 362, 110002. [Google Scholar] [CrossRef] [PubMed]
- Ünal, İ.; Cansız, D.; Beler, M.; Sezer, Z.; Güzel, E.; Emekli-Alturfan, E. Sodium-dependent Glucose Co-Transporter-2 Inhibitor Empagliflozin Exerts Neuroprotective Effects in Rotenone-Induced Parkinson’s Disease Model in Zebrafish; Mechanism Involving Ketogenesis and Autophagy. Brain Res. 2023, 15, 148536. [Google Scholar] [CrossRef] [PubMed]
- Alshehri, M.M.; Danazumi, A.U.; Alshammari, M.K.; Bello, R.O.; Alghazwni, M.K.; Alshehri, A.M.; Alshlali, O.M.; Umar, H.I. Repurposing the inhibitors of MMP-9 and SGLT-2 against ubiquitin specific protease 30 in Parkinson’s disease: Computational modelling studies. J. Biomol. Struct. Dyn. 2023, 3, 1–12. [Google Scholar] [CrossRef] [PubMed]
- Mui, J.V.; Zhou, J.; Lee, S.; Leung, K.S.K.; Lee, T.T.L.; Chou, O.H.I.; Tsang, S.L.; Wai, A.K.C.; Liu, T.; Wong, W.T.; et al. Sodium-Glucose Cotransporter 2 (SGLT2) Inhibitors vs. Dipeptidyl Peptidase-4 (DPP4) Inhibitors for New-Onset Dementia: A Propensity Score-Matched Population-Based Study With Competing Risk Analysis. Front Cardiovasc. Med. 2021, 8, 747620. [Google Scholar] [CrossRef]
- Tharmaraja, T.; Ho, J.S.Y.; Sia, C.H.; Lim, N.A.; Chong, Y.F.; Lim, A.Y.L.; Rathakrishnan, R.R.; Yeo, L.L.L.; Sharma, V.K.; Tan, B.Y.Q. Sodium-glucose cotransporter 2 inhibitors and neurological disorders: A scoping review. Ther. Adv. Chronic. Dis. 2022, 13, 20406223221086996. [Google Scholar] [CrossRef]
- Inzucchi, S.; Rosenstock, J.; Umpierrez, G. Type 2 Diabetes and Insulin Secretagogues. J. Clin. Endocrinol. Metab. 2012, 97, 37A. [Google Scholar] [CrossRef] [PubMed]
- Wu, C.Y.; Iskander, C.; Wang, C.; Xiong, L.Y.; Shah, B.R.; Edwards, J.D.; Kapral, M.K.; Herrmann, N.; Lanctôt, K.L.; Masellis, M.; et al. Association of sulfonylureas with the risk of dementia: A population-based cohort study. J. Am. Geriatr. Soc. 2023, 71, 3059–3070. [Google Scholar] [CrossRef] [PubMed]
- Lechin, F.; van der Dijs, B.; Pardey-Maldonado, B.; Rivera, J.E.; Lechin, M.E.; Baez, S. Amantadine reduces glucagon and enhances insulin secretion throughout the oral glucose tolerance test: Central plus peripheral nervous system mechanisms. Diabetes Metab. Syndr. Obes. 2009, 2, 203–213. [Google Scholar] [CrossRef]
- Sanni, O.; Terre’Blanche, G. Dual A1 and A2A adenosine receptor antagonists, methoxy substituted 2-benzylidene-1-indanone, suppresses intestinal postprandial glucose and attenuates hyperglycaemia in fructose-streptozotocin diabetic rats. BMC Endocr. Disord. 2023, 23, 97. [Google Scholar] [CrossRef] [PubMed]
- Meltzer, H.Y.; Roth, B.L. Lorcaserin and pimavanserin: Emerging selectivity of serotonin receptor subtype-targeted drugs. J. Clin. Investig. 2013, 123, 4986–4991. [Google Scholar] [CrossRef] [PubMed]
- Meyer, J.M.; Correll, C.U. Increased Metabolic Potential, Efficacy, and Safety of Emerging Treatments in Schizophrenia. CNS Drugs 2023, 37, 545–570. [Google Scholar] [CrossRef]
- Coffey, B.; Jankovic, J.; Claassen, D.O.; Jimenez-Shahed, J.; Gertz, B.J.; Garofalo, E.A.; Stamler, D.A.; Wieman, M.; Savola, J.M.; Gordon, M.F.; et al. Efficacy and Safety of Fixed-Dose Deutetrabenazine in Children and Adolescents for Tics Associated With Tourette Syndrome: A Randomized Clinical Trial. JAMA Netw. Open 2021, 4, e2129397. [Google Scholar] [CrossRef] [PubMed]
- Huntington Study Group; Frank, S.; Testa, C.M.; Stamler, D.; Kayson, E.; Davis, C.; Edmondson, M.C.; Kinel, S.; Leavitt, B.; Oakes, D.; et al. Effect of Deutetrabenazine on Chorea Among Patients With Huntington Disease: A Randomized Clinical Trial. JAMA 2016, 316, 40–50. [Google Scholar] [CrossRef]
- Hainer, V.; Kabrnova, K.; Aldhoon, B.; Kunesova, M.; Wagenknecht, M. Serotonin and norepinephrine reuptake inhibition and eating behavior. Ann. N. Y. Acad. Sci. 2006, 1083, 252–269. [Google Scholar] [CrossRef] [PubMed]
- Tilbrook, D.; Jacob, J.; Parsons, P.; Edwards, C.; Loewen, K.; Kelly, L. Opioid use disorder and type 2 diabetes mellitus: Effect of participation in buprenorphine-naloxone substitution programs on glycemic control. Can. Fam. Physician 2017, 63, e350–e354. [Google Scholar]
- Fagerholm, V.; Haaparanta, M.; Scheinin, M. α2-adrenoceptor regulation of blood glucose homeostasis. Basic Clin. Pharmacol. Toxicol. 2011, 108, 365–370. [Google Scholar] [CrossRef]
- Raingeard, I.; Courtet, P.; Renard, E.; Bringer, J. Naltrexone improves blood glucose control in type 1 diabetic women with severe and chronic eating disorders. Diabetes Care 2004, 27, 847–848. [Google Scholar] [CrossRef] [PubMed]
Mechanism | Effect | Consequences |
---|---|---|
Interference with insulin-containing grain trafficking (Dopamine-containing vesicles) | Blunt insulin release |
|
Impaired intra-pancreatic dopamine catabolism (Monoaminoxidases) | Catecholamine-induced (alpha and D2/D3 receptors) suppression of insulin synthesis and secretion |
|
Meal-induced intestinal synthesis of dopamine | Anti-incretin effect |
|
Enhancement of alpha-cell activity (High-dose dopamine) | Glucagon secretion |
|
Suppression of prolactin release | Suppression of prolactin-induced insulin release |
|
Reduction in growth hormone | Amelioration of insulin release and peripheral insulin resistance |
|
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Lisco, G.; De Tullio, A.; Iovino, M.; Disoteo, O.; Guastamacchia, E.; Giagulli, V.A.; Triggiani, V. Dopamine in the Regulation of Glucose Homeostasis, Pathogenesis of Type 2 Diabetes, and Chronic Conditions of Impaired Dopamine Activity/Metabolism: Implication for Pathophysiological and Therapeutic Purposes. Biomedicines 2023, 11, 2993. https://doi.org/10.3390/biomedicines11112993
Lisco G, De Tullio A, Iovino M, Disoteo O, Guastamacchia E, Giagulli VA, Triggiani V. Dopamine in the Regulation of Glucose Homeostasis, Pathogenesis of Type 2 Diabetes, and Chronic Conditions of Impaired Dopamine Activity/Metabolism: Implication for Pathophysiological and Therapeutic Purposes. Biomedicines. 2023; 11(11):2993. https://doi.org/10.3390/biomedicines11112993
Chicago/Turabian StyleLisco, Giuseppe, Anna De Tullio, Michele Iovino, Olga Disoteo, Edoardo Guastamacchia, Vito Angelo Giagulli, and Vincenzo Triggiani. 2023. "Dopamine in the Regulation of Glucose Homeostasis, Pathogenesis of Type 2 Diabetes, and Chronic Conditions of Impaired Dopamine Activity/Metabolism: Implication for Pathophysiological and Therapeutic Purposes" Biomedicines 11, no. 11: 2993. https://doi.org/10.3390/biomedicines11112993