Abstract
Neurodegenerative diseases (NDDs) represent a heterogeneous group of disorders characterized by progressive neuronal loss, which results in significant deficits in memory, cognition, motor skills, and sensory functions. As the prevalence of NDDs rises, there is an urgent unmet need for effective therapies. Current drug development approaches primarily target single pathological features of the disease, which could explain the limited efficacy observed in late-stage clinical trials. Originally developed for the treatment of obesity and diabetes, incretin-based therapies, particularly long-acting GLP-1 receptor (GLP-1R) agonists and GLP-1R–gastric inhibitory polypeptide receptor (GIPR) dual agonists, are emerging as promising treatments for NDDs. Despite limited conclusive preclinical evidence, their pleiotropic ability to reduce neuroinflammation, enhance neuronal energy metabolism and promote synaptic plasticity positions them as potential disease-modifying NDD interventions. In anticipation of results from larger clinical trials, continued advances in next-generation incretin mimetics offer the potential for improved brain access and enhanced neuroprotection, paving the way for incretin-based therapies as a future cornerstone in the management of NDDs.
This is a preview of subscription content, access via your institution
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$32.99 / 30 days
cancel any time
Subscribe to this journal
Receive 12 digital issues and online access to articles
$119.00 per year
only $9.92 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to full article PDF
Prices may be subject to local taxes which are calculated during checkout





Similar content being viewed by others
References
Wilson, D. M. et al. Hallmarks of neurodegenerative diseases. Cell 186, 693–714 (2023).
Breijyeh, Z. & Karaman, R. Comprehensive review on Alzheimer’s disease: causes and treatment. Molecules 25, 5789 (2020).
2023 Alzheimer’s disease facts and figures. Alzheimers Dement. 19, 1598–1695, (2023).
WHO. Parkinson disease https://www.who.int/news-room/fact-sheets/detail/parkinson-disease (World Health Organization, 2023).
Calabresi, P. et al. Alpha-synuclein in Parkinson’s disease and other synucleinopathies: from overt neurodegeneration back to early synaptic dysfunction. Cell Death Dis. 14, 176 (2023).
Kirkeby, A. et al. Preclinical quality, safety, and efficacy of a human embryonic stem cell-derived product for the treatment of Parkinson’s disease, STEM-PD. Cell Stem Cell 30, 1299–1314.e9 (2023).
Hanzel, C. E. et al. Neuronal driven pre-plaque inflammation in a transgenic rat model of Alzheimer’s disease. Neurobiol. Aging 35, 2249–2262 (2014).
Cummings, J. et al. Alzheimer’s disease drug development pipeline: 2024. Alzheimers Dement. 10, e12465 (2024).
Drucker, D. J. The benefits of GLP-1 drugs beyond obesity. Science 385, 258–260 (2024).
Procaccini, C. et al. Role of metabolism in neurodegenerative disorders. Metabolism 65, 1376–1390 (2016).
Ott, A. et al. Diabetes mellitus and the risk of dementia: The Rotterdam Study. Neurology 53, 1937–1942, (1999).
Crane, P. K. et al. Glucose levels and risk of dementia. N. Engl. J. Med. 369, 540–548 (2013).
Hassing, L. B. et al. Overweight in midlife and risk of dementia: a 40-year follow-up study. Int. J. Obes. 33, 893–898 (2009).
Kivipelto, M. et al. Obesity and vascular risk factors at midlife and the risk of dementia and Alzheimer disease. Arch. Neurol. 62, 1556–1560 (2005).
Beydoun, M. A., Beydoun, H. A. & Wang, Y. Obesity and central obesity as risk factors for incident dementia and its subtypes: a systematic review and meta-analysis. Obes. Rev. 9, 204–218 (2008).
Eskelinen, M. H. et al. Fat intake at midlife and cognitive impairment later in life: a population-based CAIDE study. Int. J. Geriatr. Psychiatry 23, 741–747 (2008).
Kalmijn, S. et al. Dietary fat intake and the risk of incident dementia in the Rotterdam Study. Ann. Neurol. 42, 776–782 (1997).
Han, K., Kim, B., Lee, S. H. & Kim, M. K. A nationwide cohort study on diabetes severity and risk of Parkinson disease. NPJ Parkinsons Dis. 9, 11 (2023).
Athauda, D. et al. The impact of type 2 diabetes in Parkinson’s disease. Mov. Disord. 37, 1612–1623 (2022).
Yue, X. et al. Risk of Parkinson disease in diabetes mellitus: an updated meta-analysis of population-based cohort studies. Medicine 95, e3549 (2016).
Golimstok, A. et al. Cardiovascular risk factors and frontotemporal dementia: a case-control study. Transl. Neurodegener. 3, 13 (2014).
Hou, W. H., Li, C. Y., Chang, H. H., Sun, Y. & Tsai, C. C. A population-based cohort study suggests an increased risk of multiple sclerosis incidence in patients with type 2 diabetes mellitus. J. Epidemiol. 27, 235–241 (2017).
Branigan, G. L., Torrandell-Haro, G., Vitali, F., Brinton, R. D. & Rodgers, K. Age and sex differences on anti-hyperglycemic medication exposure and risk of newly diagnosed multiple sclerosis in propensity score matched type 2 diabetics. Heliyon 8, e11196 (2022).
D’Ovidio, F. et al. The role of pre-morbid diabetes on developing amyotrophic lateral sclerosis. Eur. J. Neurol. 25, 164–170 (2018).
Jawaid, A. et al. ALS disease onset may occur later in patients with pre-morbid diabetes mellitus. Eur. J. Neurol. 17, 733–739 (2010).
Tsai, C. P., Hu, C. & Lee, C. T. Finding diseases associated with amyotrophic lateral sclerosis: a total population-based case-control study. Amyotroph. Lateral Scler. Frontotemporal. Degener. 20, 82–89 (2019).
Sun, Y., Lu, C. J., Chen, R. C., Hou, W. H. & Li, C. Y. Risk of amyotrophic lateral sclerosis in patients with diabetes: a nationwide population-based cohort study. J. Epidemiol. 25, 445–451 (2015).
Paganoni, S. et al. Pre-morbid type 2 diabetes mellitus is not a prognostic factor in amyotrophic lateral sclerosis. Muscle Nerve 52, 339–343 (2015).
Mariosa, D., Kamel, F., Bellocco, R., Ye, W. & Fang, F. Association between diabetes and amyotrophic lateral sclerosis in Sweden. Eur. J. Neurol. 22, 1436–1442 (2015).
Gao, L., Cui, Z., Shen, L. & Ji, H. F. Shared genetic etiology between type 2 diabetes and Alzheimer’s disease identified by bioinformatics analysis. J. Alzheimers Dis. 50, 13–17 (2016).
Hao, K. et al. Shared genetic etiology underlying Alzheimer’s disease and type 2 diabetes. Mol. Aspects Med. 43–44, 66–76 (2015).
Zhu, Z., Lin, Y., Li, X., Driver, J. A. & Liang, L. Shared genetic architecture between metabolic traits and Alzheimer’s disease: a large-scale genome-wide cross-trait analysis. Hum. Genet. 138, 271–285 (2019).
Zeng, R. et al. Dissecting shared genetic architecture between obesity and multiple sclerosis. eBioMedicine 93, 104647 (2023).
Bomfim, T. R. et al. An anti-diabetes agent protects the mouse brain from defective insulin signaling caused by Alzheimer’s disease- associated Aβ oligomers. J. Clin. Invest. 122, 1339–1353 (2012).
Moloney, A. M. et al. Defects in IGF-1 receptor, insulin receptor and IRS-1/2 in Alzheimer’s disease indicate possible resistance to IGF-1 and insulin signalling. Neurobiol. Aging 31, 224–243 (2010).
Bassil, F. et al. Impaired brain insulin signalling in Parkinson’s disease. Neuropathol. Appl. Neurobiol. 48, e12760 (2022).
Kellar, D. & Craft, S. Brain insulin resistance in Alzheimer’s disease and related disorders: mechanisms and therapeutic approaches. Lancet Neurol. 19, 758–766 (2020).
Ruiz-Pozo, V. A. et al. The Molecular mechanisms of the relationship between insulin resistance and Parkinson’s disease pathogenesis. Nutrients 15, 3585 (2023).
Ayromlou, H. et al. Insulin resistance is associated with cognitive dysfunction in multiple sclerosis patients: a cross-sectional study. J. Neuroendocrinol. 35, e13288 (2023).
Milstein, J. L. & Ferris, H. A. The brain as an insulin-sensitive metabolic organ. Mol. Metab. 52, 101234 (2021).
Szablewski, L. Brain glucose transporters: role in pathogenesis and potential targets for the treatment of Alzheimer’s disease. Int. J. Mol. Sci. 22, 8142 (2021).
Sharma, M., Yadav, Y. & Dey, C. S. Neuronal insulin signaling and resistance: a balancing act of kinases and phosphatases. J. Endocrinol. 260, e230151 (2024).
Zhao, W. Q. et al. Amyloid beta oligomers induce impairment of neuronal insulin receptors. FASEB J. 22, 246–260 (2008).
Lourenco, M. V. et al. TNF-alpha mediates PKR-dependent memory impairment and brain IRS-1 inhibition induced by Alzheimer’s beta-amyloid oligomers in mice and monkeys. Cell Metab. 18, 831–843 (2013).
Luciunaite, A. et al. Soluble Aβ oligomers and protofibrils induce NLRP3 inflammasome activation in microglia. J. Neurochem. 155, 650–661 (2020).
Hotamisligil, G. S., Budavari, A., Murray, D. & Spiegelman, B. M. Reduced tyrosine kinase activity of the insulin receptor in obesity-diabetes. Central role of tumor necrosis factor-alpha. J. Clin. Invest. 94, 1543–1549 (1994).
Pomytkin, I. et al. Insulin receptor in the brain: mechanisms of activation and the role in the CNS pathology and treatment. CNS Neurosci. Ther. 24, 763–774 (2018).
Carvalho, C. & Cardoso, S. Diabetes–Alzheimer’s disease link: targeting mitochondrial dysfunction and redox imbalance. Antioxid. Redox. Signal. 34, 631–649 (2021).
Schubert, M. et al. Role for neuronal insulin resistance in neurodegenerative diseases. Proc. Natl Acad. Sci. USA 101, 3100–3105 (2004).
García-Cáceres, C. et al. Astrocytic insulin signaling couples brain glucose uptake with nutrient availability. Cell 166, 867–880 (2016).
Ferris, H. A. et al. Loss of astrocyte cholesterol synthesis disrupts neuronal function and alters whole-body metabolism. Proc. Natl Acad. Sci. USA 114, 1189–1194 (2017).
Chen, W. et al. Loss of insulin signaling in astrocytes exacerbates Alzheimer-like phenotypes in a 5xFAD mouse model. Proc. Natl Acad. Sci. USA 120, e2220684120 (2023).
Chen, W., Liu, X., Munoz, V. R. & Kahn, C. R. Loss of insulin signaling in microglia impairs cellular uptake of abeta and neuroinflammatory response exacerbating Alzheimer-like neuropathology. Preprint at bioRxiv https://doi.org/10.1101/2024.08.22.609112 (2024).
Kwon, H. S. & Koh, S. H. Neuroinflammation in neurodegenerative disorders: the roles of microglia and astrocytes. Transl. Neurodegener. 9, 42 (2020).
Botella Lucena, P. & Heneka, M. T. Inflammatory aspects of Alzheimer’s disease. Acta Neuropathol. 148, 31 (2024).
Zhang, W., Xiao, D., Mao, Q. & Xia, H. Role of neuroinflammation in neurodegeneration development. Signal Transduct. Target Ther. 8, 267 (2023).
Tarkowski, E., Andreasen, N., Tarkowski, A. & Blennow, K. Intrathecal inflammation precedes development of Alzheimer’s disease. J. Neurol. Neurosurg. Psychiatry 74, 1200–1205 (2003).
in t’ Veld, B. A. et al. Nonsteroidal antiinflammatory drugs and the risk of Alzheimer’s disease. N. Engl. J. Med. 345, 1515–1521 (2001).
De Felice, F. G. & Ferreira, S. T. Inflammation, defective insulin signaling, and mitochondrial dysfunction as common molecular denominators connecting type 2 diabetes to Alzheimer disease. Diabetes 63, 2262–2272 (2014).
Elliott, R. et al. Glucagon-like peptide-1 (7–36) amide and glucose-dependent insulinotropic polypeptide secretion in response to nutrient ingestion in man: acute post-prandial and 24-h secretion patterns. J. Endocrinol. 138, 159–166 (1993).
Drucker, D. J. & Holst, J. J. The expanding incretin universe: from basic biology to clinical translation. Diabetologia 66, 1765–1779 (2023).
El, K. et al. GIP mediates the incretin effect and glucose tolerance by dual actions on α cells and β cells. Sci. Adv. 7, eabf1948 (2021).
Turton, M. et al. A role for glucagon-like peptide-1 in the central regulation of feeding. Nature 379, 69–72 (1996).
Flint, A., Raben, A., Astrup, A. & Holst, J. J. Glucagon-like peptide 1 promotes satiety and suppresses energy intake in humans. J. Clin. Invest. 101, 515–520 (1998).
Liskiewicz, A. & Müller, T. D. Regulation of energy metabolism through central GIPR signaling. Peptides 176, 171198 (2024).
Finan, B. et al. Reappraisal of GIP pharmacology for metabolic diseases. Trends Mol. Med. 22, 359–376 (2016).
Mentlein, R., Gallwitz, B. & Schmidt, W. E. Dipeptidyl‐peptidase IV hydrolyses gastric inhibitory polypeptide, glucagon‐like peptide‐1 (7–36) amide, peptide histidine methionine and is responsible for their degradation in human serum. Eur. J. Biochem. 214, 829–835 (1993).
Kusminski, C. M. et al. Transforming obesity: the advancement of multi-receptor drugs. Cell 187, 3829–3853 (2024).
Larsen, P. J., Tang-Christensen, M. & Holst, J. J. & Ørskov, C. Distribution of glucagon-like peptide-1 and other preproglucagon-derived peptides in the rat hypothalamus and brainstem. Neuroscience 77, 257–270 (1997).
Trapp, S. & Richards, J. E. The gut hormone glucagon-like peptide-1 produced in brain: is this physiologically relevant? Curr. Opin. Pharmacol. 13, 964–969 (2013).
Cork, S. C. et al. Distribution and characterisation of Glucagon-like peptide-1 receptor expressing cells in the mouse brain. Mol. Metab. 4, 718–731 (2015).
Fukuda, M. The role of GIP receptor in the CNS for the pathogenesis of obesity. Diabetes 70, 1929–1937 (2021).
Adriaenssens, A. E. et al. Glucose-dependent insulinotropic polypeptide receptor-expressing cells in the hypothalamus regulate food intake. Cell Metab. 30, 987–996.e6 (2019).
Kaplan, A. M. & Vigna, S. R. Gastric inhibitory polypeptide (GIP) binding sites in rat brain. Peptides 15, 297–302 (1994).
Usdin, T., Mezey, E., Button, D., Brownstein, M. & Bonner, T. Gastric inhibitory polypeptide receptor, a member of the secretin-vasoactive intestinal peptide receptor family, is widely distributed in peripheral organs and the brain. Endocrinology 133, 2861–2870 (1993).
Trapp, S. & Brierley, D. I. Brain GLP‐1 and the regulation of food intake: GLP‐1 action in the brain and its implications for GLP‐1 receptor agonists in obesity treatment. Br. J. Pharmacol. 179, 557–570 (2022).
Fortin, S. M. et al. The locus coeruleus contributes to the anorectic, nausea, and autonomic physiological effects of glucagon-like peptide-1. Sci. Adv. 9, eadh0980 (2023).
Graham, D. L. et al. A novel mouse model of glucagon-like peptide-1 receptor expression: a look at the brain. J. Comp. Neurol. 528, 2445–2470 (2020).
Smith, C. et al. A comparative transcriptomic analysis of glucagon-like peptide-1 receptor- and glucose-dependent insulinotropic polypeptide receptor-expressing cells in the hypothalamus. Appetite 174, 106022 (2022).
Steuernagel, L. et al. HypoMap—a unified single-cell gene expression atlas of the murine hypothalamus. Nat. Metab. 4, 1402–1419 (2022).
Tadross, J. A. et al. A comprehensive spatio-cellular map of the human hypothalamus. Nature 639, 708–716 (2025).
Muller, T. D. et al. Glucagon-like peptide 1 (GLP-1). Mol. Metab. 30, 72–130 (2019).
Kurtzhals, P., Østergaard, S., Nishimura, E. & Kjeldsen, T. Derivatization with fatty acids in peptide and protein drug discovery. Nat. Rev. Drug Discov. 22, 59–80 (2023).
Knudsen, L. B. Inventing liraglutide, a glucagon-like peptide-1 analogue, for the treatment of diabetes and obesity. ACS Pharmacol. Transl. Sci. 2, 468–484 (2019).
Knudsen, L. B. & Lau, J. The discovery and development of liraglutide and semaglutide. Front. Endocrinol. 10, 155 (2019).
Griffith, D. A. et al. A small-molecule oral agonist of the human glucagon-like peptide-1 receptor. J. Med. Chem. 65, 8208–8226 (2022).
Wharton, S. et al. Daily oral GLP-1 receptor agonist orforglipron for adults with obesity. N. Engl. J. Med. 389, 877–888 (2023).
Wang, Z. J. et al. Semaglutide ameliorates cognition and glucose metabolism dysfunction in the 3xTg mouse model of Alzheimer’s disease via the GLP-1R/SIRT1/GLUT4 pathway. Neuropharmacology 240, 109716 (2023).
Qin, W. et al. Neuronal SIRT1 activation as a novel mechanism underlying the prevention of Alzheimer disease amyloid neuropathology by calorie restriction. J. Biol. Chem. 281, 21745–21754 (2006).
Xie, X.-Y., Mo, Z.-H., Chen, K., He, H.-H. & Xie, Y.-H. Glucagon-like peptide-1 improves proliferation and differentiation of endothelial progenitor cells via upregulating VEGF generation. Med. Sci. Monit. 17, BR35 (2011).
Jais, A. et al. Myeloid-cell-derived VEGF maintains brain glucose uptake and limits cognitive impairment in obesity. Cell 165, 882–895 (2016).
Gejl, M. et al. Blood–brain glucose transfer in Alzheimer’s disease: effect of GLP-1 analog treatment. Sci. Rep. 7, 17490 (2017).
Fontanella, R. A. et al. Tirzepatide prevents neurodegeneration through multiple molecular pathways. J. Transl. Med. 22, 114 (2024).
Athauda, D. et al. Utility of neuronal-derived exosomes to examine molecular mechanisms that affect motor function in patients with Parkinson disease: a secondary analysis of the Exenatide-PD Trial. JAMA Neurol 76, 420–429, (2019).
Paladugu, L. et al. Liraglutide has anti-inflammatory and anti-amyloid properties in Streptozotocin-induced and 5xFAD mouse models of Alzheimer’s disease. Int. J. Mol. Sci. 22, 860 (2021).
Hogan, A. E. et al. Glucagon-like peptide 1 analogue therapy directly modulates innate immune-mediated inflammation in individuals with type 2 diabetes mellitus. Diabetologia 57, 781–784 (2014).
Wong, C. K. et al. Divergent roles for the gut intraepithelial lymphocyte GLP-1R in control of metabolism, microbiota, and T cell-induced inflammation. Cell Metab. 34, 1514–1531(2022).
Ellingsgaard, H. et al. Interleukin-6 enhances insulin secretion by increasing glucagon-like peptide-1 secretion from L cells and alpha cells. Nat. Med. 17, 1481–1489 (2011).
Kahles, F. et al. GLP-1 secretion is increased by inflammatory stimuli in an IL-6-dependent manner, leading to hyperinsulinemia and blood glucose lowering. Diabetes 63, 3221–3229 (2014).
Heng, T. S., Painter, M. W. & Immunological Genome Project, C. The Immunological Genome Project: networks of gene expression in immune cells. Nat. Immunol. 9, 1091–1094 (2008).
Wong, C. K. et al. Central glucagon-like peptide 1 receptor activation inhibits Toll-like receptor agonist-induced inflammation. Cell Metab. 36, 130–143 (2023).
Wu, H. Y., Tang, X. Q., Mao, X. F. & Wang, Y. X. Autocrine interleukin-10 mediates glucagon-like peptide-1 receptor-induced spinal microglial beta-endorphin expression. J. Neurosci. 37, 11701–11714 (2017).
Kopp, K. O., Glotfelty, E. J., Li, Y. & Greig, N. H. Glucagon-like peptide-1 (GLP-1) receptor agonists and neuroinflammation: implications for neurodegenerative disease treatment. Pharmacol. Res. 186, 106550 (2022).
Heneka, M. T. et al. NLRP3 is activated in Alzheimer’s disease and contributes to pathology in APP/PS1 mice. Nature 493, 674–678 (2013).
Ising, C. et al. NLRP3 inflammasome activation drives tau pathology. Nature 575, 669–673 (2019).
Ammar, R. A., Mohamed, A. F., Kamal, M. M., Safar, M. M. & Abdelkader, N. F. Neuroprotective effect of liraglutide in an experimental mouse model of multiple sclerosis: role of AMPK/SIRT1 signaling and NLRP3 inflammasome. Inflammopharmacology 30, 919–934 (2022).
Kim, S., Moon, M. & Park, S. Exendin-4 protects dopaminergic neurons by inhibition of microglial activation and matrix metalloproteinase-3 expression in an animal model of Parkinson’s disease. J. Endocrinol. 202, 431–439 (2009).
Zhang, L., Zhang, L., Li, L. & Holscher, C. Neuroprotective effects of the novel GLP-1 long acting analogue semaglutide in the MPTP Parkinson’s disease mouse model. Neuropeptides 71, 70–80 (2018).
Zhang, L., Zhang, L., Li, L. & Holscher, C. Semaglutide is neuroprotective and reduces alpha-synuclein levels in the chronic MPTP mouse model of Parkinson’s disease. J. Parkinsons Dis. 9, 157–171 (2019).
Li, Y. et al. Exendin-4 ameliorates motor neuron degeneration in cellular and animal models of amyotrophic lateral sclerosis. PLoS ONE 7, e32008 (2012).
Knippenberg, S., Thau, N., Dengler, R., Brinker, T. & Petri, S. Intracerebroventricular injection of encapsulated human mesenchymal cells producing glucagon-like peptide 1 prolongs survival in a mouse model of ALS. PLoS ONE 7, e36857 (2012).
Verma, S. et al. Effects of once-weekly semaglutide 2.4 mg on C-reactive protein in adults with overweight or obesity (STEP 1, 2, and 3): exploratory analyses of three randomised, double-blind, placebo-controlled, phase 3 trials. eClinicalMedicine 55, 101737 (2023).
Wilson, J. M. et al. The dual glucose-dependent insulinotropic polypeptide and glucagon-like peptide-1 receptor agonist tirzepatide improves cardiovascular risk biomarkers in patients with type 2 diabetes: a post hoc analysis. Diabetes Obes. Metab. 24, 148–153 (2022).
Bomba, M. et al. Exenatide exerts cognitive effects by modulating the BDNF–TrkB neurotrophic axis in adult mice. Neurobiol. Aging 64, 33–43 (2018).
Bomba, M. et al. Exenatide reverts the high-fat-diet-induced impairment of BDNF signaling and inflammatory response in an animal model of Alzheimer’s disease. J. Alzheimers Dis. 70, 793–810 (2019).
Li, Y., Liu, W., Li, L. & Hölscher, C. D-Ala2-GIP-glu-PAL is neuroprotective in a chronic Parkinson’s disease mouse model and increases BNDF expression while reducing neuroinflammation and lipid peroxidation. Eur. J. Pharmacol. 797, 162–172 (2017).
Parkhurst, C. N. et al. Microglia promote learning-dependent synapse formation through brain-derived neurotrophic factor. Cell 155, 1596–1609 (2013).
McClean, P. L., Jalewa, J. & Holscher, C. Prophylactic liraglutide treatment prevents amyloid plaque deposition, chronic inflammation and memory impairment in APP/PS1 mice. Behav. Brain Res. 293, 96–106 (2015).
McClean, P. L. & Holscher, C. Liraglutide can reverse memory impairment, synaptic loss and reduce plaque load in aged APP/PS1 mice, a model of Alzheimer’s disease. Neuropharmacology 76, 57–67 (2014).
Wang, X. et al. Exendin-4 antagonizes Aβ1–42-induced attenuation of spatial learning and memory ability. Exp. Ther. Med. 12, 2885–2892 (2016).
Perry, T. et al. A novel neurotrophic property of glucagon-like peptide 1: a promoter of nerve growth factor-mediated differentiation in PC12 cells. J. Pharmacol. Exp. Ther. 300, 958–966 (2002).
Bertilsson, G. 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. 86, 326–338 (2008).
Ising, C. & Heneka, M. T. Functional and structural damage of neurons by innate immune mechanisms during neurodegeneration. Cell Death Dis. 9, 120 (2018).
Watson, K. T. et al. Neural correlates of liraglutide effects in persons at risk for Alzheimer’s disease. Behav. Brain Res. 356, 271–278 (2019).
Perry, T., Haughey, N. J., Mattson, M. P., Egan, J. M. & Greig, N. H. Protection and reversal of excitotoxic neuronal damage by glucagon-like peptide-1 and exendin-4. J. Pharmacol. Exp. Ther. 302, 881–888 (2002).
Liu, W. et al. Neuroprotective effects of lixisenatide and liraglutide in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mouse model of Parkinson’s disease. Neuroscience 303, 42–50 (2015).
Jia, X. T. et al. Exendin-4, a glucagon-like peptide 1 receptor agonist, protects against amyloid-beta peptide-induced impairment of spatial learning and memory in rats. Physiol. Behav. 159, 72–79 (2016).
Li, Y., Liu, W., Li, L. & Hölscher, C. Neuroprotective effects of a GIP analogue in the MPTP Parkinson’s disease mouse model. Neuropharmacology 101, 255–263 (2016).
Wang, Z. J. et al. Semaglutide promotes the transition of microglia from M1 to M2 type to reduce brain inflammation in APP/PS1/tau mice. Neuroscience 563, 222–234 (2024).
Yang, S. et al. Tirzepatide shows neuroprotective effects via regulating brain glucose metabolism in APP/PS1 mice. Peptides 179, 171271 (2024).
Yamamoto, E. A. et al. The perivascular space is a conduit for cerebrospinal fluid flow in humans: a proof-of-principle report. Proc. Natl Acad. Sci. USA 121, e2407246121 (2024).
Huang, S. Y. et al. Glymphatic system dysfunction predicts amyloid deposition, neurodegeneration, and clinical progression in Alzheimer’s disease. Alzheimers Dement. 20, 3251–3269 (2024).
Rasmussen, M. K., Mestre, H. & Nedergaard, M. The glymphatic pathway in neurological disorders. Lancet Neurol. 17, 1016–1024 (2018).
Beschorner, N. & Nedergaard, M. Glymphatic system dysfunction in neurodegenerative diseases. Curr. Opin. Neurol. 37, 182–188 (2024).
Lohela, T. J., Lilius, T. O. & Nedergaard, M. The glymphatic system: implications for drugs for central nervous system diseases. Nat. Rev. Drug Discov. 21, 763–779 (2022).
Wu, W. et al. Modulation of glymphatic system by visual circuit activation alleviates memory impairment and apathy in a mouse model of Alzheimer’s disease. Nat. Commun. 16, 63 (2025).
Murdock, M. H. et al. Multisensory gamma stimulation promotes glymphatic clearance of amyloid. Nature 627, 149–156 (2024).
Sasaki, K. et al. GLP-1 receptor signaling restores aquaporin 4 subcellular polarization in reactive astrocytes and promotes amyloid beta clearance in a mouse model of Alzheimer’s disease. Biochem. Biophys. Res. Commun. 741, 151016 (2024).
Lv, C. et al. Cerebral glucagon-like peptide-1 receptor activation alleviates traumatic brain injury by glymphatic system regulation in mice. CNS Neurosci. Ther. 29, 3876–3888 (2023).
During, M. J. et al. Glucagon-like peptide-1 receptor is involved in learning and neuroprotection. Nat. Med. 9, 1173–1179 (2003).
McClean, P. L., Parthsarathy, V., Faivre, E. & Holscher, C. The diabetes drug liraglutide prevents degenerative processes in a mouse model of Alzheimer’s disease. J. Neurosci. 31, 6587–6594 (2011).
Han, W. N. et al. Liraglutide protects against amyloid-beta protein-induced impairment of spatial learning and memory in rats. Neurobiol. Aging 34, 576–588 (2013).
Xiong, H. et al. The neuroprotection of liraglutide on Alzheimer-like learning and memory impairment by modulating the hyperphosphorylation of tau and neurofilament proteins and insulin signaling pathways in mice. J. Alzheimers Dis. 37, 623–635 (2013).
Hansen, H. H. et al. The GLP-1 receptor agonist liraglutide improves memory function and increases hippocampal CA1 neuronal numbers in a senescence-accelerated mouse model of Alzheimer’s disease. J. Alzheimers Dis. 46, 877–888 (2015).
Zheng, J. et al. GLP-1 improves the supportive ability of astrocytes to neurons by promoting aerobic glycolysis in Alzheimer’s disease. Mol. Metab. 47, 101180 (2021).
Zhang, M. et al. Glucagon-like peptide-1 analogs mitigate neuroinflammation in Alzheimer’s disease by suppressing NLRP2 activation in astrocytes. Mol. Cell. Endocrinol. 542, 111529 (2022).
Cai, H. Y. et al. Lixisenatide rescues spatial memory and synaptic plasticity from amyloid beta protein-induced impairments in rats. Neuroscience 277, 6–13 (2014).
Zhang, Y. et al. Semaglutide ameliorates Alzheimer’s disease and restores oxytocin in APP/PS1 mice and human brain organoid models. Biomed. Pharmacother. 180, 117540 (2024).
Chen, S. et al. Liraglutide improves water maze learning and memory performance while reduces hyperphosphorylation of tau and neurofilaments in APP/PS1/Tau triple transgenic mice. Neurochem. Res. 42, 2326–2335 (2017).
Qi, L. et al. Subcutaneous administration of liraglutide ameliorates learning and memory impairment by modulating tau hyperphosphorylation via the glycogen synthase kinase-3beta pathway in an amyloid beta protein induced alzheimer disease mouse model. Eur. J. Pharmacol. 783, 23–32 (2016).
Guo, X. et al. Tirzepatide ameliorates spatial learning and memory impairment through modulation of aberrant insulin resistance and inflammation response in diabetic rats. Front. Pharmacol. 14, 1146960 (2023).
Li, T. et al. A GLP-1/GIP/Gcg receptor triagonist improves memory behavior, as well as synaptic transmission, neuronal excitability and Ca2+ homeostasis in 3xTg-AD mice. Neuropharmacology 170, 108042 (2020).
Forny Germano, L. et al. The GLP-1 medicines semaglutide and tirzepatide do not alter disease-related pathology, behaviour or cognitive function in 5XFAD and APP/PS1 mice. Mol. Metab. 89, 102019 (2024).
Gejl, M. et al. In Alzheimer’s disease, 6-month treatment with GLP-1 analog prevents decline of brain glucose metabolism: randomized, placebo-controlled, double-blind clinical trial. Front Aging Neurosci 8, 108 (2016).
Cukierman-Yaffe, T. et al. Effect of dulaglutide on cognitive impairment in type 2 diabetes: an exploratory analysis of the REWIND trial. Lancet Neurol. 19, 582–590 (2020).
Norgaard, C. H. et al. Treatment with glucagon-like peptide-1 receptor agonists and incidence of dementia: Data from pooled double-blind randomized controlled trials and nationwide disease and prescription registers. Alzheimers Dement. 8, e12268 (2022).
Edison, P. et al. Evaluation of liraglutide in the treatment of Alzheimer’s disease. Alzheimer’s Dement. 17, e057848 (2021).
Li, Y. 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. USA 106, 1285–1290 (2009).
Harkavyi, A. et al. Glucagon-like peptide 1 receptor stimulation reverses key deficits in distinct rodent models of Parkinson’s disease. J. Neuroinflammation 5, 19 (2008).
Yu, Y. W. et al. Glucose-dependent insulinotropic polypeptide mitigates 6-OHDA-induced behavioral impairments in Parkinsonian rats. Int. J. Mol. Sci. 19, 1153 (2018).
Feng, P. et al. Two novel dual GLP-1/GIP receptor agonists are neuroprotective in the MPTP mouse model of Parkinson’s disease. Neuropharmacology 133, 385–394 (2018).
Aviles-Olmos, I. et al. Exenatide and the treatment of patients with Parkinson’s disease. J. Clin. Invest. 123, 2730–2736 (2013).
Aviles-Olmos, I. et al. Motor and cognitive advantages persist 12 months after exenatide exposure in Parkinson’s disease. J. Parkinsons Dis. 4, 337–344 (2014).
Athauda, D. et al. Exenatide once weekly versus placebo in Parkinson’s disease: a randomised, double-blind, placebo-controlled trial. Lancet 390, 1664–1675 (2017).
Hogg, E. et al. A phase II, randomized, double-blinded, placebo-controlled trial of liraglutide in Parkinson’s disease. Lancet https://doi.org/10.2139/ssrn.4212371 (2022).
Meissner, W. G. et al. Trial of lixisenatide in early Parkinson’s disease. N. Engl. J. Med. 390, 1176–1185 (2024).
Vijiaratnam, N. et al. Exenatide once a week versus placebo as a potential disease-modifying treatment for people with Parkinson’s disease in the UK: a phase 3, multicentre, double-blind, parallel-group, randomised, placebo-controlled trial. Lancet 405, 627–636 (2025).
McGarry, A. et al. Safety, tolerability, and efficacy of NLY01 in early untreated Parkinson’s disease: a randomised, double-blind, placebo-controlled trial. Lancet Neurol. 23, 37–45 (2024).
Keerie, A. et al. The GLP-1 receptor agonist, liraglutide, fails to slow disease progression in SOD1(G93A) and TDP-43(Q331K) transgenic mouse models of ALS. Sci. Rep. 11, 17027 (2021).
Motataianu, A. et al. Exploring the role of metabolic hormones in amyotrophic lateral sclerosis. Int. J. Mol. Sci. 25, 5059 (2024).
Kuyucu, E., Gumus, B., Erbas, O., Oltulu, F. & Bora, A. Exenatide promotes regeneration of injured rat sciatic nerve. Neural Regen. Res. 12, 637–643, (2017).
Yamamoto, K. et al. Therapeutic effect of exendin-4, a long-acting analogue of glucagon-like peptide-1 receptor agonist, on nerve regeneration after the crush nerve injury. BioMed Res. Int. 2013, 315848 (2013).
Takaku, S., Tsukamoto, M., Niimi, N., Yako, H. & Sango, K. Exendin-4 promotes schwann cell survival/migration and myelination in vitro. Int. J. Mol. Sci. 22, 2971 (2021).
Lee, C. H. et al. Activation of glucagon-like peptide-1 receptor promotes neuroprotection in experimental autoimmune encephalomyelitis by reducing neuroinflammatory responses. Mol. Neurobiol. 55, 3007–3020 (2018).
DellaValle, B. et al. Glucagon-like peptide-1 analog, liraglutide, delays onset of experimental autoimmune encephalitis in lewis rats. Front. Pharmacol. 7, 433 (2016).
Chiou, H. C. et al. Dulaglutide modulates the development of tissue-infiltrating TH1/TH17 cells and the pathogenicity of encephalitogenic TH1 cells in the central nervous system. Int. J. Mol. Sci. 20, 1584 (2019).
Ong, L. K. Beyond thE Primary Infarction: Focus on Mechanisms Related to Secondary Neurodegeneration after Stroke. Int. J. Mol. Sci. 23, 16024 (2022).
Verges, B. et al. Protection against stroke with glucagon-like peptide-1 receptor agonists: a comprehensive review of potential mechanisms. Cardiovasc. Diabetol. 21, 242 (2022).
Teramoto, S. et al. Exendin-4, a glucagon-like peptide-1 receptor agonist, provides neuroprotection in mice transient focal cerebral ischemia. J. Cereb. Blood Flow Metab. 31, 1696–1705 (2011).
Basalay, M. V., Davidson, S. M. & Yellon, D. M. Neuroprotection in rats following ischaemia-reperfusion injury by GLP-1 analogues-liraglutide and semaglutide. Cardiovasc. Drugs Ther. 33, 661–667 (2019).
Marlet, I. R., Olmestig, J. N. E., Vilsboll, T., Rungby, J. & Kruuse, C. Neuroprotective mechanisms of glucagon-like peptide-1-based therapies in ischaemic stroke: a systematic review based on pre-clinical studies. Basic Clin. Pharmacol. Toxicol. 122, 559–569 (2018).
Maskery, M. P. et al. Glucagon-like peptide-1 receptor agonists as neuroprotective agents for ischemic stroke: a systematic scoping review. J. Cereb. Blood Flow Metab. 41, 14–30 (2021).
Bellastella, G. et al. Glucagon-like peptide-1 receptor agonists and prevention of stroke systematic review of cardiovascular outcome trials with meta-analysis. Stroke 51, 666–669 (2020).
Sattar, N. 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. 9, 653–662 (2021).
Lin, D. S., Lee, J. K. & Chen, W. J. Major adverse cardiovascular and limb events in patients with diabetes treated with GLP-1 receptor agonists vs DPP-4 inhibitors. Diabetologia 64, 1949–1962 (2021).
Bassil, F. et al. Insulin resistance and exendin-4 treatment for multiple system atrophy. Brain 140, 1420–1436 (2017).
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. 180, 114187 (2020).
Rhea, E. M. et al. Brain uptake pharmacokinetics of albiglutide, dulaglutide, tirzepatide, and DA5-CH in the search for new treatments of Alzheimer’s and Parkinson’s diseases. Tissue Barriers 12, 2292461 (2023).
Secher, A. et al. The arcuate nucleus mediates GLP-1 receptor agonist liraglutide-dependent weight loss. J. Clin. Invest. 124, 4473–4488 (2014).
Gabery, S. et al. Semaglutide lowers body weight in rodents via distributed neural pathways. JCI Insight 5, e133429 (2020).
Skovbjerg, G. et al. Uncovering CNS access of lipidated exendin-4 analogues by quantitative whole-brain 3D light sheet imaging. Neuropharmacology 238, 109637 (2023).
Adriaenssens, A. et al. Hypothalamic and brainstem glucose-dependent insulinotropic polypeptide receptor neurons employ distinct mechanisms to affect feeding. JCI Insight 8, e164921 (2023).
Garcia-Caceres, C. et al. Role of astrocytes, microglia, and tanycytes in brain control of systemic metabolism. Nat. Neurosci. 22, 7–14 (2019).
Imbernon, M. et al. Tanycytes control hypothalamic liraglutide uptake and its anti-obesity actions. Cell Metab. 34, 1054–1063(2022).
Sweeney, M. D., Sagare, A. P. & Zlokovic, B. V. Blood–brain barrier breakdown in Alzheimer disease and other neurodegenerative disorders. Nat. Rev. Neurol. 14, 133–150 (2018).
Hisadome, K., Reimann, F., Gribble, F. M. & Trapp, S. Leptin directly depolarizes preproglucagon neurons in the nucleus tractus solitarius: electrical properties of glucagon-like peptide 1 neurons. Diabetes 59, 1890–1898 (2010).
Brierley, D. I. et al. Central and peripheral GLP-1 systems independently suppress eating. Nat. Metab. 3, 258–273 (2021).
Llewellyn-Smith, I. J., Reimann, F., Gribble, F. M. & Trapp, S. Preproglucagon neurons project widely to autonomic control areas in the mouse brain. Neuroscience 180, 111–121 (2011).
Hsu, T. M., Hahn, J. D., Konanur, V. R., Lam, A. & Kanoski, S. E. Hippocampal GLP-1 receptors influence food intake, meal size, and effort-based responding for food through volume transmission. Neuropsychopharmacology 40, 327–337 (2015).
Montaner, M. et al. A neuronal circuit driven by GLP-1 in the olfactory bulb regulates insulin secretion. Nat. Commun. 15, 6941 (2024).
Thiebaud, N. et al. The incretin hormone glucagon-like peptide 1 increases mitral cell excitability by decreasing conductance of a voltage-dependent potassium channel. J. Physiol. 594, 2607–2628 (2016).
Zueco, J. A. et al. Coexpression of glucagon-like peptide-(GLP-1) receptor, vasopressin, and oxytocin mRNAs in neurons of the rat hypothalamic supraoptic and paraventricular nuclei: effect of GLP-17–36 amide on vasopressin and oxytocin release. J. Neurochem. 72, 10–16 (1999).
Lin, Y. T., Chen, C. C., Huang, C. C., Nishimori, K. & Hsu, K. S. Oxytocin stimulates hippocampal neurogenesis via oxytocin receptor expressed in CA3 pyramidal neurons. Nat. Commun. 8, 537 (2017).
Chaudhuri, A. et al. Exenatide exerts a potent antiinflammatory effect. J. Clin. Endocrinol. Metab. 97, 198–207 (2012).
Kappe, C., Tracy, L. M., Patrone, C., Iverfeldt, K. & Sjoholm, A. GLP-1 secretion by microglial cells and decreased CNS expression in obesity. J. Neuroinflammation 9, 276 (2012).
Jin, H., Li, M., Jeong, E., Castro-Martinez, F. & Zuker, C. S. A body–brain circuit that regulates body inflammatory responses. Nature 630, 695–703 (2024).
Cummings, J. L. et al. Evoke and evoke+: design of two large-scale, double-blind, placebo-controlled, phase 3 studies evaluating efficacy, safety, and tolerability of semaglutide in early-stage symptomatic Alzheimer’s disease. Alzheimers Res. Ther. 17, 14 (2025).
Willard, F. S. et al. Tirzepatide is an imbalanced and biased dual GIP and GLP-1 receptor agonist. JCI Insight 5, e140532 (2020).
Borner, T. et al. GIP receptor agonism attenuates GLP-1 receptor agonist–induced nausea and emesis in preclinical models. Diabetes 70, 2545–2553 (2021).
Yusta, B. et al. GLP-1R agonists modulate enteric immune responses through the intestinal intraepithelial lymphocyte GLP-1R. Diabetes 64, 2537–2549 (2015).
Quarta, C. et al. Molecular integration of incretin and glucocorticoid action reverses immunometabolic dysfunction and obesity. Cell Metab. 26, 620–632 (2017).
Decarie-Spain, L. et al. GLP-1/dexamethasone inhibits food reward without inducing mood and memory deficits in mice. Neuropharmacology 151, 55–63 (2019).
Finan, B. et al. Targeted estrogen delivery reverses the metabolic syndrome. Nat. Med. 18, 1847–1856 (2012).
Ge, F. et al. Ovariectomy induces microglial cell activation and inflammatory response in rat prefrontal cortices to accelerate the chronic unpredictable stress‐mediated anxiety and depression. BioMed Res. Int. 2020, 3609758 (2020).
Rentzeperi, E., Pegiou, S., Koufakis, T., Grammatiki, M. & Kotsa, K. Sex differences in response to treatment with glucagon-like peptide 1 receptor agonists: opportunities for a tailored approach to diabetes and obesity care. J. Pers. Med. 12, 454 (2022).
Bianco, A., Antonacci, Y. & Liguori, M. Sex and gender differences in neurodegenerative diseases: challenges for therapeutic opportunities. Int. J. Mol. Sci. 24, 6354 (2023).
Liu, J., Chang, L., Song, Y., Li, H. & Wu, Y. The role of NMDA receptors in Alzheimer’s disease. Front. Neurosci. 13, 43 (2019).
Petersen, J. et al. GLP-1-directed NMDA receptor antagonism for obesity treatment. Nature 629, 1133–1141 (2024).
Wu, D. et al. The blood–brain barrier: structure, regulation, and drug delivery. Signal Transduct. Target.Ther. 8, 217 (2023).
Terstappen, G. C., Meyer, A. H., Bell, R. D. & Zhang, W. Strategies for delivering therapeutics across the blood–brain barrier. Nat. Rev. Drug Discovery. 20, 362–383 (2021).
Godschall, E. N. et al. A brain reward circuit inhibited by next-generation weight loss drugs. Preprint at bioRxiv https://doi.org/10.1101/2024.12.12.628169 (2024).
Acknowledgements
We thank previous and current members of the Clemmensen Group for scientific discussions. This work was supported by the Novo Nordisk Foundation (NNF22OC0073778). The Novo Nordisk Foundation Center for Basic Metabolic Research is an independent research centre, based at the University of Copenhagen, Denmark, and is partially funded by an unconditional donation from the Novo Nordisk Foundation (www.cbmr.ku.dk) (grant numbers NNF18CC0034900 and NNF23SA0084103).
Author information
Authors and Affiliations
Contributions
A.V. and C.C. drafted the manuscript and created the visual elements; M.T.H. critically revised the manuscript for important intellectual content. All authors provided input and approved the final version of the manuscript.
Corresponding author
Ethics declarations
Competing interests
C.C. is a cofounder of Ousia Pharma, a biotechnology company developing therapeutics for treatment of obesity. M.T.H. is a scientific advisory board member at Alector the Dementia Discovery Fund and Muna Therapeutics and has received honoraria for oral presentations from Pfizer, Novartis, Roche, Abbvie and Biogen. A.V. does not declare any competing interests.
Peer review
Peer review information
Nature Metabolism thanks Carol Troy, Stefan Trapp, Xu Chen and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editor: Christoph Schmitt, in collaboration with the Nature Metabolism editorial team.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.
About this article
Cite this article
Vear, A., Heneka, M.T. & Clemmensen, C. Incretin-based therapeutics for the treatment of neurodegenerative diseases. Nat Metab 7, 679–696 (2025). https://doi.org/10.1038/s42255-025-01263-4
Received:
Accepted:
Published:
Issue date:
DOI: https://doi.org/10.1038/s42255-025-01263-4