GLP-1 Analogs, SGLT-2, and DPP-4 Inhibitors: A Triad of Hope for Alzheimer’s Disease Therapy
Abstract
:1. Introduction
2. Metabolic and Molecular Links between Diabetes and Alzheimer’s Disease
2.1. Insulin Resistance and Brain Glucose Metabolism
2.2. Oxidative Stress and Inflammation
2.3. Intestinal Dysbiosis
2.4. Amylin and Amyloid-β (Aβ) Protein Pathologies
2.5. Tau Pathology and Neurofibrillary Tangles
2.6. Neurotransmitter Imbalance
2.7. Blood–Brain Barrier Disorders
3. From Blood Sugar Control to Brain Health: The Potential Role of Antidiabetic Drugs in Alzheimer’s Disease
3.1. GLP-1 Receptor Agonists
3.1.1. Mechanism of Action GLP-1 Receptor Agonists
3.1.2. Physiological and Pharmacokinetic Attributes of GLP-1 Receptor Agonists in Central Nervous System—Mechanisms and Benefits
3.1.3. Molecular Mechanisms Underlying the Neuroprotective Effects of GLP-1 Agonists’ Improvement of Insulin Sensitivity
Anti-Inflammatory Effects
Preventing Neuronal Cell Death and Neurotoxic Damage
Mitigating Oxidative Stress and Mitochondrial Dysfunction
Mitigating Aβ Aggregation and Tau Protein Hyperphosphorylation
3.2. Dipeptidyl Peptidase-4 (DPP-4) Inhibitors
3.2.1. Mechanism of Action of Dipeptidyl Peptidase-4 (DPP-4) Inhibitors—Pharmacokinetic and Pharmacological Characterization
3.2.2. Molecular Mechanisms Underlying the Neuroprotective Effects of DPP-4 Inhibitors
Improvement of Insulin Sensitivity
Anti-Inflammatory Effects
Preventing Neuronal Cell Death and Neurotoxic Damage
Mitigating Oxidative Stress and Mitochondrial Dysfunction
Mitigating Aβ Aggregation and Tau Protein Hyperphosphorylation
3.3. Sodium-Glucose Cotransporter 2 (SGLT-2) Inhibitors
3.3.1. Mechanism of Action of Sodium–Glucose Cotransporter 2 (SGLT-2) Inhibitors—Pharmacokinetic and Pharmacological Characterization
3.3.2. Physiological and Pharmacokinetic Attributes of SGLT-2 Inhibitors in the Central Nervous System—Mechanisms and Benefits
3.3.3. Molecular Mechanisms Underlying the Neuroprotective Effects of SGLT-2 Inhibitors’ Improvement of Insulin Sensitivity
Anti-Inflammatory Effects
Preventing Neuronal Cell Death and Neurotoxic Damage
mTOR Activation and Its Impact on Catabolic Pathways
Mitigating AB Aggregation and Tau Protein Hyperphosphorylation
4. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Nguyen, T.T.; Nguyen, T.T.D.; Nguyen, T.K.O.; Vo, T.K. Advances in developing therapeutic strategies for Alzheimer’s disease. Biomed. Pharmacother. 2021, 139, 111623. [Google Scholar] [CrossRef]
- Zhang, X.X.; Tian, Y.; Wang, Z.T.; Ma, Y.H.; Tan, L.; Yu, J.T. The epidemiology of Alzheimer’s disease modifiable risk factors and prevention. J. Prev. Alzheimers Dis. 2021, 8, 313–321. [Google Scholar] [CrossRef] [PubMed]
- Clark, K.; Leung, Y.Y.; Lee, W.P.; Voight, B.; Wang, L.S. Polygenic risk scores in Alzheimer’s disease genetics: Methodology, applications, inclusion, and diversity. J. Alzheimers Dis. 2022, 89, 1–12. [Google Scholar] [CrossRef] [PubMed]
- Silva, M.V.F.; Loures, C.D.M.G.; Alves, L.C.V.; de Souza, L.C.; Borges, K.B.G.; Carvalho, M.D.G. Alzheimer’s disease: Risk factors and potentially protective measures. J. Biomed. Sci. 2019, 26, 33. [Google Scholar] [CrossRef]
- Dave, B.P.; Shah, Y.B.; Maheshwari, K.G.; Mansuri, K.A.; Prajapati, B.S.; Postwala, H.I.; Chorawala, M.R. Pathophysiological Aspects and Therapeutic Armamentarium of Alzheimer’s Disease: Recent Trends and Future Development. Cell. Mol. Neurobiol. 2023, 1–38. [Google Scholar] [CrossRef]
- Dewanjee, S.; Chakraborty, P.; Bhattacharya, H.; Chacko, L.; Singh, B.; Chaudhary, A.; Javvaji, K.; Pradhan, S.R.; Vallamkondu, J.; Dey, A.; et al. Altered glucose metabolism in Alzheimer’s disease: Role of mitochondrial dysfunction and oxidative stress. Free Radic. Biol. Med. 2022, 193, 134–157. [Google Scholar] [CrossRef]
- Kevadiya, B.D.; Ottemann, B.M.; Thomas, M.B.; Mukadam, I.; Nigam, S.; McMillan, J.; Gorantla, S.; Bronich, T.K.; Edagwa, B.; Gendelman, H.E. Neurotheranostics as personalized medicines. Adv. Drug Deliv. Rev. 2019, 148, 252–289. [Google Scholar] [CrossRef]
- Tremblay, A.J.; Lamarche, B.; Deacon, C.F.; Weisnagel, S.J.; Couture, P. Effects of sitagliptin therapy on markers of low-grade inflammation and cell adhesion molecules in patients with type 2 diabetes. Metabolism 2014, 63, 1141–1148. [Google Scholar] [CrossRef] [PubMed]
- Liu, X.; Men, P.; Wang, B.; Cai, G.; Zhao, Z. Effect of dipeptidyl-peptidase-4 inhibitors on C-reactive protein in patients with type 2 diabetes: A systematic review and meta-analysis. Lipids Health Dis. 2019, 18, 144. [Google Scholar] [CrossRef]
- Piątkowska-Chmiel, I.; Gawrońska-Grzywacz, M.; Popiołek, Ł.; Herbet, M.; Dudka, J. The novel adamantane derivatives as potential mediators of inflammation and neural plasticity diabetietes mice with cognitive impairment. Sci. Rep. 2022, 12, 6708. [Google Scholar] [CrossRef]
- Kim, Y.J.; Kim, Y.J.; Lee, S.H.; Kim, H.J. DPP-4 inhibitor vildagliptin enhances the expression of BDNF and synapsin I in the hippocampus of diabetic mice. Neurosci. Lett. 2016, 619, 46–51. [Google Scholar]
- Freeman, A.M.; Acevedo, L.A.; Pennings, N. Insulin Resistance; StatPearls Publishing: St. Petersburg, FA, USA, 2023. [Google Scholar]
- Arnold, S.E.; Arvanitakis, Z.; Macauley-Rambach, S.L.; Koenig, A.M.; Wang, H.Y.; Ahima, R.S.; Craft, S.; Gandy, S.; Buettner, C.; Stoeckel, L.E.; et al. Brain insulin resistance in type 2 diabetes and Alzheimer disease: Concepts and conundrums. Nat. Rev. Neurol. 2018, 14, 168–181. [Google Scholar] [CrossRef] [PubMed]
- De la Monte, S.M. Contributions of brain insulin resistance and deficiency in amyloid-related neurodegeneration in Alzheimer’s disease. Drugs 2012, 72, 49–66. [Google Scholar] [CrossRef] [PubMed]
- Hoyer, S. Is sporadic Alzheimer disease the brain type of non-insulin dependent diabetes mellitus? A challenging hypothesis. J. Neural Transm. 1998, 105, 415–422. [Google Scholar] [CrossRef]
- Szablewski, L. Glucose transporters in brain: In health and in Alzheimer’s disease. J. Alzheimers Dis. 2017, 55, 1307–1320. [Google Scholar] [CrossRef] [PubMed]
- Aliev, G.; Smith, M.A.; Obrenovich, M.E.; de la Torre, J.C.; Perry, G. Role of vascular hypoperfusion-induced oxidative stress and mitochondria failure in the pathogenesis of Azheimer disease. Neurotox. Res. 2003, 5, 491–504. [Google Scholar] [CrossRef]
- Wang, W.; Zhao, F.; Ma, X.; Perry, G.; Zhu, X. Mitochondria dysfunction in the pathogenesis of Alzheimer’s disease: Recent advances. Mol. Neurodegener. 2020, 15, 30. [Google Scholar] [CrossRef]
- Benzinger, T.L.; Blazey, T.; Jack Jr, C.R.; Koeppe, R.A.; Su, Y.; Xiong, C.; Raichle, M.E.; Snyder, A.Z.; Ances, B.M.; Bateman, R.J.; et al. Regional variability in biomarker imaging in autosomal dominant Alzheimer’s disease. Proc. Natl. Acad. Sci. USA 2013, 110, e4502–e4509. [Google Scholar] [CrossRef]
- Altmann, A.; Ng, B.; Landau, S.M.; Jagust, W.J.; Greicius, M.D. Alzheimer’s Disease Neuroimaging Initiative. Regional brain hypometabolism is unrelated to regional amyloid plaque burden. Brain 2015, 138 Pt 12, 3734–3746. [Google Scholar] [CrossRef]
- Rivera, E.J.; Goldin, A.; Fulmer, N.; Tavares, R.; Wands, J.R.; de la Monte, S.M. Insulin and insulin-like growth factor expression and function deteriorate with progression of Alzheimer’s disease: Link to brain reductions in acetylcholine. J. Alzheimers Dis. 2015, 8, 247–268. [Google Scholar] [CrossRef]
- Baker, L.D.; Cross, D.J.; Minoshima, S.; Belongia, D.; Watson, G.S.; Craft, S. Insulin resistance and Alzheimer-like reductions in regional cerebral glucose metabolism for cognitively normal adults with prediabetes or early type 2 diabetes. Arch. Neurol. 2011, 68, 51–57. [Google Scholar] [CrossRef] [PubMed]
- Park, S.A. A common pathogenic mechanism linking type-2 diabetes and Alzheimer’s disease: Evidence from animal models. J. Clin. Neurol. 2011, 7, 10–18. [Google Scholar] [CrossRef] [PubMed]
- Kimura, N. Diabetes Mellitus Induces Alzheimer’s Disease Pathology: Histopathological Evidence from Animal Models. Int. J. Mol. Sci. 2016, 17, 503. [Google Scholar] [CrossRef]
- Shankar, G.M.; Li, S.; Mehta, T.H.; Garcia-Munoz, A.; Shepardson, N.E.; Smith, I.; Brett, F.M.; Farrell, M.A.; Rowan, M.J.; Lemere, C.A.; et al. Amyloid-beta protein dimers isolated directly from Alzheimer’s brains impair synaptic plasticity and memory. Nat. Med. 2008, 14, 837–842. [Google Scholar] [CrossRef] [PubMed]
- Sędzikowska, A.; Szablewski, L. Insulin and Insulin Resistance in Alzheimer’s Disease. Int. J. Mol. Sci. 2021, 22, 9987. [Google Scholar] [CrossRef]
- Pandini, G.; Pace, V.; Copani, A.; Squatrito, S.; Milardi, D.; Vigneri, R. Insulin has multiple antiamyloidogenic effects on human neuronal cells. Endocrinology 2013, 154, 375–387. [Google Scholar] [CrossRef]
- Zhao, W.Q.; Lacor, P.N.; Chen, H.; Lambert, M.P.; Quon, M.J.; Krafft, G.A.; Klein, W.L. Insulin receptor dysfunction impairs cellular clearance of neurotoxic oligomeric a{beta}. J. Biol. Chem. 2009, 284, 18742–18753. [Google Scholar] [CrossRef]
- Currais, A.; Prior, M.; Lo, D.; Jolivalt, C.; Schubert, D.; Maher, P. Diabetes exacerbates amyloid and neurovascular pathology in aging-accelerated mice. Aging Cell 2012, 11, 1017–1026. [Google Scholar] [CrossRef]
- Devi, L.; Alldred, M.J.; Ginsberg, S.D.; Ohno, M. Mechanisms underlying insulin deficiency-induced acceleration of β-amyloidosis in a mouse model of Alzheimer’s disease. PLoS ONE 2012, 7, e32792. [Google Scholar] [CrossRef]
- Muriach, M.; Flores-Bellver, M.; Romero, F.J.; Barcia, J.M. Diabetes and the brain: Oxidative stress, inflammation, and autophagy. Oxidative Med. Cell. Longev. 2014, 2014, 102158. [Google Scholar] [CrossRef]
- Grillo, C.A.; Piroli, G.G.; Rosell, D.R.; Hoskin, E.K.; McEwen, B.S.; Reagan, L.P. Region specific increases in oxidative stress and superoxide dismutase in the hippocampus of diabetic rats subjected to stress. Neuroscience 2003, 121, 133–140. [Google Scholar] [CrossRef] [PubMed]
- Ulusu, N.N.; Sahilli, M.; Avci, A.; Canbolat, O.; Ozansoy, G.; Ari, N.; Bali, M.; Stefek, M.; Stolc, S.; Gajdosik, A.; et al. Pentose phosphate pathway, glutathione-dependent enzymes and antioxidant defense during oxidative stress in diabetic rodent brain and peripheral organs: Effects of stobadine and vitamin E. Neurochem. Res. 2003, 28, 815–823. [Google Scholar] [CrossRef]
- Varga, Z.V.; Giricz, Z.; Liaudet, L.; Haskó, G.; Ferdinandy, P.; Pacher, P. Interplay of oxidative, nitrosative/nitrative stress, inflammation, cell death and autophagy in diabetic cardiomyopathy. Biochim. Biophys. Acta (BBA)-Mol. Basis Dis. 2015, 1852, 232–242. [Google Scholar] [CrossRef]
- Shukla, A.; Jung, M.; Stern, M.; Fukagawa, N.K.; Taatjes, D.J.; Sawyer, D.; Van Houten, B.; Mossman, B.T. Asbestos induces mitochondrial DNA damage and dysfunction linked to the development of apoptosis. Am. J. Physiol. Lung Cell. Mol. Physiol. 2003, 285, L1018–L1025. [Google Scholar] [CrossRef]
- Merad-Boudia, M.; Nicole, A.; Santiard-Baron, D.; Saillé, C.; Ceballos-Picot, I. Mitochondrial impairment as an early event in the process of apoptosis induced by glutathione depletion in neuronal cells: Relevance to Parkinson’s disease. Biochem. Pharmacol. 1998, 56, 645–655. [Google Scholar] [CrossRef] [PubMed]
- Leuner, K.; Schütt, T.; Kurz, C.; Eckert, S.H.; Schiller, C.; Occhipinti, A.; Mai, S.; Jendrach, M.; Eckert, G.P.; Kruse, S.E.; et al. Mitochondrion-derived reactive oxygen species lead to enhanced amyloid beta formation. Antioxid. Redox Signal. 2012, 16, 1421–1433. [Google Scholar] [CrossRef]
- Butterfield, D.A.; Halliwell, B. Oxidative stress, dysfunctional glucose metabolism and Alzheimer disease. Nat. Rev. Neurosci. 2019, 20, 148–160. [Google Scholar] [CrossRef] [PubMed]
- Wang, X.; Wang, W.; Li, L.; Perry, G.; Lee, H.G.; Zhu, X. Oxidative stress and mitochondrial dysfunction in Alzheimer’s disease. Biochim. Biophys. Acta (BBA)-Mol. Basis Dis. 2014, 1842, 1240–1247. [Google Scholar] [CrossRef]
- Huang, Y.J.; Jin, M.H.; Pi, R.B.; Zhang, J.J.; Ouyang, Y.; Chao, X.J.; Chen, M.H.; Liu, P.Q.; Yu, J.C.; Ramassamy, C.; et al. Acrolein induces Alzheimer’s disease-like pathologies in vitro and in vivo. Toxicol. Lett. 2013, 217, 184–191. [Google Scholar] [CrossRef] [PubMed]
- Nunomura, A.; Moreira, P.I.; Castellani, R.J.; Lee, H.G.; Zhu, X.; Smith, M.A.; Perry, G. Oxidative damage to RNA in aging and neurodegenerative disorders. Neurotox. Res. 2012, 22, 231–248. [Google Scholar] [CrossRef]
- Tohgi, H.; Yonezawa, H.; Takahashi, S.; Sato, N.; Kato, E.; Kudo, M.; Hatano, K.; Sasaki, T. Cerebral blood flow and oxygen metabolism in senile dementia of Alzheimer’s type and vascular dementia with deep white matter changes. Neuroradiology 1998, 40, 131–137. [Google Scholar] [CrossRef] [PubMed]
- Buchan, R.J.; Nagata, K.; Yokoyama, E.; Langman, P.; Yuya, H.; Hirata, Y.; Hatazawa, J.; Kanno, I. Regional correlations between the EEG and oxygen metabolism in dementia of Alzheimer’s type. Electroencephalogr. Clin. Neurophysiol. 1997, 103, 409–417. [Google Scholar] [CrossRef] [PubMed]
- Meffert, M.K.; Baltimore, D. Physiological functions for brain NF-κB. Trends Neurosci. 2005, 28, 37–43. [Google Scholar] [CrossRef] [PubMed]
- Vaughan, S.; Jat, P.S. Deciphering the role of nuclear factor-κB in cellular senescence. Aging 2011, 3, 913. [Google Scholar] [CrossRef]
- Patel, S.; Santani, D. Role of NF-κB in the pathogenesis of diabetes and its associated complications. Pharmacol. Rep. 2009, 61, 595–603. [Google Scholar] [CrossRef]
- Incalza, M.A.; D’Oria, R.; Natalicchio, A.; Perrini, S.; Laviola, L.; Giorgino, F. Oxidative stress and reactive oxygen species in endothelial dysfunction associated with cardiovascular and metabolic diseases. Vasc. Pharmacol. 2018, 100, 1–19. [Google Scholar] [CrossRef]
- Southam, K.A.; Vincent, A.J.; Small, D.H. Do microglia default on network maintenance in Alzheimer’s disease? J. Alzheimers Dis. 2016, 51, 657–669. [Google Scholar] [CrossRef]
- Forster, M.J.; Dubey, A.; Dawson, K.M.; Stutts, W.A.; Lal, H.; Sohal, R.S. Age-related losses of cognitive function and motor skills in mice are associated with oxidative protein damage in the brain. Proc. Natl. Acad. Sci. USA 1996, 93, 4765–4769. [Google Scholar] [CrossRef]
- Lovell, M.A.; Markesbery, W.R. Oxidative DNA damage in mild cognitive impairment and late-stage Alzheimer’s disease. Nucleic Acids Res. 2007, 35, 7497–7504. [Google Scholar] [CrossRef]
- Gella, A.; Durany, N. Oxidative stress in Alzheimer disease. Cell Adhes. Migr. 2009, 3, 88–93. [Google Scholar] [CrossRef]
- Chen, C.; Ahn, E.H.; Kang, S.S.; Liu, X.; Alam, A.; Ye, K. Gut dysbiosis contributes to amyloid pathology, associated with C/EBPβ/AEP signaling activation in Alzheimer’s disease mouse model. Sci. Adv. 2020, 6, eaba0466. [Google Scholar] [CrossRef] [PubMed]
- Vallianou, N.G.; Stratigou, T.; Tsagarakis, S. Microbiome and diabetes: Where are we now? Diabetes Res. Clin. Pract. 2018, 146, 111–118. [Google Scholar] [CrossRef]
- Grasset, E.; Burcelin, R. The gut microbiota to the brain axis in the metabolic control. Rev. Endocr. Metab. Disord. 2019, 20, 427–438. [Google Scholar] [CrossRef]
- Yan, W.; Pang, M.; Yu, Y.; Gou, X.; Si, P.; Zhawatibai, A.; Zhang, Y.; Zhang, M.; Guo, T.; Yi, X.; et al. The neuroprotection of liraglutide on diabetic cognitive deficits is associated with improved hippocampal synapses and inhibited neuronal apoptosis. Life Sci. 2019, 231, 116566. [Google Scholar] [CrossRef]
- Shabbir, U.; Tyagi, A.; Elahi, F.; Aloo, S.O.; Oh, D.-H. The Potential Role of Polyphenols in Oxidative Stress and Inflammation Induced by Gut Microbiota in Alzheimer’s Disease. Antioxidants 2021, 10, 1370. [Google Scholar] [CrossRef]
- Wang, Y.; Zhang, Z.; Li, B.; He, B.; Li, L.; Nice, E.C.; Zhang, W.; Xu, J. New Insights into the Gut Microbiota in Neurodegenerative Diseases from the Perspective of Redox Homeostasis. Antioxidants 2022, 11, 2287. [Google Scholar] [CrossRef] [PubMed]
- Dalile, B.; Van Oudenhove, L.; Vervliet, B.; Verbeke, K. The role of short-chain fatty acids in microbiota–gut–brain communication. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 461–478. [Google Scholar] [CrossRef] [PubMed]
- Yarandi, S.S.; Peterson, D.A.; Treisman, G.J.; Moran, T.H.; Pasricha, P.J. Modulatory effects of gut microbiota on the central nervous system: How gut could play a role in neuropsychiatric health and diseases. J. Neurogastroenterol. Motil. 2016, 22, 201. [Google Scholar] [CrossRef]
- Oleskin, A.V.; Shenderov, B.A. Neuromodulatory effects and targets of the SCFAs and gasotransmitters produced by the human symbiotic microbiota. Microb. Ecol. Health Dis. 2016, 27, 30971. [Google Scholar] [CrossRef]
- Tiso, M.; Schechter, A.N. Nitrate reduction to nitrite, nitric oxide and ammonia by gut bacteria under physiological conditions. PLoS ONE 2015, 10, e0119712. [Google Scholar]
- Ayaz, B.D.; Zubcevic, J. Gut microbiota and neuroinflammation in pathogenesis of hypertension: A potential role for hydrogen sulfide. Pharmacol. Res. 2020, 153, 104677. [Google Scholar] [CrossRef] [PubMed]
- Tilg, H.; Moschen, A.R. Microbiota and diabetes: An evolving relationship. Gut 2014, 63, 1513–1521. [Google Scholar] [CrossRef]
- Sircana, A.; Framarin, L.; Leone, N.; Berrutti, M.; Castellino, F.; Parente, R.; De Michieli, F.; Paschetta, E.; Musso, G. Altered gut microbiota in type 2 diabetes: Just a coincidence? Curr. Diabetes Rep. 2018, 18, 98. [Google Scholar] [CrossRef]
- Arnoriaga-Rodriguez, M.; Mayneris-Perxachs, J.; Burokas, A.; Contreras-Rodriguez, O.; Blasco, G.; Coll, C.; Biarnes, C.; Miranda-Olivos, R.; Latorre, J.; Moreno-Navarrete, J.M.; et al. Obesity impairs short-term and working memory through gut microbial metabolism of aromatic amino acids. Cell Metab. 2020, 32, 548–560. [Google Scholar] [CrossRef] [PubMed]
- Li, S.; Wei, J.; Chen, T. Gut microbiota in the occurrence, development and treatment of gut-brain disorders. Front. Cell. Infect. Microbiol. 2021, 11, 1286. [Google Scholar] [CrossRef]
- Cattaneo, A.; Cattane, N.; Galluzzi, S.; Provasi, S.; Lopizzo, N.; Festari, C.; Ferrari, C.; Guerra, U.P.; Paghera, B.; Muscio, C.; et al. Association of brain amyloidosis with pro-inflammatory gut bacterial taxa and peripheral inflammation markers in cognitively impaired elderly. Neurobiol. Aging 2017, 49, 60–68. [Google Scholar] [CrossRef] [PubMed]
- Strandwitz, P.; Kim, K.H.; Terekhova, D.; Liu, J.K.; Sharma, A.; Levering, J.; McDonald, D.; Dietrich, D.; Ramadhar, T.R.; Lekbua, A.; et al. GABA-modulating bacteria of the human gut microbiota. Nat. Microbiol. 2019, 4, 396–403. [Google Scholar] [CrossRef]
- Huang, D.; Liu, D.; Yin, J.; Qian, T.; Shrestha, S.; Ni, H. Glutamate-glutamine and GABA in brain of normal aged and patients with cognitive impairment. Eur. Radiol. 2017, 27, 2698–2705. [Google Scholar] [CrossRef] [PubMed]
- Kowalski, K.; Mulak, A. Brain-gut-microbiota axis in Alzheimer’s disease. J. Neurogastroenterol. Motil. 2019, 25, 48. [Google Scholar] [CrossRef] [PubMed]
- Corrigan, R.R.; Labrador, L.; Grizzanti, J.; Mey, M.; Piontkivska, H.; Casadesús, G. Neuroprotective Mechanisms of Amylin Receptor Activation, Not Antagonism, in the APP/PS1 Mouse Model of Alzheimer’s Disease. J. Alzheimers Dis. 2023, 91, 1495–1514. [Google Scholar] [CrossRef] [PubMed]
- Oskarsson, M.E.; Paulsson, J.F.; Schultz, S.W.; Ingelsson, M.; Westermark, P.; Westermark, G.T. In vivo seeding and cross-seeding of localized amyloidosis: A molecular link between type 2 diabetes and Alzheimer disease. Am. J. Pathol. 2015, 185, 834–846. [Google Scholar] [CrossRef] [PubMed]
- Fawver, J.N.; Ghiwot, Y.; Koola, C.; Carrera, W.; Rodriguez-Rivera, J.; Hernandez, C.; TDineley, K.; Kong, Y.; Li, J.; Jhamandas, J.; et al. Islet amyloid polypeptide (IAPP): A second amyloid in Alzheimer’s disease. Curr. Alzheimer Res. 2014, 11, 928–940. [Google Scholar] [CrossRef]
- Jackson, K.; Barisone, G.A.; Diaz, E.; Jin, L.W.; DeCarli, C.; Despa, F. Amylin deposition in the brain: A second amyloid in Alzheimer disease? Ann. Neurol. 2013, 74, 517–526. [Google Scholar] [CrossRef]
- Wijesekara, N.; Ahrens, R.; Sabale, M.; Wu, L.; Ha, K.; Verdile, G.; Fraser, P.E. Amyloid-β and islet amyloid pathologies link Alzheimer’s disease and type 2 diabetes in a transgenic model. FASEB J. 2017, 31, 5409–5418. [Google Scholar] [CrossRef]
- Du, H.; Guo, L.; Yan, S.; Sosunov, A.A.; McKhann, G.M.; Yan, S.S. Early deficits in synaptic mitochondria in an Alzheimer’s disease mouse model. Proc. Natl. Acad. Sci. USA 2010, 107, 18670–18675. [Google Scholar] [CrossRef] [PubMed]
- Du, H.; Guo, L.; Yan, S.S. Synaptic mitochondrial pathology in Alzheimer’s disease. Antioxid. Redox Signal. 2012, 16, 1467–1475. [Google Scholar] [CrossRef]
- Kosik, K.S.; Joachim, C.L.; Selkoe, D.J. Microtubule-associated protein tau (tau) is a major antigenic component of paired helical filaments in Alzheimer disease. Proc. Natl. Acad. Sci. USA 1986, 83, 4044–4048. [Google Scholar] [CrossRef]
- Jucker, M.; Walker, L.C. Self-propagation of pathogenic protein aggregates in neurodegenerative diseases. Nature 2013, 501, 45–51. [Google Scholar] [CrossRef] [PubMed]
- Clodfelder-Miller, B.J.; Zmijewska, A.A.; Johnson, G.V.; Jope, R.S. Tau is hyperphosphorylated at multiple sites in mouse brain in vivo after streptozotocin-induced insulin deficiency. Diabetes 2006, 55, 3320–3325. [Google Scholar] [CrossRef]
- Zhang, Y.; Huang, N.Q.; Yan, F. Diabetes mellitus and Alzheimer’s disease: GSK-3β as a potential link. Behav. Brain Res. 2018, 339, 57–65. [Google Scholar] [CrossRef]
- Mehla, J.; Chauhan, B.C.; Chauhan, N.B. Experimental induction of type 2 diabetes in aging-accelerated mice triggered Alzheimer-like pathology and memory deficits. J. Alzheimers Dis. 2014, 39, 145–162. [Google Scholar] [CrossRef]
- Mahboobi, H.; Golmirzaei, J.; HGan, S.; Jalalian, M.; AKamal, M. Humanin: A possible linkage between Alzheimer’s disease and type 2 diabetes. CNS Neurol. Disord.-Drug Targets (Former Curr. Drug Targets-CNS Neurol. Disord.) 2014, 13, 543–552. [Google Scholar] [CrossRef]
- Michailidis, M.; Moraitou, D.; Tata, D.A.; Kalinderi, K.; Papamitsou, T.; Papaliagkas, V. Alzheimer’s disease as type 3 diabetes: Common pathophysiological mechanisms between Alzheimer’s disease and type 2 diabetes. Int. J. Mol. Sci. 2022, 23, 2687. [Google Scholar] [CrossRef]
- Gasparini, L.; Xu, H. Potential roles of insulin and IGF-1 in Alzheimer’s disease. Trends Neurosci. 2003, 26, 404–406. [Google Scholar] [CrossRef] [PubMed]
- Terr, A.V., Jr.; Buccafusco, J.J. The cholinergic hypothesis of age and Alzheimer’s disease-related cognitive deficits: Recent challenges and their implications for novel drug development. J. Pharmacol. Exp. Ther. 2003, 306, 821–827. [Google Scholar] [CrossRef]
- Markowska, A.L.; Mooney, M.; Sonntag, W.E. Insulin-like growth factor-1 ameliorates age-related behavioral deficits. Neuroscience 1998, 87, 559–569. [Google Scholar] [CrossRef] [PubMed]
- Carro, E.; Trejo, J.L.; Gomez-Isla, T.; LeRoith, D.; Torres-Aleman, I. Serum insulin-like growth factor I regulates brain amyloid-β levels. Nat. Med. 2002, 8, 1390–1397. [Google Scholar] [CrossRef] [PubMed]
- d’Almeida, O.C.; Violante, I.R.; Quendera, B.; Moreno, C.; Gomes, L.; Castelo-Branco, M. The neurometabolic profiles of GABA and Glutamate as revealed by proton magnetic resonance spectroscopy in type 1 and type 2 diabetes. PLoS ONE 2020, 15, e0240907. [Google Scholar] [CrossRef]
- Bowman, G.L.; Kaye, J.A.; Moore, M.; Waichunas, D.; Carlson, N.E.; Quinn, J.F. Blood–brain barrier impairment in Alzheimer disease: Stability and functional significance. Neurology 2007, 68, 1809–1814. [Google Scholar] [CrossRef] [PubMed]
- Bell, R.D.; Zlokovic, B.V. Neurovascular mechanisms and blood–brain barrier disorder in Alzheimer’s disease. Acta Neuropathol. 2009, 118, 103–113. [Google Scholar] [CrossRef]
- Toledo, J.B.; Arnold, S.E.; Raible, K.; Brettschneider, J.; Xie, S.X.; Grossman, M.; Monsell, S.E.; Kukull, W.A.; Trojanowski, J.Q. Contribution of cerebrovascular disease in autopsy confirmed neurodegenerative disease cases in the National Alzheimer’s Coordinating Centre. Brain 2013, 136, 2697–2706. [Google Scholar] [CrossRef] [PubMed]
- Shin, Y.; Choi, S.H.; Kim, E.; Bylykbashi, E.; Kim, J.A.; Chung, S.; Kim, D.Y.; Kamm, R.D.; Tanzi, R.E. Blood–brain barrier dysfunction in a 3D in vitro model of Alzheimer’s disease. Adv. Sci. 2019, 6, 1900962. [Google Scholar] [CrossRef]
- Yamazaki, Y.; Kanekiyo, T. Blood-brain barrier dysfunction and the pathogenesis of Alzheimer’s disease. Int. J. Mol. Sci. 2017, 18, 1965. [Google Scholar] [CrossRef]
- Dallaire-Théroux, C.; Callahan, B.L.; Potvin, O.; Saikali, S.; Duchesne, S. Radiological-Pathological Correlation in Alzheimer’s Disease: Systematic Review of Antemortem Magnetic Resonance Imaging Findings. J. Alzheimers Dis. 2017, 57, 575–601. [Google Scholar] [CrossRef] [PubMed]
- Klohs, J. An integrated view on vascular dysfunction in Alzheimer’s disease. Neurodegener. Dis. 2020, 19, 109–127. [Google Scholar] [CrossRef]
- Graziano, M.P.; Hey, P.J.; Borkowski, D.; Chicchi, G.G.; Strader, C.D. Cloning and functional expression of a human glucagon-like peptide-1 receptor. Biochem. Biophys. Res. Commun. 1993, 196, 141–146. [Google Scholar] [CrossRef]
- Baggio, L.L.; Drucker, D.J. Biology of incretins: GLP-1 and GIP. Gastroenterology 2007, 132, 2131–2157. [Google Scholar] [CrossRef]
- Reed, J.; Bain, S.; Kanamarlapudi, V. Recent advances in understanding the role of glucagon-like peptide 1. F1000Research 2020, 9, 239. [Google Scholar] [CrossRef] [PubMed]
- Drucker, D.J. Mechanisms of action and therapeutic application of glucagon-like peptide-1. Cell Metab. 2018, 27, 740–756. [Google Scholar] [CrossRef] [PubMed]
- Leitner, D.R.; Frühbeck, G.; Yumuk, V.; Schindler, K.; Micic, D.; Woodward, E.; Toplak, H. Obesity and Type 2 Diabetes: Two Diseases with a Need for Combined Treatment Strategies—EASO Can Lead the Way. Obes. Facts 2017, 10, 483–492. [Google Scholar] [CrossRef]
- Del Olmo-Garcia, M.I.; Merino-Torres, J.F. GLP-1 Receptor Agonists and Cardiovascular Disease in Patients with Type 2 Diabetes. J. Diabetes Res. 2018, 2018, 4020492. [Google Scholar] [CrossRef]
- Reuser, A.J.; Wisselaar, H.A. An evaluation of the potential side-effects of alpha-glucosidase inhibitors used for the management of diabetes mellitus. Eur. J. Clin. Investig. 1994, 3 (Suppl. S24), 19–24. [Google Scholar] [CrossRef]
- Buse, J.B.; Wexler, D.J.; Tsapas, A.; Rossing, P.; Mingrone, G.; Mathieu, C.; D’Alessio, D.A.; Davies, M.J. Update to: Management of Hyperglycemia in Type 2 Diabetes, 2018. A Consensus Report by the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD). Diabetes Care 2019, 43, 487–493. [Google Scholar] [CrossRef]
- Shaefer, C.F., Jr.; Kushner, P.; Aguilar, R. User’s guide to mechanism of action and clinical use of GLP-1 receptor agonists. Postgrad. Med. 2015, 127, 818–826. [Google Scholar] [CrossRef]
- Dong, M.; Wen, S.; Zhou, L. The Relationship Between the Blood-Brain-Barrier and the Central Effects of Glucagon-Like Peptide-1 Receptor Agonists and Sodium-Glucose Cotransporter-2 Inhibitors. Diabetes Metab. Syndr. Obes. Targets Ther. 2022, 15, 2583–2597. [Google Scholar] [CrossRef]
- Malm-Erjefält, M.; Bjørnsdottir, I.; Vanggaard, J.; Helleberg, H.; Larsen, U.; Oosterhuis, B.; van Lier, J.J.; Zdravkovic, M.; Olsen, A.K. Metabolism and excretion of the once-daily human glucagon-like peptide-1 analog liraglutide in healthy male subjects and its in vitro degradation by dipeptidyl peptidase IV and neutral endopeptidase. Drug Metab. Dispos. 2010, 38, 1944–1953. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Campbell, J.E.; Drucker, D.J. Pharmacology, physiology, and mechanisms of incretin hormone action. Cell Metab. 2013, 17, 819–837. [Google Scholar] [CrossRef]
- Fineman, M.; Flanagan, S.; Taylor, K.; Aisporna, M.; Shen, L.Z.; Mace, K.F.; Walsh, B.; Diamant, M.; Cirincione, B.; Kothare, P.; et al. Pharmacokinetics and pharmacodynamics of exenatide extended-release after single and multiple dosing. Clin. Pharmacokinet. 2011, 50, 65–74. [Google Scholar] [CrossRef] [PubMed]
- Ahren, B. GLP-1-based therapy of type 2 diabetes: GLP-1 mimetics and DPP-IV inhibitors. Curr. Diabetes Rep. 2007, 7, 340–347. [Google Scholar] [CrossRef] [PubMed]
- Yoo, B.K.; Triller, D.M.; Yoo, D.J. Formulary forum: Exenatide: A new option for the treatment of type 2 diabetes. Ann Pharmacother. 2016, 40, 1777–1784. [Google Scholar] [CrossRef] [PubMed]
- Kalra, S.; Bhattacharya, S.; Kapoor, N. Contemporary classification of glucagon-like peptide 1 receptor agonists (GLP1RAs). Diabetes Ther. 2021, 12, 2133–2147. [Google Scholar] [CrossRef] [PubMed]
- Drucker, D.J. The biology of incretin hormones. Cell Metab. 2006, 3, 153–165. [Google Scholar] [CrossRef] [PubMed]
- Tamaki, C.; Takeuchi, M.; Iwamoto, N.; Glaesner, W. Pharmacological profile and clinical trial results of a long-acting, once weekly human GLP-1 receptor agonist Dulaglutide (Genetical Recombination). Nippon Yakurigaku Zasshi 2015, 146, 215–224. [Google Scholar] [CrossRef]
- Alorfi, N.M.; Algarni, A.S. Clinical impact of semaglutide, a glucagon-like peptide-1 receptor agonist, on obesity management: A review. Clin. Pharmacol. 2022, 14, 61–67. [Google Scholar] [CrossRef]
- Mahapatra, M.K.; Karuppasamy, M.; Sahoo, B.M. Therapeutic potential of semaglutide, a newer GLP-1 receptor agonist, in abating obesity, non-alcoholic steatohepatitis and neurodegenerative diseases: A narrative review. Pharm. Res. 2022, 39, 1233–1248. [Google Scholar] [CrossRef] [PubMed]
- Overgaard, R.V.; Hertz, C.L.; Ingwersen, S.H.; Navarria, A.; Drucker, D.J. Levels of circulating semaglutide determine reductions in HbA1c and body weight in people with type 2 diabetes. Cell Rep. Med. 2021, 2, 100387. [Google Scholar] [CrossRef]
- Sfairopoulos, D.; Liatis, S.; Tigas, S.; Liberopoulos, E. Clinical pharmacology of glucagon-like peptide-1 receptor agonists. Hormones 2018, 17, 333–350. [Google Scholar] [CrossRef]
- Hunter, K.; Holscher, C. Drugs developed to treat diabetes, liraglutide and lixisenatide, cross the blood brain barrier and enhance neurogenesis. BMC Neurosci. 2012, 13, 33. [Google Scholar] [CrossRef] [PubMed]
- Scott, L.J. Lixisenatide: A review of its use in patients with type 2 diabetes mellitus. BioDrugs 2013, 27, 509–523. [Google Scholar] [CrossRef] [PubMed]
- Holscher, 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] [PubMed]
- Zhang, Z.Q.; Hölscher, C. GIP has neuroprotective effects in Alzheimer and Parkinson’s disease models. Peptides 2020, 125, 170184. [Google Scholar] [CrossRef] [PubMed]
- 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-kappaB 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]
- McGovern, S.F.; Hunter, K.; Holscher, C. Effects of the glucagon-like polypeptide-1 analogue (Val8)GLP-1 on learning, progenitor cell proliferation and neurogenesis in the C57B/16 mouse brain. Brain Res. 2012, 1473, 204–213. [Google Scholar] [CrossRef] [PubMed]
- Kanoski, S.E.; Fortin, S.M.; Arnold, M.; Grill, H.J.; Hayes, M.R. Peripheral and central GLP-1 receptor populations mediate the anorectic effects of peripherally administered GLP-1 receptor agonists, liraglutide and exendin-4. Endocrinology 2011, 152, 3103–3112. [Google Scholar] [CrossRef]
- Paternoster, S.; Falasca, M. Dissecting the physiology and pathophysiology of glucagon-like Peptide-1. Front. Endocrinol. 2018, 9, 584. [Google Scholar] [CrossRef] [PubMed]
- Sandoval, D.A.; Bagnol, D.; Woods, S.C.; D’Alessio, D.A.; Seeley, R.J. Arcuate glucagon-like peptide 1 receptors regulate glucose homeostasis but not food intake. Diabetes 2008, 57, 2046–2054. [Google Scholar] [CrossRef]
- Knudsen, L.B.; Lau, J. The discovery and development of liraglutide and semaglutide. Front. Endocrinol. 2019, 10, 155. [Google Scholar] [CrossRef]
- Secher, A.; Jelsing, J.; Baquero, A.F.; Hecksher-Sørensen, J.; Cowley, M.A.; Dalbøge, L.S.; Hansen, G.; Grove, K.L.; Pyke, C.; Raun, K.; et al. The arcuate nucleus mediates GLP-1 receptor agonist liraglutide-dependent weight loss. J. Clin. Investig. 2014, 124, 4473–4488. [Google Scholar] [CrossRef]
- Shi, X.; Chacko, S.; Li, F.; Li, D.; Burrin, D.; Chan, L.; Guan, X. Acute activation of GLP-1-expressing neurons promotes glucose homeostasis and insulin sensitivity. Mol. Metab. 2017, 6, 1350–1359. [Google Scholar] [CrossRef]
- Gaykema, R.P.; Newmyer, B.A.; Ottolini, M.; Raje, V.; Warthen, D.M.; Lambeth, P.S.; Niccum, M.; Yao, T.; Huang, Y.; Schulman, I.G.; et al. Activation of murine pre-proglucagon-producing neurons reduces food intake and body weight. J. Clin. Investig. 2017, 127, 1031–1045. [Google Scholar] [CrossRef]
- Kastin, A.; Akerstrom, V. Entry of exendin-4 into the brain is rapid but may be limited at high doses. Int. J. Obes. 2003, 27, 313–318. [Google Scholar] [CrossRef] [PubMed]
- Li, L.; Zhang, Z.F.; Holscher, C.; Gao, C.; Jiang, Y.H.; Liu, Y.Z. (Val⁸) glucagon-like peptide-1 prevents tau hyperphosphorylation, impairment of spatial learning and ultra-structural cellular damage induced by streptozotocin in rat brains. Eur. J. Pharmacol. 2012, 674, 280–286. [Google Scholar] [CrossRef]
- Sharma, M.K.; Jalewa, J.; Hölscher, C. Neuroprotective and anti-apoptotic effects of liraglutide on SH-SY5Y cells exposed to methylglyoxal stress. J. Neurochem. 2014, 128, 459–471. [Google Scholar] [CrossRef]
- Monti, G.; Gomes Moreira, D.; Richner, M.; Mutsaers, H.A.M.; Ferreira, N.; Jan, A. GLP-1 Receptor Agonists in Neurodegeneration: Neurovascular Unit in the Spotlight. Cells 2022, 11, 2023. [Google Scholar] [CrossRef] [PubMed]
- Paladugu, L.; Gharaibeh, A.; Kolli, N.; Learman, C.; Hall, T.C.; Li, L.; Rossignol, J.; Maiti, P.; Dunbar, G.L. Liraglutide has anti-inflammatory and anti-amyloid properties in streptozotocin-induced and 5xFAD mouse models of Alzheimer’s disease. Int. J. Mol. Sci. 2021, 22, 860. [Google Scholar] [CrossRef]
- Park, J.S.; Kam, T.I.; Lee, S.; Park, H.; Oh, Y.; Kwon, S.H.; Song, J.J.; Kim, D.; Kim, H.; Jhaldiyal, A.; et al. Blocking microglial activation of reactive astrocytes is neuroprotective in models of Alzheimer’s disease. Acta Neuropathol. Commun. 2021, 9, 78. [Google Scholar] [CrossRef]
- Jantrapirom, S.; Nimlamool, W.; Chattipakorn, N.; Chattipakorn, S.; Temviriyanukul, P.; Inthachat, W. Liraglutide suppresses tau hyperphosphorylation, amyloid beta accumulation through regulating neuronal insulin signaling and BACE-1 Activity. Int. J. Mol. Sci. 2020, 21, 1725. [Google Scholar] [CrossRef] [PubMed]
- Park, K.H.; Barrett, T. Gliosis precedes amyloid-β deposition and pathological tau accumulation in the neuronal cell cycle re-entry mouse model of Alzheimer’s disease. J. Alzheimers Dis. Rep. 2020, 4, 243–253. [Google Scholar] [CrossRef] [PubMed]
- Wang, Y.; Chen, S.; Xu, Z.; Chen, S.; Yao, W.; Gao, X. GLP-1 receptor agonists downregulate aberrant GnT-III expression in Alzheimer’s disease models through the Akt/GSK-3beta/beta-catenin signaling. Neuropharmacology 2018, 131, 190–199. [Google Scholar] [CrossRef]
- Yuan, Z.; Li, D.; Feng, P.; Xue, G.; Ji, C.; Li, G.; Hölscher, C. A novel GLP-1/GIP dual agonist is more effective than liraglutide in reducing inflammation and enhancing GDNF release in the MPTP mouse model of Parkinson’s disease. Eur. J. Pharmacol. 2017, 812, 82–90. [Google Scholar] [CrossRef]
- Müller, T.D.; Finan, B.; Bloom, S.R.; D’Alessio, D.; Drucker, D.J.; Flatt, P.R.; Fritsche, A.; Gribble, F.; Grill, H.J.; Habener, J.F.; et al. Glucagon-like peptide 1 (GLP-1). Mol. Metab. 2019, 30, 72–130. [Google Scholar] [CrossRef]
- Batista, A.F.; Forny-Germano, L.; Clarke, J.R.; Lyra e Silva, N.M.; Brito-Moreira, J.; Boehnke, S.E.; Winterborn, A.; Coe, B.C.; Lablans, A.; Vital, J.F.; et al. The diabetes drug liraglutide reverses cognitive impairment in mice and attenuates insulin receptor and synaptic pathology in a non-human primate model of Alzheimer’s disease. J. Pathol. 2018, 245, 85–100. [Google Scholar] [CrossRef]
- Lee, C.H.; Jeon, S.J.; Cho, K.S.; Moon, E.; Sapkota, A.; Jun, H.S.; Ryu, J.H.; Choi, J.W. Activation of glucagon-like peptide-1 receptor promotes neuroprotection in experimental autoimmune encephalomyelitis by reducing neuroinflammatory responses. Mol. Neurobiol. 2018, 55, 3007–3020. [Google Scholar] [CrossRef]
- Solmaz, V.; Cinar, B.P.; Yigitturk, G.; Cavusoglu, T.; Taskiran, D.; Erbas, O. Exenatide reduces TNF-alpha expression and improves hippocampal neuron numbers and memory in streptozotocin treated rats. Eur. J. Pharmacol. 2015, 765, 482–487. [Google Scholar] [CrossRef] [PubMed]
- Bomba, M.; Granzotto, A.; Castelli, V.; Onofrj, M.; Lattanzio, R.; Cimini, A.; Sensi, S.L. 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. 2019, 70, 793–810. [Google Scholar] [CrossRef] [PubMed]
- Parthsarathy, V.; Holscher, 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]
- 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. 2015, 293, 96–106. [Google Scholar] [CrossRef]
- Novo Nordisk A/S, A Research Study Investigating Semaglutide in People With Early Alzheimer’s Disease (EVOKE), NCT04777396. Available online: https://classic.clinicaltrials.gov/ct2/show/study/NCT04777396 (accessed on 1 September 2023).
- Goel, P.; Chakrabarti, S.; Goel, K.; Bhutani, K.; Chopra, T.; Bali, S. Neuronal cell death mechanisms in Alzheimer’s disease: An insight. Front. Mol. Neurosci. 2022, 15, 937133. [Google Scholar] [CrossRef]
- Velmurugan, K.; Bouchard, R.; Mahaffey, G.; Pugazhenthi, S. Neuroprotective actions of glucagon-like peptide-1 in differentiated human neuroprogenitor cells. J. Neurochem. 2012, 123, 919–931. [Google Scholar] [CrossRef]
- 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. 2002, 302, 881–888. [Google Scholar] [CrossRef] [PubMed]
- Qiu, C.; Wang, Y.P.; Pan, X.D.; Liu, X.Y.; Chen, Z.; Liu, L.B. Exendin-4 protects Aβ(1-42) oligomer-induced PC12 cell apoptosis. Am. J. Transl. Res. 2016, 8, 3540–3548. [Google Scholar] [PubMed]
- During, M.J.; Cao, L.; Zuzga, D.S.; Francis, J.S.; Fitzsimons, H.L.; Jiao, X.; Bland, R.J.; Klugmann, M.; Banks, W.A.; Drucker, D.J.; et al. Glucagon-like peptide-1 receptor is involved in learning and neuroprotection. Nat. Med. 2003, 9, 1173–1179. [Google Scholar] [CrossRef]
- Yang, J.L.; Chen, W.Y.; Chen, Y.P.; Kuo, C.Y.; Chen, S.D. Activation of GLP-1 Receptor Enhances Neuronal Base Excision Repair via PI3K-AKT-Induced Expression of Apurinic/Apyrimidinic Endonuclease 1. Theranostics 2016, 6, 2015–2027. [Google Scholar] [CrossRef] [PubMed]
- Shi, L.; Zhang, Z.; Li, L.; Hölscher, C. A novel dual GLP-1/GIP receptor agonist alleviates cognitive decline by re-sensitizing insulin signaling in the Alzheimer icv. STZ rat model. Behav. Brain Res. 2017, 327, 65–74. [Google Scholar] [CrossRef] [PubMed]
- Liu, X.-Y.; Wang, L.-X.; Chen, Z.; Liu, L.-B. Liraglutide prevents beta-amyloidinduced neurotoxicity in SH-SY5Y cells via a PI3K-dependent signaling pathway. Neurol. Res. 2016, 38, 313–319. [Google Scholar] [CrossRef] [PubMed]
- Panagaki, T.; Michael, M.; Holscher, C. Liraglutide restores chronic ER stress, autophagy impairments and apoptotic signalling in SH-SY5Y cells. Sci. Rep. 2017, 7, 16158. [Google Scholar] [CrossRef]
- Bianchi, M.; D’Oria, V.; Braghini, M.R.; Petrini, S.; Manco, M. Liraglutide treatment ameliorates neurotoxicity induced by stable silencing of Pin1. Int. J. Mol. Sci. 2019, 20, 5064. [Google Scholar] [CrossRef]
- Chang, Y.F.; Zhang, D.; Hu, W.M.; Liu, D.X.; Li, L. Semaglutide-mediated protection against abeta correlated with enhancement of autophagy and inhibition of apotosis. J. Clin. Neurosci. 2020, 81, 234–239. [Google Scholar] [CrossRef]
- Femminella, G.D.; Frangou, E.; Love, S.B.; Busza, G.; Holmes, C.; Ritchie, C.; Lawrence, R.; McFarlane, B.; Tadros, G.; Ridha, B.H.; et al. Evaluating the effects of the novel GLP-1 analogue liraglutide in Alzheimer’s disease: Study protocol for a randomized controlled trial (ELAD study). Trials 2019, 20, 191. [Google Scholar] [CrossRef]
- Nørgaard, C.H.; Friedrich, S.; Hansen, C.T.; Gerds, T.; Ballard, C.; Møller, D.V.; Knudsen, L.B.; Kvist, K.; Zinman, B.; Holm, E.; 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. 2022, 8, e12268. [Google Scholar] [CrossRef] [PubMed]
- Akimoto, H.; Negishi, A.; Oshima, S.; Wakiyama, H.; Okita, M.; Horii, N.; Inoue, N.; Ohshima, S.; Kobayashi, D. Antidiabetic Drugs for the Risk of Alzheimer Disease in Patients With Type 2 DM Using FAERS. Am. J. Alzheimers Dis. Other Dement. 2020, 35, 1533317519899546. [Google Scholar] [CrossRef] [PubMed]
- Tobore, T.O. On the central role of mitochondrial dysfunction and oxidative stress in Alzheimer’s disease. Neurol. Sci. 2019, 40, 1527–1540. [Google Scholar] [CrossRef] [PubMed]
- Chen, S.; Liu, A.R.; An, F.M.; Yao, W.B.; Gao, X.D. Amelioration of neurodegenerative changes in cellular and rat models of diabetes-related Alzheimer’s disease by exendin-4. Age 2012, 34, 1211–1224. [Google Scholar] [CrossRef]
- An, J.; Zhou, Y.; Zhang, M.; Xie, Y.; Ke, S.; Liu, L.; Pan, X.; Chen, Z. Exenatide alleviates mitochondrial dysfunction and cognitive impairment in the 5×FAD mouse model of Alzheimer’s disease. Behav. Brain Res. 2019, 370, 111932. [Google Scholar] [CrossRef]
- Yusta, B.; Baggio, L.L.; Estall, J.L.; Koehler, J.A.; Holland, D.P.; Li, H.; Pipeleers, D.; Ling, Z.; Drucker, D.J. GLP-1 receptor activation improves β cell function and survival following induction of endoplasmic reticulum stress. Cell Metab. 2006, 4, 391–406. [Google Scholar] [CrossRef]
- Shimoda, M.; Kanda, Y.; Hamamoto, S.; Tawaramoto, K.; Hashiramoto, M.; Matsuki, M.; Kaku, K. The human glucagon-like peptide-1 analogue liraglutide preserves pancreatic beta cells via regulation of cell kinetics and suppression of oxidative and endoplasmic reticulum stress in a mouse model of diabetes. Diabetologia 2011, 54, 1098–1108. [Google Scholar] [CrossRef]
- Spielman, L.J.; Gibson, D.L.; Klegeris, A. Incretin hormones regulate microglia oxidative stress, survival and expression of trophic factors. Eur. J. Cell Biol. 2017, 96, 240–253. [Google Scholar] [CrossRef]
- Zheng, C.; Zhou, M.; Sun, J.; Xiong, H.; Peng, P.; Gu, Z.; Deng, Y. The protective effects of liraglutide on AD-like neurodegeneration induced by oxidative stress in human neuroblastoma SH-SY5Y cells. Chem. Biol. Interact. 2019, 310, 108688. [Google Scholar] [CrossRef] [PubMed]
- Duarte, A.I.; Candeias, E.; Alves, I.N.; Mena, D.; Silva, D.F.; Machado, N.J.; Campos, E.J.; Santos, M.S.; Oliveira, C.R.; Moreira, P.I. Liraglutide protects against brain amyloid-beta1-42 accumulation in female mice with early Alzheimer’s disease-like pathology by partially rescuing Oxidative/Nitrosative stress and inflammation. Int. J. Mol. Sci. 2020, 21, 1746. [Google Scholar] [CrossRef]
- Xie, Y.; Zheng, J.; Li, S.; Li, H.; Zhou, Y.; Zheng, W.; Zhang, M.; Liu, L.; Chen, Z. GLP-1 improves the neuronal supportive ability of astrocytes in Alzheimer’s disease by regulating mitochondrial dysfunction via the cAMP/PKA pathway. Biochem. Pharmacol. 2021, 188, 114578. [Google Scholar] [CrossRef] [PubMed]
- Perry, T.; Lahiri, D.K.; Sambamurti, K.; Chen, D.; Mattson, M.P.; Egan, J.M.; Greig, N.H. Glucagon-like peptide-1 decreases endogenous amyloid-beta peptide (Abeta) levels and protects hippocampal neurons from death induced by Abeta and iron. J. Neurosci. Res. 2003, 72, 603–612. [Google Scholar] [CrossRef] [PubMed]
- Xu, W.; Yang, Y.; Yuan, G.; Zhu, W.; Ma, D.; Hu, S. Exendin-4, a glucagon-like peptide-1 receptor agonist, reduces Alzheimer disease-associated tau hyperphosphorylation in the hippocampus of rats with type 2 diabetes. J. Investig. Med. 2015, 63, 267–272. [Google Scholar] [CrossRef]
- McClean, P.L.; Hölscher, C. Liraglutide can reverse memory impairment, synaptic loss and reduce plaque load in aged APP/PS1 mice, a model of Alzheimer’s disease. Neuropharmacology 2014, 76 Pt A, 57–67. [Google Scholar] [CrossRef]
- Cai, H.Y.; Hölscher, C.; Yue, X.H.; Zhang, S.X.; Wang, X.H.; Qiao, F.; Yang, W.; Qi, J.S. Lixisenatide rescues spatial memory and synaptic plasticity from amyloid beta protein-induced impairments in rats. Neuroscience 2014, 277, 6–13. [Google Scholar] [CrossRef]
- Zhang, S.X.; Cai, H.Y.; Ma, X.W.; Yuan, L.; Zhang, J.; Wang, Z.J.; Li, Y.F.; Qi, J.S. GLP-1 analogue CJC-1131 prevents amyloid β protein-induced impirments of spatial memory and synaptic plasticity in rats. Behav. Brain Res. 2017, 326, 237–243. [Google Scholar] [CrossRef] [PubMed]
- Zhou, M.; Chen, S.; Peng, P.; Gu, Z.; Yu, J.; Zhao, G.; Deng, Y. Dulaglutide ameliorates STZ-induced AD-like impairment of learning and memory ability by modulating hyperphosphorylation of tau and NFs through GSK3beta. Biochem. Biophys. Res. Commun. 2019, 511, 154–160. [Google Scholar] [CrossRef]
- Kong, J.; Wan, L.; Wang, Y.; Zhang, H.; Zhang, W. Liraglutide attenuates Abeta42 generation in APPswe/SH-SY5Y cells through the regulation of autophagy. Neuropsychiatr. Dis. Treat. 2020, 16, 1817–1825. [Google Scholar] [CrossRef]
- Holubová, M.; Hrubá, L.; Popelová, A.; Bencze, M.; Pražienková, V.; Gengler, S.; Kratochvílová, H.; Haluzík, M.; Železná, B.; Kuneš, J.; et al. Liraglutide and a lipidized analog of prolactin-releasing peptide show neuroprotective effects in a mouse model of beta-amyloid pathology. Neuropharmacology 2019, 144, 377–387. [Google Scholar] [CrossRef]
- Ma, D.L.; Chen, F.Q.; Xu, W.J.; Yue, W.Z.; Yuan, G.; Yang, Y. Early intervention with glucagon-like peptide 1 analog liraglutide prevents tau hyperphosphorylation in diabetic db/db mice. J. Neurochem. 2015, 135, 301–308. [Google Scholar] [CrossRef]
- Song, X.; Sun, Y.; Wang, Z.; Su, Y.; Wang, Y.; Wang, X. Exendin-4 alleviates β-Amyloid peptide toxicity via DAF-16 in a Caenorhabditis elegans model of Alzheimer’s disease. Front. Aging Neurosci. 2022, 14, 955113. [Google Scholar] [CrossRef] [PubMed]
- Goetzl, E.J.; Kapogiannis, D.; Schwartz, J.B.; Lobach, I.V.; Goetzl, L.; Abner, E.L.; Jicha, G.A.; Karydas, A.M.; Boxer, A.; Miller, B.L. Decreased synaptic proteins in neuronal exosomes of frontotemporal dementia and Alzheimer’s disease. FASEB Off. Publ. Fed. Am. Soc. Exp. Biol. 2016, 30, 4141–4148. [Google Scholar] [CrossRef]
- Hopsu-Havu, V.K.; Glenner, G.G. A new dipeptide naphthylamidase hydrolyzing glycyl-prolyl-beta-naphthylamide. Histochem. Histochem. Histochim. 1966, 7, 197–201. [Google Scholar] [CrossRef]
- Ambhore, J.P.; Laddha, P.R.; Nandedkar, A.; Ajmire, P.V.; Chumbhale, D.S.; Navghare, A.B.; Kuchake, V.G.; Chaudhari, P.J.; Adhao, V.S. Medicinal chemistry of non-peptidomimetic dipeptidyl peptidase IV (DPP IV) inhibitors for treatment of Type-2 diabetes mellitus: Insights on recent development. J. Mol. Struct. 2023, 1284, 135249. [Google Scholar] [CrossRef]
- Mentlein, R. Dipeptidyl-peptidase IV (CD26)--role in the inactivation of regulatory peptides. Regul. Pept. 1999, 85, 9–24. [Google Scholar] [CrossRef]
- Patel, B.D.; Ghate, M.D. Recent approaches to medicinal chemistry and therapeutic potential of dipeptidyl peptidase-4 (DPP-4) inhibitors. Eur. J. Med. Chem. 2014, 74, 574–605. [Google Scholar] [CrossRef] [PubMed]
- Baetta, R.; Corsini, A. Pharmacology of dipeptidyl peptidase-4 inhibitors: Similarities and differences. Drugs 2011, 71, 1441–1467. [Google Scholar] [CrossRef] [PubMed]
- Scheen, A.J. Pharmacokinetics of dipeptidylpeptidase-4 inhibitors. Diabetes Obes. Metab. 2010, 12, 648–658. [Google Scholar] [CrossRef]
- Keating, G.M. Alogliptin: A review of its use in patients with type 2 diabetes mellitus. Drugs 2015, 75, 777–796. [Google Scholar] [CrossRef]
- Gutch, M.; Joshi, A.; Kumar, S.; Agarwal, A.; Pahan, R.K.; Razi, S.M. Gemigliptin: Newer Promising Gliptin for Type 2 Diabetes Mellitus. Indian J. Endocrinol. Metab. 2017, 21, 898–902. [Google Scholar] [CrossRef]
- Gejl, M.; Gjedde, A.; Egefjord, L.; Møller, A.; Hansen, S.B.; Vang, K.; Rodell, A.; Brændgaard, H.; Gottrup, H.; Schacht, A.; 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. 2016, 8, 108. [Google Scholar] [CrossRef] [PubMed]
- D’Amico, M.; Di Filippo, C.; Marfella, R.; Abbatecola, A.M.; Ferraraccio, F.; Rossi, F.; Paolisso, G. Long-term inhibition of dipeptidyl peptidase-4 in Alzheimer’s prone mice. Exp. Gerontol. 2010, 45, 202–207. [Google Scholar] [CrossRef]
- Siddiqui, N.; Ali, J.; Parvez, S.; Najmi, A.K.; Akhtar, M. Neuroprotective Role of DPP-4 Inhibitor Linagliptin Against Neurodegeneration, Neuronal Insulin Resistance and Neuroinflammation Induced by Intracerebroventricular Streptozotocin in Rat Model of Alzheimer’s Disease. Neurochem. Res. 2023, 48, 2714–2730. [Google Scholar] [CrossRef]
- Siddiqui, N.; Ali, J.; Parvez, S.; Zameer, S.; Najmi, A.K.; Akhtar, M. Linagliptin, a DPP-4 inhibitor, ameliorates Aβ (1-42) peptides induced neurodegeneration and brain insulin resistance (BIR) via insulin receptor substrate-1 (IRS-1) in rat model of Alzheimer’s disease. Neuropharmacology 2021, 195, 108662. [Google Scholar] [CrossRef] [PubMed]
- Elbaz, E.M.; Senousy, M.A.; El-Tanbouly, D.M.; Sayed, R.H. Neuroprotective effect of linagliptin against cuprizone-induced demyelination and behavioural dysfunction in mice: A pivotal role of AMPK/SIRT1 and JAK2/STAT3/NF-κB signalling pathway modulation. Toxicol. Appl. Pharmacol. 2018, 352, 153–161. [Google Scholar] [CrossRef]
- Ma, Q.H.; Jiang, L.F.; Mao, J.L.; Xu, W.X.; Huang, M. Vildagliptin prevents cognitive deficits and neuronal apoptosis in a rat model of Alzheimer’s disease. Mol. Med. Rep. 2018, 17, 4113–4119. [Google Scholar] [CrossRef]
- Zhang, D.D.; Shi, N.; Fang, H.; Ma, L.; Wu, W.P.; Zhang, Y.Z.; Tian, J.L.; Tian, L.B.; Kang, K.; Chen, S. Vildagliptin, a DPP4 inhibitor, alleviates diabetes-associated cognitive deficits by decreasing the levels of apoptosis-related proteins in the rat hippocampus. Exp. Ther. Med. 2018, 15, 5100–5106. [Google Scholar] [CrossRef]
- Kosaraju, J.; Gali, C.C.; Khatwal, R.B.; Dubala, A.; Chinni, S.; Holsinger, R.D. Saxagliptin: A dipeptidyl peptidase-4 inhibitor ameliorates streptozotocin induced Alzheimer’s disease. Neuropharmacology 2013, 72, 291–300. [Google Scholar] [CrossRef] [PubMed]
- White, C.L.; Pistell, P.J.; Purpera, M.N.; Gupta, S.; Fernandez-Kim, S.O.; Hise, T.L.; Keller, J.N.; Ingram, D.K.; Morrison, C.D.; Bruce-Keller, A.J. Effects of high fat diet on Morris maze performance, oxidative stress, and inflammation in rats: Contributions of maternal diet. Neurobiol. Dis. 2009, 35, 3–13. [Google Scholar] [CrossRef]
- Pintana, H.; Apaijai, N.; Chattipakorn, N.; Chattipakorn, S.C. DPP-4 inhibitors improve cognition and brain mitochondrial function of insulin-resistant rats. J. Endocrinol. 2013, 218, 1–11. [Google Scholar] [CrossRef]
- Isik, A.T.; Soysal, P.; Yay, A.; Usarel, C. The effects of sitagliptin, a DPP-4 inhibitor, on cognitive functions in elderly diabetic patients with or without Alzheimer’s disease. Diabetes Res. Clin. Pract. 2017, 123, 192–198. [Google Scholar] [CrossRef]
- Jeong, S.H.; Kim, H.R.; Kim, J.; Kim, H.; Hong, N.; Jung, J.H.; Baik, K.; Cho, H.; Lyoo, C.H.; Ye, B.S.; et al. Association of dipeptidyl peptidase-4 inhibitor use and amyloid burden in patients with diabetes and AD-related cognitive impairment. Neurology 2021, 97, e1110. [Google Scholar] [CrossRef]
- Rieg, T.; Vallon, V. Development of SGLT1 and SGLT2 inhibitors. Diabetologia 2018, 61, 2079–2086. [Google Scholar] [CrossRef]
- Bailey, C.J. Renal glucose reabsorption inhibitors to treat diabetes. Trends Pharmacol. Sci. 2011, 32, 63–71. [Google Scholar] [CrossRef]
- Dardi, I.; Kouvatsos, T.; Jabbour, S.A. SGLT2 inhibitors. Biochem. Pharmacol. 2016, 101, 27–39. [Google Scholar] [CrossRef]
- Scheen, A.J. Pharmacokinetics, Pharmacodynamics and Clinical Use of SGLT2 Inhibitors in Patients with Type 2 Diabetes Mellitus and Chronic Kidney Disease. Clin. Pharmacokinet. 2015, 54, 691–708. [Google Scholar] [CrossRef]
- Scheen, A.J. Pharmacokinetic and pharmacodynamic profile of empagliflozin, a sodium glucose co-transporter 2 inhibitor. Clin. Pharmacokinet. 2014, 53, 213–225. [Google Scholar] [CrossRef]
- Plosker, G.L. Dapagliflozin: A review of its use in type 2 diabetes mellitus. Drugs 2012, 72, 2289–2312. [Google Scholar] [CrossRef] [PubMed]
- Kasichayanula, S.; Liu, X.; LaCreta, F.; Griffen, S.C.; Boulton, D.W. Clinical pharmacokinetics and pharmacodynamics of dapagliflozin, a selective inhibitor of sodium-glucose co-transporter type 2. Clin. Pharmacokinet. 2014, 53, 17–27. [Google Scholar] [CrossRef] [PubMed]
- Tahara, A.; Takasu, T.; Yokono, M.; Imamura, M.; Kurosaki, E. Characterization and comparison of sodium-glucose cotransporter 2 inhibitors in pharmacokinetics, pharmacodynamics, and pharmacologic effects. J. Pharmacol. Sci. 2016, 130, 159–169. [Google Scholar] [CrossRef] [PubMed]
- Plosker, G.L. Canagliflozin: A review of its use in patients with type 2 diabetes mellitus. Drugs 2014, 74, 807–824. [Google Scholar] [CrossRef]
- Filippas-Ntekouan, S.; Tsimihodimos, V.; Filippatos, T.; Dimitriou, T.; Elisaf, M. SGLT-2 inhibitors: Pharmacokinetics characteristics and effects on lipids. Expert Opin. Drug Metab. Toxicol. 2018, 14, 1113–1121. [Google Scholar] [CrossRef]
- Poole, R.M.; Dungo, R.T. Ipragliflozin: First global approval. Drugs 2014, 74, 611–617. [Google Scholar] [CrossRef] [PubMed]
- Poole, R.M.; Prossler, J.E. Tofogliflozin: First global approval. Drugs 2014, 74, 939–944. [Google Scholar] [CrossRef]
- Zell, M.; Husser, C.; Kuhlmann, O.; Schwab, D.; Uchimura, T.; Kemei, T.; Kawashima, K.; Yamane, M.; Pähler, A. Metabolism and mass balance of SGLT2 inhibitor tofogliflozin following oral administration to humans. Xenobiotica 2014, 44, 369–378. [Google Scholar] [CrossRef] [PubMed]
- Sasaki, T.; Seino, Y.; Fukatsu, A.; Sakai, S.; Samukawa, Y. Safety, pharmacokinetics, and pharmacodynamics of single and multiple luseogliflozin dosing in healthy Japanese males: A randomized, single-blind, placebo-controlled trial. Adv. Ther. 2014, 31, 345–361. [Google Scholar] [CrossRef] [PubMed]
- Schwab, D.; Portron, A.; Backholer, Z.; Lausecker, B.; Kawashima, K. A novel double-tracer technique to characterize absorption, distribution, metabolism and excretion (ADME) of [14C]tofogliflozin after oral administration and concomitant intravenous microdose administration of [13C]tofogliflozin in humans. Clin. Pharmacokinet. 2013, 52, 463–473. [Google Scholar] [CrossRef] [PubMed]
- Merovci, A.; Solis-Herrera, C.; Daniele, G.; Eldor, R.; Fiorentino, T.V.; Tripathy, D.; Xiong, J.; Perez, Z.; Norton, L.; Abdul-Ghani, M.A.; et al. Dapagliflozin improves muscle insulin sensitivity but enhances endogenous glucose production. J. Clin. Investig. 2014, 124, 509–514. [Google Scholar] [CrossRef]
- Yaribeygi, H.; Sathyapalan, T.; Maleki, M.; Jamialahmadi, T.; Sahebkar, A. Molecular mechanisms by which SGLT2 inhibitors can induce insulin sensitivity in diabetic milieu: A mechanistic review. Life Sci. 2020, 240, 117090. [Google Scholar] [CrossRef]
- List, J.F.; Whaley, J.M. Glucose dynamics and mechanistic implications of SGLT2 inhibitors in animals and humans. Kidney Int. Suppl. 2011, 120, S20–S27. [Google Scholar] [CrossRef] [PubMed]
- Khat, D.Z.; Husain, M. Molecular Mechanisms Underlying the Cardiovascular Benefits of SGLT2i and GLP-1RA. Curr. Diabetes Rep. 2018, 18, 45. [Google Scholar] [CrossRef] [PubMed]
- Pinto, L.C.; Rados, D.V.; Remonti, L.R.; Kramer, C.K.; Leitao, C.B.; Gross, J.L. Efficacy of SGLT2 inhibitors in glycemic control, weight loss and blood pressure reduction: A systematic review and meta-analysis. Diabetol. Metab. Syndr. 2015, 7 (Suppl. S1), A58. [Google Scholar] [CrossRef]
- Cai, X.; Ji, L.; Chen, Y.; Yang, W.; Zhou, L.; Han, X.; Zhang, S.; Ji, L. Comparisons of weight changes between sodium-glucose cotransporter 2 inhibitors treatment and glucagon-like peptide-1 analogs treatment in type 2 diabetes patients: A meta-analysis. J. Diabetes Investig. 2017, 8, 510–517. [Google Scholar] [CrossRef]
- Clamp, L.D.; Hume, D.J.; Lambert, E.V.; Kroff, J. Enhanced insulin sensitivity in successful, long-term weight loss maintainers compared with matched controls with no weight loss history. Nutr. Diabetes 2017, 7, e282. [Google Scholar] [CrossRef] [PubMed]
- Chen, H. Cellular inflammatory responses: Novel insights for obesity and insulin resistance. Pharmacol. Res. 2006, 53, 469–477. [Google Scholar] [CrossRef] [PubMed]
- Shamansurova, Z.; Tan, P.; Ahmed, B.; Pepin, E.; Seda, O.; Lavoie, J.L. Adipose tissue (P)RR regulates insulin sensitivity, fat mass and body weight. Mol. Metab. 2016, 5, 959–969. [Google Scholar] [CrossRef] [PubMed]
- Xu, L.; Ota, T. Emerging roles of SGLT2 inhibitors in obesity and insulin resistance: Focus on fat browning and macrophage polarization. Adipocyte 2018, 7, 121–128. [Google Scholar] [CrossRef] [PubMed]
- Xu, L.; Nagata, N.; Nagashimada, M.; Zhuge, F.; Ni, Y.; Chen, G.; Mayoux, E.; Kaneko, S.; Ota, T. SGLT2 Inhibition by Empagliflozin Promotes Fat Utilization and Browning and Attenuates Inflammation and Insulin Resistance by Polarizing M2 Macrophages in Diet-induced Obese Mice. eBioMedicine 2017, 20, 137–149. [Google Scholar] [CrossRef]
- Koike, Y.; Shirabe, S.I.; Maeda, H.; Yoshimoto, A.; Arai, K.; Kumakura, A.; Hirao, K.; Terauchi, Y. Effect of canagliflozin on the overall clinical state including insulin resistance in Japanese patients with type 2 diabetes mellitus. Diabetes Res. Clin. Pract. 2019, 149, 140–146. [Google Scholar] [CrossRef]
- Suridjan, I.; Pollock, B.G.; Verhoeff, N.P.; Voineskos, A.N.; Chow, T.; Rusjan, P.M.; Lobaugh, N.J.; Houle, S.; Mulsant, B.H.; Mizrahi, R. In-vivo imaging of grey and white matter neuroinflammation in Alzheimer’s disease: A positron emission tomography study with a novel radioligand, [18F]-FEPPA. Mol. Psychiatry 2015, 20, 1579–1587. [Google Scholar] [CrossRef]
- Walker, K.A.; Gottesman, R.F.; Wu, A.; Knopman, D.S.; Gross, A.L.; Mosley, T.H., Jr.; Selvin, E.; Windham, B.G. Systemic inflammation during midlife and cognitive change over 20 years: The ARIC Study. Neurology 2019, 92, e1256–e1267. [Google Scholar] [CrossRef] [PubMed]
- Lathe, R.; Sapronova, A.; Kotelevtsev, Y. Atherosclerosis and Alzheimer-diseases with a common cause? Inflammation, oxysterols, vasculature. BMC Geriatr. 2014, 14, 36. [Google Scholar] [CrossRef] [PubMed]
- Kelley, N.; Jeltema, D.; Duan, Y.; He, Y. The NLRP3 Inflammasome: An Overview of Mechanisms of Activation and Regulation. Int. J. Mol. Sci. 2019, 20, 3328. [Google Scholar] [CrossRef]
- Tejera, D.; Mercan, D.; Sanchez-Caro, J.M.; Hanan, M.; Greenberg, D.; Soreq, H.; Latz, E.; Golenbock, D.; Heneka, M.T. Systemic inflammation impairs microglial Aβ clearance through NLRP 3 inflammasome. EMBO J. 2019, 38, e101064. [Google Scholar] [CrossRef] [PubMed]
- Lonnemann, N.; Hosseini, S.; Marchetti, C.; Skouras, D.B.; Stefanoni, D.; D’Alessandro, A.; Dinarello, C.A.; Korte, M. The NLRP3 inflammasome inhibitor OLT1177 rescues cognitive impairment in a mouse model of Alzheimer’s disease. Proc. Natl. Acad. Sci. USA 2020, 117, 32145–32154. [Google Scholar] [CrossRef]
- Heerspink, H.J.; Perco, P.; Mulder, S.; Leierer, J.; Hansen, M.K.; Heinzel, A.; Mayer, G. Canagliflozin reduces inflammation and fibrosis biomarkers: A potential mechanism of action for beneficial effects of SGLT2 inhibitors in diabetic kidney disease. Diabetologia 2019, 62, 1154–1166. [Google Scholar] [CrossRef]
- Lee, N.; Heo, Y.J.; Choi, S.E.; Jeon, J.Y.; Han, S.J.; Kim, D.J.; Kang, Y.; Lee, K.W.; Kim, H.J. Anti-inflammatory Effects of Empagliflozin and Gemigliptin on LPS-Stimulated Macrophage via the IKK/NF-κB, MKK7/JNK, and JAK2/STAT1 Signalling Pathways. J. Immunol. Res. 2021, 2021, 9944880. [Google Scholar] [CrossRef]
- Wiciński, M.; Wódkiewicz, E.; Górski, K.; Walczak, M.; Malinowski, B. Perspective of SGLT2 inhibition in treatment of conditions connected to neuronal loss: Focus on Alzheimer’s disease and ischemia-related brain injury. Pharmaceuticals 2020, 13, 379. [Google Scholar] [CrossRef]
- Noel, J.A.; Hougen, I.; Sood, M.M. The Intersection of SGLT2 Inhibitors, Cognitive Impairment, and CKD. Front. Neurol. 2022, 13, 823569. [Google Scholar] [CrossRef]
- Yamazaki, Y.; Harada, S.; Wada, T.; Hagiwara, T.; Yoshida, S.; Tokuyama, S. Sodium influx through cerebral sodium-glucose transporter type 1 exacerbates the development of cerebral ischemic neuronal damage. Eur. J. Pharmacol. 2017, 799, 103–110. [Google Scholar] [CrossRef]
- Ishida, N.; Saito, M.; Sato, S.; Koepsell, H.; Taira, E.; Hirose, M. SGLT1 participates in the development of vascular cognitive impairment in a mouse model of small vessel disease. Neurosci. Lett. 2020, 727, 134929. [Google Scholar] [CrossRef] [PubMed]
- Sim, A.Y.; Barua, S.; Kim, J.Y.; Lee, Y.H.; Lee, J.E. Role of dpp-4 and sglt2 inhibitors connected to alzheimer disease in type 2 diabetes mellitus. Front. Neurosci. 2021, 15, 708547. [Google Scholar] [CrossRef]
- Usman, M.S.; Siddiqi, T.J.; Memon, M.M.; Khan, M.S.; Rawasia, W.F.; Talha Ayub, M.; Sreenivasan, J.; Golzar, Y. Sodium-glucose co-transporter 2 inhibitors and cardiovascular outcomes: A systematic review and meta-analysis. Eur. J. Prev. Cardiol. 2018, 25, 495–502. [Google Scholar] [CrossRef]
- Hierro-Bujalance, C.; Infante-Garcia, C.; Del Marco, A.; Herrera, M.; Carranza-Naval, M.J.; Suarez, J.; Alves-Martinez, P.; Lubian-Lopez, S.; Garcia-Alloza, M. Empagliflozin reduces vascular damage and cognitive impairment in a mixed murine model of Alzheimer’s disease and type 2 diabetes. Alzheimers Res. Ther. 2020, 12, 40. [Google Scholar] [CrossRef] [PubMed]
- Uddin, M.S.; Rahman, M.A.; Kabir, M.T.; Behl, T.; Mathew, B.; Perveen, A.; Barreto, G.E.; Bin-Jumah, M.N.; Abdel-Daim, M.M.; Ashraf, G.M. Multifarious roles of mTOR signaling in cognitive aging and cerebrovascular dysfunction of Alzheimer’s disease. IUBMB Life 2020, 72, 1843–1855. [Google Scholar] [CrossRef]
- 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] [PubMed]
- Esterline, R.; Oscarsson, J.; Burns, J. A role for sodium glucose cotransporter 2 inhibitors (SGLT2is) in the treatment of Alzheimer’s disease? Int. Rev. Neurobiol. 2020, 155, 113–140. [Google Scholar] [CrossRef] [PubMed]
- Packer, M. Longevity genes, cardiac ageing, and the pathogenesis of cardiomyopathy: Implications for understanding the effects of current and future treatments for heart failure. Eur. Heart J. 2020, 41, 3856–3861. [Google Scholar] [CrossRef]
- Stanciu, G.D.; Rusu, R.N.; Bild, V.; Filipiuc, L.E.; Tamba, B.I.; Ababei, D.C. Systemic actions of sglt2 inhibition on chronic mtor activation as a shared pathogenic mechanism between alzheimer’s disease and diabetes. Biomedicines 2021, 9, 576. [Google Scholar] [CrossRef]
- Van Skike, C.E.; Jahrling, J.B.; Olson, A.B.; Sayre, N.L.; Hussong, S.A.; Ungvari, Z.; Lechleiter, J.D.; Galvan, V. Inhibition of mTOR protects the blood-brain barrier in models of Alzheimer’s disease and vascular cognitive impairment. Am. J. Physiol. Heart Circ. Physiol. 2018, 314, H693–H703. [Google Scholar] [CrossRef]
- Emedina, M.; Avila, J. The role of extracellular Tau in the spreading of neurofibrillary pathology. Front. Cell. Neurosci. 2014, 8, 113. [Google Scholar]
- Holtzman, D.M.; Morris, J.C.; Goate, A.M. Alzheimer’s Disease: The Challenge of the Second Century. Sci. Transl. Med. 2011, 3, 77. [Google Scholar] [CrossRef] [PubMed]
- Sa-Nguanmoo, P.; Tanajak, P.; Kerdphoo, S.; Jaiwongkam, T.; Pratchayasakul, W.; Chattipakorn, N.; Chattipakorn, S.C. SGLT2-inhibitor and DPP-4 inhibitor improve brain function via attenuating mitochondrial dysfunction, insulin resistance, inflammation, and apoptosis in HFD-induced obese rats. Toxicol. Appl. Pharmacol. 2017, 333, 43–50. [Google Scholar] [CrossRef] [PubMed]
- Mone, P.; Lombardi, A.; Gambardella, J.; Pansini, A.; Macina, G.; Morgante, M.; Frullone, S.; Santulli, G. EmpaglifloziImproves Cognitive Impairment in Frail Older Adults With Type 2 Diabetes and Heart Failure With Preserved Ejection Fraction. Diabetes Care 2022, 45, 1247–1251. [Google Scholar] [CrossRef] [PubMed]
- Arafa, N.M.S.; Ali, E.H.A.; Hassan, M.K. Canagliflozin prevents scopolamine-induced memory impairment in rats: Comparison with galantamine hydrobromide action. Chem. Interact. 2017, 277, 195–203. [Google Scholar] [CrossRef] [PubMed]
- Shaikh, S.; Rizvi, S.M.D.; Shakil, S.; Riyaz, S.; Biswas, D.; Jahan, R. Forxiga (dapagliflozin): Plausible role in the treatment of diabetes-associated neurological disorders. Biotechnol. Appl. Biochem. 2016, 63, 145–150. [Google Scholar] [CrossRef]
- Langbaum, J.B.; Chen, K.; Lee, W.; Reschke, C.; Bandy, D.; Fleisher, A.S.; Alexander, G.E.; Foster, N.L.; Weiner, M.W.; Koeppe, R.A.; et al. Categorical and correlational analyses of baseline fluorodeoxyglucose positron emission tomography images from the Alzheimer’s Disease Neuroimaging Initiative (ADNI). Neuroimage 2009, 45, 1107–1116. [Google Scholar] [CrossRef]
Parameters | Glucagon-like Peptide-1 (GLP-1) Receptor Agonists | ||||
---|---|---|---|---|---|
Liraglutide | Exenatide | Dulaglutide | Semaglutide | Lixisenatide | |
Molecular Weight [kDa] | 3.8 | 3.7 | 59.7 | 4.1 | 3.3 |
Cmax | 1.6 ng/mL | 300 ug/mL | 51.6 ng/mL | 3.4 ng/mL | 35 ng/mL |
The volume of distribution [L] | 11–17 | 28 | 3.09–6 | 8–9.4 | 100 |
Tmax [h] | 9–12 | 2.4 | 90 | 165–184 | 3 |
Bioavailability [%] | 55 | 22–25 | 47–65 | 89 | N/A |
Ability to cross the BBB | YES | YES | NO | NO | YES |
References | [106,107,108] | [106,109,110,111,112,113,114] | [106,115] | [106,116,117,118] | [106,110,119,120,121] |
Parameters | DPP-4 Inhibitors | |||||
---|---|---|---|---|---|---|
Sitagliptin | Vildagliptin | Saxagliptin | Alogliptin | Linagliptin | Gemigliptin | |
Molecular Weight [g/mol] | 407.31 | 303.4 | 315.417 | 339.399 | 472.553 | 489.370 |
Cmax | 491.7 (135.9) ng/mL | 308 ± 67 ng/mL | 640 (21) ng/mL | 57.8 ng/mL | 11–12 nmol/L | 62.7 ng/mL |
The volume of distribution [L] | 198 | 71 | 151 | 417 L | 1110 L | fill |
Tmax [h] | 2,5 | 1.75 | 2 | 1,5 | 2 | 1.8 |
Bioavailability [%] | 87% | 85% | 75% | 100% | 30% | 63% |
Ability to cross the BBB | YES | YES | YES | YES | YES | N/A |
Half-life | 12.4 h | 2.5 h, 3.1 h (active metabolite) | 21 h | 12 h | 17.1 | |
The primary route of elimination | Urine | Bile duct, urine | Urine, feces | Feces, urine | Urine, feces | |
References | [189,190,191,192] |
Parameters | SGLT-2 Inhibitors | |||||
---|---|---|---|---|---|---|
Dapagliphlozin | Canagliphlozin | Empagliphlozin | Ipragliphlozin | Luseogliphlozin | Tofogliphlozin | |
Molecular Weight [kDa] | 408.9 | 444.5 | 450.9 | 404.5 | 434.5 | 386.4 |
Cmax | 2465 nmol/L for the 100 mg dose 7828 nmol/L for the 300 mg dose | 2465 nmol/L for the 100 mg dose 7828 nmol/L for the 300 mg dose | 259 nmol/L for the 10 mg dosxe 687 nmol/L for the 25 mg dose | 1277 ± 360a | NA | NA |
The volume of distribution [L] | 118 | 119 | 74 | NA | 39 | 50 |
Tmax [h] | 1.5 | 1.5 | 1 | 1.5 | 1.5 | 0.75 |
Bioavailability [%] | 78% | 65% | 86% | 90.2% | NA | 97.5 |
Ability to cross the BBB | YES | YES | YES | YES | YES | YES |
Half-life | 12.9 h | 10.6 h for the 100 mg dose 13.1 h for the 300 mg dose | 12.4 h | 14.97 h | NA | NA |
The primary route of elimination | feces/urine | |||||
References | [210,211,212,213,214] | [120,210,214] | [210,211,214] | [210,214,215,216] | [210,214,217] | [210,214,218,219,220] |
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
Złotek, M.; Kurowska, A.; Herbet, M.; Piątkowska-Chmiel, I. GLP-1 Analogs, SGLT-2, and DPP-4 Inhibitors: A Triad of Hope for Alzheimer’s Disease Therapy. Biomedicines 2023, 11, 3035. https://doi.org/10.3390/biomedicines11113035
Złotek M, Kurowska A, Herbet M, Piątkowska-Chmiel I. GLP-1 Analogs, SGLT-2, and DPP-4 Inhibitors: A Triad of Hope for Alzheimer’s Disease Therapy. Biomedicines. 2023; 11(11):3035. https://doi.org/10.3390/biomedicines11113035
Chicago/Turabian StyleZłotek, Magdalena, Antonina Kurowska, Mariola Herbet, and Iwona Piątkowska-Chmiel. 2023. "GLP-1 Analogs, SGLT-2, and DPP-4 Inhibitors: A Triad of Hope for Alzheimer’s Disease Therapy" Biomedicines 11, no. 11: 3035. https://doi.org/10.3390/biomedicines11113035
APA StyleZłotek, M., Kurowska, A., Herbet, M., & Piątkowska-Chmiel, I. (2023). GLP-1 Analogs, SGLT-2, and DPP-4 Inhibitors: A Triad of Hope for Alzheimer’s Disease Therapy. Biomedicines, 11(11), 3035. https://doi.org/10.3390/biomedicines11113035