Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2382 2023-12-15 12:33:55 |
2 Reference format revised. Meta information modification 2382 2023-12-18 07:06:51 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Mohamed-Mohamed, H.; García-Morales, V.; Sánchez Lara, E.M.; González-Acedo, A.; Pardo-Moreno, T.; Tovar-Gálvez, M.I.; Melguizo-Rodríguez, L.; Ramos-Rodríguez, J.J. Diabetes Mellitus, Alzheimer’s Disease and Vascular Dementia. Encyclopedia. Available online: https://encyclopedia.pub/entry/52811 (accessed on 27 April 2024).
Mohamed-Mohamed H, García-Morales V, Sánchez Lara EM, González-Acedo A, Pardo-Moreno T, Tovar-Gálvez MI, et al. Diabetes Mellitus, Alzheimer’s Disease and Vascular Dementia. Encyclopedia. Available at: https://encyclopedia.pub/entry/52811. Accessed April 27, 2024.
Mohamed-Mohamed, Himan, Victoria García-Morales, Encarnación María Sánchez Lara, Anabel González-Acedo, Teresa Pardo-Moreno, María Isabel Tovar-Gálvez, Lucía Melguizo-Rodríguez, Juan José Ramos-Rodríguez. "Diabetes Mellitus, Alzheimer’s Disease and Vascular Dementia" Encyclopedia, https://encyclopedia.pub/entry/52811 (accessed April 27, 2024).
Mohamed-Mohamed, H., García-Morales, V., Sánchez Lara, E.M., González-Acedo, A., Pardo-Moreno, T., Tovar-Gálvez, M.I., Melguizo-Rodríguez, L., & Ramos-Rodríguez, J.J. (2023, December 15). Diabetes Mellitus, Alzheimer’s Disease and Vascular Dementia. In Encyclopedia. https://encyclopedia.pub/entry/52811
Mohamed-Mohamed, Himan, et al. "Diabetes Mellitus, Alzheimer’s Disease and Vascular Dementia." Encyclopedia. Web. 15 December, 2023.
Diabetes Mellitus, Alzheimer’s Disease and Vascular Dementia
Edit

Type 2 diabetes mellitus (T2D) is a metabolic disease reaching pandemic levels worldwide. In parallel, Alzheimer’s disease (AD) and vascular dementia (VaD) are the two leading causes of dementia in an increasingly long-living Western society. Numerous epidemiological studies support the role of T2D as a risk factor for the development of dementia. 

Alzheimer’s disease dementia vascular dementia diabetes insulin neurodegeneration neuron death neuroinflammation

1. Introduction

Diabetes mellitus (DM) is a chronic disease characterized by elevated blood glucose levels due to the body’s inability to produce sufficient insulin or use it efficiently. Although there are different types of DM, the most common are type 1 diabetes (T1D) and type 2 diabetes (T2D) [1].
On the other hand, dementia is a syndrome that encompasses a heterogeneous group of diseases of the central nervous system (CNS), characterized by cognitive impairment [2]. In terms of incidence, the main types of dementia are Alzheimer’s disease (AD), vascular dementia (VaD), dementia with Lewy bodies and frontotemporal lobar dementia [3]. According to a WHO report, dementia currently affects around 50 million people, mostly from low- and middle-income countries, and about 10 million cases are registered every year [4]. It is estimated that by 2030, 82 million people will have dementia [4], and this number will increase to 130 million by 2050 [5].
AD is a neurodegenerative disease of the central nervous system and is characterized by a progressive deterioration of higher brain functions, affecting the ability to make decisions and execute them [6][7]. AD patients survive an average of 7 years, and slightly less than 3% of those affected live more than 14 years after diagnosis [8]. In terms of epidemiology, AD is the leading cause of dementia and occurs most frequently in people over 65 years of age (with a 10% prevalence), representing between 60% and 75% of all dementia cases [9][10]. The major risk factor for AD is age, and as life expectancy progressively increases, so does the number of people affected by this disease. Currently, more than 50 million people worldwide suffer from AD, with an associated economic cost of 30,000 EUR/year/patient in developed countries [11]. Projections for 2050 suggest that the number of people affected could exceed 107 million [9]. These rates represent an unbearable economic and social drain.

2. Link between Diabetes Mellitus, Alzheimer’s Disease and Vascular Dementia

Over the last 15 years, the relationship between T2D, AD and VaD has been extensively studied. Based on published studies, the data suggest that T2D, among other metabolic pathologies, could contribute to the development of a neurodegenerative process that would be a precursor to the development of dementia. Following this idea, DM seems to play a role at this level. The bibliography shows that the incidence of both pathologies increases with age, and it is common to find them coexisting. In fact, some authors claim that 45% of people with T2D suffer from mild cognitive impairment [12], raising the chance of developing dementia by 1.5–2.5 times [13]. In contrast, people with established dementia show insulin disturbances, with altered fasting glucose levels [14].
It also appears that the risk of dementia increases as patients develop other risk factors, including heart disease, hyperlipidemia, hypercholesterolemia or smoking, although T2D is important for its synergistic capacity [13][15][16]. In this relationship, insulin levels and insulin resistance are best correlated with the severity and progression of VaD [17][18]. Furthermore, it seems that DM promotes vascular damage caused by high levels of glucose in the blood, which is accentuated if it coexists with other pathologies that affect blood vessels, such as arteriosclerosis and hypertension [19][20].
The relationship between T2D and VaD can be based on processes that converge between the two pathologies, described below.

2.1. Insulin Receptors in Central Nervous System

Insulin receptors at the central level are located in astrocytes and neuronal synapses and are very abundant in regions of the basal forebrain, such as the septum (origin of cortical and hippocampal cholinergic innervation) [21], and areas particularly relevant for learning and memory processes, such as the cortex and the hippocampus [22]. This wide distribution of insulin receptors explains the involvement of insulin in cognitive processes [23], probably mediated by relevant neurotransmitters in AD, such as norepinephrine and acetylcholine [24][25]. Insulin also contributes to synaptic plasticity mediated by insulin receptors [26]. Similarly, insulin regulates glucose metabolism as the main nutrient of the CNS, directly involved in learning and memory processes [27][28]. In fact, the acute administration of insulin to humans and rodents produces an improvement in cognitive processes and memory [29][30][31][32][33] as well as an increase in the expression of insulin receptors in the dentate gyrus, leading to better performance in spatial memory tests [32][34]. Indeed, some authors have postulated that insulin could counterbalance AD pathology [35].

2.2. Type 2 Diabetes Progression Correlated with Pancreatic Amilin Deposition as Brain Aβ Deposition Correlated with Alzheimer’s Disease Progression

The progression of T2D is correlated with pancreatic amylin deposition, which is similar to that of brain Aβ. In addition, insulin, amylin and Aβ are degraded peripherally by the insulin-degrading enzyme (IDE) [36]. This suggests that these substrates may compete at this level [37]. It has been postulated that substrate imbalance may influence the pathogenesis of AD and T2D [38].
From another point of view, insulin resistance detected in AD patients could be mediated by a decrease in the activity of the enzymes responsible for its degradation. Both the insulin-degrading enzyme (IDE) and neprelisine (NEP) are involved in insulin degradation as well as Aβ and amylin degradation [39]. Accordingly, it has been postulated that an imbalance of substrates can affect the degradation rate of other substrates and possibly influence the pathogenesis of T2D and AD [38][40][41][42]. A decrease in IDE expression may result in reduced insulin and Aβ degradation in the brain [41][43]. Consequently, it is conceivable that actions promoted by elevated insulin levels, or its deficiency, may also be a link between T2D and AD.

2.3. Insulin-Like Growth Factor

Insulin and the insulin-like growth factor (IGF) have similar structures and play a significant role in the regulation of aging. IGF acts as a cellular growth factor but also plays a hormonal role in regulating growth and metabolism at the systemic level [44]. IGF is the main prenatal and postnatal growth factor. This is why low levels of insulin during gestation can lead to slower growth and low height and weight of the offspring [33][45]. In contrast, an increase in insulin levels at an early stage, as occurs in maternal T2D, leads to large and overweight children, among other complications [46].
In this context, insulin plays a crucial role in the development of neuronal complexity, as well as in neurogenesis in early neonatal development. In this sense, insulin deprivation during early life may result in reduced neural network development, whereas exogenous administration of insulin may promote increased neural development and complexity [33][47]. A more complex neural network is a protective factor against the development of dementia [48]. Based on the aforementioned, a gestational environment with altered insulin levels could condition the possible development of dementia in the later stages of life [49][50].
Additionally, plasma inulin can cross the BBB in a soluble form and gain access to neurons, microvasculature and even immature neuronal bodies [51]. Insulin plays a key role as a growth-regulating hormone during the early stages of life. In animal models, it has been shown that an imbalance in insulin levels also impairs neurogenesis in early life [33], as well as in more mature and long-lived phases [52]. In addition, the coexistence of T2D with AD pathology reduces neurogenesis and thus neuronal replacement from stages prior to cognitive decline [53]. Insulin is also involved in the regulation of neuronal and synaptic functions in the hippocampus, cortex and cerebellum, protecting neurons from neurodegeneration and cell death [54][55][56][57].

2.4. Insulin Promotes Typical Features of Alzheimer’s Disease

Tau pathology is one of the main features of AD, and many studies have reported the impact of insulin dysfunction and diabetes on it [58][59][60]. Indeed, high insulin levels in the brain, induced by prediabetes or T2D, may increase the hyperphosphorylation of tau protein [61]. It is feasible that higher levels of phosphorylated tau observed in hyperinsulinemia and T2D states could be mediated by insulin receptors at the central level [59]. Additionally, T1D hypoinsulinemia may increase tau hyperphosphorylation [62]. In this regard, a clinical phase I study with the drug SCR-1693 showed a reduction in tau protein phosphorylation levels associated with an improvement in cognitive and central insulin resistance [63].
Inflammation of the CNS is increasingly regarded to play a role in cognitive disorders such as dementia [64]. Insulin promotes the expression of proinflammatory cytokines such as α-TNF and IL6 [65], which are the most important proinflammatory cytokines. Moreover, these cytokines negatively affect the metabolism of Aβ oligomers [66].
On the other hand, one of the mechanisms proposed to link insulin to cognitive impairment is its role in Aβ metabolism [37][41]. In this regard, insulin promotes the amyloidogenic pathway by modulating β and γ-secretases [40][67][68]. Additionally, insulin inhibits or hinders the passage of Aβ through the BBB [69]. Following this idea, insulin would prevent the clearance of Aβ and promote its accumulation in the brain [70]. In return, Aβ interferes with insulin signaling in the CNS; in fact, soluble Aβ oligomers may disrupt insulin signaling in hippocampus neuronal cultures [71]

2.5. Role of Prediabetes and Diabetes Mechanism in the Neurodegenerative Process of Alzheimer’s Disease and Vascular Dementia

Previous epidemiological and clinical studies support a close relationship between T2D and AD [72][73]; however, the underlying linking mechanisms are not yet fully understood. It also remains unclear whether hyperinsulinemia and insulin resistance, indicative of a prediabetic state prior to T2D, may induce or accelerate central pathology in AD, in a similar manner to that induced by T2D. Indeed, glucose and insulin play a crucial role in maintaining normal brain activity, and alterations of insulin-dependent functions could be associated with central pathological conditions observed in AD [66][72][74].
The available scientific evidence on this relationship supports that the first pathological event in the DM disorder, as a promoter of dementia, is insulin resistance in the CNS [17][51][75]. Insulin plays an important role in cell growth and differentiation, as well as in protein synthesis [51][76]. Additionally, insulin inhibits catabolic processes such as glycolysis, lipolysis and proteolysis [76]. The wide distribution of insulin receptors throughout the CNS underscores its importance in central glucose homeostasis processes and its role in cognitive processes and neuronal development [51]. In this sense, it has been described that alterations in insulin balance in the CNS accelerate the brain aging process, increasing vascular damage and primarily affecting synaptic plasticity [76][77][78]. Vascular damage is often one of the first central events in diabetes [79], even in the prediabetic stages, and compensatory high insulin levels are sufficient to cause this damage [80]. Simultaneously with vascular damage, brain aging occurs, which is translated into reduced neuronal arborization and synaptic density. The loss of neuronal and synaptic density has been proposed as one of the best pathological markers for the assessment of AD. There is ample scientific evidence that different states of Aβ aggregation, from the most soluble forms to the formation of senile plaques, are neurotoxic [81][82][83]. It has been reported that synaptic loss promotes cell dedifferentiation and ultimately neuronal death [84].
In the second stage, scholars propose that the sequence of events involves the inflammatory process and glial activation. It has been suggested that the insulin resistance typical of prediabetes and T2D may exacerbate the inflammatory process when it interacts with the presence of Aβ [66]. Previous studies report that the presence of Aβ is sufficient to promote microglial activation and the inflammatory process [85][86]. Vascular damage and the T2D-induced imbalance of Aβ pathology toward more soluble forms have been shown to induce an increase in cytotoxic and pro-inflammatory cytokines and increase microglial activity [87][88]. This inflammatory process would increase oxidative molecular species, generating a harmful environment that would promote neurodegenerative processes [89]. Microglial cells, faced with vascular damage and the accumulation of toxic substances such as Aβ, begin to produce proinflammatory cytokines (IL-1β, IL-6, IL-18 and tumor necrosis factor-α (TNF-α)), chemokines (CCL1, CCL5, CXCL1), small messenger molecules (prostaglandins, NO) and ROS [90]. Although most cells involved in this process of neuroinflammation are microglia and astrocytes, capillary endothelial cells are also involved, as well as some infiltrating blood cells, which are more frequent when there is tissue damage in the BBB [90][91]. This pro-inflammatory ecosystem could lead to synaptic dysfunction, neuronal death and inhibition of neurogenesis [53][87][92].

3. Conclusions

Growing evidence suggests that T2D may increase the risk of developing AD. Several studies have found that insulin resistance and metabolic dysfunction associated with T2D negatively affect the brain and increase the accumulation of Aβ. Also, T2D promotes chronic inflammation, oxidative stress and vascular dysfunction in the brain which links T2D with AD and VaD.
Understanding the relationship between DM and AD is essential to prevent the development of AD through metabolic control. Likewise, knowledge of the involvement of DM in the progress and development of AD can help us address new therapeutic strategies for its treatment. In this sense, treatment to control T2D, such as metformin, has shown a beneficial effect, like neuroprotective, anti-inflammatory and antioxidant action in animal models [93][94][95]. However, limited studies in humans have shown a more discrete positive relationship with the use of metformin as a treatment for AD [96]. The possible beneficial effects of metformin as a treatment for Alzheimer’s disease are still being studied and are a hot topic (for a review, see [97]).
Another emerging candidate for a therapeutic approach to AD an VaD is the application of intranasal insulin. This treatment has been shown to have beneficial effects in reducing inflammation and improving immune function. In addition, intranasal insulin improves cognitive status and helps to maintain cognitive abilities [98][99]. Other studies have shown that intranasal insulin as a treatment for AD has neuroprotective effects, and its use can maintain the insulin brain signaling to improve cognitive health [100] by reducing the P-tau/Aβ42 ratio [101] and preserving the white matter volume in deep and frontal regions, which correlates with AD progression [102]. However, recent long-term studies in humans have shown limited efficacy of intranasal insulin as a treatment for AD [101][103]. The current disagreement in the literature shows the need to carry out studies that demonstrate the usefulness of this treatment in the control of AD.
Finally, as the understanding of the relationship between T2D, AD and VaD increases, there are new opportunities for prevention and treatment. Improving glycemic control and promoting healthy lifestyles should be explored further. Thus, early attention to risk factors associated with dementia, such as T2D, may play a crucial role in preventing or delaying dementia.

References

  1. American Diabetes Association. 2. Classification and Diagnosis of Diabetes: Standards of Medical Care in Diabetes-2019. Diabetes Care 2019, 42 (Suppl. S1), S13–S28.
  2. Santos, M.A.O.; Bezerra, L.S.; Correia, C.d.C.; Bruscky, I.S. Neuropsychiatric symptoms in vascular dementia: Epidemiologic and clinical aspects. Dement. Neuropsychol. 2018, 12, 40–44.
  3. Cunningham, E.L.; McGuinness, B.; Herron, B.; Passmore, A.P. Dementia. Ulst. Med. J. 2015, 84, 79–87.
  4. OMS. Dementia; 2019.
  5. Garre-Olmo, J. Epidemiology of Alzheimer’s disease and other dementias. Rev. Neurol. 2018, 66, 377–386.
  6. Garcia-Morales, V.; Gonzalez-Acedo, A.; Melguizo-Rodriguez, L.; Pardo-Moreno, T.; Costela-Ruiz, V.J.; Montiel-Troya, M.; Ramos-Rodriguez, J.J. Current Understanding of the Physiopathology, Diagnosis and Therapeutic Approach to Alzheimer’s Disease. Biomedicines 2021, 9, 1910.
  7. Pardo-Moreno, T.; González-Acedo, A.; Rivas-Domínguez, A.; García-Morales, V.; García-Cozar, F.J.; Ramos-Rodríguez, J.J.; Melguizo-Rodríguez, L. Therapeutic Approach to Alzheimer’s Disease: Current Treatments and New Perspectives. Pharmaceutics 2022, 14, 1117.
  8. Molsa, P.K.; Marttila, R.J.; Rinne, U.K. Long-term survival and predictors of mortality in Alzheimer’s disease and multi-infarct dementia. Acta Neurol. Scand. 1995, 91, 159–164.
  9. Niu, H.; Álvarez-Álvarez, I.; Guillén-Grima, F.; Aguinaga-Ontoso, I. Prevalence and incidence of Alzheimer’s disease in Europe: A meta-analysis. Neurologia 2017, 32, 523–532.
  10. Ayodele, T.; Rogaeva, E.; Kurup, J.T.; Beecham, G.; Reitz, C. Early-Onset Alzheimer’s Disease: What Is Missing in Research? Curr. Neurol. Neurosci. Rep. 2021, 21, 4.
  11. Lleo, A. Alzheimer’s disease: An ignored condition. Med. Clin. 2018, 150, 432–433.
  12. You, Y.; Liu, Z.; Chen, Y.; Xu, Y.; Qin, J.; Guo, S.; Huang, J.; Tao, J. The prevalence of mild cognitive impairment in type 2 diabetes mellitus patients: A systematic review and meta-analysis. Acta Diabetol. 2021, 58, 671–685.
  13. Ninomiya, T. Epidemiological Evidence of the Relationship Between Diabetes and Dementia. Adv. Exp. Med. Biol. 2019, 1128, 13–25.
  14. Lee, D.Y.; Kim, J.; Park, S.; Park, S.Y.; Yu, J.H.; Seo, J.A.; Kim, N.H.; Yoo, H.J.; Kim, S.G.; Choi, K.M.; et al. Fasting Glucose Variability and the Risk of Dementia in Individuals with Diabetes: A Nationwide Cohort Study. Diabetes Metab. J. 2022, 46, 923–935.
  15. Xue, M.; Xu, W.; Ou, Y.-N.; Cao, X.-P.; Tan, M.-S.; Tan, L.; Yu, J.-T. Diabetes mellitus and risks of cognitive impairment and dementia: A systematic review and meta-analysis of 144 prospective studies. Ageing Res. Rev. 2019, 55, 100944.
  16. Biessels, G.J.; Despa, F. Cognitive decline and dementia in diabetes mellitus: Mechanisms and clinical implications. Nat. Rev. Endocrinol. 2018, 14, 591–604.
  17. Bhattamisra, S.K.; Shin, L.Y.; Saad, H.I.B.M.; Rao, V.; Candasamy, M.; Pandey, M.; Choudhury, H. Interlink Between Insulin Resistance and Neurodegeneration with an Update on Current Therapeutic Approaches. CNS Neurol. Disord. Drug Targets 2020, 19, 174–183.
  18. Cetinkalp, S.; Simsir, I.Y.; Ertek, S. Insulin Resistance in Brain and Possible Therapeutic Approaches. Curr. Vasc. Pharmacol. 2014, 12, 553–564.
  19. Nagar, S.D.; Qian, J.; Boerwinkle, E.; Cicek, M.; Clark, C.R.; Cohn, E.; Gebo, K.; Loperena, R.; Mayo, K.; Mockrin, S.; et al. Investigation of hypertension and type 2 diabetes as risk factors for dementia in the All of Us cohort. Sci. Rep. 2022, 12, 19797.
  20. Liu, C.L.; Lin, M.Y.; Hwang, S.J.; Liu, C.K.; Lee, H.L.; Wu, M.T. Factors associated with type 2 diabetes in patients with vascular dementia: A population-based cross-sectional study. BMC Endocr. Disord. 2018, 18, 45.
  21. Bilotta, F.; Lauretta, M.P.; Tewari, A.; Haque, M.; Hara, N.; Uchino, H.; Rosa, G. Insulin and the Brain: A Sweet Relationship With Intensive Care. J. Intensive Care Med. 2017, 32, 48–58.
  22. Kleinridders, A.; Ferris, H.A.; Cai, W.; Kahn, C.R. Insulin Action in Brain Regulates Systemic Metabolism and Brain Function. Diabetes 2014, 63, 2232–2243.
  23. Soto, M.; Cai, W.; Konishi, M.; Kahn, C.R. Insulin signaling in the hippocampus and amygdala regulates metabolism and neurobehavior. Proc. Natl. Acad. Sci. USA 2019, 116, 6379–6384.
  24. Roberson, M.R.; Harrell, L.E. Cholinergic activity and amyloid precursor protein metabolism. Brain Res. Rev. 1997, 25, 50–69.
  25. Schliebs, R.; Arendt, T. The significance of the cholinergic system in the brain during aging and in Alzheimer’s disease. J. Neural Transm. 2006, 113, 1625–1644.
  26. Abbott, M.A.; Wells, D.G.; Fallon, J.R. The insulin receptor tyrosine kinase substrate p58/53 and the insulin receptor are components of CNS synapses. J. Neurosci. 1999, 19, 7300–7308.
  27. Bingham, E.M.; Hopkins, D.; Smith, D.; Pernet, A.; Hallett, W.; Reed, L.; Marsden, P.K.; Amiel, S.A. The role of insulin in human brain glucose metabolism: An 18fluoro-deoxyglucose positron emission tomography study. Diabetes 2002, 51, 3384–3390.
  28. Mullins, R.J.; Mustapic, M.; Goetzl, E.J.; Kapogiannis, D. Exosomal biomarkers of brain insulin resistance associated with regional atrophy in Alzheimer’s disease. Hum. Brain Mapp. 2017, 38, 1933–1940.
  29. Craft, S.; Baker, L.D.; Montine, T.J.; Minoshima, S.; Watson, G.S.; Claxton, A.; Arbuckle, M.; Callaghan, M.; Tsai, E.; Plymate, S.R.; et al. Intranasal insulin therapy for Alzheimer disease and amnestic mild cognitive impairment: A pilot clinical trial. Arch. Neurol. 2012, 69, 29–38.
  30. Kamat, P.K.; Kalani, A.; Rai, S.; Tota, S.K.; Kumar, A.; Ahmad, A.S. Streptozotocin Intracerebroventricular-Induced Neurotoxicity and Brain Insulin Resistance: A Therapeutic Intervention for Treatment of Sporadic Alzheimer’s Disease (sAD)-Like Pathology. Mol. Neurobiol. 2016, 53, 4548–4562.
  31. Kern, W.; Peters, A.; Fruehwald-Schultes, B.; Deininger, E.; Born, J.; Fehm, H.L. Improving Influence of Insulin on Cognitive Functions in Humans. Neuroendocrinology 2001, 74, 270–280.
  32. Rajasekar, N.; Nath, C.; Hanif, K.; Shukla, R. Intranasal insulin improves cerebral blood flow, Nrf-2 expression and BDNF in STZ (ICV)-induced memory impaired rats. Life Sci. 2017, 173, 1–10.
  33. Ramos-Rodriguez, J.J.; Sanchez-Sotano, D.; Doblas-Marquez, A.; Infante-Garcia, C.; Lubian-Lopez, S.; Garcia-Alloza, M. Intranasal insulin reverts central pathology and cognitive impairment in diabetic mother offspring. Mol. Neurodegener. 2017, 12, 57.
  34. Zhao, W.; Chen, H.; Xu, H.; Moore, E.; Meiri, N.; Quon, M.J.; Alkon, D.L. Brain insulin receptors and spatial memory. Correlated changes in gene expression, tyrosine phosphorylation, and signaling molecules in the hippocampus of water maze trained rats. J. Biol. Chem. 1999, 274, 34893–34902.
  35. Semprini, R.; Koch, G.; Belli, L.; Lorenzo, F.D.; Ragonese, M.; Manenti, G.; Sorice, G.P.; Martorana, A. Insulin and the Future Treatment of Alzheimer’s Disease. CNS Neurol. Disord. Drug Targets 2016, 15, 660–664.
  36. Sousa, L.; Guarda, M.; Meneses, M.J.; Macedo, M.P.; Miranda, H.V. Insulin-degrading enzyme: An ally against metabolic and neurodegenerative diseases. J. Pathol. 2021, 255, 346–361.
  37. Qiu, W.Q.; Folstein, M.F. Insulin, insulin-degrading enzyme and amyloid-beta peptide in Alzheimer’s disease: Review and hypothesis. Neurobiol. Aging 2006, 27, 190–198.
  38. Götz, J.; Ittner, L.M.; Lim, Y.-A. Common features between diabetes mellitus and Alzheimer’s disease. Cell. Mol. Life Sci. 2009, 66, 1321–1325.
  39. Gasiorowski, K.; Brokos, B.; Leszek, J.; Tarasov, V.; Ashraf, G.; Aliev, G. Insulin Resistance in Alzheimer Disease: p53 and MicroRNAs as Important Players. Curr. Top Med. Chem. 2017, 17, 1429–1437.
  40. Eckman, E.A.; Eckman, C.B. Abeta-degrading enzymes: Modulators of Alzheimer’s disease pathogenesis and targets for therapeutic intervention. Biochem. Soc. Trans. 2005, 33 Pt 5, 1101–1105.
  41. Farris, W.; Mansourian, S.; Chang, Y.; Lindsley, L.; Eckman, E.A.; Frosch, M.P.; Eckman, C.B.; Tanzi, R.E.; Selkoe, D.J.; Guenette, S. Insulin-degrading enzyme regulates the levels of insulin, amyloid beta-protein, and the beta-amyloid precursor protein intracellular domain in vivo. Proc. Natl. Acad. Sci. USA 2003, 100, 4162–4167.
  42. Liu, Z.; Zhu, H.; Fang, G.G.; Walsh, K.; Mwamburi, M.; Wolozin, B.; Abdul-Hay, S.O.; Ikezu, T.; Leissring, M.A.; Qiu, W.Q. Characterization of insulin degrading enzyme and other amyloid-beta degrading proteases in human serum: A role in Alzheimer’s disease? J. Alzheimers Dis. 2012, 29, 329–340.
  43. Zhao, L.; Teter, B.; Morihara, T.; Lim, G.P.; Ambegaokar, S.S.; Ubeda, O.J.; Frautschy, S.A.; Cole, G.M. Insulin-degrading enzyme as a downstream target of insulin receptor signaling cascade: Implications for Alzheimer’s disease intervention. J. Neurosci. 2004, 24, 11120–11126.
  44. Stoeckel, L.E. Brain insulin resistance as a contributing factor to dementia and psychiatric disease. Exp. Neurol. 2020, 326, 113205.
  45. Harvey, N.C.; Holroyd, C.; Ntani, G.; Javaid, K.; Cooper, P.; Moon, R.; Cole, Z.; Tinati, T.; Godfrey, K.; Dennison, E.; et al. Vitamin D supplementation in pregnancy: A systematic review. Health Technol. Assess. 2014, 18, 1–190.
  46. Moon, J.H.; Jang, H.C. Gestational Diabetes Mellitus: Diagnostic Approaches and Maternal-Offspring Complications. Diabetes Metab. J. 2022, 46, 3–14.
  47. Abbasi, Z.; Seno, M.M.G.; Fereidoni, M. A Neonatal Mild Defect in Brain Insulin Signaling Predisposes a Subclinical Model of Sporadic Alzheimer’s to Develop the Disease. J. Mol. Neurosci. 2021, 71, 1473–1484.
  48. De Strooper, B.; Karran, E. The Cellular Phase of Alzheimer’s Disease. Cell 2016, 164, 603–615.
  49. Ross, M.G.; Desai, M.; Khorram, O.; McKnight, R.A.; Lane, R.H.; Torday, J. Gestational programming of offspring obesity: A potential contributor to Alzheimer’s disease. Curr. Alzheimer Res. 2007, 4, 213–217.
  50. Cordner, Z.A.; Khambadkone, S.G.; Boersma, G.J.; Song, L.; Summers, T.N.; Moran, T.H.; Tamashiro, K.L. Maternal high-fat diet results in cognitive impairment and hippocampal gene expression changes in rat offspring. Exp. Neurol. 2019, 318, 92–100.
  51. Akhtar, A.; Sah, S. Insulin signaling pathway and related molecules: Role in neurodegeneration and Alzheimer’s disease. Neurochem. Int. 2020, 135, 104707.
  52. Ramos-Rodriguez, J.J.; Molina-Gil, S.; Ortiz-Barajas, O.; Jimenez-Palomares, M.; Perdomo, G.; Cozar-Castellano, I.; Lechuga-Sancho, A.M.; Garcia-Alloza, M. Central Proliferation and Neurogenesis Is Impaired in Type 2 Diabetes and Prediabetes Animal Models. PLoS ONE 2014, 9, e89229.
  53. Hierro-Bujalance, C.; del Marco, A.; Ramos-Rodríguez, J.J.; Infante-Garcia, C.; Gomez-Santos, S.B.; Herrera, M.; Garcia-Alloza, M. Cell proliferation and neurogenesis alterations in Alzheimer’s disease and diabetes mellitus mixed murine models. J. Neurochem. 2020, 154, 673–692.
  54. Zhao, W.-Q.; Alkon, D.L. Role of insulin and insulin receptor in learning and memory. Mol. Cell. Endocrinol. 2001, 177, 125–134.
  55. Chiu, S.-L.; Chen, C.-M.; Cline, H.T. Insulin Receptor Signaling Regulates Synapse Number, Dendritic Plasticity, and Circuit Function In Vivo. Neuron 2008, 58, 708–719.
  56. Morrison, C.D. Leptin signaling in brain: A link between nutrition and cognition? Biochim. Biophys. Acta 2009, 1792, 401–408.
  57. Ferrario, C.R.; Reagan, L. Insulin-mediated synaptic plasticity in the CNS: Anatomical, functional and temporal contexts. Neuropharmacology 2018, 136 Pt B, 182–191.
  58. El Khoury, N.B.; Gratuze, M.; Papon, M.-A.; Bretteville, A.; Planel, E. Insulin dysfunction and Tau pathology. Front. Cell. Neurosci. 2014, 8, 22.
  59. Mittal, K.; Katare, D. Shared links between type 2 diabetes mellitus and Alzheimer’s disease: A review. Diabetes Metab. Syndr. 2016, 10 (Suppl. S1), S144–S149.
  60. Park, C. Cognitive effects of insulin in the central nervous system. Neurosci. Biobehav. Rev. 2001, 25, 311–323.
  61. Ma, N.; Liang, Y.; Yue, L.; Liu, P.; Xu, Y.; Zhu, C. The identities of insulin signaling pathway are affected by overexpression of Tau and its phosphorylation form. Front. Aging Neurosci. 2022, 14, 1057281.
  62. Schechter, R.; Beju, D.; Miller, K.E. The effect of insulin deficiency on tau and neurofilament in the insulin knockout mouse. Biochem. Biophys. Res. Commun. 2005, 334, 979–986.
  63. Bi, A.; An, W.; Wang, C.; Hua, Y.; Fang, F.; Dong, X.; Chen, R.; Zhang, Z.; Luo, L. SCR-1693 inhibits tau phosphorylation and improves insulin resistance associated cognitive deficits. Neuropharmacology 2020, 168, 108027.
  64. Bevan-Jones, W.R.; Surendranathan, A.; Passamonti, L.; Rodríguez, P.V.; Arnold, R.; Mak, E.; Su, L.; Coles, J.P.; Fryer, T.D.; Hong, Y.T.; et al. Neuroimaging of Inflammation in Memory and Related Other Disorders (NIMROD) study protocol: A deep phenotyping cohort study of the role of brain inflammation in dementia, depression and other neurological illnesses. BMJ Open 2017, 7, e013187.
  65. Mitrou, P.; Boutati, E.; Lambadiari, V.; Tsegka, A.; Raptis, A.E.; Tountas, N.; Economopoulos, T.; Raptis, S.A.; Dimitriadis, G. Insulin resistance in hyperthyroidism: The role of IL6 and TNF alpha. Eur. J. Endocrinol. 2010, 162, 121–126.
  66. Bosco, D.; Fava, A.; Plastino, M.; Montalcini, T.; Pujia, A. Possible implications of insulin resistance and glucose metabolism in Alzheimer’s disease pathogenesis. J. Cell. Mol. Med. 2011, 15, 1807–1821.
  67. Wacławczyk, D.; Silberring, J.; Grasso, G. The insulin-degrading enzyme as a link between insulin and neuropeptides metabolism. J. Enzym. Inhib. Med. Chem. 2021, 36, 183–187.
  68. Fewlass, D.C.; Noboa, K.; Pi-Sunyer, F.X.; Johnston, J.M.; Yan, S.D.; Tezapsidis, N. Obesity-related leptin regulates Alzheimer’s Abeta. FASEB J. 2004, 18, 1870–1878.
  69. Rhea, E.M.; Banks, W.A. Role of the Blood-Brain Barrier in Central Nervous System Insulin Resistance. Front. Neurosci. 2019, 13, 521.
  70. Banks, W.A.; Jaspan, J.B.; Huang, W.; Kastin, A.J. Transport of Insulin Across the Blood-Brain Barrier: Saturability at Euglycemic Doses of Insulin. Peptides 1997, 18, 1423–1429.
  71. Zhao, W.; De Felice, F.G.; Fernandez, S.; Chen, H.; Lambert, M.P.; Quon, M.J.; Krafft, G.A.; Klein, W.L. Amyloid beta oligomers induce impairment of neuronal insulin receptors. FASEB J. 2008, 22, 246–260.
  72. Craft, S. The role of metabolic disorders in Alzheimer disease and vascular dementia: Two roads converged. Arch. Neurol. 2009, 66, 300–305.
  73. 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 2014, 63, 2262–2272.
  74. Garcia-Alloza, M. Streptozotocin as a tool to induce central pathology and cognitive impairment in rodents. In Streptozotocin: Uses, Mechanism of Action and SideEffects; Gauthier, E.L., Ed.; New Developments in Medical Research; Nova Biomedical: New York, NY, USA, 2014.
  75. Burillo, J.; Marqués, P.; Jiménez, B.; González-Blanco, C.; Benito, M.; Guillén, C. Insulin Resistance and Diabetes Mellitus in Alzheimer’s Disease. Cells 2021, 10, 1236.
  76. Sędzikowska, A.; Szablewski, L. Insulin and Insulin Resistance in Alzheimer’s Disease. Int. J. Mol. Sci. 2021, 22, 9987.
  77. Ramos-Rodriguez, J.J.; Spires-Jones, T.; Pooler, A.M.; Lechuga-Sancho, A.M.; Bacskai, B.J.; Garcia-Alloza, M. Progressive Neuronal Pathology and Synaptic Loss Induced by Prediabetes and Type 2 Diabetes in a Mouse Model of Alzheimer’s Disease. Mol. Neurobiol. 2016, 54, 3428–3438.
  78. Spinelli, M.; Fusco, S.; Grassi, C. Brain Insulin Resistance and Hippocampal Plasticity: Mechanisms and Biomarkers of Cognitive Decline. Front. Neurosci. 2019, 13, 788.
  79. Venkat, P.; Chopp, M.; Chen, J. Blood–Brain Barrier Disruption, Vascular Impairment, and Ischemia/Reperfusion Damage in Diabetic Stroke. J. Am. Heart Assoc. 2017, 6, e005819.
  80. Ramos-Rodriguez, J.J.; Ortiz-Barajas, O.; Gamero-Carrasco, C.; de la Rosa, P.R.; Infante-Garcia, C.; Zopeque-Garcia, N.; Lechuga-Sancho, A.M.; Garcia-Alloza, M. Prediabetes-induced vascular alterations exacerbate central pathology in APPswe/PS1dE9 mice. Psychoneuroendocrinology 2014, 48, 123–135.
  81. Koffie, R.M.; Meyer-Luehmann, M.; Hashimoto, T.; Adams, K.W.; Mielke, M.L.; Garcia-Alloza, M.; Micheva, K.D.; Smith, S.J.; Kim, M.L.; Lee, V.M.; et al. Oligomeric amyloid beta associates with postsynaptic densities and correlates with excitatory synapse loss near senile plaques. Proc. Natl. Acad. Sci. USA 2009, 106, 4012–4017.
  82. Meyer-Luehmann, M.; Spires-Jones, T.L.; Prada, C.; Garcia-Alloza, M.; de Calignon, A.; Rozkalne, A.; Koenigsknecht-Talboo, J.; Holtzman, D.M.; Bacskai, B.J.; Hyman, B.T. Rapid appearance and local toxicity of amyloid-beta plaques in a mouse model of Alzheimer’s disease. Nature 2008, 451, 720–724.
  83. 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.
  84. Wang, Y.-C.; Lauwers, E.; Verstreken, P. Presynaptic protein homeostasis and neuronal function. Curr. Opin. Genet. Dev. 2017, 44, 38–46.
  85. Morgan, D. Modulation of microglial activation state following passive immunization in amyloid depositing transgenic mice. Neurochem. Int. 2006, 49, 190–194.
  86. Garcia-Alloza, M.; Ferrara, B.J.; Dodwell, S.A.; Hickey, G.A.; Hyman, B.T.; Bacskai, B.J. A limited role for microglia in antibody mediated plaque clearance in APP mice. Neurobiol. Dis. 2007, 28, 286–292.
  87. Sankar, S.B.; Infante-Garcia, C.; Weinstock, L.D.; Ramos-Rodriguez, J.J.; Hierro-Bujalance, C.; Fernandez-Ponce, C.; Wood, L.B.; Garcia-Alloza, M. Amyloid beta and diabetic pathology cooperatively stimulate cytokine expression in an Alzheimer’s mouse model. J. Neuroinflamm. 2020, 17, 38.
  88. Dey, A.; Hao, S.; Erion, J.R.; Wosiski-Kuhn, M.; Stranahan, A.M. Glucocorticoid sensitization of microglia in a genetic mouse model of obesity and diabetes. J. Neuroimmunol. 2014, 269, 20–27.
  89. Onyango, A.N. Cellular Stresses and Stress Responses in the Pathogenesis of Insulin Resistance. Oxidative Med. Cell. Longev. 2018, 2018, 4321714.
  90. Disabato, D.J.; Quan, N.; Godbout, J. Neuroinflammation: The devil is in the details. J. Neurochem. 2016, 139 (Suppl. S2), 136–153.
  91. Heneka, M.T.; Kummer, M.P.; Latz, E. Innate immune activation in neurodegenerative disease. Nat. Rev. Immunol. 2014, 14, 463–477.
  92. Mishra, A.; Kim, H.J.; Shin, A.H.; Thayer, S.A. Synapse loss induced by interleukin-1beta requires pre- and post-synaptic mechanisms. J. Neuroimmune. Pharmacol. 2012, 7, 571–578.
  93. Oliveira, W.H.; Braga, C.F.; Lós, D.B.; Araújo, S.M.R.; França, M.R.; Duarte-Silva, E.; Rodrigues, G.B.; Rocha, S.W.S.; Peixoto, C.A. Metformin prevents p-tau and amyloid plaque deposition and memory impairment in diabetic mice. Exp. Brain Res. 2021, 239, 2821–2839.
  94. Xu, X.; Sun, Y.; Cen, X.; Shan, B.; Zhao, Q.; Xie, T.; Wang, Z.; Hou, T.; Xue, Y.; Zhang, M.; et al. Metformin activates chaperone-mediated autophagy and improves disease pathologies in an Alzheimer disease mouse model. Protein Cell 2021, 12, 769–787.
  95. Farr, S.A.; Roesler, E.; Niehoff, M.L.; Roby, D.A.; McKee, A.; Morley, J.E. Metformin Improves Learning and Memory in the SAMP8 Mouse Model of Alzheimer’s Disease. J. Alzheimer’s Dis. 2019, 68, 1699–1710.
  96. Zheng, J.; Xu, M.; Walker, V.; Yuan, J.; Korologou-Linden, R.; Robinson, J.; Huang, P.; Burgess, S.; Yeung, S.L.A.; Luo, S.; et al. Evaluating the efficacy and mechanism of metformin targets on reducing Alzheimer’s disease risk in the general population: A Mendelian randomisation study. Diabetologia 2022, 65, 1664–1675.
  97. Al-Kuraishy, H.M.; Al-Gareeb, A.I.; Saad, H.M.; Batiha, G.E.-S. Long-term use of metformin and Alzheimer’s disease: Beneficial or detrimental effects. Inflammopharmacology 2023, 31, 1107–1115.
  98. AboEl-Azm, Y.H.; El-Samahy, M.; Hendi, N.I.; Arar, A.; Yasen, N.S.; Ramadan, S.; Zedan, E.M.; Al-Dardery, N.M.; Khaity, A. Safety and efficacy of intranasal insulin in patients with Alzheimer’s disease: A systematic review and meta-analysis. J. Clin. Transl. Res. 2023, 9, 222–235.
  99. Kellar, D.; Register, T.; Lockhart, S.N.; Aisen, P.; Raman, R.; Rissman, R.A.; Brewer, J.; Craft, S. Intranasal insulin modulates cerebrospinal fluid markers of neuroinflammation in mild cognitive impairment and Alzheimer’s disease: A randomized trial. Sci. Rep. 2022, 12, 1346.
  100. Novak, V.; Milberg, W.; Hao, Y.; Munshi, M.; Novak, P.; Galica, A.; Manor, B.; Roberson, P.; Craft, S.; Abduljalil, A. Enhancement of Vasoreactivity and Cognition by Intranasal Insulin in Type 2 Diabetes. Diabetes Care 2014, 37, 751–759.
  101. Craft, S.; Claxton, A.; Baker, L.D.; Hanson, A.J.; Cholerton, B.; Trittschuh, E.H.; Dahl, D.; Caulder, E.; Neth, B.; Montine, T.J.; et al. Effects of Regular and Long-Acting Insulin on Cognition and Alzheimer’s Disease Biomarkers: A Pilot Clinical Trial. J. Alzheimer’s Dis. 2017, 57, 1325–1334.
  102. Kellar, D.; Lockhart, S.N.; Aisen, P.; Raman, R.; Rissman, R.A.; Brewer, J.; Craft, S. Intranasal Insulin Reduces White Matter Hyperintensity Progression in Association with Improvements in Cognition and CSF Biomarker Profiles in Mild Cognitive Impairment and Alzheimer’s Disease. J. Prev. Alzheimer’s Dis. 2021, 8, 240–248.
  103. Craft, S.; Raman, R.; Chow, T.W.; Rafii, M.S.; Sun, C.K.; Rissman, R.A.; Donohue, M.C.; Brewer, J.B.; Jenkins, C.; Harless, K.; et al. Safety, Efficacy, and Feasibility of Intranasal Insulin for the Treatment of Mild Cognitive Impairment and Alzheimer Disease Dementia: A Randomized Clinical Trial. JAMA Neurol. 2020, 77, 1099–1109.
More
Information
Subjects: Nursing
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , , , ,
View Times: 74
Revisions: 2 times (View History)
Update Date: 18 Dec 2023
1000/1000