Abstract
To maintain energy supply to the brain, a direct energy source called adenosine triphosphate (ATP) is produced by oxidative phosphorylation and aerobic glycolysis of glucose in the mitochondria and cytoplasm. Brain glucose metabolism is reduced in many neurodegenerative diseases, including Alzheimer’s disease (AD), where it appears presymptomatically in a progressive and region-specific manner. Following dysregulation of energy metabolism in AD, many cellular repair/regenerative processes are activated to conserve the energy required for cell viability. Glucose metabolism plays an important role in the pathology of AD and is closely associated with the tricarboxylic acid cycle, type 2 diabetes mellitus, and insulin resistance. The glucose intake in neurons is from endothelial cells, astrocytes, and microglia. Damage to neurocentric glucose also damages the energy transport systems in AD. Gut microbiota is necessary to modulate bidirectional communication between the gastrointestinal tract and brain. Gut microbiota may influence the process of AD by regulating the immune system and maintaining the integrity of the intestinal barrier. Furthermore, some therapeutic strategies have shown promising therapeutic effects in the treatment of AD at different stages, including the use of antidiabetic drugs, rescuing mitochondrial dysfunction, and epigenetic and dietary intervention. This review discusses the underlying mechanisms of alterations in energy metabolism in AD and provides potential therapeutic strategies in the treatment of AD.
Key words: Alzheimer’s disease, energy metabolism, nerve cells, gut–brain axis
Introduction
The brain, accounting for approximately 2% of the adult body weight, consumes over 20% of energy under physiological conditions.[1] Brain energy metabolism mainly relies on adenosine triphosphate (ATP), which is used by Na+/K+-ATPase and Ca2+-ATPase to maintain transmembrane ion gradients during neuronal signal transduction.[2, 3] Among the brain neurons, excitatory (glutamatergic) neurons utilize 80%–85% of the brain’s total ATP, while the remaining neurons are inhibitory.[1, 4] ATP released from neurons, astrocytes, and microglia is also involved in a multitude of processes including immune response, axonal transport, microglial motility, DNA repair, and protein production.[5] Ninety-five percent or more of the ATP in the brain is produced by glucose metabolism, which is absorbed from the neurovascular unit, including brain capillary endothelial cells, pericytes, astrocytes, oligodendrocytes, microglia, and neurons.[6] In normal conditions, glucose uptake is driven by the energy requirements of the activated neurons in different regions of the brain. Glucose transport in the cortex, hippocampus, and cerebellum is associated with glucose transporters including glucose transporter 1 (GLUT1) (the capillary endothelium, membrane of astrocytes, and oligodendrocytes), GLUT2 (plasma membrane of astrocytes), GLUT3, and GLUT4 (neurons).[7, 8, 9] Glucose reaches the neurons either by diffusing directly or by being channeled by the end-feet of astrocytes surrounding the capillary walls. In astrocytes, glucose is metabolized to ATP or converted to lactate, an alternative energy source.[10] The ATP is predominantly generated by oxidative phosphorylation of glucose via the tricarboxylic acid cycle (TCA cycle) within the mitochondria.[11]
Research has shown that aberrant brain energy metabolism is involved in the progression of disorders of the central nervous system (CNS), such as Alzheimer’s disease (AD), Parkinson’s disease (PD), Huntington’s disease (HD), and frontotemporal dementia (FTD).[12, 13, 14] AD is the most common neurodegenerative disease and is characterized by extracellular senile plaques composed of amyloid-β (Aβ) and intracellular neurofibrillary tangles (abnormal phosphorylated tau aggregates) within the neocortex.[15] Additional pathological changes, beyond Aβ and tau, are incompletely understood. Evidence has demonstrated that impaired brain energy metabolism is involved in the progression of AD, and may occur before clinical symptoms arise, driving, and being driven by cognitive dysfunction in a destructive cycle.[16, 17] For example, positron emission tomography (PET) studies showed that glucose uptake is 10%–12% lower in the entorhinal cortex and parietal lobes of patients with mild cognitive impairment (MCI), which becomes anatomically widespread in the progression of AD.[1] Lower glucose uptake, TCA activity, mitochondrial function, and energetic transport of astrocytes and oligodendrocytes are associated with AD; therefore, weight loss and type 2 diabetes mellitus (T2DM) are considered a particular type of AD.[18, 19] Neuroinflammation is one of the pathological features of AD, rapidly consuming glucose and depleting energy to the neurons.[20, 21] The regional brain metabolism impairment may distinguish AD from other pattern dementia, including FTD, PD, or Lewy body disease.[22]
Energy balance of nerve cell microenvironment in AD
Neuron
The brain, one of the most energy-dependent organs in the body, relies primarily on glucose for its energy requirements under physiological conditions. Dysfunction of energy supply has been demonstrated in CNS disorders, including AD. Cerebral glucose metabolism in patients with AD is significantly diminished compared to that in controls. However, cognitive dysfunction is correlated with a reduced glucose metabolism in the regions dependent on glucose metabolism such as the posterior cingulate and parietal, temporal, and prefrontal cortices.[18, 23] In AD, energy-related metabolisms, such as TCA cycle, oxidative phosphorylation, ATPase, glycolysis, ketone body, and creatine metabolisms, are significantly dysregulated.[18, 24] Increasing evidence has shown that syndrome components including T2DM display increasing risk of MCI and AD.[25, 26] Insulin resistance plays an important role in the progression of T2DM, which is observed in approximately 80% of the patients with AD.[27] Insulin signaling pathways, including altered levels of insulin and insulin-like growth factor (IGF), are impaired in the brains of AD patients.[28, 29] Insulin-related signaling pathways play a key role in energy homeostasis, neuronal survival, and memory processes, all of which are critical for learning and cognitive functions in the cortical and hippocampal areas.[30] Thus, insulin may directly modulate cognitive ability. Insulin signaling dysfunction impairs Aβ degradation or vice versa and contributes to neurotransmitter release, neuronal survival, and cognitive damage, thereby leading to AD.[31] Soluble Aβ oligomers bind to membrane insulin receptors in the neurons, leading to cytotoxicity.[32] The insulin-degrading enzyme (IDE) degrades both insulin and Aβ and has a higher affinity to bind to insulin. In mice lacking IDE, Aβ degradation was reduced, leading to Aβ deposits in the brain.[33] In addition to Aβ metabolism, insulin and IGF1 regulate the expression and phosphorylation of tau by activation of related kinases,[34, 35] including the tau kinases (glycogen synthase kinase[GSK]-3β, c-Jun N-terminal kinase [JNK], and adenosine 5’-monophosphate-activated protein kinase [AMPK]) and the tau phosphatases (PP2A and PP1), which play an important role in tau pathology.[36] In insulin receptor substrate 2 (IRS-2)-deficient mice, tau phosphorylation is increased by disrupting the tau kinases.[37] Altered insulin signaling in the brain promotes tau cleavage and restricts alternative splicing of tau.[38, 39, 40]
A number of energy-related enzymes, which play an important role in the function of the TCA cycle, are dysregulated in AD. The dysfunction of the AMPK signaling pathway may be involved in AD as AMPK is a key sensor and regulator of energy metabolism. Increased AMPK phosphorylation has been observed in the brains of patients with AD and mice models.[41, 42] Neurofibrillary tangles, aggregates of hyperphosphorylated tau protein, are the main pathological markers of AD and are regulated at numerous sites by AMPK.[43, 44] In contrast, activation of AMPK decreases the accumulation of Aβ both in vitro and in vivo.[45] Thus, AMPK modulates energy metabolism, affecting both tauopathy and amyloidogenesis in AD. The mammalian target of rapamycin (mTOR) signaling pathway also regulates protein synthesis, mitochondrial function, and energy homeostasis, which affect aging and neurodegeneration. Several studies have shown that the mTOR signaling pathway is aberrantly upregulated in the brains of patients with AD and mouse models during neurodegeneration, resulting in association with Braak stages and/or cognitive decline in patients with AD.[46, 47, 48] The upstream signaling pathway of mTOR, PI3K/Akt axis, was also impaired in AD.[49, 50] Reports have shown that continuous activation of PI3K/Akt/mTOR signaling in neurons inhibits the insulin receptor substrate 1 (IRS1) in the brains of MCI, patients with AD, and AD models, demonstrating energy metabolism dysfunction in AD pathology.[51, 52] Poly(ADP-ribose) polymerase-1 (PARP-1) plays an important role in maintaining genome stability, transcriptional regulation, and long-term potentiation.[53] Under pathological conditions, excessive activation of PARP-1 results in nicotinamide adenine dinucleotide (NAD+) depletion, inducing abnormal energy metabolism in the mitochondria by regulating the expression of mitochondrial proteins.[54] PARP-1 activity and PAR accumulation are enhanced in AD, particularly in the neurons of the frontal and temporal lobes.[55] In the mitochondria, Aβ contributes to PARP-1 over-activation, inducing energy metabolism dysfunction and alterations in mitochondrial membrane potential by superoxide radical production.[56, 57] Also, TCA substrates prevent PARP-1-mediated neuronal damage by inhibiting the oxidative stress induced by Aβ.[58, 59]
Recent studies have shown that vascular mechanisms also play an important role in the metabolic alterations in AD. [60, 61, 62] In aged individuals, cerebral amyloid angiopathy occurs in the brain vasculature and is more severe in AD.[63] The neurovascular unit consists of neurons, astrocytes, and vasculature. It is required to maintain basic energy metabolism and brain function. In AD, Aβ, which is involved in vascular pathology, induces progressive neurovascular unit dysfunction, characterized by vascular reactivity failure, smooth muscle cell loss, and vessel integrity breakdown.[64] Functional hyperemia, which disturbs neurodegeneration, is also perturbed in AD.[65, 66]
In the adult brains of animals and humans, neurogenesis is mainly located in the subventricular zone (SVZ) of the lateral ventricle and the subgranular zone (SGZ) of the dentate gyrus (DG) in the hippocampus.[67, 68, 69] Hippocampal neurogenesis contributes to the progression of learning and memory and inhibition of neurogenesis hampers trace-related memory formation.[70, 71] Energy metabolism affects the proliferation of neural stem cells and the differentiation of newborn cells, modulating neurogenesis in the AD brain.[72, 73] In neurogenesis, the expression of energy metabolism-related proteins, including insulin-like growth factor binding protein 3, cytochrome c oxidase, and acetyl-coenzyme A synthetase 1, is dysregulated in AD.[74, 75, 76] These data indicate that abnormal metabolism in neurons, related to energy-related enzymes, may facilitate the process of AD.
Vascular endothelial cells
To sustain tissue homeostasis, blood vessels formed by endothelial cells (ECs) contribute to the delivery of oxygen and nutrients and the removal of metabolites.[77] To meet the energy requirements of tissue cells, ECs migrate from existing vessels forming new blood vessels in a process called angiogenesis.[78] In nutrient exchange, various organotypic metabolism mechanisms occur, for example, the brain and skeletal muscles rely on glucose as the main energy substrate, whereas the heart and brown adipose tissue favor fatty acids.[79] In the brain vasculature, nutrients are transported in the form of a continuous, nonfenestrated, tightly sealed endothelium.[80]
The main components of the neurovascular system, the ECs, play an important role in maintaining the integrity of the blood–brain barrier (BBB). In AD, neurovascular system impairment leads to reduced brain perfusion and disruption to the BBB leading to the entry of neurotoxic metabolites, resulting in synapse loss.[81, 82] It is also associated with the upregulation of angiogenic factors and receptors, including ETS-related gene (ERG), FMS-related tyrosine kinase 1 (FLT1), and von Willebrand factor (VWF).[83] The ECs fuel their own energy from glycolytic breakdown of glucose or lactate, even under quiescent conditions.[84] The GLUT1 in ECs facilitates glucose diffusion, which is regulated by vascular endothelial growth factor(VEGF).[85] Levels of GLUT1 in the BBB are higher than those in other organs, and reduced GLUT1 levels lead to decreased glucose delivery and lower cerebrospinal fluid (CSF) glucose levels.[86, 87] Reduced GLUT1 levels may be characterized by seizures, movement disorders, and neurodevelopmental delays.[87, 88] The downregulation of GLUT1 is related to microvascular impairment and BBB dysfunction, exacerbating AD.[89, 90] Growing evidence suggests that mitochondrial dysfunction precedes the onset of AD,[91] by causing vascular degeneration and hypoperfusion, and cognitive dysfunction.[92] Mitochondrial dysfunction produces reactive oxygen species (ROS), which contributes to ECs apoptosis, BBB damage, and degeneration.[93] The activation of NADPH oxidase(NOX) may also contribute to EC dysfunction.[93, 94] For example, NOX2 knockout in ECs abrogates ROS production and vascular dysfunction in AD.[95, 96] In conclusion, vascular endothelial cells transport energy elements and metabolic waste bidirectionally between the blood and the brain in a receptor-dependent manner, modulating the development of AD.
Astrocyte
Astrocytes, structure support cells, play critical roles in contacting neurons and maintaining a milieu for proper neuronal function, such as regulating ion channels, providing metabolites for neurons, and sustaining the integrity of the BBB.[97] Communication between astrocytes and neurons contributes to the regulation of brain signaling and synaptic functions. Astrocytes also release nutrients, such as the bioenergetic substrate lactate, glycogen-derived lactate, and pyruvate, which protect neurons from nutritional damage and maintain homeostatic synaptic plasticity.[4, 98, 99] Aging results in a shift of brain energy metabolism into an age-dependent astrocytic metabolic form.[100] Lactate released from anaerobic glycolysis in astrocytes becomes the main energy source for neurons.[2, 101] However, the age-dependent astrocytic metabolic shift exacerbates the brain hypometabolic state, which is caused by mitochondrial oxidative metabolism in astrocytes.[102] In the hippocampus and cerebellum, the transport and metabolism of glucose are faster in astrocytes than those in neurons. Therefore, astrocytes may have a greater impact on glucose metabolism in the brain than neurons.[103] Glucose metabolism is the main energy source for brain function. However, glucose transport and utilization dysfunction are found in cognitively normal individuals with AD risk genes and in patients diagnosed with early AD.[104, 105] Although the precise mechanisms contributing to the disorder of glucose metabolism in AD is still unclear, the GLUT1 in astrocytes may play an important role in glucose transport. The GLUT1 is reduced in the brain of patients with AD [106] exacerbating the pathophysiological progression of AD.[89] However, increasing the expression of GLUT1 in AD mice decreased the Aβ content.[89]
The change in the morphology and function of astrocytes in the brains of AD mice and humans is known as astrogliosis. The Aβ may be responsible for the activation of glial cells.[107, 108] Studies showed that a reduction in glucose metabolism contributes to plaque formation. In astrocytes, glycolysis plays an important role in amyloid accumulation and cytotoxicity, and the inhibition of glycolysis leads to the accumulation of Aβ and Aβ-induced cytotoxicity.[109] In postmortems of the brain tissue of patients with AD, astrocytes were surrounded by plaques,[110] which may reduce amyloid plaque deposits.[111] Bioenergetic dysfunction in astrocytes may contribute to amyloid accumulation and cytotoxicity via glycolysis, and inhibition of astrocytic 6-phosphofructo-2-kinase/ fructose-2,6-biphosphatase 3 (PFKFB3) may lead to Aβ accumulation and cytotoxicity.[109] Astrocytes can internalize different forms of Aβ into lysosome-like granules, as shown in the brain tissue of patients with AD.[112, 113]
Cholesterol synthesis in astrocytes is maintained at a low rate due to the BBB. Astrocytes have higher cholesterol levels in their membranes, crucial for cholesterol metabolism, than neurons.[114] The higher cholesterol content in the astrocyte membrane is susceptible to an Aβ-induced Ca2+-dependent influx.[115] Consequently, the increased production of nicotinamide adenine dinucleotide phosphate (NADPH)-oxidase and free radicals activates PARP, thereby impairing the NAD+ cycle.[116, 117] Apolipoprotein E (ApoE) is secreted mainly from astrocytes and plays an important role in both cholesterol metabolism and the degradation and clearance of Aβ. The uptake of Aβ may be associated with surface ApoE receptors, including low-density lipoprotein receptors (LDLR) and low-density lipoprotein receptor-related protein 1 (LRP-1),[107] ultimately leading to oxidative stress and neuronal degeneration. Thus, metabolic dysfunction in astrocytes leads to Aβ accumulation, oxidative stress, and damage to cholesterol metabolism, contributing to AD.
Microglia
As the resident macrophages, microglia prevents damage to the neurons by clearing toxic proteins, such as toxins, infectious agents, and pathogens. The hyperactivation of microglia plays a key role in neuroinflammation, neurodegenerative diseases, and neuronal energy dysfunction.[118, 119] Energy from microglia is rooted in oxidative phosphorylation; however, a shift to an aerobic glycolysis-predominant phenotype occurs in neurodegenerative diseases via the upregulation of GLUT1 and GLUT4 expression.[120, 121] When neuroinflammation is sustainably induced, activated microglia require more energy, thereby reducing the available energy to the neurons.[122] Recent studies showed that microglia dysfunction is correlated with an increased risk of AD, whereas microglia recovery may slow disease progression.[123, 124] Unlike astrocytes, microglia do not directly provide energy to neurons. However, during inflammation, local neurons retrieve lactate from the activated microglia.[125]
Increasing evidence indicates that neuroinflammation and oxidative stress as a consequence of microglial activation are associated with AD pathophysiology.[123] In AD, the role of microglia is dynamically regulated depending on the stage of the disease.[126] However, the relationship between microglial bioenergetics, neuroinflammation, and brain energy in AD is poorly understood. Studies using multiple-tracer PET showed that glucose hypermetabolism of inflammatory cells was observed in the brains of patients with AD and not in healthy individuals.[127] Microglial energy metabolism contributed to neuroinflammation by regulating glycolytic flux and enzyme expression in mitochondria.[64] Furthermore, the triggering receptor expressed on myeloid cells 2 (TREM2) was identified as a risk factor for AD in coding variants.[128] Microglia TREM2 mice exhibited dysfunctional ATP levels and biosynthetic pathways, altering the cellular energetic and biosynthetic metabolism processes in AD.[129] These results demonstrated that microglia energy metabolism dysfunction limits the energy available to neurons and activates neuroinflammation, accelerating the pathology of AD.
Gut–brain axis dysfunction in energy metabolism of AD
The gut microbiota modulates the overall energy homeostasis, inhibits intestinal surface pathogen adhesion, synthesizes vitamin K, salvages energy from short-chain fatty acid production, regulates the immune system, and maintains the integrity of the intestinal barrier.[130, 131] Recent studies demonstrated that gut microbiota plays pivotal roles in adjusting bidirectional communication between the gastrointestinal tract and the brain.[132, 133] Studies showed that exogenous butyrate generated by the gut microbiota facilitated the confirmation of dendritic spines, long-term potentiation, and cognitive formation.[134] Gut microbiota also synthesizes neurotransmitters essential for brain activity, including gamma-aminobutyric acid, butyrate, 5-hydroxytryptamine, dopamine, serotonin, and histamine, which can be released into the bloodstream and can cross the BBB.[135, 136, 137, 138] Disruptions and changes in the gut microbiota (dysbiosis) are involved in the pathogenesis of many CNS diseases, including autism, PD, schizophrenia, multiple sclerosis, and AD.[139, 140, 141] In individuals with AD, reduced microbiota levels can decrease the level of the by-product, butyrate, in the brain, aggravating cognitive dysfunction.[142] In AD mouse models, the Firmicutes numbers were reduced and the Bacteroides numbers increased in the intestine, leading to amyloid deposition. [143] Correspondingly, Aβ pathology was alleviated in the cerebrum in germ-free AD mice.[143] Several species of gram-negative bacteria produce lipopolysaccharide (LPS), contributing to the prolonged elevation of Aβ in the hippocampus of AD patients, resulting in cognitive dysfunction.[144] Recently, studies showed that treatment with a probiotic mixture containing Bifidobacterium longum and different Lactobacillus strains positively influenced the cognitive function and metabolic status of patients with AD.[145] In addition, Helicobacter pylori (H. pylori) induced high levels of amino acids, activating the mammalian target of rapamycin complex 1 (mTORC1), modulating AD.[146] Furthermore, H. pylori modulates the hyperphosphorylation of tau proteins in AD.[147] Thus, the gut–brain axis provides another potential explanation for energy metabolism dysfunction in AD.
Potential therapies for AD based on brain energy metabolism
At present, there are many different metabolic pathways and processes that reduce brain energy metabolism dysfunction in AD, including supporting mitochondrial function, maintaining the stability of the TCA cycle, increasing insulin sensitivity, and restoring downstream signaling.[1] A proprietary tricyclic pyrone, CP2, enhanced mitochondrial biogenesis by binding to and inhibiting complex I, thereby improving cognitive function in transgenic AD mice.[148] In AD, mitochondrial division decoupled from the normal fission–fusion cycle is increased, and inhibiting mitochondrial fission improves mitochondrial biogenesis thereby improving functioning of complex I.[149] The ratio of NAD+/NADH is also a marker of brain energy status, reflecting the redox state of cells. Furthermore, a higher ratio of NAD+/NADH or NAD+ precursor nicotinamide riboside mitigates cognitive impairment in AD by improving the energetic status of the brain.[17, 150] Clinical trials demonstrated that patients with AD following ketogenic diets containing medium-chain triglycerides and very low carbohydrate ketogenic diets showed increased cognitive ability in most adherent patients, in contrast to the control group.[151, 152] The mechanisms might be associated with the retrieval of TCA cycle activity, which increases the levels of acetyl-CoA, thereby fueling aerobic glycolysis in the AD brain.[153, 154, 155] Peripheral insulin sensitivity, which depends on energy intake or use, is correlated with energy metabolism in the brain. Liraglutide is a GLP 1 receptor agonist approved for the treatment of insulin resistance, obesity, and T2DM.[156] Liraglutide enhances glucose uptake in the brain; however, it has no effect on cognitive outcomes.[157] Dipeptidyl peptidase 4 (DPP4) inhibitors, other T2DM medications, prolong the activation of GLP 1, and sitagliptin improves cognitive damage in older individuals suffering from diabetes with or without AD.[158, 159]
Conclusion
Here, we demonstrated that brain energy metabolism dysfunction, including reduced glucose uptake, insulin resistance, impaired TCA cycle, and glycolysis, is involved in the pathology of AD, which impairs axonal transport, mitochondrial function, and ATP production. In addition, brain energy metabolism may serve as a potential biomarker for AD treatment. Based on the multiple brain energetic pathways, many pharmacological agents targeting metabolic enzymes, receptors, or proteins are used to ameliorate cognitive function by enhancing the energy metabolism level in AD. This simultaneously clears aggregated Aβ/ tau and/ or suppresses the reactive oxygen response and neuroinflammation.[160, 161] Metformin and ketone-based interventions restored brain energy metabolism and delayed the onset and progression of AD by improving neuronal integrity, synaptic remodeling, and neuronal–glial interactions.[162, 163, 164] Hormone-based interventions delayed the onset of neuropathology and cognitive decline by modulating appetite and energy expenditure, and epigenetic modification strategies have shown promising effects in improving brain energetics.[19, 165] Further investigations using imaging, metabolite, and hormone approaches will provide a precise understanding of the AD pathology.[166, 167]
In conclusion, aberrant energy metabolism in the brain plays a critical role in cognitive dysfunction in patients with AD, while maintaining energy homeostasis might be a promising treatment to delay the onset and progression of AD.
Funding Statement
This work was supported by the grants from the National Key Research and Development Program of China (2018YFC1704400), the National Nature Science Foundation of China (82001122, 81630028, and 81920108017), the Natural Science Foundation of Jiangsu Province (BK20200129 and BK20211005), and the Fundamental Research Funds for the Central Universities (021414380519).
Footnotes
Conflict of Interest
The authors declare that there are no conflicts of interest.
References
- 1.Cunnane SC, Trushina E, Morland C, Prigione A, Casadesus G, Andrews ZB. Brain energy rescue: An emerging therapeutic concept for neurodegenerative disorders of ageing. Nat Rev Drug Discov. 2020;19:609–33. doi: 10.1038/s41573-020-0072-x. et al. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Magistretti PJ, Allaman I. Lactate in the brain: From metabolic end-product to signalling molecule. Nat Rev Neurosci. 2018;19:235–49. doi: 10.1038/nrn.2018.19. [DOI] [PubMed] [Google Scholar]
- 3.Oyarzabal A, Marin-Valencia I. Synaptic energy metabolism and neuronal excitability, in sickness and health. J Inherit Metab Dis. 2019;42:220–36. doi: 10.1002/jimd.12071. [DOI] [PubMed] [Google Scholar]
- 4.Sola C, Filliol I, Gutierrez MC, Mokrousov I, Vincent V, Rastogi N. Spoligotype database of mycobacterium tuberculosis: Biogeographic distribution of shared types and epidemiologic and phylogenetic perspectives. Emerg Infect Dis. 2001;7:390–6. doi: 10.3201/eid0703.010304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Engl E, Attwell D. Non-signalling energy use in the brain. J Physiol. 2015;593:3417–29. doi: 10.1113/jphysiol.2014.282517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Hirunpattarasilp C, Attwell D, Freitas F. The role of pericytes in brain disorders: From the periphery to the brain. J Neurochem. 2019;150:648–65. doi: 10.1111/jnc.14725. [DOI] [PubMed] [Google Scholar]
- 7.Ashrafi G, Wu Z, Farrell RJ, Ryan TA. Glut4 mobilization supports energetic demands of active synapses. Neuron. 2017;93:606–15. doi: 10.1016/j.neuron.2016.12.020. e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Tups A, Benzler J, Sergi D, Ladyman SR, Williams LM. Central regulation of glucose homeostasis. Compr Physiol. 2017;7:741–64. doi: 10.1002/cphy.c160015. [DOI] [PubMed] [Google Scholar]
- 9.Myers MG. Affinati AH, Richardson N, Schwartz MW. Central nervous system regulation of organismal energy and glucose homeostasis. Nat Metab. 2021;3:737–50. doi: 10.1038/s42255-021-00408-5. Jr. [DOI] [PubMed] [Google Scholar]
- 10.Jammal L, Whalley B, Barkai E. Learning-induced modulation of the effect of neuroglial transmission on synaptic plasticity. J Neurophysiol. 2018;119:2373–9. doi: 10.1152/jn.00101.2018. [DOI] [PubMed] [Google Scholar]
- 11.Bordone MP, Salman MM, Titus HE, Amini E, Andersen JV, Chakraborti B. The energetic brain - a review from students to students. J Neurochem. 2019;151:139–65. doi: 10.1111/jnc.14829. et al. [DOI] [PubMed] [Google Scholar]
- 12.Minhas PS, Latif-Hernandez A, McReynolds MR, Durairaj AS, Wang Q, Rubin A. Restoring metabolism of myeloid cells reverses cognitive decline in ageing. Nature. 2021;590:122–8. doi: 10.1038/s41586-020-03160-0. et al. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Bartke A, Brannan S, Hascup E, Hascup K, Darcy J. Energy metabolism and aging. World J Mens Health. 2021;39:222–32. doi: 10.5534/wjmh.200112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Anderson CC, Marentette JO, Rauniyar AK, Prutton KM, Khatri M, Matheson C. Maneb alters central carbon metabolism and thiol redox status in a toxicant model of Parkinson’s disease. Free Radic Biol Med. 2021;162:65–76. doi: 10.1016/j.freeradbiomed.2020.11.028. et al. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Jack CR. Bennett DA, Blennow K, Carrillo MC, Dunn B, Haeberlein SB. Nia-aa research framework: Toward a biological definition of Alzheimer’s disease. Alzheimers Dement. 2018;14:535–62. doi: 10.1016/j.jalz.2018.02.018. Jr. et al. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Johnson ECB, Dammer EB, Duong DM, Ping L, Zhou M, Yin L. Large-scale proteomic analysis of Alzheimer’s disease brain and cerebrospinal fluid reveals early changes in energy metabolism associated with microglia and astrocyte activation. Nat Med. 2020;26:769–80. doi: 10.1038/s41591-020-0815-6. et al. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Butterfield DA, Halliwell B. Oxidative stress, dysfunctional glucose metabolism and alzheimer disease. Nature reviews. Neuroscience. 2019;20:148–60. doi: 10.1038/s41583-019-0132-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.van der Velpen V, Teav T, Gallart-Ayala H, Mehl F, Konz I, Clark C. Systemic and central nervous system metabolic alterations in Alzheimer’s disease. Alzheimers Res Ther. 2019;11:93. doi: 10.1186/s13195-019-0551-7. et al. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Demarest TG, Varma VR, Estrada D, Babbar M, Basu S, Mahajan UV. Biological sex and DNA repair deficiency drive Alzheimer’s disease via systemic metabolic remodeling and brain mitochondrial dysfunction. Acta Neuropathol. 2020;140:25–47. doi: 10.1007/s00401-020-02152-8. et al. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Briston T, Hicks AR. Mitochondrial dysfunction and neurodegenerative proteinopathies: Mechanisms and prospects for therapeutic intervention. Biochem Soc Trans. 2018;46:829–42. doi: 10.1042/BST20180025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Ryu JC, Zimmer ER, Rosa-Neto P, Yoon SO. Consequences of metabolic disruption in alzheimer’s disease pathology. Neurotherapeutics. 2019;16:600–10. doi: 10.1007/s13311-019-00755-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Wilson H, Pagano G, Politis M. Dementia spectrum disorders: Lessons learnt from decades with pet research. J Neural Transm (Vienna) 2019;126:233–51. doi: 10.1007/s00702-019-01975-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Weise CM, Chen K, Chen Y, Kuang X, Savage CR, Reiman EM. Left lateralized cerebral glucose metabolism declines in amyloid-beta positive persons with mild cognitive impairment. Neuroimage Clin. 2018;20:286–96. doi: 10.1016/j.nicl.2018.07.016. et al. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Sertbas M, Ulgen K, Cakir T. Systematic analysis of transcription-level effects of neurodegenerative diseases on human brain metabolism by a newly reconstructed brain-specific metabolic network. FEBS Open Bio. 2014;4:542–53. doi: 10.1016/j.fob.2014.05.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Biessels GJ, Staekenborg S, Brunner E, Brayne C, Scheltens P. Risk of dementia in diabetes mellitus: A systematic review. Lancet Neurol. 2006;5:64–74. doi: 10.1016/S1474-4422(05)70284-2. [DOI] [PubMed] [Google Scholar]
- 26.Diniz Pereira J, Gomes Fraga V, Morais Santos AL, Carvalho MDG, Caramelli P, Braga Gomes K. Alzheimer’s disease and type 2 diabetes mellitus: A systematic review of proteomic studies. J Neurochem. 2021;156:753–76. doi: 10.1111/jnc.15166. [DOI] [PubMed] [Google Scholar]
- 27.Shi L, Buckley NJ, Bos I, Engelborghs S, Sleegers K, Frisoni GB. Plasma proteomic biomarkers relating to alzheimer’s disease: A meta-analysis based on our own studies. Front Aging Neurosci. 2021;13:712545. doi: 10.3389/fnagi.2021.712545. et al. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Gabbouj S, Ryhanen S, Marttinen M, Wittrahm R, Takalo M, Kemppainen S. Altered insulin signaling in alzheimer’s disease brain - special emphasis on pi3k-akt pathway. Front Neurosci. 2019;13:629. doi: 10.3389/fnins.2019.00629. et al. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Sim AY, Barua S, Kim JY, Lee YH, Lee JE. Role of dpp-4 and sglt2 inhibitors connected to alzheimer disease in type 2 diabetes mellitus. Front Neurosci. 2021;15:708547. doi: 10.3389/fnins.2021.708547. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Szablewski L. Brain glucose transporters: Role in pathogenesis and potential targets for the treatment of alzheimer’s disease. Int J Mol Sci. 2021;22:8142. doi: 10.3390/ijms22158142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Bomfim TR, Forny-Germano L, Sathler LB, Brito-Moreira J, Houzel JC, Decker H. An anti-diabetes agent protects the mouse brain from defective insulin signaling caused by alzheimer’s disease- associated abeta oligomers. J Clin Invest. 2012;122:1339–53. doi: 10.1172/JCI57256. et al. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.De Felice FG. Alzheimer’s disease and insulin resistance: Translating basic science into clinical applications. J Clin Invest. 2013;123:531–9. doi: 10.1172/JCI64595. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Pedros I, Petrov D, Allgaier M, Sureda F, Barroso E, Beas-Zarate C. Early alterations in energy metabolism in the hippocampus of appswe/ ps1de9 mouse model of alzheimer’s disease. Biochim Biophys Acta. 2014;1842:1556–66. doi: 10.1016/j.bbadis.2014.05.025. et al. [DOI] [PubMed] [Google Scholar]
- 34.Rebelos E, Rinne JO, Nuutila P, Ekblad LL. Brain glucose metabolism in health, obesity, and cognitive decline-does insulin have anything to do with it? A narrative review. J Clin Med. 2021;10:1532. doi: 10.3390/jcm10071532. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Chatterjee S, Mudher A. Alzheimer’s disease and type 2 diabetes: A critical assessment of the shared pathological traits. Front Neurosci. 2018;12:383. doi: 10.3389/fnins.2018.00383. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Yin X, Zhao C, Qiu Y, Zhou Z, Bao J, Qian W. Dendritic/post-synaptic tau and early pathology of alzheimer’s disease. Front Mol Neurosci. 2021;14:671779. doi: 10.3389/fnmol.2021.671779. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Schubert M, Brazil DP, Burks DJ, Kushner JA, Ye J, Flint CL. Insulin receptor substrate-2 deficiency impairs brain growth and promotes tau phosphorylation. J Neurosci. 2003;23:7084–92. doi: 10.1523/JNEUROSCI.23-18-07084.2003. et al. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Kim B, Backus C, Oh S, Feldman EL. Hyperglycemia-induced tau cleavage in vitro and in vivo: A possible link between diabetes and alzheimer’s disease. J Alzheimers Dis. 2013;34:727–39. doi: 10.3233/JAD-121669. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Poddar MK, Banerjee S, Chakraborty A, Dutta D. Metabolic disorder in alzheimer’s disease. Metab Brain Dis. 2021;36:781–813. doi: 10.1007/s11011-021-00673-z. [DOI] [PubMed] [Google Scholar]
- 40.Trujillo-Estrada L, Nguyen C, da Cunha C, Cai L, Forner S, Martini AC. Tau underlies synaptic and cognitive deficits for type 1, but not type 2 diabetes mouse models. Aging Cell. 2019;18:e12919. doi: 10.1111/acel.12919. et al. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Ma T, Chen Y, Vingtdeux V, Zhao H, Viollet B, Marambaud P. Inhibition of amp-activated protein kinase signaling alleviates impairments in hippocampal synaptic plasticity induced by amyloid beta. J Neurosci. 2014;34:12230–8. doi: 10.1523/JNEUROSCI.1694-14.2014. et al. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Zhao F, Wang C, Zhu X. Isoform-specific roles of ampk catalytic alpha subunits in alzheimer’s disease. J Clin Invest. 2020;130:3403–5. doi: 10.1172/JCI137908. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Wang X, Zimmermann HR, Lockhart SN, Craft S, Ma T. Decreased levels of blood ampkalpha1 but not ampkalpha2 isoform in patients with mild cognitive impairment and alzheimer’s disease: A pilot study. J Alzheimers Dis. 2020;76:217–24. doi: 10.3233/JAD-191189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Zimmermann HR, Yang W, Kasica NP, Zhou X, Wang X, Beckelman BC. Brain-specific repression of ampkalpha1 alleviates pathophysiology in alzheimer’s model mice. J Clin Invest. 2020;130:3511–27. doi: 10.1172/JCI133982. et al. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Ali T, Rehman SU, Khan A, Badshah H, Abid NB, Kim MW. Adiponectin-mimetic novel nonapeptide rescues aberrant neuronal metabolic-associated memory deficits in alzheimer’s disease. Mol Neurodegener. 2021;16:23. doi: 10.1186/s13024-021-00445-4. et al. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Querfurth H, Lee HK. Mammalian/mechanistic target of rapamycin (mtor) complexes in neurodegeneration. Mol Neurodegener. 2021;16:44. doi: 10.1186/s13024-021-00428-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Sun YX, Ji X, Mao X, Xie L, Jia J, Galvan V. Differential activation of mtor complex 1 signaling in human brain with mild to severe alzheimer’s disease. J Alzheimers Dis. 2014;38:437–44. doi: 10.3233/JAD-131124. et al. [DOI] [PubMed] [Google Scholar]
- 48.Perluigi M, Di Domenico F, Barone E, Butterfield DA. Mtor in alzheimer disease and its earlier stages: Links to oxidative damage in the progression of this dementing disorder. Free Radic Biol Med. 2021;169:382–96. doi: 10.1016/j.freeradbiomed.2021.04.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Yoon JH, Lee N, Youn K, Jo MR, Kim HR, Lee DS. Dieckol ameliorates abeta production via pi3k/akt/gsk-3beta regulated app processing in sweapp n2a cell. Mar Drugs. 2021;19:152. doi: 10.3390/md19030152. et al. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Wei W, Norton DD, Wang X, Kusiak JW. Abeta 17-42 in alzheimer’s disease activates jnk and caspase-8 leading to neuronal apoptosis. Brain. 2002;125:2036–43. doi: 10.1093/brain/awf205. [DOI] [PubMed] [Google Scholar]
- 51.Gupta A, Dey CS. Pten, a widely known negative regulator of insulin/ pi3k signaling, positively regulates neuronal insulin resistance. Mol Biol Cell. 2012;23:3882–98. doi: 10.1091/mbc.E12-05-0337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Mencer S, Kartawy M, Lendenfeld F, Soluh H, Tripathi MK, Khaliulin I. Proteomics of autism and alzheimer’s mouse models reveal common alterations in mtor signaling pathway. Transl Psychiatry. 2021;11:480. doi: 10.1038/s41398-021-01578-2. et al. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Tzekaki EE, Tsolaki M, Geromichalos GD, Pantazaki Alpha A. Extra virgin olive oil consumption from mild cognitive impairment patients attenuates oxidative and nitrative stress reflecting on the reduction of the parp levels and DNA damage. Exp Gerontol. 2021;156:111621. doi: 10.1016/j.exger.2021.111621. [DOI] [PubMed] [Google Scholar]
- 54.Lapucci A, Pittelli M, Rapizzi E, Felici R, Moroni F, Chiarugi A. Poly(adpribose) polymerase-1 is a nuclear epigenetic regulator of mitochondrial DNA repair and transcription. Mol Pharmacol. 2011;79:932–40. doi: 10.1124/mol.110.070110. [DOI] [PubMed] [Google Scholar]
- 55.Strosznajder JB, Czapski GA, Adamczyk A, Strosznajder RP. Poly(adpribose) polymerase-1 in amyloid beta toxicity and alzheimer’s disease. Mol Neurobiol. 2012;46:78–84. doi: 10.1007/s12035-012-8258-9. [DOI] [PubMed] [Google Scholar]
- 56.Litwiniuk A, Baranowska-Bik A, Domanska A, Kalisz M, Bik W. Contribution of mitochondrial dysfunction combined with nlrp3 inflammasome activation in selected neurodegenerative diseases. Pharmaceuticals (Basel) 2021;14:1221. doi: 10.3390/ph14121221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Pagani L, Eckert A. Amyloid-beta interaction with mitochondria. Int J Alzheimers Dis. 2011;2011:925050. doi: 10.4061/2011/925050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Abeti R, Abramov AY, Duchen MR. Beta-amyloid activates parp causing astrocytic metabolic failure and neuronal death. Brain. 2011;134:1658–72. doi: 10.1093/brain/awr104. [DOI] [PubMed] [Google Scholar]
- 59.Kurokin I, Lauer AA, Janitschke D, Winkler J, Theiss EL, Griebsch LV. Targeted lipidomics of mitochondria in a cellular alzheimer’s disease model. Biomedicines. 2021;9:1062. doi: 10.3390/biomedicines9081062. et al. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Schneider JA, Arvanitakis Z, Leurgans SE, Bennett DA. The neuropathology of probable alzheimer disease and mild cognitive impairment. Ann Neurol. 2009;66:200–8. doi: 10.1002/ana.21706. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Aamodt EB, Schellhorn T, Stage E, Sanjay AB, Logan PE, Svaldi DO. Predicting the emergence of major neurocognitive disorder within three months after a stroke. Front Aging Neurosci. 2021;13:705889. doi: 10.3389/fnagi.2021.705889. et al. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Cheng YW, Chiu MJ, Chen YF, Cheng TW, Lai YM, Chen TF. The contribution of vascular risk factors in neurodegenerative disorders: From mild cognitive impairment to alzheimer’s disease. Alzheimers Res Ther. 2020;12:91. doi: 10.1186/s13195-020-00658-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Portnoy G, Kantor D, Bar-Natan E. Adaptation of a psychiatric service to missile war. Gen Hosp Psychiatry. 1993;15:418–9. doi: 10.1016/0163-8343(93)90012-d. [DOI] [PubMed] [Google Scholar]
- 64.Zhang X, Alshakhshir N, Zhao L. Glycolytic metabolism, brain resilience, and alzheimer’s disease. Front Neurosci. 2021;15:662242. doi: 10.3389/fnins.2021.662242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Li L, Tong XK, Hosseini Kahnouei M, Vallerand D, Hamel E, Girouard H. Impaired hippocampal neurovascular coupling in a mouse model of alzheimer’s disease. Front Physiol. 2021;12:715446. doi: 10.3389/fphys.2021.715446. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Gonzalez-Molina LA, Villar-Vesga J, Henao-Restrepo J, Villegas A, Lopera F, Cardona-Gomez GP. Extracellular vesicles from 3xtg-ad mouse and alzheimer’s disease patient astrocytes impair neuroglial and vascular components. Front Aging Neurosci. 2021;13:593927. doi: 10.3389/fnagi.2021.593927. et al. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Wiskott L, Rasch MJ, Kempermann G. A functional hypothesis for adult hippocampal neurogenesis: Avoidance of catastrophic interference in the dentate gyrus. Hippocampus. 2006;16:329–43. doi: 10.1002/hipo.20167. [DOI] [PubMed] [Google Scholar]
- 68.Goncalves JT, Schafer ST, Gage FH. Adult neurogenesis in the hippocampus: From stem cells to behavior. Cell. 2016;167:897–914. doi: 10.1016/j.cell.2016.10.021. [DOI] [PubMed] [Google Scholar]
- 69.Clark SB. Computer searches: Effect on animal research. Science. 1988;240:587. doi: 10.1126/science.3363340. [DOI] [PubMed] [Google Scholar]
- 70.Jin WN, Shi K, He W, Sun JH, Van Kaer L, Shi FD. Neuroblast senescence in the aged brain augments natural killer cell cytotoxicity leading to impaired neurogenesis and cognition. Nat Neurosci. 2021;24:61–73. doi: 10.1038/s41593-020-00745-w. et al. [DOI] [PubMed] [Google Scholar]
- 71.Lensu S, Waselius T, Makinen E, Kettunen H, Virtanen A, Tiirola M. Irradiation of the head reduces adult hippocampal neurogenesis and impairs spatial memory, but leaves overall health intact in rats. Eur J Neurosci. 2021;53:1885–904. doi: 10.1111/ejn.15102. et al. [DOI] [PubMed] [Google Scholar]
- 72.Hamilton LK, Dufresne M, Joppe SE, Petryszyn S, Aumont A, Calon F. Aberrant lipid metabolism in the forebrain niche suppresses adult neural stem cell proliferation in an animal model of alzheimer’s disease. Cell Stem Cell. 2015;17:397–411. doi: 10.1016/j.stem.2015.08.001. et al. [DOI] [PubMed] [Google Scholar]
- 73.Ribeiro MF, Genebra T, Rego AC, Rodrigues CMP, Sola S. Amyloid beta peptide compromises neural stem cell fate by irreversibly disturbing mitochondrial oxidative state and blocking mitochondrial biogenesis and dynamics. Mol Neurobiol. 2019;56:3922–36. doi: 10.1007/s12035-018-1342-z. [DOI] [PubMed] [Google Scholar]
- 74.Geschwind DH, Ou J, Easterday MC, Dougherty JD, Jackson RL, Chen Z. A genetic analysis of neural progenitor differentiation. Neuron. 2001;29:325–39. doi: 10.1016/s0896-6273(01)00209-4. et al. [DOI] [PubMed] [Google Scholar]
- 75.Yu J, Xiong C, Zhuo B, Wen Z, Shen J, Liu C. Analysis of local chromatin states reveals gene transcription potential during mouse neural progenitor cell differentiation. Cell Rep. 2020;32:107953. doi: 10.1016/j.celrep.2020.107953. et al. [DOI] [PubMed] [Google Scholar]
- 76.Lee CM, Zhou L, Liu J, Shi J, Geng Y, Liu M. Single-cell rnaseq analysis revealed long-lasting adverse effects of tamoxifen on neurogenesis in prenatal and adult brains. Proc Natl Acad Sci U S A. 2020;117:19578–89. doi: 10.1073/pnas.1918883117. et al. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Potente M, Gerhardt H, Carmeliet P. Basic and therapeutic aspects of angiogenesis. Cell. 2011;146:873–87. doi: 10.1016/j.cell.2011.08.039. [DOI] [PubMed] [Google Scholar]
- 78.Adams RH, Alitalo K. Molecular regulation of angiogenesis and lymphangiogenesis. Nat Rev Mol Cell Biol. 2007;8:464–78. doi: 10.1038/nrm2183. [DOI] [PubMed] [Google Scholar]
- 79.Kummitha CM, Kalhan SC, Saidel GM, Lai N. Relating tissue/ organ energy expenditure to metabolic fluxes in mouse and human: Experimental data integrated with mathematical modeling. Physiol Rep. 2014;2:e12159. doi: 10.14814/phy2.12159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Dight J, Zhao J, Styke C, Khosrotehrani K, Patel J. Resident vascular endothelial progenitor definition and function: the age of reckoning. Angiogenesis. 2021;25:15–33. doi: 10.1007/s10456-021-09817-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Sweeney MD, Sagare AP, Zlokovic BV. Blood-brain barrier breakdown in alzheimer disease and other neurodegenerative disorders. Nat Rev Neurol. 2018;14:133–50. doi: 10.1038/nrneurol.2017.188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Bennett RE, Robbins AB, Hu M, Cao X, Betensky RA, Clark T. Tau induces blood vessel abnormalities and angiogenesis-related gene expression in p301l transgenic mice and human alzheimer’s disease. Proc Natl Acad Sci U S A. 2018;115:E1289–98. doi: 10.1073/pnas.1710329115. et al. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Lau SF, Cao H, Fu AKY, Ip NY. Single-nucleus transcriptome analysis reveals dysregulation of angiogenic endothelial cells and neuroprotective glia in alzheimer’s disease. Proc Natl Acad Sci U S A. 2020;117:25800–9. doi: 10.1073/pnas.2008762117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Fitzgerald G, Soro-Arnaiz I, De Bock K. The warburg effect in endothelial cells and its potential as an anti-angiogenic target in cancer. Front Cell Dev Biol. 2018;6:100. doi: 10.3389/fcell.2018.00100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Tang M, Gao G, Rueda CB, Yu H, Thibodeaux DN, Awano T. Brain microvasculature defects and glut1 deficiency syndrome averted by early repletion of the glucose transporter-1 protein. Nat Commun. 2017;8:14152. doi: 10.1038/ncomms14152. et al. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Nolan DJ, Ginsberg M, Israely E, Palikuqi B, Poulos MG, James D. Molecular signatures of tissue-specific microvascular endothelial cell heterogeneity in organ maintenance and regeneration. Dev Cell. 2013;26:204–19. doi: 10.1016/j.devcel.2013.06.017. et al. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.De Vivo DC, Trifiletti RR, Jacobson RI, Ronen GM, Behmand RA, Harik SI. Defective glucose transport across the blood-brain barrier as a cause of persistent hypoglycorrhachia, seizures, and developmental delay. N Engl J Med. 1991;325:703–9. doi: 10.1056/NEJM199109053251006. [DOI] [PubMed] [Google Scholar]
- 88.Seidner G, Alvarez MG, Yeh JI, O’Driscoll KR, Klepper J, Stump TS. Glut-1 deficiency syndrome caused by haploinsufficiency of the blood-brain barrier hexose carrier. Nat Genet. 1998;18:188–91. doi: 10.1038/ng0298-188. et al. [DOI] [PubMed] [Google Scholar]
- 89.Winkler EA, Nishida Y, Sagare AP, Rege SV, Bell RD, Perlmutter D. Glut1 reductions exacerbate alzheimer’s disease vasculo-neuronal dysfunction and degeneration. Nat Neurosci. 2015;18:521–30. doi: 10.1038/nn.3966. et al. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Veys K, Fan Z, Ghobrial M, Bouche A, Garcia-Caballero M, Vriens K. Role of the glut1 glucose transporter in postnatal cns angiogenesis and blood-brain barrier integrity. Circ Res. 2020;127:466–82. doi: 10.1161/CIRCRESAHA.119.316463. et al. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Yao J, Irwin RW, Zhao L, Nilsen J, Hamilton RT, Brinton RD. Mitochondrial bioenergetic deficit precedes alzheimer’s pathology in female mouse model of alzheimer’s disease. Proc Natl Acad Sci U S A. 2009;106:14670–5. doi: 10.1073/pnas.0903563106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Wang W, Yin J, Ma X, Zhao F, Siedlak SL, Wang Z. Inhibition of mitochondrial fragmentation protects against alzheimer’s disease in rodent model. Hum Mol Genet. 2017;26:4118–31. doi: 10.1093/hmg/ddx299. et al. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Han BH, Zhou ML, Johnson AW, Singh I, Liao F, Vellimana AK. Contribution of reactive oxygen species to cerebral amyloid angiopathy, vasomotor dysfunction, and microhemorrhage in aged tg2576 mice. Proc Natl Acad Sci U S A. 2015;112:E881–90. doi: 10.1073/pnas.1414930112. et al. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Drummond GR, Selemidis S, Griendling KK, Sobey CG. Combating oxidative stress in vascular disease: Nadph oxidases as therapeutic targets. Nat Rev Drug Discov. 2011;10:453–71. doi: 10.1038/nrd3403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Park L, Anrather J, Girouard H, Zhou P, Iadecola C. Nox2-derived reactive oxygen species mediate neurovascular dysregulation in the aging mouse brain. J Cereb Blood Flow Metab. 2007;27:1908–18. doi: 10.1038/sj.jcbfm.9600491. [DOI] [PubMed] [Google Scholar]
- 96.Fragoso-Morales LG, Correa-Basurto J, Rosales-Hernandez MC. Implication of nicotinamide adenine dinucleotide phosphate (nadph) oxidase and its inhibitors in alzheimer’s disease murine models. Antioxidants (Basel) 2021;10:218. doi: 10.3390/antiox10020218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Wang DD, Bordey A. The astrocyte odyssey. Prog Neurobiol. 2008;86:342–67. doi: 10.1016/j.pneurobio.2008.09.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Wang Y, Fu WY, Cheung K, Hung KW, Chen C, Geng H. Astrocytesecreted il-33 mediates homeostatic synaptic plasticity in the adult hippocampus. Proc Natl Acad Sci U S A. 2021;118:e2020810118. doi: 10.1073/pnas.2020810118. et al. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Nagai J, Yu X, Papouin T, Cheong E, Freeman MR, Monk KR. Behaviorally consequential astrocytic regulation of neural circuits. Neuron. 2021;109:576–96. doi: 10.1016/j.neuron.2020.12.008. et al. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Pamies D, Sartori C, Schvartz D, Gonzalez-Ruiz V, Pellerin L, Nunes C. Neuroinflammatory response to tnfalpha and il1beta cytokines is accompanied by an increase in glycolysis in human astrocytes in vitro. Int J Mol Sci. 2021;22:4065. doi: 10.3390/ijms22084065. et al. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Dienel GA. Brain glucose metabolism: Integration of energetics with function. Physiol Rev. 2019;99:949–1045. doi: 10.1152/physrev.00062.2017. [DOI] [PubMed] [Google Scholar]
- 102.Yin F, Sancheti H, Liu Z, Cadenas E. Mitochondrial function in ageing: Coordination with signalling and transcriptional pathways. J Physiol. 2016;594:2025–42. doi: 10.1113/JP270541. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Jakoby P, Schmidt E, Ruminot I, Gutierrez R, Barros LF, Deitmer JW. Higher transport and metabolism of glucose in astrocytes compared with neurons: A multiphoton study of hippocampal and cerebellar tissue slices. Cereb Cortex. 2014;24:222–31. doi: 10.1093/cercor/bhs309. [DOI] [PubMed] [Google Scholar]
- 104.Iadecola C. Sugar and alzheimer’s disease: A bittersweet truth. Nat Neurosci. 2015;18:477–8. doi: 10.1038/nn.3986. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Akramifard H, Balafar MA, Razavi SN, Ramli AR. Early detection of alzheimer’s disease based on clinical trials, three-dimensional imaging data, and personal information using autoencoders. J Med Signals Sens. 2021;11:120–30. doi: 10.4103/jmss.JMSS_11_20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Kalaria RN, Harik SI. Reduced glucose transporter at the blood-brain barrier and in cerebral cortex in alzheimer disease. J Neurochem. 1989;53:1083–8. doi: 10.1111/j.1471-4159.1989.tb07399.x. [DOI] [PubMed] [Google Scholar]
- 107.Osborn LM, Kamphuis W, Wadman WJ, Hol EM. Astrogliosis: An integral player in the pathogenesis of alzheimer’s disease. Prog Neurobiol. 2016;144:121–41. doi: 10.1016/j.pneurobio.2016.01.001. [DOI] [PubMed] [Google Scholar]
- 108.Leng K, Li E, Eser R, Piergies A, Sit R, Tan M. Molecular characterization of selectively vulnerable neurons in alzheimer’s disease. Nat Neurosci. 2021;24:276–87. doi: 10.1038/s41593-020-00764-7. et al. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Fu W, Shi D, Westaway D, Jhamandas JH. Bioenergetic mechanisms in astrocytes may contribute to amyloid plaque deposition and toxicity. J Biol Chem. 2015;290:12504–13. doi: 10.1074/jbc.M114.618157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Kurt MA, Davies DC, Kidd M. Beta-amyloid immunoreactivity in astrocytes in alzheimer’s disease brain biopsies: An electron microscope study. Exp Neurol. 1999;158:221–8. doi: 10.1006/exnr.1999.7096. [DOI] [PubMed] [Google Scholar]
- 111.Yamamoto N, Fujii Y, Kasahara R, Tanida M, Ohora K, Ono Y. Simvastatin and atorvastatin facilitates amyloid beta-protein degradation in extracellular spaces by increasing neprilysin secretion from astrocytes through activation of mapk/erk1/2 pathways. Glia. 2016;64:952–62. doi: 10.1002/glia.22974. et al. [DOI] [PubMed] [Google Scholar]
- 112.Legname G, Scialo C. On the role of the cellular prion protein in the uptake and signaling of pathological aggregates in neurodegenerative diseases. Prion. 2020;14:257–70. doi: 10.1080/19336896.2020.1854034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Funato H, Yoshimura M, Yamazaki T, Saido TC, Ito Y, Yokofujita J. Astrocytes containing amyloid beta-protein (abeta)-positive granules are associated with abeta40-positive diffuse plaques in the aged human brain. Am J Pathol. 1998;152:983–92. et al. [PMC free article] [PubMed] [Google Scholar]
- 114.Pfrieger FW, Ungerer N. Cholesterol metabolism in neurons and astrocytes. Prog Lipid Res. 2011;50:357–71. doi: 10.1016/j.plipres.2011.06.002. [DOI] [PubMed] [Google Scholar]
- 115.Ionov M, Burchell V, Klajnert B, Bryszewska M, Abramov AY. Mechanism of neuroprotection of melatonin against beta-amyloid neurotoxicity. Neuroscience. 2011;180:229–37. doi: 10.1016/j.neuroscience.2011.02.045. [DOI] [PubMed] [Google Scholar]
- 116.Abramov AY, Canevari L, Duchen MR. Beta-amyloid peptides induce mitochondrial dysfunction and oxidative stress in astrocytes and death of neurons through activation of nadph oxidase. J Neurosci. 2004;24:565–75. doi: 10.1523/JNEUROSCI.4042-03.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Malkov A, Popova I, Ivanov A, Jang SS, Yoon SY, Osypov A. Abeta initiates brain hypometabolism, network dysfunction and behavioral abnormalities via nox2-induced oxidative stress in mice. Commun Biol. 2021;4:1054. doi: 10.1038/s42003-021-02551-x. et al. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Hickman S, Izzy S, Sen P, Morsett L, El Khoury J. Microglia in neurodegeneration. Nat Neurosci. 2018;21:1359–69. doi: 10.1038/s41593-018-0242-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Prinz M, Jung S, Priller J. Microglia biology: One century of evolving concepts. Cell. 2019;179:292–11. doi: 10.1016/j.cell.2019.08.053. [DOI] [PubMed] [Google Scholar]
- 120.Aldana BI. Microglia-specific metabolic changes in neurodegeneration. J Mol Biol. 2019;431:1830–42. doi: 10.1016/j.jmb.2019.03.006. [DOI] [PubMed] [Google Scholar]
- 121.Boland B, Yu WH, Corti O, Mollereau B, Henriques A, Bezard E. Promoting the clearance of neurotoxic proteins in neurodegenerative disorders of ageing. Nat Rev Drug Discov. 2018;17:660–88. doi: 10.1038/nrd.2018.109. et al. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Deczkowska A, Keren-Shaul H, Weiner A, Colonna M, Schwartz M, Amit I. Disease-associated microglia: A universal immune sensor of neurodegeneration. Cell. 2018;173:1073–81. doi: 10.1016/j.cell.2018.05.003. [DOI] [PubMed] [Google Scholar]
- 123.Sobue A, Komine O, Hara Y, Endo F, Mizoguchi H, Watanabe S. Microglial gene signature reveals loss of homeostatic microglia associated with neurodegeneration of alzheimer’s disease. Acta Neuropathol Commun. 2021;9:1. doi: 10.1186/s40478-020-01099-x. et al. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Fassler M, Rappaport MS, Cuno CB, George J. Engagement of trem2 by a novel monoclonal antibody induces activation of microglia and improves cognitive function in alzheimer’s disease models. J Neuroinflammation. 2021;18:19. doi: 10.1186/s12974-020-01980-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Barros LF, Brown A, Swanson RA. Glia in brain energy metabolism: A perspective. Glia. 2018;66:1134–7. doi: 10.1002/glia.23316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Keren-Shaul H, Spinrad A, Weiner A, Matcovitch-Natan O, Dvir-Szternfeld R, Ulland TK. A unique microglia type associated with restricting development of alzheimer’s disease. Cell. 2017;169:1276–90. doi: 10.1016/j.cell.2017.05.018. et al. e17. [DOI] [PubMed] [Google Scholar]
- 127.Backes H, Walberer M, Ladwig A, Rueger MA, Neumaier B, Endepols H. Glucose consumption of inflammatory cells masks metabolic deficits in the brain. Neuroimage. 2016;128:54–62. doi: 10.1016/j.neuroimage.2015.12.044. et al. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Ulrich JD, Ulland TK, Colonna M, Holtzman DM. Elucidating the role of trem2 in alzheimer’s disease. Neuron. 2017;94:237–48. doi: 10.1016/j.neuron.2017.02.042. [DOI] [PubMed] [Google Scholar]
- 129.Ulland TK, Song WM, Huang SC, Ulrich JD, Sergushichev A, Beatty WL. Trem2 maintains microglial metabolic fitness in alzheimer’s disease. Cell. 2017;170:649–63. doi: 10.1016/j.cell.2017.07.023. et al. e13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Backhed F, Ley RE, Sonnenburg JL, Peterson DA, Gordon JI. Host-bacterial mutualism in the human intestine. Science. 2005;307:1915–20. doi: 10.1126/science.1104816. [DOI] [PubMed] [Google Scholar]
- 131.Kayama H, Takeda K. Manipulation of epithelial integrity and mucosal immunity by host and microbiota-derived metabolites. Eur J Immunol. 2020;50:921–31. doi: 10.1002/eji.201948478. [DOI] [PubMed] [Google Scholar]
- 132.Kowalski K, Mulak A. Brain-gut-microbiota axis in alzheimer’s disease. J Neurogastroenterol Motil. 2019;25:48–60. doi: 10.5056/jnm18087. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Sochocka M, Donskow-Lysoniewska K, Diniz BS, Kurpas D, Brzozowska E, Leszek J. The gut microbiome alterations and inflammation-driven pathogenesis of alzheimer’s disease-a critical review. Mol Neurobiol. 2019;56:1841–51. doi: 10.1007/s12035-018-1188-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Olson CA, Vuong HE, Yano JM, Liang QY, Nusbaum DJ, Hsiao EY. The gut microbiota mediates the anti-seizure effects of the ketogenic diet. Cell. 2018;174:497. doi: 10.1016/j.cell.2018.06.051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Baj A, Moro E, Bistoletti M, Orlandi V, Crema F, Giaroni C. Glutamatergic signaling along the microbiota-gut-brain axis. Int J Mol Sci. 2019;20:1482. doi: 10.3390/ijms20061482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Franceschi F, Ojetti V, Candelli M, Covino M, Cardone S, Potenza A. Microbes and alzheimer’ disease: Lessons from h. Pylori and gut microbiota. Eur Rev Med Pharmacol Sci. 2019;23:426–30. doi: 10.26355/eurrev_201901_16791. et al. [DOI] [PubMed] [Google Scholar]
- 137.Jameson KG, Hsiao EY. Linking the gut microbiota to a brain neurotransmitter. Trends Neurosci. 2018;41:413–4. doi: 10.1016/j.tins.2018.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Kim T. Can gamma entrainment of the brain rhythms prevent or alleviate Alzheimer’s disease? J Transl Intern Med. 2021;9:231–3. doi: 10.2478/jtim-2021-0048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.De Angelis M, Francavilla R, Piccolo M, De Giacomo A, Gobbetti M. Autism spectrum disorders and intestinal microbiota. Gut Microbes. 2015;6:207–13. doi: 10.1080/19490976.2015.1035855. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Berer K, Mues M, Koutrolos M, Rasbi ZA, Boziki M, Johner C. Commensal microbiota and myelin autoantigen cooperate to trigger autoimmune demyelination. Nature. 2011;479:538–41. doi: 10.1038/nature10554. et al. [DOI] [PubMed] [Google Scholar]
- 141.Spielman LJ, Gibson DL, Klegeris A. Unhealthy gut, unhealthy brain: The role of the intestinal microbiota in neurodegenerative diseases. Neurochem Int. 2018;120:149–63. doi: 10.1016/j.neuint.2018.08.005. [DOI] [PubMed] [Google Scholar]
- 142.Naveed M, Mubeen S, Khan A, Ibrahim S, Meer B. Plasma biomarkers: Potent screeners of alzheimer’s disease. Am J Alzheimers Dis Other Demen. 2019;34:290–301. doi: 10.1177/1533317519848239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Harach T, Marungruang N, Duthilleul N, Cheatham V, Mc Coy KD, Frisoni G. Reduction of abeta amyloid pathology in appps1 transgenic mice in the absence of gut microbiota. Sci Rep. 2017;7:41802. doi: 10.1038/srep41802. et al. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Kahn MS, Kranjac D, Alonzo CA, Haase JH, Cedillos RO, McLinden KA. Prolonged elevation in hippocampal abeta and cognitive deficits following repeated endotoxin exposure in the mouse. Behav Brain Res. 2012;229:176–84. doi: 10.1016/j.bbr.2012.01.010. et al. [DOI] [PubMed] [Google Scholar]
- 145.Akbari E, Asemi Z, Daneshvar Kakhaki R, Bahmani F, Kouchaki E, Tamtaji OR. Effect of probiotic supplementation on cognitive function and metabolic status in alzheimer’s disease: A randomized, double-blind and controlled trial. Front Aging Neurosci. 2016;8:256. doi: 10.3389/fnagi.2016.00256. et al. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Cuomo P, Papaianni M, Sansone C, Iannelli A, Iannelli D, Medaglia C. An in vitro model to investigate the role of helicobacter pylori in type 2 diabetes, obesity, alzheimer’s disease and cardiometabolic disease. Int J Mol Sci. 2020;21:8369. doi: 10.3390/ijms21218369. et al. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Shabbir U, Arshad MS, Sameen A, Oh DH. Crosstalk between gut and brain in alzheimer’s disease: The role of gut microbiota modulation strategies. Nutrients. 2021;13:690. doi: 10.3390/nu13020690. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Zhang L, Zhang S, Maezawa I, Trushin S, Minhas P, Pinto M. Modulation of mitochondrial complex i activity averts cognitive decline in multiple animal models of familial alzheimer’s disease. EBioMedicine. 2015;2:294–305. doi: 10.1016/j.ebiom.2015.03.009. et al. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Baek SH, Park SJ, Jeong JI, Kim SH, Han J, Kyung JW. Inhibition of drp1 ameliorates synaptic depression, abeta deposition, and cognitive impairment in an alzheimer’s disease model. J Neurosci. 2017;37:5099–110. doi: 10.1523/JNEUROSCI.2385-16.2017. et al. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Hou Y, Lautrup S, Cordonnier S, Wang Y, Croteau DL, Zavala E. Nad(+) supplementation normalizes key alzheimer’s features and DNA damage responses in a new ad mouse model with introduced DNA repair deficiency. Proc Natl Acad Sci U S A. 2018;115:E1876–85. doi: 10.1073/pnas.1718819115. et al. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Taylor MK, Sullivan DK, Mahnken JD, Burns JM, Swerdlow RH. Feasibility and efficacy data from a ketogenic diet intervention in alzheimer’s disease. Alzheimers Dement (N Y) 2018;4:28–36. doi: 10.1016/j.trci.2017.11.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Ota M, Matsuo J, Ishida I, Takano H, Yokoi Y, Hori H. Effects of a medium-chain triglyceride-based ketogenic formula on cognitive function in patients with mild-to-moderate alzheimer’s disease. Neurosci Lett. 2019;690:232–6. doi: 10.1016/j.neulet.2018.10.048. et al. [DOI] [PubMed] [Google Scholar]
- 153.Wilkins HM, Koppel S, Carl SM, Ramanujan S, Weidling I, Michaelis ML. Oxaloacetate enhances neuronal cell bioenergetic fluxes and infrastructure. J Neurochem. 2016;137:76–87. doi: 10.1111/jnc.13545. et al. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Roy M, Beauvieux MC, Naulin J, El Hamrani D, Gallis JL, Cunnane SC. Rapid adaptation of rat brain and liver metabolism to a ketogenic diet: An integrated study using (1)h- and (13)c-nmr spectroscopy. J Cereb Blood Flow Metab. 2015;35:1154–62. doi: 10.1038/jcbfm.2015.29. et al. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Mattson MP, Moehl K, Ghena N, Schmaedick M, Cheng A. Intermittent metabolic switching, neuroplasticity and brain health. Nat Rev Neurosci. 2018;19:63–80. doi: 10.1038/nrn.2017.156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Goldenberg RM, Steen O. Semaglutide: Review and place in therapy for adults with type 2 diabetes. Can J Diabetes. 2019;43:136–45. doi: 10.1016/j.jcjd.2018.05.008. [DOI] [PubMed] [Google Scholar]
- 157.Sayed NH, Fathy N, Kortam MA, Rabie MA, Mohamed AF, Kamel AS. Vildagliptin attenuates huntington’s disease through activation of glp-1 receptor/pi3k/akt/bdnf pathway in 3-nitropropionic acid rat model. Neurotherapeutics. 2020;17:252–68. doi: 10.1007/s13311-019-00805-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Chalichem NSS, Gonugunta C, Krishnamurthy PT, Duraiswamy B. Dpp4 inhibitors can be a drug of choice for type 3 diabetes: A mini review. Am J Alzheimers Dis Other Demen. 2017;32:444–51. doi: 10.1177/1533317517722005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Isik AT, 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;123:192–8. doi: 10.1016/j.diabres.2016.12.010. [DOI] [PubMed] [Google Scholar]
- 160.de Freitas Silva M, Dias KST, Gontijo VS, Ortiz CJC, Viegas C. Multi-target directed drugs as a modern approach for drug design towards alzheimer’s disease: An update. Curr Med Chem. 2018;25:3491–525. doi: 10.2174/0929867325666180111101843. Jr. [DOI] [PubMed] [Google Scholar]
- 161.Cummings J, Ritter A, Rothenberg K. Advances in management of neuropsychiatric syndromes in neurodegenerative diseases. Curr Psychiatry Rep. 2019;21:79. doi: 10.1007/s11920-019-1058-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Kashiwaya Y, Bergman C, Lee JH, Wan R, King MT, Mughal MR. A ketone ester diet exhibits anxiolytic and cognition-sparing properties, and lessens amyloid and tau pathologies in a mouse model of alzheimer’s disease. Neurobiol Aging. 2013;34:1530–9. doi: 10.1016/j.neurobiolaging.2012.11.023. et al. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Zilberter M, Ivanov A, Ziyatdinova S, Mukhtarov M, Malkov A, Alpar A. Dietary energy substrates reverse early neuronal hyperactivity in a mouse model of alzheimer’s disease. J Neurochem. 2013;125:157–71. doi: 10.1111/jnc.12127. et al. [DOI] [PubMed] [Google Scholar]
- 164.Morland C, Andersson KA, Haugen OP, Hadzic A, Kleppa L, Gille A. Exercise induces cerebral vegf and angiogenesis via the lactate receptor hcar1. Nat Commun. 2017;8:15557. doi: 10.1038/ncomms15557. et al. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Webber CJ, Lei SE, Wolozin B. The pathophysiology of neurodegenerative disease: Disturbing the balance between phase separation and irreversible aggregation. Prog Mol Biol Transl Sci. 2020;174:187–223. doi: 10.1016/bs.pmbts.2020.04.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Insel PS, Ossenkoppele R, Gessert D, Jagust W, Landau S, Hansson O. Time to amyloid positivity and preclinical changes in brain metabolism, atrophy, and cognition: Evidence for emerging amyloid pathology in alzheimer’s disease. Front Neurosci. 2017;11:281. doi: 10.3389/fnins.2017.00281. et al. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Neth BJ, Mintz A, Whitlow C, Jung Y, Solingapuram Sai K, Register TC. Modified ketogenic diet is associated with improved cerebrospinal fluid biomarker profile, cerebral perfusion, and cerebral ketone body uptake in older adults at risk for alzheimer’s disease: A pilot study. Neurobiol Aging. 2020;86:54–63. doi: 10.1016/j.neurobiolaging.2019.09.015. et al. [DOI] [PMC free article] [PubMed] [Google Scholar]