Next Article in Journal
Osteogenic Differentiation of Human Periodontal Ligament Stromal Cells Influences Their Immunosuppressive Potential toward Allogenic CD4+ T Cells
Next Article in Special Issue
Minding the Gap: Exploring Neuroinflammatory and Microglial Sex Differences in Alzheimer’s Disease
Previous Article in Journal
STXBP6 Gene Mutation: A New Form of SNAREopathy Leads to Developmental Epileptic Encephalopathy
Previous Article in Special Issue
Phototherapy of Alzheimer’s Disease: Photostimulation of Brain Lymphatics during Sleep: A Systematic Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Insulin-like Growth Factor I in Mechanisms of Resilience and Vulnerability to Sporadic Alzheimer’s Disease

by
Jonathan A. Zegarra-Valdivia
1,2,3,
Jaime Pignatelli
2,4,
Angel Nuñez
5 and
Ignacio Torres Aleman
1,2,6,*
1
Achucarro Basque Center for Neuroscience, 48940 Leioa, Spain
2
Biomedical Research Networking Center on Neurodegenerative Diseases (CIBERNED), 28029 Madrid, Spain
3
School of Medicine, Universidad Señor de Sipán, Chiclayo 14000, Peru
4
Cajal Institute (CSIC), 28002 Madrid, Spain
5
Department of Anatomy, Histology and Neuroscience, Universidad Autónoma de Madrid, 28049 Madrid, Spain
6
Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(22), 16440; https://doi.org/10.3390/ijms242216440
Submission received: 11 October 2023 / Revised: 6 November 2023 / Accepted: 14 November 2023 / Published: 17 November 2023
(This article belongs to the Special Issue Recent Advances in Alzheimer’s Disease)

Abstract

:
Despite decades of intense research, disease-modifying therapeutic approaches for Alzheimer’s disease (AD) are still very much needed. Apart from the extensively analyzed tau and amyloid pathological cascades, two promising avenues of research that may eventually identify new druggable targets for AD are based on a better understanding of the mechanisms of resilience and vulnerability to this condition. We argue that insulin-like growth factor I (IGF-I) activity in the brain provides a common substrate for the mechanisms of resilience and vulnerability to AD. We postulate that preserved brain IGF-I activity contributes to resilience to AD pathology as this growth factor intervenes in all the major pathological cascades considered to be involved in AD, including metabolic impairment, altered proteostasis, and inflammation, to name the three that are considered to be the most important ones. Conversely, disturbed IGF-I activity is found in many AD risk factors, such as old age, type 2 diabetes, imbalanced diet, sedentary life, sociality, stroke, stress, and low education, whereas the Apolipoprotein (Apo) E4 genotype and traumatic brain injury may also be influenced by brain IGF-I activity. Accordingly, IGF-I activity should be taken into consideration when analyzing these processes, while its preservation will predictably help prevent the progress of AD pathology. Thus, we need to define IGF-I activity in all these conditions and develop a means to preserve it. However, defining brain IGF-I activity cannot be solely based on humoral or tissue levels of this neurotrophic factor, and new functionally based assessments need to be developed.

1. Introduction

Mutations in amyloid precursor protein (APP) and preselinins are associated with familial Alzheimer’s disease (fAD), which constitutes around 1% of AD cases [1], and are the cause of overproduction of amyloid β (Aβ) peptides [2,3]. Excess production and reduced clearance of Aβ peptides have been postulated for many years as the major pathogenic pathway in AD [4]. fAD patients usually start to show symptoms at around the fourth decade of life [5], which suggests that overproduction of Aβ over the decades is required to start AD symptoms. In sporadic AD (sAD) associated with old age, it is now considered that Aβ accumulation starts at least 20 years before AD symptoms become evident, at around >65 years of age [6]. Hence, apparently, fewer years of Aβ accumulation are required in sAD to develop symptoms, compared to fAD. Among several potential explanations, it might be that a younger brain combats Aβ accumulation more efficiently than an older brain, as specific mechanisms of resilience to cognitive deterioration have been described [7,8], which may weaken along with age. Alternatively, it is possible that in sAD, other pathogenic pathways contribute to Aβ accumulation to reach a pathological threshold earlier. This threshold is specific for each individual [9], and a sizable proportion of elders (around 30%) show Aβ accumulation without AD symptoms [10]. As sAD is considered a multifactorial disease resulting from genetic/environmental interactions [11], while the former are, at present, difficult to overcome, environmental risk factors are possible to curtail. Indeed, lifestyle interventions are now implemented in personalized medicine protocols for AD patients [12], and constitute the basis of current therapeutic proposals [13] apart from pharmacotherapy.
Based on our observations and those from many other laboratories, we postulate that among other processes involved in the transition from prodromic to fully fledged sAD, such as vascular, metabolic, or immune disturbances [14], an additional underlying process that may help explain why a given lifestyle factor modulates sAD risk is brain insulin-like growth factor I (IGF-I) activity. We already postulated that loss of IGF-I function due to reduced IGF-I receptor sensitivity, IGF-I deficiency, or both, favors the development of pathogenic events related to AD [15]. In turn, AD pathology may also contribute to disrupted IGF-I activity. We later discussed in more detail the role of insulin peptides (ILPs) in the link between lifestyle and sAD risk [16], providing a mechanistic framework for this connection. Namely, inflammation, oxidative stress, altered proteostasis, impaired Aβ clearance, tau hyperphosphorylation, disturbed metabolism, and reduced cytoprotection would be processes resulting from impaired ILP activity. We now elaborate further this proposal, postulating a specific link between IGF-I activity and mechanisms of resilience and vulnerability in AD.
Many clinical studies link altered IGF-I activity and sAD risk (see, for example, [17,18]), but no definitive connection has been yet confirmed. We now propose that most of the established risk factors for sAD, including old age, type 2 diabetes, sedentary life, loneliness, unbalanced diets, stroke, and post-traumatic stress disorders (PTSD) are associated with impaired IGF-I function. Other well-established risk factors such as ApoE4 genotype may also be related to IGF-I dysfunction, although the evidence is not as robust. Traumatic brain injury (TBI), which also disturbs brain IGF-I activity, may also be a risk factor for sAD, but the evidence in this regard is not yet firm. At any rate, not only we [19] but also others already suggested a link between altered IGF-I activity and diverse AD co-morbidities and risk factors [20,21]. However, we would like to point out that other disturbances, besides altered IGF-I activity, will likely be involved in the connection between the above-mentioned risk factors and AD.
This growth factor shows a wide repertoire of actions in the brain, and its alteration will impact on many aspects known to be affected in AD. For a more detailed discussion of this topic, we refer readers to a previous review [16]. In brief, neurotrophic activity of IGF-I involves adult neurogenesis [22], re-innervation after insult [23], reduction in of inflammation [24] and oxidative stress [25], promotion of glucose uptake [26], and many others [27]. Its pro-cognitive actions include numerous effects on neuronal plasticity [28], cognition [29], and mood [30]. IGF-I is also involved in key homeostatic processes, including energy allocation [31] and the sleep/wake cycle [32]. The circadian activity of IGF-I [33] is likely involved in the latter. Table 1 shows a summary of major IGF-I activities in relation to AD and supporting references.

2. IGF-I and AD Resilience

The concept of AD resilience has been coined to explain the presence of AD pathology in cognitively intact individuals [39]. The specific mechanism underlying AD resilience is still undetermined and is often related to the concept of cognitive reserve (see below). Resilience to AD seems in part to be genetically determined as it shows a sex-dependent inheritable architecture [40], and this is not surprising considering the heavy genetic make-up of AD risk [41]. This genetic component may help uncover novel targets of resilience, such as the recently reported reelin, a protein functionally related to ApoE [42], a major genetic risk factor for sAD. However, the bulk of mechanisms of AD resilience are not genetic, and novel proposals are needed.
Accordingly, several lines of research are trying to shed light on AD resilience, as it appears very promising to develop novel routes of AD therapy. For example, early life context [43], aerobic glycolysis [44], efficient microglial phagocytosis [45], and dendritic spine plasticity [46] have all been suggested to contribute to resilience/vulnerability to AD. Therefore, understanding the underlying mechanisms will unveil new potential targets in AD prevention. In this vein, while no general consensus has yet been reached, and the major conclusions indicate that further work is needed to firmly establish a causal link between circulating IGF-I levels and cognition [47], available information allows us to suggest that preserved brain IGF-I activity also contributes to resilience to AD pathology. Thus, all the major characteristics found in individuals resilient to AD can be explained in the light of preserved brain IGF-I activity. These include conserved neuronal numbers, synaptic markers, and axonal architecture, as well as cytokine profiles consisting of higher anti-inflammatory signals and neurotrophic factors, and lower cytokine mediators of microglial recruitment [48,49]. Indeed, recent ideas supporting a multifactorial approach to treating cognitive loss in dementia [13] can be accommodated in our proposal if we consider these multi-pronged measures as a means to preserve brain IGF-I activity, such as through behavior (Figure 1).

Mechanisms of IGF-I-Dependent AD Resilience

Potentiation of neurotrophic activity, most often BDNF [51], has already been invoked as a mechanism of AD resilience [52], but specific mechanisms and factors need to be defined. Since the neuroprotective actions of IGF-I are pleiotropic [27,47], all the major characteristics found in AD resilience can be readily explained through them. These variety of IGF-I effects involve different pathways, as explained in detail elsewhere [16]. Importantly, other neurotrophic pleiotropic factors, such as melatonin, have also been implicated in AD resilience through longevity signals, such as Sirt1, or anti-inflammatory pathways involving NFκB [53]. Therefore, it is very likely that different neurotrophic activities, and not only IGF-I, are involved in resilience to AD.
As for the mechanisms underlying IGF-I-mediated AD resilience, we first focus on cell-based processes that affect all types of brain cells [54]. Among them, synapse loss is considered a major structural disturbance associated with cognitive deterioration in sAD [55]. Thus, IGF-I is involved in physiological synaptogenesis during development [56], in adult brains [57], and in synapse repletion after an insult [58]. Importantly, dendritic spines, a major site of cortical synapses, provide AD resilience [46], while IGF-I promotes dendritogenesis [59] and is intricately involved in synaptic physiology [60,61].
Another process that is emerging as an important event in cellular changes in AD is neuro-inflammation, classically associated with astrocytes and microglia as the main cellular effectors [62,63]. We must remember that inflammation is primarily a homeostatic response to pathology, and when it becomes maladaptive, for as yet poorly described reasons, it constitutes a key factor in driving sAD pathology [64,65], leading to the alteration of structural and functional brain networks seen in AD, as recently reported [66]. This “double-edge sword” process [67] is also modulated by IGF-I acting through a calcineurin-NFκB pathway in astrocytes that reversibly drives AD pathology in AD mice [24]. Naturally, neuro-inflammation also impacts on many other cellular activities, such as astrocyte phagocytosis [68], microglial reactivity [69] and proliferation [70], and activity of brain resident macrophages [71], and it also interacts with the brain angiotensin anti-inflammatory pathway [72,73]. The involvement of IGF-I in the response to neuro-inflammatory processes associated with brain damage in general attests to an important role of IGF-I in neuro-inflammation [72]. Conversely, neuro-inflammation associated with AD will contribute to IGF-I resistance in a “vicious circle” often described in the AD pathological cascade.
Other cell-associated processes in AD pathology, such as excess oxidative stress [74], which is probably directly involved in AD-related cell demise [75], are also counteracted by IGF-I [25]. Since an efficient mechanism of prevention of oxidative stress has been suggested to work in the brain of individuals showing AD resilience [76], antioxidant actions of IGF-I in brain tissue could be forming part of this resilience. Moreover, tau hyperphosphorylation in neurons, a hallmark of AD, can also be ameliorated by IGF-I through its capacity to inhibit tau kinases such as GSK-3 [77]. Indeed, IGF-I null mice show brain tau hyperphosphorylation [78]. Finally, disturbed proteostasis, a common trait in many neurodegenerative diseases [79] and considered a major culprit in AD [80], is also related to brain IGF-I actions affecting brain Aβ clearance, catabolism, and neurotoxicity [81,82,83].
At the system level, dysregulated neural circuit activity [84,85] and an altered astrocytic network [86,87], or both disturbances interacting with each other [88,89], are postulated to participate in the initiation and maintenance of the AD pathogenic cascade. While diverse explanations have been proposed, including early alterations of peptidergic systems [90,91], tau accumulation [92], or early loss of inhibitory tone [93], impaired brain IGF-I activity may also be involved. Although the evidence is less robust than its relation to cell-based processes related to AD pathology, it is well documented that IGF-I regulates neuronal activity at various levels. Thus, IGF-I modulates neuronal excitability [60] and excitatory/inhibitory balance [29,94], which also includes its actions through astrocytes [37], a type of glial cell known to modulate neuronal circuits. Indeed, we recently argued that regulation of neuronal activity by IGF-I is so widespread that it may underlie its role as an interoceptive neuromodulatory signal involved in brain states [47].
Another system-level disturbance associated with AD is insulin resistance, as seen in type 2 diabetes [95]. In this case, the evidence linking IGF-I activity with insulin sensitivity and brain insulin actions is robust [96,97], even though the hierarchical structure of these relationships is not yet defined. Finally, a vascular-related disturbance underlying sAD pathology was invoked decades ago, and is of potential relevance to this disease, if only because vascular disturbances are commonly associated with AD pathological hallmarks [98]. Again, brain IGF-I is instrumental in brain vascular function [99].
Both higher education and physical activity are associated to better mental health and are claimed to promote resilience to AD [100,101,102,103]. Thus, a straightforward connection between preserved brain IGF-I activity and AD resilience can be established. Indeed, both increased mental activity associated with environmental enrichment [104] and higher physical activity associated with exercise promote brain IGF-I function [105,106].
The aforementioned processes illustrate the pleiotropic actions of IGF-I on brain cells since multiple aspects of cell physiology appear to be targeted by IGF-I. This is a key characteristic of brain IGF-I function that probably is present in other organs.

3. IGF-I and AD Risk

We will discuss now those instances where disturbed brain IGF-I activity likely helps explain its association with AD risk. The underlying mechanisms usually relate to reduced IGF-I activity, resistance to IGF-I actions, or even both.

3.1. Old Age

Age is the most important risk factor for sAD [1] and is associated with a decline in the activity of IGF-I [107] in the form of deficiency [108] and resistance [109], affecting also the brain [110]. This reduced activity is sufficient to explain the lower IGF-I-dependent resources to combat age-associated deleterious changes that may contribute to the development of the pathogenic cascade in AD. Reduced IGF-I input during aging compromises health span in general [111], while in the brain, this deficiency impacts on vascular function [112], neuro-vascular coupling [113], cognition [114], mood balance [115], and sensory perception [116,117]. A particular characteristic of aging that is presently gaining attention in its relation to AD pathology is that it is frequently associated with disturbed circadian rhythms in the brain and in peripheral organs [118], mostly because AD patients show altered sleep/wake timing [119]. Of note, Aβ clearance takes place mostly during sleep [120], while IGF-I regulates the expression of circadian clock genes [121], and its production depends on them [33]. Accordingly, we recently observed that IGF-I also modulates the sleep/wake cycle [122] and other circadian behaviors [123].
Despite an obvious association between reduced IGF-I activity during aging and deleterious changes in brain function found in sAD, current mainstream thinking poses brain IGF-I activity as detrimental in AD pathology [124,125]. Thus, whether age-associated lowering IGF-I activity is adaptive or maladaptive for AD, along with other brain maladies [126,127], is still debated. We recently addressed this controversy, favoring the proposal that the IGF-I receptor (IGF-IR) is a dependence receptor [128] with ligand-independent actions that are counter-regulated by IGF-I [129]. We reasoned that in old age, IGF-I activity is decreased, and ligand-independent actions of IGF-IR remains unchecked. However, more work is needed to clarify the potential causal role of IGF-I in AD pathology.

3.2. Type 2 Diabetes

Loss of insulin sensitivity underlying type 2 diabetes (T2D) is linked to age [130] and affects also IGF-I sensitivity [131], as both ILPs are functionally interrelated in the control of glucose handling [31,132] and probably in other functions [128]. Since different lines of evidence support that T2D is a risk factor for sAD [133], in recent years, a causal connection between T2D and AD has been extensively explored and discussed [95,134], although no firm conclusions have been reached yet. Thus, it is not clear whether T2D favors AD [135,136], or vice versa [137,138], or whether both conditions evolve in parallel [139]. Among the favored mechanisms underlying this connection, T2D-induced brain Aβ accumulation [140], tau hyperphosphorylation [141], Aβ-induced loss of insulin sensitivity [142], T2D-associated oxidative stress and inflammation [134], subclinical blood–brain barrier (BBB) breakdown [143], and defective insulin signaling [144] have been proposed.
We consider that brain disturbances associated with T2D, most prominently cognitive deterioration [145], may be related not only to the underlying metabolic alterations but also to a dysfunctional ILP system (which includes IGF-binding proteins) that will interfere with essential homeostasis processes, such as central control of energy allocation [97,146], inflammation/oxidative stress, feeding, or the sleep/wake cycle, as discussed above.

3.3. Imbalanced Diet

A link between diet and AD has long been recognized in epidemiological studies [147], although recent evidence questions a direct cause–effect relationship since diverse contradictory observations [148,149] no longer allow us to unequivocally ascribe obesity as a risk factor for AD, although a majority of analyses favor this connection [150]. As type 2 diabetes, metabolic syndrome, and imbalanced diets are closely linked, metabolic alterations associated with improper feeding behavior still need to be considered important contributors in the path to AD, as pilot experiments have suggested [151]. At any rate, diet habits long considered to be protective, such as the Mediterranean diet, are probably beneficial in attenuating AD risk [152]. Among the potential pathogenic drivers in the link between diet and AD, neuroinflammation is considered the best candidate [153] as it is a disturbance associated with reduced IGF-I activity [154].

3.4. Sedentary Life

Among the modifiable lifestyle factors included in preventive schemes for sAD and many other maladies, an active life is commonly considered. There is now ample experimental and epidemiological evidence that physical activity is an effective measure to preserve cognitive abilities [155], with therapeutic application [156]. Experimental evidence suggests that the therapeutic efficacy of physical activity differs from that provided by mental activity [157]. However, this distinction may not be relevant for our argument, as it seems that brain activity per se [105], regardless of what triggers it, stimulates brain uptake of circulating IGF-I. Conversely, since serum IGF-I is a mediator of exercise neuroprotection [158], we suggested that disturbed IGF-I action in the brain contributes to the deleterious effects of a sedentary life through a loss of homeostatic repair mechanisms [159].

3.5. Low Education

Years in school are inversely associated with the risk of dementia [160] and rate of cognitive decline [161]. The concept of cognitive reserve [162], with more functional resources available, provides a commonly used explanation for this link [163]. This concept helps interpret the fact that around 30% of people with AD pathology show normal cognition [164], with explanations varying from larger brains or increased neuronal plasticity in individuals with higher mental activity linked to educational status. While attempts to identify the processes purportedly connecting cognitive reserve and sAD risk are still not satisfactory [165], mendelian randomization analyses point to cognitive performance associated with cognitive reserve as a direct cause of protection against AD [166]. Significantly, both brain growth [167] and neuronal plasticity are directly related to brain IGF-I activity [168]. From our point of view, higher education is associated with sustained higher mental activity, which will preserve brain IGF-I activity.

3.6. Stroke

Cerebrovascular accidents are considered the “silent pandemic” as they are the second cause of death worldwide [169]. Unfortunately, they are widely held as an sAD risk factor [170], and vascular pathology is the most common co-morbidity observed in AD brains [98]. Among the potential mechanistic links between stroke and sAD, defects in interstitial fluid drainage of Aβ peptides [171], development of cerebral amyloid angiopathy [172], tissue hypoxia [173], and excitotoxicity [174] have all been argued as pathological disturbances resulting from stroke that favor the development of sAD. In line with our proposal that IGF-I activity is responsible for the link between stroke and sAD, IGF-I activity in stroke patients is altered [175], while disturbed IGF-I activity underlies cerebrovascular dysfunction in AD mice [176]. In addition, insulin resistance, a consequence of IGF-I dysfunction [132,177], also links stroke with AD [178].

3.7. Post-Traumatic Stress Disorder

This condition is another instance that may help explain the increasing world incidence of sAD. Hence, post-traumatic stress disorder (PTSD) also shows an increased incidence worldwide, being linked to conflicts, natural disasters, and climate change [179], and may also be a risk factor for sAD, although this is still not firmly settled yet [180,181]. This is not surprising as stress in general is linked to AD pathology [182]. In this vein, as low serum IGF-I is linked to higher vulnerability to stress in humans and mice [30], and vulnerability to stress is also linked to AD risk [183], a direct link between stress and AD risk is readily justified by low IGF-I activity.
Notwithstanding a link between stress in general, IGF-I, and AD, in the particular case of PTSD, sleep disturbances associated with this condition have been argued to have a link with AD [184]. However, no AD-like pathology is found in PTSD patients [185]. Since sleep is associated with Aβ clearance [120], sleep disturbances are found in mice with reduced IGF-I activity in hypothalamic orexin neurons [32], and these mice develop PTSD-like features upon exposure to trauma [94], we propose that the link between PTSD and AD is mediated by faulty IGF-I activity in the hypothalamus. At any rate, PTSD exacerbates AD pathology in mouse models [186].
We next discuss those factors with weaker evidence that brain IGF-I activity underlies their connection with sAD risk.

3.8. ApoE4

The E4 allele of ApoE is a major genetic contributor to sAD [187]. Only a few scattered observations link ApoE4 with IGF-I, with both seemingly interacting with each other. Therefore, no robust evidence is yet available regarding a role of IGF-I in the genetic risk posed by ApoE4. Higher levels of serum IGF-I are associated with the ApoE4 genotype in a UK BioBank sample of 400,000 individuals [188], while a modifying role of ApoE4 has been ascribed to the connection between serum IGF-I levels and brain network activity in a >13,000 large cohort of the UK Biobank [189]. It has also been pointed out a modifying effect of IGF-I polymorphisms on the genetic risk of AD [190], whereas the ApoE4 genotype modulates brain responses to insulin [191], whose effects are related to IGF-I. Further, the ApoE genotype modulates the brain IGF system [192].

3.9. Traumatic Brain Injury

An association of traumatic brain injury (TBI) with a greater risk of developing dementia has also been postulated, and TBI is considered a risk factor for sAD [193], although there is still no general agreement [194]. Increased Aβ production early after TBI [195], supported by clinical observations [196], provides an easy explanation for the underlying IGF-I dysfunction, as IGF-I has been postulated to participate in Aβ clearance [81]. Indeed, a previous proposal already suggested that IGF-I underlies the link between TBI and AD [197], and recent data tend to substantiate this claim as many of the newly reported alterations purportedly linking both conditions may involve IGF-I. Thus, the resultant vascular injuries [198], axon damage favoring Aβ and hyperphosphorylated tau accumulation [199], or neuro-inflammation [200] may readily associate with IGF-I dysfunction. However, not all the changes recently reported, such as enhanced production of acetylated tau [201] or TDP-43 [202]; disturbed cathepsin B [203] or delta-secretase function [204]; and specific post-TBI disturbances identified in patients such as diffuse axonal injury, which provide an explanation for the neurodegenerative changes probably anteceding sAD [205], can yet be associated with IGF-I.

4. Outlook

Defining what we could consider a preserved IGF-I activity is challenging. A range of serum IGF-I values in the normal population has not yet been recorded since technical shortcomings have not been entirely solved [206,207,208]. Further, due to the complex biology of the IGF system, which includes IGF-I and -II together with six binding proteins, serum concentrations are, in all likelihood, insufficient to define IGF-I activity in target organs. In an attempt to respond to this limitation, clinical assays, such as “free IGF-I” [209] or “IGF-I binding activity” [210], have been proposed, but their clinical validity is not yet confirmed at a general level. Therefore, this first goal is still distant. Moreover, for a better functional account of the actual IGF-I input to the brain, we suggest that other approaches should be implemented. As an example, we already proposed [211] an exercise-based procedure to probe IGF-I activity in the brain by combining a bout of exercise (similar to procedures already used in the clinical setting to determine cardiovascular fitness) with electrophysiological (EEG) recordings of brain activity (Figure 2). This protocol is intended to be used in clinical practice and is based on previous findings of exercise-induced IGF-I entrance into the brain [106] and IGF-I-induced changes in EEG patterns [212].

5. Summary

The shortcomings of current concepts in AD research are now widely accepted [213]. Among the elegant proposals [214] and elaborated suggestions [213] formulated to provide a working framework to elucidate the etiopathogenic processes in sAD, we put forward a reductionist approach for furthering research into the development of novel AD therapies based on the notion that IGF-I activity in the brain may serve as a biomarker of resilience/vulnerability to AD pathology. Although faulty brain IGF-I activity will not be the sole factor leading to a multifactorial, highly complex disease such as sAD, we propose that it should be taken into account as a potential therapeutic aid in conjunction with novel avenues of treatment.

Funding

This research was funded by the Spanish Ministry of Science (PID2019-104376RB-I00 (AEI/FEDER, UE).

Acknowledgments

We thank past members of the lab for their contributions and the long-term funding support from Ciberned and the Spanish Ministerio de Ciencia e Innovación.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Mayeux, R. Epidemiology of neurodegeneration. Annu. Rev. Neurosci. 2003, 26, 81–104. [Google Scholar] [CrossRef] [PubMed]
  2. Citron, M.; Oltersdorf, T.; Haass, C.; McConlogue, L.; Hung, A.Y.; Seubert, P.; Vigo-Pelfrey, C.; Lieberburg, I.; Selkoe, D.J. Mutation of the β-amyloid precursor protein in familial Alzheimer’s disease increases β-protein production. Nature 1992, 360, 672–674. [Google Scholar] [CrossRef] [PubMed]
  3. Sherrington, R.; Rogaev, E.I.; Liang, Y.; Rogaeva, E.A.; Levesque, G.; Ikeda, M.; Chi, H.; Lin, C.; Li, G.; Holman, K.; et al. Cloning of a gene bearing missense mutations in early-onset familial Alzheimer’s disease. Nature 1995, 375, 754–760. [Google Scholar] [CrossRef] [PubMed]
  4. Hardy, J.; Selkoe, D.J. The amyloid hypothesis of Alzheimer’s disease: Progress and problems on the road to therapeutics. Science 2002, 297, 353–356. [Google Scholar] [CrossRef]
  5. Ryman, D.C.; Acosta-Baena, N.; Aisen, P.S.; Bird, T.; Danek, A.; Fox, N.C.; Goate, A.; Frommelt, P.; Ghetti, B.; Langbaum, J.B.; et al. Symptom onset in autosomal dominant Alzheimer disease: A systematic review and meta-analysis. Neurology 2014, 83, 253–260. [Google Scholar] [CrossRef]
  6. Jack, C.R., Jr.; Bennett, D.A.; Blennow, K.; Carrillo, M.C.; Dunn, B.; Haeberlein, S.B.; Holtzman, D.M.; Jagust, W.; Jessen, F.; Karlawish, J.; et al. NIA-AA Research Framework: Toward a biological definition of Alzheimer’s disease. Alzheimer’s Dement. 2018, 14, 535–562. [Google Scholar] [CrossRef]
  7. Udeochu, J.C.; Amin, S.; Huang, Y.; Fan, L.; Torres, E.R.S.; Carling, G.K.; Liu, B.; McGurran, H.; Coronas-Samano, G.; Kauwe, G.; et al. Tau activation of microglial cGAS–IFN reduces MEF2C-mediated cognitive resilience. Nat. Neurosci. 2023, 26, 737–750. [Google Scholar] [CrossRef]
  8. Barker, S.J.; Raju, R.M.; Milman, N.E.P.; Wang, J.; Davila-Velderrain, J.; Gunter-Rahman, F.; Parro, C.C.; Bozzelli, P.L.; Abdurrob, F.; Abdelaal, K.; et al. MEF2 is a key regulator of cognitive potential and confers resilience to neurodegeneration. Sci. Transl. Med. 2021, 13, eabd7695. [Google Scholar] [CrossRef]
  9. Vogel, J.W.; Vachon-Presseau, E.; Pichet Binette, A.; Tam, A.; Orban, P.; La Joie, R.; Savard, M.; Picard, C.; Poirier, J.; Bellec, P.; et al. Brain properties predict proximity to symptom onset in sporadic Alzheimer’s disease. Brain 2018, 141, 1871–1883. [Google Scholar] [CrossRef]
  10. Arenaza-Urquijo, E.M.; Vemuri, P. Resistance vs resilience to Alzheimer disease: Clarifying terminology for preclinical studies. Neurology 2018, 90, 695–703. [Google Scholar] [CrossRef]
  11. Blennow, K.; de Leon, M.J.; Zetterberg, H. Alzheimer’s disease. Lancet 2006, 368, 387–403. [Google Scholar] [CrossRef] [PubMed]
  12. Kosik, K.S. PErsonalized medicine for effective alzheimer disease treatment. JAMA Neurol. 2015, 72, 497–498. [Google Scholar] [CrossRef] [PubMed]
  13. Rao, R.V.; Subramaniam, K.G.; Gregory, J.; Bredesen, A.L.; Coward, C.; Okada, S.; Kelly, L.; Bredesen, D.E. Rationale for a Multi-Factorial Approach for the Reversal of Cognitive Decline in Alzheimer’s Disease and MCI: A Review. Int. J. Mol. Sci. 2023, 24, 1659. [Google Scholar] [CrossRef]
  14. Aisen, P.S.; Cummings, J.; Jack, C.R., Jr.; Morris, J.C.; Sperling, R.; Frölich, L.; Jones, R.W.; Dowsett, S.A.; Matthews, B.R.; Raskin, J.; et al. On the path to 2025: Understanding the Alzheimer’s disease continuum. Alzheimer’s Res. Ther. 2017, 9, 60. [Google Scholar] [CrossRef]
  15. Carro, E.; Torres-Aleman, I. The role of insulin and insulin-like growth factor I in the molecular and cellular mechanisms underlying the pathology of Alzheimer’s disease. Eur. J. Pharmacol. 2004, 490, 127–133. [Google Scholar] [CrossRef]
  16. Fernandez, A.M.; Santi, A.; Torres Aleman, I. Insulin peptides as mediators of the impact of life style in Alzheimer’s disease. Brain Plast. 2018, 4, 3–15. [Google Scholar] [CrossRef]
  17. Almeida, O.P.; Hankey, G.J.; Yeap, B.B.; Paul Chubb, S.A.; Gollege, J.; Flicker, L. Risk of prevalent and incident dementia associated with insulin-like growth factor and insulin-like growth factor-binding protein 3. Mol. Psychiatry 2018, 23, 1825–1829. [Google Scholar] [CrossRef] [PubMed]
  18. de Bruijn, R.F.; Janssen, J.A.; Brugts, M.P.; van Duijn, C.M.; Hofman, A.; Koudstaal, P.J.; Ikram, M.A. Insulin-Like Growth Factor-I Receptor Stimulating Activity is Associated with Dementia. J. Alzheimer’s Dis. 2014, 42, 137–142. [Google Scholar] [CrossRef]
  19. Zegarra-Valdivia, J.A.; Santi, A.; Fernandez de Sevilla, M.E.; Nunez, A.; Torres Aleman, I. Serum Insulin-Like Growth Factor I Deficiency Associates to Alzheimer’s Disease Co-Morbidities. J. Alzheimer’s Dis. 2019, 69, 979–987. [Google Scholar] [CrossRef]
  20. Amtul, Z.; Hill, D.J.; Arany, E.J.; Cechetto, D.F. Altered Insulin/Insulin-Like Growth Factor Signaling in a Comorbid Rat model of Ischemia and β-Amyloid Toxicity. Sci. Rep. 2018, 8, 5136. [Google Scholar] [CrossRef]
  21. Galle, S.A.; Geraedts, I.K.; Deijen, J.B.; Milders, M.V.; Drent, M.L. The Interrelationship between Insulin-Like Growth Factor 1, Apolipoprotein E ε4, Lifestyle Factors, and the Aging Body and Brain. J. Prev. Alzheimer’s Dis. 2020, 7, 265–273. [Google Scholar] [CrossRef] [PubMed]
  22. Aberg, M.A.; Aberg, N.D.; Hedbacker, H.; Oscarsson, J.; Eriksson, P.S. Peripheral infusion of IGF-I selectively induces neurogenesis in the adult rat hippocampus. J. Neurosci. 2000, 20, 2896–2903. [Google Scholar] [CrossRef] [PubMed]
  23. Fernandez, A.M.; de la Vega, A.G.; Torres-Aleman, I. Insulin-like growth factor I restores motor coordination in a rat model of cerebellar ataxia. Proc. Natl. Acad. Sci. USA 1998, 95, 1253–1258. [Google Scholar] [CrossRef]
  24. Fernandez, A.M.; Jimenez, S.; Mecha, M.; Davila, D.; Guaza, C.; Vitorica, J.; Torres-Aleman, I. Regulation of the phosphatase calcineurin by insulin-like growth factor I unveils a key role of astrocytes in Alzheimer’s pathology. Mol. Psychiatry 2012, 17, 705–718. [Google Scholar] [CrossRef]
  25. Heck, S.; Lezoualc’h, F.; Engert, S.; Behl, C. Insulin-like growth factor-1-mediated neuroprotection against oxidative stress is associated with activation of nuclear factor kappaB. J. Biol. Chem. 1999, 274, 9828–9835. [Google Scholar] [CrossRef] [PubMed]
  26. Cheng, C.M.; Reinhardt, R.R.; Lee, W.H.; Joncas, G.; Patel, S.C.; Bondy, C.A. Insulin-like growth factor 1 regulates developing brain glucose metabolism. Proc. Natl. Acad. Sci. USA 2000, 97, 10236–10241. [Google Scholar] [CrossRef]
  27. Fernandez, A.M.; Torres-Aleman, I. The many faces of insulin-like peptide signalling in the brain. Nat. Rev. Neurosci. 2012, 13, 225–239. [Google Scholar] [CrossRef] [PubMed]
  28. Liu, Z.; Chen, Z.; Shang, C.; Yan, F.; Shi, Y.; Zhang, J.; Qu, B.; Han, H.; Wang, Y.; Li, D.; et al. IGF1-Dependent Synaptic Plasticity of Mitral Cells in Olfactory Memory during Social Learning. Neuron 2017, 95, 106–122.e5. [Google Scholar] [CrossRef] [PubMed]
  29. Trejo, J.L.; Piriz, J.; Llorens-Martin, M.V.; Fernandez, A.M.; Bolos, M.; LeRoith, D.; Nunez, A.; Torres-Aleman, I. Central actions of liver-derived insulin-like growth factor I underlying its pro-cognitive effects. Mol. Psychiatry 2007, 12, 1118–1128. [Google Scholar] [CrossRef]
  30. Santi, A.; Bot, M.; Aleman, A.; Penninx, B.W.J.H.; Aleman, I.T. Circulating insulin-like growth factor I modulates mood and is a biomarker of vulnerability to stress: From mouse to man. Transl. Psychiatry 2018, 8, 142. [Google Scholar] [CrossRef]
  31. Fernandez, A.M.; Hernandez, E.; Guerrero-Gomez, D.; Miranda-Vizuete, A.; Torres Aleman, I. A network of insulin peptides regulate glucose uptake by astrocytes: Potential new druggable targets for brain hypometabolism. Neuropharmacology 2018, 136 Pt B, 216–222. [Google Scholar] [CrossRef]
  32. Zegarra-Valdivia, J.A.; Pignatelli, J.; Fernandez de Sevilla, M.E.; Fernandez, A.M.; Munive, V.; Martinez-Rachadell, L.; Nunez, A.; Torres Aleman, I. Insulin-like growth factor I modulates sleep through hypothalamic orexin neurons. FASEB J. 2020, 34, 15975–15990. [Google Scholar] [CrossRef]
  33. Chaudhari, A.; Gupta, R.; Patel, S.; Velingkaar, N.; Kondratov, R. Cryptochromes regulate IGF-1 production and signaling through control JAK2 dependent STAT5B phosphorylation. Mol. Biol. Cell 2017, 28, 834–842. [Google Scholar] [CrossRef]
  34. Aberg, M.A.; Aberg, N.D.; Palmer, T.D.; Alborn, A.M.; Carlsson-Skwirut, C.; Bang, P.; Rosengren, L.E.; Olsson, T.; Gage, F.H.; Eriksson, P.S. IGF-I has a direct proliferative effect in adult hippocampal progenitor cells. Mol. Cell Neurosci. 2003, 24, 23–40. [Google Scholar] [CrossRef] [PubMed]
  35. Fernandez, A.M.; Fernandez, S.; Carrero, P.; Garcia-Garcia, M.; Torres-Aleman, I. Calcineurin in reactive astrocytes plays a key role in the interplay between proinflammatory and anti-inflammatory signals. J. Neurosci. 2007, 27, 8745–8756. [Google Scholar] [CrossRef] [PubMed]
  36. Ayadi, A.E.; Zigmond, M.J.; Smith, A.D. IGF-1 protects dopamine neurons against oxidative stress: Association with changes in phosphokinases. Exp. Brain Res. 2016, 234, 1863–1873. [Google Scholar] [CrossRef] [PubMed]
  37. Noriega-Prieto, J.A.; Maglio, L.E.; Zegarra-Valdivia, J.A.; Pignatelli, J.; Fernandez, A.M.; Martinez-Rachadell, L.; Fernandes, J.; Nunez, A.; Araque, A.; Torres-Aleman, I.; et al. Astrocytic IGF-IRs Induce Adenosine-Mediated Inhibitory Downregulation and Improve Sensory Discrimination. J. Neurosci. 2021, 41, 4768–4781. [Google Scholar] [CrossRef] [PubMed]
  38. Hernandez-Garzon, E.; Fernandez, A.M.; Perez-Alvarez, A.; Genis, L.; Bascunana, P.; Fernandez de la Rosa, R.; Delgado, M.; Angel Pozo, M.; Moreno, E.; McCormick, P.J.; et al. The insulin-like growth factor I receptor regulates glucose transport by astrocytes. Glia 2016, 64, 1962–1971. [Google Scholar] [CrossRef]
  39. Gómez-Isla, T.; Frosch, M.P. Lesions without symptoms: Understanding resilience to Alzheimer disease neuropathological changes. Nat. Rev. Neurol. 2022, 18, 323–332. [Google Scholar] [CrossRef]
  40. Eissman, J.M.; Dumitrescu, L.; Mahoney, E.R.; Smith, A.N.; Mukherjee, S.; Lee, M.L.; Scollard, P.; Choi, S.E.; Bush, W.S.; Engelman, C.D.; et al. Sex differences in the genetic architecture of cognitive resilience to Alzheimer’s disease. Brain 2022, 145, 2541–2554. [Google Scholar] [CrossRef]
  41. Bellenguez, C.; Küçükali, F.; Jansen, I.E.; Kleineidam, L.; Moreno-Grau, S.; Amin, N.; Naj, A.C.; Campos-Martin, R.; Grenier-Boley, B.; Andrade, V.; et al. New insights into the genetic etiology of Alzheimer’s disease and related dementias. Nat. Genet. 2022, 54, 412–436. [Google Scholar] [CrossRef] [PubMed]
  42. Lopera, F.; Marino, C.; Chandrahas, A.S.; O’Hare, M.; Villalba-Moreno, N.D.; Aguillon, D.; Baena, A.; Sanchez, J.S.; Vila-Castelar, C.; Ramirez Gomez, L.; et al. Resilience to autosomal dominant Alzheimer’s disease in a Reelin-COLBOS heterozygous man. Nat. Med. 2023, 29, 1243–1252. [Google Scholar] [CrossRef]
  43. Lesuis, S.L.; Hoeijmakers, L.; Korosi, A.; de Rooij, S.R.; Swaab, D.F.; Kessels, H.W.; Lucassen, P.J.; Krugers, H.J. Vulnerability and resilience to Alzheimer’s disease: Early life conditions modulate neuropathology and determine cognitive reserve. Alzheimer’s Res. Ther. 2018, 10, 95. [Google Scholar] [CrossRef] [PubMed]
  44. Goyal, M.S.; Blazey, T.; Metcalf, N.V.; McAvoy, M.P.; Strain, J.F.; Rahmani, M.; Durbin, T.J.; Xiong, C.; Benzinger, T.L.; Morris, J.C.; et al. Brain aerobic glycolysis and resilience in Alzheimer disease. Proc. Natl. Acad. Sci. USA 2023, 120, e2212256120. [Google Scholar] [CrossRef] [PubMed]
  45. Fracassi, A.; Marcatti, M.; Tumurbaatar, B.; Woltjer, R.; Moreno, S.; Taglialatela, G. TREM2-induced activation of microglia contributes to synaptic integrity in cognitively intact aged individuals with Alzheimer’s neuropathology. Brain Pathol. 2023, 33, e13108. [Google Scholar] [CrossRef]
  46. Boros, B.D.; Greathouse, K.M.; Gentry, E.G.; Curtis, K.A.; Birchall, E.L.; Gearing, M.; Herskowitz, J.H. Dendritic spines provide cognitive resilience against Alzheimer’s disease. Ann. Neurol. 2017, 82, 602–614. [Google Scholar] [CrossRef]
  47. Nuñez, A.; Zegarra-Valdivia, J.; Fernandez de Sevilla, D.; Pignatelli, J.; Torres Aleman, I. The neurobiology of insulin-like growth factor I: From neuroprotection to modulation of brain states. Mol. Psychiatry 2023, 28, 3220–3230. [Google Scholar] [CrossRef]
  48. Neuner, S.M.; Telpoukhovskaia, M.; Menon, V.; O’Connell, K.M.S.; Hohman, T.J.; Kaczorowski, C.C. Translational approaches to understanding resilience to Alzheimer’s disease. Trends Neurosci. 2022, 45, 369–383. [Google Scholar] [CrossRef]
  49. Perez-Nievas, B.G.; Stein, T.D.; Tai, H.C.; Dols-Icardo, O.; Scotton, T.C.; Barroeta-Espar, I.; Fernandez-Carballo, L.; de Munain, E.L.; Perez, J.; Marquie, M.; et al. Dissecting phenotypic traits linked to human resilience to Alzheimer’s pathology. Brain 2013, 136 Pt 8, 2510–2526. [Google Scholar] [CrossRef]
  50. Stefaniak, O.; Dobrzyńska, M.; Drzymała-Czyż, S.; Przysławski, J. Diet in the Prevention of Alzheimer’s Disease: Current Knowledge and Future Research Requirements. Nutrients 2022, 14, 4564. [Google Scholar] [CrossRef]
  51. Choi, S.H.; Bylykbashi, E.; Chatila, Z.K.; Lee, S.W.; Pulli, B.; Clemenson, G.D.; Kim, E.; Rompala, A.; Oram, M.K.; Asselin, C.; et al. Combined adult neurogenesis and BDNF mimic exercise effects on cognition in an Alzheimer’s mouse model. Science 2018, 361, eaan8821. [Google Scholar] [CrossRef] [PubMed]
  52. Pham, A.Q.; Dore, K. Novel approaches to increase synaptic resilience as potential treatments for Alzheimer’s disease. Semin. Cell Dev. Biol. 2023, 139, 84–92. [Google Scholar] [CrossRef] [PubMed]
  53. Corpas, R.; Griñán-Ferré, C.; Palomera-Ávalos, V.; Porquet, D.; García de Frutos, P.; Franciscato Cozzolino, S.M.; Rodríguez-Farré, E.; Pallàs, M.; Sanfeliu, C.; Cardoso, B.R. Melatonin induces mechanisms of brain resilience against neurodegeneration. J. Pineal Res. 2018, 65, e12515. [Google Scholar] [CrossRef]
  54. De Strooper, B.; Karran, E. The Cellular Phase of Alzheimer’s Disease. Cell 2016, 164, 603–615. [Google Scholar] [CrossRef] [PubMed]
  55. DeKosky, S.T.; Scheff, S.W. Synapse loss in frontal cortex biopsies in Alzheimer’s disease: Correlation with cognitive severity. Ann. Neurol. 1990, 27, 457–464. [Google Scholar] [CrossRef]
  56. O’Kusky, J.R.; Ye, P.; D’Ercole, A.J. Insulin-like growth factor-I promotes neurogenesis and synaptogenesis in the hippocampal dentate gyrus during postnatal development. J. Neurosci. 2000, 20, 8435–8442. [Google Scholar] [CrossRef]
  57. Nieto-Estevez, V.; Defterali, C.; Vicario-Abejon, C. IGF-I: A Key Growth Factor that Regulates Neurogenesis and Synaptogenesis from Embryonic to Adult Stages of the Brain. Front. Neurosci. 2016, 10, 52. [Google Scholar] [CrossRef]
  58. Lee, M.; Kim, E.J.; Kim, M.J.; Yum, M.S.; Yeom, J.; Kim, K. Insulin-Like Growth Factor-1 Promotes Synaptogenesis Signaling, a Major Dysregulated Pathway in Malformation of Cortical Development, in a Rat Model. Mol. Neurobiol. 2023, 60, 3299–3310. [Google Scholar] [CrossRef]
  59. Niblock, M.M.; Brunso-Bechtold, J.K.; Riddle, D.R. Insulin-like growth factor I stimulates dendritic growth in primary somatosensory cortex. J. Neurosci. 2000, 20, 4165–4176. [Google Scholar] [CrossRef]
  60. Gazit, N.; Vertkin, I.; Shapira, I.; Helm, M.; Slomowitz, E.; Sheiba, M.; Mor, Y.; Rizzoli, S.; Slutsky, I. IGF-1 Receptor Differentially Regulates Spontaneous and Evoked Transmission via Mitochondria at Hippocampal Synapses. Neuron 2016, 89, 583–597. [Google Scholar] [CrossRef]
  61. Katsenelson, M.; Shapira, I.; Abbas, E.; Jevdokimenko, K.; Styr, B.; Ruggiero, A.; Aïd, S.; Fornasiero, E.F.; Holzenberger, M.; Rizzoli, S.O.; et al. IGF-1 receptor regulates upward firing rate homeostasis via the mitochondrial calcium uniporter. Proc. Natl. Acad. Sci. USA 2022, 119, e2121040119. [Google Scholar] [CrossRef] [PubMed]
  62. Patani, R.; Hardingham, G.E.; Liddelow, S.A. Functional roles of reactive astrocytes in neuroinflammation and neurodegeneration. Nat. Rev. Neurol. 2023, 19, 395–409. [Google Scholar] [CrossRef] [PubMed]
  63. Aguzzi, A.; Barres, B.A.; Bennett, M.L. Microglia: Scapegoat, saboteur, or something else? Science 2013, 339, 156–161. [Google Scholar] [CrossRef] [PubMed]
  64. Heppner, F.L.; Ransohoff, R.M.; Becher, B. Immune attack: The role of inflammation in Alzheimer disease. Nat. Rev. Neurosci. 2015, 16, 358–372. [Google Scholar] [CrossRef]
  65. Parachikova, A.; Agadjanyan, M.G.; Cribbs, D.H.; Blurton-Jones, M.; Perreau, V.; Rogers, J.; Beach, T.G.; Cotman, C.W. Inflammatory changes parallel the early stages of Alzheimer disease. Neurobiol. Aging 2007, 28, 1821–1833. [Google Scholar] [CrossRef] [PubMed]
  66. Leng, F.; Hinz, R.; Gentleman, S.; Hampshire, A.; Dani, M.; Brooks, D.J.; Edison, P. Neuroinflammation is independently associated with brain network dysfunction in Alzheimer’s disease. Mol. Psychiatry 2023, 28, 1303–1311. [Google Scholar] [CrossRef]
  67. Wyss-Coray, T.; Mucke, L. Inflammation in neurodegenerative disease-a double-edged sword. Neuron 2002, 35, 419. [Google Scholar] [CrossRef]
  68. Pinto-Benito, D.; Paradela-Leal, C.; Ganchala, D.; de Castro-Molina, P.; Arevalo, M.A. IGF-1 regulates astrocytic phagocytosis and inflammation through the p110α isoform of PI3K in a sex-specific manner. Glia 2022, 70, 1153–1169. [Google Scholar] [CrossRef]
  69. Herrera, M.L.; Bandín, S.; Champarini, L.G.; Hereñú, C.B.; Bellini, M.J. Intramuscular insulin-like growth factor-1 gene therapy modulates reactive microglia after traumatic brain injury. Brain Res. Bull. 2021, 175, 196–204. [Google Scholar] [CrossRef]
  70. O’Donnell, S.L.; Frederick, T.J.; Krady, J.K.; Vannucci, S.J.; Wood, T.L. IGF-I and microglia/macrophage proliferation in the ischemic mouse brain. Glia 2002, 39, 85–97. [Google Scholar] [CrossRef]
  71. Ivan, D.C.; Berve, K.C.; Walthert, S.; Monaco, G.; Borst, K.; Bouillet, E.; Ferreira, F.; Lee, H.; Steudler, J.; Buch, T.; et al. Insulin-like growth factor-1 receptor controls the function of CNS-resident macrophages and their contribution to neuroinflammation. Acta Neuropathol. Commun. 2023, 11, 35. [Google Scholar] [CrossRef]
  72. Labandeira-Garcia, J.L.; Costa-Besada, M.A.; Labandeira, C.M.; Villar-Cheda, B.; Rodríguez-Perez, A.I. Insulin-Like Growth Factor-1 and Neuroinflammation. Front. Aging Neurosci. 2017, 9, 365. [Google Scholar] [CrossRef] [PubMed]
  73. Ogundele, O.M.; Lee, C.C.; Francis, J. Age-dependent alterations to paraventricular nucleus insulin-like growth factor 1 receptor as a possible link between sympathoexcitation and inflammation. J. Neurochem. 2016, 139, 706–721. [Google Scholar] [CrossRef] [PubMed]
  74. Nunomura, A.; Castellani, R.J.; Zhu, X.; Moreira, P.I.; Perry, G.; Smith, M.A. Involvement of oxidative stress in Alzheimer disease. J. Neuropathol. Exp. Neurol. 2006, 65, 631–641. [Google Scholar] [CrossRef]
  75. Markesbery, W.R.; Carney, J.M. Oxidative alterations in Alzheimer’s disease. Brain Pathol. 1999, 9, 133–146. [Google Scholar] [CrossRef] [PubMed]
  76. Fracassi, A.; Marcatti, M.; Zolochevska, O.; Tabor, N.; Woltjer, R.; Moreno, S.; Taglialatela, G. Oxidative Damage and Antioxidant Response in Frontal Cortex of Demented and Nondemented Individuals with Alzheimer’s Neuropathology. J. Neurosci. 2021, 41, 538–554. [Google Scholar] [CrossRef]
  77. Desbois-Mouthon, C.; Cadoret, A.; Blivet-Van Eggelpoël, M.J.; Bertrand, F.; Cherqui, G.; Perret, C.; Capeau, J. Insulin and IGF-1 stimulate the β-catenin pathway through two signalling cascades involving GSK-3β inhibition and Ras activation. Oncogene 2001, 20, 252–259. [Google Scholar] [CrossRef] [PubMed]
  78. Cheng, C.M.; Tseng, V.; Wang, J.; Wang, D.; Matyakhina, L.; Bondy, C.A. Tau is hyperphosphorylated in the insulin-like growth factor-I null brain. Endocrinology 2005, 146, 5086–5091. [Google Scholar] [CrossRef] [PubMed]
  79. Wilson, D.M., 3rd; Cookson, M.R.; Van Den Bosch, L.; Zetterberg, H.; Holtzman, D.M.; Dewachter, I. Hallmarks of neurodegenerative diseases. Cell 2023, 186, 693–714. [Google Scholar] [CrossRef]
  80. Sorrentino, V.; Romani, M.; Mouchiroud, L.; Beck, J.S.; Zhang, H.; D’Amico, D.; Moullan, N.; Potenza, F.; Schmid, A.W.; Rietsch, S.; et al. Enhancing mitochondrial proteostasis reduces amyloid-β proteotoxicity. Nature 2017, 552, 187–193. [Google Scholar] [CrossRef]
  81. Carro, E.; Trejo, J.L.; Gomez-Isla, T.; LeRoith, D.; Torres-Aleman, I. Serum insulin-like growth factor I regulates brain amyloid-β levels. Nat. Med. 2002, 8, 1390–1397. [Google Scholar] [CrossRef] [PubMed]
  82. Adlerz, L.; Holback, S.; Multhaup, G.; Iverfeldt, K. IGF-1-induced processing of the amyloid precursor protein family is mediated by different signaling pathways. J. Biol. Chem. 2007, 282, 10203–10209. [Google Scholar] [CrossRef] [PubMed]
  83. Selles, M.C.; Fortuna, J.T.S.; Zappa-Villar, M.F.; de Faria, Y.P.R.; Souza, A.S.; Suemoto, C.K.; Leite, R.E.P.; Rodriguez, R.D.; Grinberg, L.T.; Reggiani, P.C.; et al. Adenovirus-Mediated Transduction of Insulin-Like Growth Factor 1 Protects Hippocampal Neurons from the Toxicity of Aβ Oligomers and Prevents Memory Loss in an Alzheimer Mouse Model. Mol. Neurobiol. 2020, 57, 1473–1483. [Google Scholar] [CrossRef] [PubMed]
  84. Busche, M.A.; Eichhoff, G.; Adelsberger, H.; Abramowski, D.; Wiederhold, K.H.; Haass, C.; Staufenbiel, M.; Konnerth, A.; Garaschuk, O. Clusters of Hyperactive Neurons Near Amyloid Plaques in a Mouse Model of Alzheimer’s Disease. Science 2008, 321, 1686–1689. [Google Scholar] [CrossRef]
  85. Koelewijn, L.; Lancaster, T.M.; Linden, D.; Dima, D.C.; Routley, B.C.; Magazzini, L.; Barawi, K.; Brindley, L.; Adams, R.; Tansey, K.E.; et al. Oscillatory hyperactivity and hyperconnectivity in young APOE-ɛ4 carriers and hypoconnectivity in Alzheimer’s disease. eLife 2019, 8, e36011. [Google Scholar] [CrossRef]
  86. Lee, Y.F.; Russ, A.N.; Zhao, Q.; Perle, S.J.; Maci, M.; Miller, M.R.; Hou, S.S.; Algamal, M.; Zhao, Z.; Li, H.; et al. Optogenetic targeting of astrocytes restores slow brain rhythm function and slows Alzheimer’s disease pathology. Sci. Rep. 2023, 13, 13075. [Google Scholar] [CrossRef]
  87. Bosson, A.; Paumier, A.; Boisseau, S.; Jacquier-Sarlin, M.; Buisson, A.; Albrieux, M. TRPA1 channels promote astrocytic Ca(2+) hyperactivity and synaptic dysfunction mediated by oligomeric forms of amyloid-β peptide. Mol. Neurodegener. 2017, 12, 53. [Google Scholar] [CrossRef]
  88. Kuchibhotla, K.V.; Lattarulo, C.R.; Hyman, B.T.; Bacskai, B.J. Synchronous hyperactivity and intercellular calcium waves in astrocytes in Alzheimer mice. Science 2009, 323, 1211–1215. [Google Scholar] [CrossRef]
  89. Lines, J.; Baraibar, A.M.; Fang, C.; Martin, E.D.; Aguilar, J.; Lee, M.K.; Araque, A.; Kofuji, P. Astrocyte-neuronal network interplay is disrupted in Alzheimer’s disease mice. Glia 2022, 70, 368–378. [Google Scholar] [CrossRef]
  90. Calafate, S.; Özturan, G.; Thrupp, N.; Vanderlinden, J.; Santa-Marinha, L.; Morais-Ribeiro, R.; Ruggiero, A.; Bozic, I.; Rusterholz, T.; Lorente-Echeverría, B.; et al. Early alterations in the MCH system link aberrant neuronal activity and sleep disturbances in a mouse model of Alzheimer’s disease. Nat. Neurosci. 2023, 26, 1021–1031. [Google Scholar] [CrossRef]
  91. Harris, S.S.; Wolf, F.; De Strooper, B.; Busche, M.A. Tipping the Scales: Peptide-Dependent Dysregulation of Neural Circuit Dynamics in Alzheimer’s Disease. Neuron 2020, 107, 417–435. [Google Scholar] [PubMed]
  92. Huijbers, W.; Schultz, A.P.; Papp, K.V.; LaPoint, M.R.; Hanseeuw, B.; Chhatwal, J.P.; Hedden, T.; Johnson, K.A.; Sperling, R.A. Tau Accumulation in Clinically Normal Older Adults Is Associated with Hippocampal Hyperactivity. J. Neurosci. 2019, 39, 548–556. [Google Scholar] [CrossRef] [PubMed]
  93. Nuriel, T.; Angulo, S.L.; Khan, U.; Ashok, A.; Chen, Q.; Figueroa, H.Y.; Emrani, S.; Liu, L.; Herman, M.; Barrett, G.; et al. Neuronal hyperactivity due to loss of inhibitory tone in APOE4 mice lacking Alzheimer’s disease-like pathology. Nat. Commun. 2017, 8, 1464. [Google Scholar] [CrossRef] [PubMed]
  94. Fernández de Sevilla, M.E.; Pignatelli, J.; Zegarra-Valdivia, J.A.; Mendez, P.; Nuñez, A.; Torres Alemán, I. Insulin-like growth factor I mitigates post-traumatic stress by inhibiting AMP-kinase in orexin neurons. Mol. Psychiatry 2022, 27, 2182–2196. [Google Scholar] [CrossRef] [PubMed]
  95. Arnold, S.E.; Arvanitakis, Z.; Macauley-Rambach, S.L.; Koenig, A.M.; Wang, H.Y.; Ahima, R.S.; Craft, S.; Gandy, S.; Buettner, C.; Stoeckel, L.E.; et al. Brain insulin resistance in type 2 diabetes and Alzheimer disease: Concepts and conundrums. Nat. Rev. Neurol. 2018, 14, 168–181. [Google Scholar] [CrossRef] [PubMed]
  96. Fernandez, A.M.; Hernandez-Garzon, E.; Perez-Domper, P.; Perez-Alvarez, A.; Mederos, S.; Matsui, T.; Santi, A.; Trueba-Saiz, A.; Garcia-Guerra, L.; Pose-Utrilla, J.; et al. Insulin Regulates Astrocytic Glucose Handling Through Cooperation With IGF-I. Diabetes 2017, 66, 64–74. [Google Scholar] [CrossRef] [PubMed]
  97. Farias Quipildor, G.; Mao, K.; Beltran, P.J.; Barzilai, N.; Huffman, D.M. Modulation of Glucose Production by Central Insulin Requires IGF-1 Receptors in AgRP Neurons. Diabetes 2021, 70, 2237–2249. [Google Scholar] [CrossRef]
  98. Love, S.; Miners, J.S. Cerebrovascular disease in ageing and Alzheimer’s disease. Acta Neuropathol. 2016, 131, 645–658. [Google Scholar] [CrossRef]
  99. Lopez-Lopez, C.; LeRoith, D.; Torres-Aleman, I. Insulin-like growth factor I is required for vessel remodeling in the adult brain. Proc. Natl. Acad. Sci. USA 2004, 101, 9833–9838. [Google Scholar] [CrossRef]
  100. Lachman, M.E.; Agrigoroaei, S.; Murphy, C.; Tun, P.A. Frequent cognitive activity compensates for education differences in episodic memory. Am. J. Geriatr. Psychiatry 2010, 18, 4–10. [Google Scholar] [CrossRef]
  101. Rovio, S.; Kåreholt, I.; Helkala, E.L.; Viitanen, M.; Winblad, B.; Tuomilehto, J.; Soininen, H.; Nissinen, A.; Kivipelto, M. Leisure-time physical activity at midlife and the risk of dementia and Alzheimer’s disease. Lancet Neurol. 2005, 4, 705–711. [Google Scholar] [CrossRef] [PubMed]
  102. Ahangari, N.; Fischer, C.E.; Schweizer, T.A.; Munoz, D.G. Cognitive resilience and severe Alzheimer’s disease neuropathology. Aging Brain 2023, 3, 100065. [Google Scholar] [CrossRef] [PubMed]
  103. Baker, L.D.; Frank, L.L.; Foster-Schubert, K.; Green, P.S.; Wilkinson, C.W.; McTiernan, A.; Plymate, S.R.; Fishel, M.A.; Watson, G.S.; Cholerton, B.A.; et al. Effects of aerobic exercise on mild cognitive impairment: A controlled trial. Arch. Neurol. 2010, 67, 71–79. [Google Scholar] [CrossRef] [PubMed]
  104. Ohline, S.M.; Abraham, W.C. Environmental enrichment effects on synaptic and cellular physiology of hippocampal neurons. Neuropharmacology 2019, 145 Pt A, 3–12. [Google Scholar] [CrossRef]
  105. Nishijima, T.; Piriz, J.; Duflot, S.; Fernandez, A.M.; Gaitan, G.; Gomez-Pinedo, U.; Verdugo, J.M.; Leroy, F.; Soya, H.; Nunez, A.; et al. Neuronal activity drives localized blood-brain-barrier transport of serum insulin-like growth factor-I into the CNS. Neuron 2010, 67, 834–846. [Google Scholar] [CrossRef]
  106. Carro, E.; Nunez, A.; Busiguina, S.; Torres-Aleman, I. Circulating insulin-like growth factor I mediates effects of exercise on the brain. J. Neurosci. 2000, 20, 2926–2933. [Google Scholar] [CrossRef] [PubMed]
  107. Aleman, A.; Torres-Aleman, I. Circulating insulin-like growth factor I and cognitive function: Neuromodulation throughout the lifespan. Prog. Neurobiol. 2009, 89, 256–265. [Google Scholar] [CrossRef]
  108. Pharaoh, G.; Owen, D.; Yeganeh, A.; Premkumar, P.; Farley, J.; Bhaskaran, S.; Ashpole, N.; Kinter, M.; Van Remmen, H.; Logan, S. Disparate Central and Peripheral Effects of Circulating IGF-1 Deficiency on Tissue Mitochondrial Function. Mol. Neurobiol. 2019, 57, 1317–1331. [Google Scholar] [CrossRef]
  109. Zegarra-Valdivia, J.A.; Chaves-Coira, I.; Fernandez de Sevilla, M.E.; Martinez-Rachadell, L.; Esparza, J.; Torres-Aleman, I.; Nunez, A. Reduced Insulin-Like Growth Factor-I Effects in the Basal Forebrain of Aging Mouse. Front. Aging Neurosci. 2021, 13, 682388. [Google Scholar] [CrossRef]
  110. Muller, A.P.; Fernandez, A.M.; Haas, C.; Zimmer, E.; Portela, L.V.; Torres-Aleman, I. Reduced brain insulin-like growth factor I function during aging. Mol. Cell Neurosci. 2012, 49, 9–12. [Google Scholar] [CrossRef]
  111. Gong, Z.; Kennedy, O.; Sun, H.; Wu, Y.; Williams, G.A.; Klein, L.; Cardoso, L.; Matheny, R.W.; Hubbard, G.B.; Ikeno, Y.; et al. Reductions in serum IGF-1 during aging impair health span. Aging Cell 2014, 13, 408–418. [Google Scholar] [CrossRef] [PubMed]
  112. Bickel, M.A.; Csik, B.; Gulej, R.; Ungvari, A.; Nyul-Toth, A.; Conley, S.M. Cell non-autonomous regulation of cerebrovascular aging processes by the somatotropic axis. Front. Endocrinol. 2023, 14, 1087053. [Google Scholar] [CrossRef] [PubMed]
  113. Zyzak, D.R.; Otto, T.; Eichenbaum, H.; Gallagher, M. Cognitive decline associated with normal aging in rats: A neuropsychological approach. Learn. Mem. 1995, 2, 1–16. [Google Scholar] [CrossRef] [PubMed]
  114. Frater, J.; Lie, D.; Bartlett, P.; McGrath, J.J. Insulin-like Growth Factor 1 (IGF-1) as a marker of cognitive decline in normal ageing: A review. Ageing Res. Rev. 2018, 42, 14–27. [Google Scholar] [CrossRef]
  115. Munive, V.; Zegarra-Valdivia, J.A.; Herrero-Labrador, R.; Fernandez, A.M.; Aleman, I.T. Loss of the interaction between estradiol and insulin-like growth factor I in brain endothelial cells associates to changes in mood homeostasis during peri-menopause in mice. Aging 2019, 11, 174–184. [Google Scholar] [CrossRef]
  116. Celaya, A.M.; Rodríguez-de la Rosa, L.; Bermúdez-Muñoz, J.M.; Zubeldia, J.M.; Romá-Mateo, C.; Avendaño, C.; Pallardó, F.V.; Varela-Nieto, I. IGF-1 Haploinsufficiency Causes Age-Related Chronic Cochlear Inflammation and Increases Noise-Induced Hearing Loss. Cells 2021, 10, 1686. [Google Scholar] [CrossRef]
  117. García-Magro, N.; Zegarra-Valdivia, J.A.; Troyas-Martinez, S.; Torres-Aleman, I.; Nuñez, A. Response Facilitation Induced by Insulin-like Growth Factor-I in the Primary Somatosensory Cortex of Mice Was Reduced in Aging. Cells 2022, 11, 717. [Google Scholar] [CrossRef]
  118. Acosta-Rodriguez, V.A.; Rijo-Ferreira, F.; Green, C.B.; Takahashi, J.S. Importance of circadian timing for aging and longevity. Nat. Commun. 2021, 12, 2862. [Google Scholar] [CrossRef]
  119. Olsson, M.; Arlig, J.; Hedner, J.; Blennow, K.; Zetterberg, H. Sleep deprivation and cerebrospinal fluid biomarkers for Alzheimer’s disease. Sleep 2018, 41, zsy025. [Google Scholar] [CrossRef]
  120. Kang, J.E.; Lim, M.M.; Bateman, R.J.; Lee, J.J.; Smyth, L.P.; Cirrito, J.R.; Fujiki, N.; Nishino, S.; Holtzman, D.M. Amyloid-β dynamics are regulated by orexin and the sleep-wake cycle. Science 2009, 326, 1005–1007. [Google Scholar] [CrossRef]
  121. Crosby, P.; Hamnett, R.; Putker, M.; Hoyle, N.P.; Reed, M.; Karam, C.J.; Maywood, E.S.; Stangherlin, A.; Chesham, J.E.; Hayter, E.A.; et al. Insulin/IGF-1 Drives PERIOD Synthesis to Entrain Circadian Rhythms with Feeding Time. Cell 2019, 177, 896–909. [Google Scholar] [CrossRef] [PubMed]
  122. Zegarra-Valdivia, J.A.; Fernandes, J.; Fernandez de Sevilla, M.E.; Trueba-Saiz, A.; Pignatelli, J.; Suda, K.; Martinez-Rachadell, L.; Fernandez, A.M.; Esparza, J.; Vega, M.; et al. Insulin-like growth factor I sensitization rejuvenates sleep patterns in old mice. Geroscience 2022, 44, 2243–2257. [Google Scholar] [CrossRef] [PubMed]
  123. Pignatelli, J.; de Sevilla, M.E.F.; Sperber, J.; Horrillo, D.; Medina-Gomez, G.; Aleman, I.T. Insulin-like Growth Factor I Couples Metabolism with Circadian Activity through Hypothalamic Orexin Neurons. Int. J. Mol. Sci. 2022, 23, 4679. [Google Scholar] [CrossRef] [PubMed]
  124. Cohen, E.; Paulsson, J.F.; Blinder, P.; Burstyn-Cohen, T.; Du, D.; Estepa, G.; Adame, A.; Pham, H.M.; Holzenberger, M.; Kelly, J.W.; et al. Reduced IGF-1 signaling delays age-associated proteotoxicity in mice. Cell 2009, 139, 1157–1169. [Google Scholar] [CrossRef] [PubMed]
  125. Gontier, G.; George, C.; Chaker, Z.; Holzenberger, M.; Aid, S. Blocking IGF Signaling in Adult Neurons Alleviates Alzheimer’s Disease Pathology through Amyloid-β Clearance. J. Neurosci. 2015, 35, 11500–11513. [Google Scholar] [CrossRef] [PubMed]
  126. Biondi, O.; Branchu, J.; Ben Salah, A.; Houdebine, L.; Bertin, L.; Chali, F.; Desseille, C.; Weill, L.; Sanchez, G.; Lancelin, C.; et al. IGF-1R Reduction Triggers Neuroprotective Signaling Pathways in Spinal Muscular Atrophy Mice. J. Neurosci. 2015, 35, 12063–12079. [Google Scholar] [CrossRef]
  127. De Magalhaes Filho, C.D.; Kappeler, L.; Dupont, J.; Solinc, J.; Villapol, S.; Denis, C.; Nosten-Bertrand, M.; Billard, J.M.; Blaise, A.; Tronche, F.; et al. Deleting IGF-1 receptor from forebrain neurons confers neuroprotection during stroke and upregulates endocrine somatotropin. J. Cereb. Blood Flow. Metab. 2016, 37, 396–412. [Google Scholar] [CrossRef]
  128. Boucher, J.; Macotela, Y.; Bezy, O.; Mori, M.A.; Kriauciunas, K.; Kahn, C.R. A Kinase-Independent Role for Unoccupied Insulin and IGF-1 Receptors in the Control of Apoptosis. Sci. Signal 2010, 3, ra87. [Google Scholar] [CrossRef]
  129. Zegarra-Valdivia, J.; Nuñez, A.; Aleman, I.T. Untangling IGF-I signaling in the aging brain. Aging 2023, 15, 599–600. [Google Scholar] [CrossRef]
  130. Kirkman, M.S.; Briscoe, V.J.; Clark, N.; Florez, H.; Haas, L.B.; Halter, J.B.; Huang, E.S.; Korytkowski, M.T.; Munshi, M.N.; Odegard, P.S.; et al. Diabetes in Older Adults. Diabetes Care 2012, 35, 2650–2664. [Google Scholar] [CrossRef]
  131. Sandhu, M.S.; Heald, A.H.; Gibson, J.M.; Cruickshank, J.K.; Dunger, D.B.; Wareham, N.J. Circulating concentrations of insulin-like growth factor-I and development of glucose intolerance: A prospective observational study. Lancet 2002, 359, 1740–1745. [Google Scholar] [CrossRef]
  132. Dunger, D.B.; Ong, K.K.; Sandhu, M.S. Serum insulin-like growth factor-I levels and potential risk of type 2 diabetes. Horm. Res. 2003, 60 (Suppl. 3), 131–135. [Google Scholar] [CrossRef] [PubMed]
  133. Irie, F.; Fitzpatrick, A.L.; Lopez, O.L.; Kuller, L.H.; Peila, R.; Newman, A.B.; Launer, L.J. Enhanced risk for Alzheimer disease in persons with type 2 diabetes and APOE epsilon4: The Cardiovascular Health Study Cognition Study. Arch. Neurol. 2008, 65, 89–93. [Google Scholar] [CrossRef] [PubMed]
  134. 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. [Google Scholar] [CrossRef] [PubMed]
  135. Li, W.; Risacher, S.L.; Huang, E.; Saykin, A.J.; Alzheimer’s Disease Neuroimaging, I. Type 2 diabetes mellitus is associated with brain atrophy and hypometabolism in the ADNI cohort. Neurology 2016, 87, 595–600. [Google Scholar] [CrossRef] [PubMed]
  136. Macauley, S.L.; Stanley, M.; Caesar, E.E.; Yamada, S.A.; Raichle, M.E.; Perez, R.; Mahan, T.E.; Sutphen, C.L.; Holtzman, D.M. Hyperglycemia modulates extracellular amyloid-β concentrations and neuronal activity in vivo. J. Clin. Investig. 2015, 125, 2463–2467. [Google Scholar] [CrossRef]
  137. Janson, J.; Laedtke, T.; Parisi, J.E.; O’Brien, P.; Petersen, R.C.; Butler, P.C. Increased risk of type 2 diabetes in Alzheimer disease. Diabetes 2004, 53, 474–481. [Google Scholar] [CrossRef]
  138. Ma, Q.L.; Yang, F.; Rosario, E.R.; Ubeda, O.J.; Beech, W.; Gant, D.J.; Chen, P.P.; Hudspeth, B.; Chen, C.; Zhao, Y.; et al. β-Amyloid Oligomers Induce Phosphorylation of Tau and Inactivation of Insulin Receptor Substrate via c-Jun N-Terminal Kinase Signaling: Suppression by Omega-3 Fatty Acids and Curcumin. J. Neurosci. 2009, 29, 9078–9089. [Google Scholar] [CrossRef]
  139. de la Monte, S.M.; Tong, M.; Daiello, L.A.; Ott, B.R. Early-Stage Alzheimer’s Disease Is Associated with Simultaneous Systemic and Central Nervous System Dysregulation of Insulin-Linked Metabolic Pathways. J. Alzheimer’s Dis. 2019, 68, 657–668. [Google Scholar] [CrossRef]
  140. Alafuzoff, I.; Aho, L.; Helisalmi, S.; Mannermaa, A.; Soininen, H. Β-amyloid deposition in brains of subjects with diabetes. Neuropathol. Appl. Neurobiol. 2009, 35, 60–68. [Google Scholar] [CrossRef]
  141. Lee, C.W.; Shih, Y.H.; Wu, S.Y.; Yang, T.; Lin, C.; Kuo, Y.M. Hypoglycemia induces tau hyperphosphorylation. Curr. Alzheimer Res. 2013, 10, 298–308. [Google Scholar] [CrossRef] [PubMed]
  142. Clarke, J.R.; NM, L.E.S.; Figueiredo, C.P.; Frozza, R.L.; Ledo, J.H.; Beckman, D.; Katashima, C.K.; Razolli, D.; Carvalho, B.M.; Frazao, R.; et al. Alzheimer-associated Aβ oligomers impact the central nervous system to induce peripheral metabolic deregulation. EMBO Mol. Med. 2015, 7, 190–210. [Google Scholar] [CrossRef] [PubMed]
  143. Goldwaser, E.L.; Acharya, N.K.; Sarkar, A.; Godsey, G.; Nagele, R.G. Breakdown of the Cerebrovasculature and Blood-Brain Barrier: A Mechanistic Link Between Diabetes Mellitus and Alzheimer’s Disease. J. Alzheimer’s Dis. 2016, 54, 445–456. [Google Scholar] [CrossRef]
  144. Arvanitakis, Z.; Wang, H.Y.; Capuano, A.W.; Khan, A.; Taïb, B.; Anokye-Danso, F.; Schneider, J.A.; Bennett, D.A.; Ahima, R.S.; Arnold, S.E. Brain Insulin Signaling, Alzheimer Disease Pathology, and Cognitive Function. Ann. Neurol. 2020, 88, 513–525. [Google Scholar] [CrossRef] [PubMed]
  145. Kodl, C.T.; Seaquist, E.R. Cognitive dysfunction and diabetes mellitus. Endocr. Rev. 2008, 29, 494–511. [Google Scholar] [CrossRef] [PubMed]
  146. Muzumdar, R.H.; Ma, X.; Fishman, S.; Yang, X.; Atzmon, G.; Vuguin, P.; Einstein, F.H.; Hwang, D.; Cohen, P.; Barzilai, N. Central and opposing effects of IGF-I and IGF-binding protein-3 on systemic insulin action. Diabetes 2006, 55, 2788–2796. [Google Scholar] [CrossRef]
  147. Baumgart, M.; Snyder, H.M.; Carrillo, M.C.; Fazio, S.; Kim, H.; Johns, H. Summary of the evidence on modifiable risk factors for cognitive decline and dementia: A population-based perspective. Alzheimer’s Dement. 2015, 11, 718–726. [Google Scholar] [CrossRef]
  148. Grapsa, I.; Mamalaki, E.; Ntanasi, E.; Kosmidis, M.H.; Dardiotis, E.; Hadjigeorgiou, G.M.; Sakka, P.; Scarmeas, N.; Yannakoulia, M. Longitudinal Examination of Body Mass Index and Cognitive Function in Older Adults: The HELIAD Study. Nutrients 2023, 15, 1795. [Google Scholar] [CrossRef]
  149. Kim, K.Y.; Ha, J.; Lee, J.Y.; Kim, E. Weight loss and risk of dementia in individuals with versus without obesity. Alzheimer’s Dement. 2023. [Google Scholar] [CrossRef]
  150. Tang, X.; Zhao, W.; Lu, M.; Zhang, X.; Zhang, P.; Xin, Z.; Sun, R.; Tian, W.; Cardoso, M.A.; Yang, J.; et al. Relationship between Central Obesity and the incidence of Cognitive Impairment and Dementia from Cohort Studies Involving 5,060,687 Participants. Neurosci. Biobehav. Rev. 2021, 130, 301–313. [Google Scholar] [CrossRef]
  151. Hoscheidt, S.; Sanderlin, A.H.; Baker, L.D.; Jung, Y.; Lockhart, S.; Kellar, D.; Whitlow, C.T.; Hanson, A.J.; Friedman, S.; Register, T.; et al. Mediterranean and Western diet effects on Alzheimer’s disease biomarkers, cerebral perfusion, and cognition in mid-life: A randomized trial. Alzheimer’s Dement. 2022, 18, 457–468. [Google Scholar] [CrossRef] [PubMed]
  152. Yusufov, M.; Weyandt, L.L.; Piryatinsky, I. Alzheimer’s disease and diet: A systematic review. Int. J. Neurosci. 2017, 127, 161–175. [Google Scholar] [CrossRef] [PubMed]
  153. Więckowska-Gacek, A.; Mietelska-Porowska, A.; Wydrych, M.; Wojda, U. Western diet as a trigger of Alzheimer’s disease: From metabolic syndrome and systemic inflammation to neuroinflammation and neurodegeneration. Ageing Res. Rev. 2021, 70, 101397. [Google Scholar] [CrossRef] [PubMed]
  154. Venters, H.D.; Tang, Q.; Liu, Q.; VanHoy, R.W.; Dantzer, R.; Kelley, K.W. A new mechanism of neurodegeneration: A proinflammatory cytokine inhibits receptor signaling by a survival peptide. Proc. Natl. Acad. Sci. USA 1999, 96, 9879–9884. [Google Scholar] [CrossRef] [PubMed]
  155. Cotman, C.W.; Berchtold, N.C. Exercise: A behavioral intervention to enhance brain health and plasticity. Trends Neurosci. 2002, 25, 295–301. [Google Scholar] [CrossRef]
  156. Forbes, D.; Thiessen, E.J.; Blake, C.M.; Forbes, S.C.; Forbes, S. Exercise programs for people with dementia. Cochrane. Database Syst. Rev. 2013, 12, CD006489. [Google Scholar]
  157. Wolf, S.A.; Kronenberg, G.; Lehmann, K.; Blankenship, A.; Overall, R.; Staufenbiel, M.; Kempermann, G. Cognitive and physical activity differently modulate disease progression in the amyloid precursor protein (APP)-23 model of Alzheimer’s disease. Biol. Psychiatry 2006, 60, 1314–1323. [Google Scholar] [CrossRef]
  158. Carro, E.; Trejo, J.L.; Busiguina, S.; Torres-Aleman, I. Circulating insulin-like growth factor I mediates the protective effects of physical exercise against brain insults of different etiology and anatomy. J. Neurosci. 2001, 21, 5678–5684. [Google Scholar] [CrossRef]
  159. Trejo, J.L.; Carro, E.; Nunez, A.; Torres-Aleman, I. Sedentary life impairs self-reparative processes in the brain: The role of serum insulin-like growth factor-I. Rev. Neurosci. 2002, 13, 365–374. [Google Scholar] [CrossRef]
  160. Del Ser, T.; Hachinski, V.; Merskey, H.; Munoz, D.G. An autopsy-verified study of the effect of education on degenerative dementia. Brain 1999, 122, 2309–2319. [Google Scholar] [CrossRef]
  161. Stern, Y.; Albert, S.; Tang, M.X.; Tsai, W.Y. Rate of memory decline in AD is related to education and occupation: Cognitive reserve? Neurology 1999, 53, 1942–1947. [Google Scholar] [CrossRef]
  162. Stern, Y.; Barulli, D. Cognitive reserve. Handb. Clin. Neurol. 2019, 167, 181–190. [Google Scholar] [PubMed]
  163. Ngandu, T.; von Strauss, E.; Helkala, E.L.; Winblad, B.; Nissinen, A.; Tuomilehto, J.; Soininen, H.; Kivipelto, M. Education and dementia: What lies behind the association? Neurology 2007, 69, 1442–1450. [Google Scholar] [CrossRef]
  164. Walker, J.M.; Dehkordi, S.K.; Schaffert, J.; Goette, W.; White Iii, C.L.; Richardson, T.E.; Zare, H. The Spectrum of Alzheimer-Type Pathology in Cognitively Normal Individuals. J. Alzheimer’s Dis. 2023, 91, 683–695. [Google Scholar] [CrossRef] [PubMed]
  165. Seyedsalehi, A.; Warrier, V.; Bethlehem, R.A.I.; Perry, B.I.; Burgess, S.; Murray, G.K. Educational attainment, structural brain reserve and Alzheimer’s disease: A Mendelian randomization analysis. Brain 2023, 146, 2059–2074. [Google Scholar] [CrossRef]
  166. Hu, Y.; Zhang, Y.; Zhang, H.; Gao, S.; Wang, L.; Wang, T.; Han, Z.; Sun, B.L.; Liu, G. Cognitive performance protects against Alzheimer’s disease independently of educational attainment and intelligence. Mol. Psychiatry 2022, 27, 4297–4306. [Google Scholar] [CrossRef] [PubMed]
  167. Beck, K.D.; Powell-Braxton, L.; Widmer, H.R.; Valverde, J.; Hefti, F. Igf1 gene disruption results in reduced brain size, CNS hypomyelination, and loss of hippocampal granule and striatal parvalbumin-containing neurons. Neuron 1995, 14, 717–730. [Google Scholar] [CrossRef]
  168. Tu, X.; Jain, A.; Parra Bueno, P.; Decker, H.; Liu, X.; Yasuda, R. Local autocrine plasticity signaling in single dendritic spines by insulin-like growth factors. Sci. Adv. 2023, 9, eadg0666. [Google Scholar] [CrossRef]
  169. Katan, M.; Luft, A. Global Burden of Stroke. Semin. Neurol. 2018, 38, 208–211. [Google Scholar] [CrossRef]
  170. Honig, L.S.; Tang, M.X.; Albert, S.; Costa, R.; Luchsinger, J.; Manly, J.; Stern, Y.; Mayeux, R. Stroke and the risk of Alzheimer disease. Arch. Neurol. 2003, 60, 1707–1712. [Google Scholar] [CrossRef]
  171. Arbel-Ornath, M.; Hudry, E.; Eikermann-Haerter, K.; Hou, S.; Gregory, J.L.; Zhao, L.; Betensky, R.A.; Frosch, M.P.; Greenberg, S.M.; Bacskai, B.J. Interstitial fluid drainage is impaired in ischemic stroke and Alzheimer’s disease mouse models. Acta Neuropathol. 2013, 126, 353–364. [Google Scholar] [CrossRef] [PubMed]
  172. Ghiso, J.; Frangione, B. Cerebral amyloidosis, amyloid angiopathy, and their relationship to stroke and dementia. J. Alzheimer’s Dis. 2001, 3, 65–73. [Google Scholar] [CrossRef] [PubMed]
  173. Sun, X.; He, G.; Qing, H.; Zhou, W.; Dobie, F.; Cai, F.; Staufenbiel, M.; Huang, L.E.; Song, W. Hypoxia facilitates Alzheimer’s disease pathogenesis by up-regulating BACE1 gene expression. Proc. Natl. Acad. Sci. USA 2006, 103, 18727–18732. [Google Scholar] [CrossRef] [PubMed]
  174. Yu, S.P.; Jiang, M.Q.; Shim, S.S.; Pourkhodadad, S.; Wei, L. Extrasynaptic NMDA receptors in acute and chronic excitotoxicity: Implications for preventive treatments of ischemic stroke and late-onset Alzheimer’s disease. Mol. Neurodegener. 2023, 18, 43. [Google Scholar] [CrossRef] [PubMed]
  175. Hayes, C.A.; Valcarcel-Ares, M.N.; Ashpole, N.M. Preclinical and clinical evidence of IGF-1 as a prognostic marker and acute intervention with ischemic stroke. J. Cereb. Blood Flow. Metab. 2021, 41, 2475–2491. [Google Scholar] [CrossRef] [PubMed]
  176. Lopez-Lopez, C.; Dietrich, M.O.; Metzger, F.; Loetscher, H.; Torres-Aleman, I. Disturbed cross talk between insulin-like growth factor I and AMP-activated protein kinase as a possible cause of vascular dysfunction in the amyloid precursor protein/presenilin 2 mouse model of Alzheimer’s disease. J. Neurosci. 2007, 27, 824–831. [Google Scholar] [CrossRef]
  177. Friedrich, N.; Thuesen, B.; Jørgensen, T.; Juul, A.; Spielhagen, C.; Wallaschofksi, H.; Linneberg, A. The association between IGF-I and insulin resistance: A general population study in Danish adults. Diabetes Care 2012, 35, 768–773. [Google Scholar] [CrossRef]
  178. Zhou, M.; Li, H.; Wang, Y.; Pan, Y.; Wang, Y. Causal effect of insulin resistance on small vessel stroke and Alzheimer’s disease: A Mendelian randomization analysis. Eur. J. Neurol. 2022, 29, 698–706. [Google Scholar] [CrossRef]
  179. Shalev, A.; Liberzon, I.; Marmar, C. Post-Traumatic Stress Disorder. N. Engl. J. Med. 2017, 376, 2459–2469. [Google Scholar] [CrossRef]
  180. Greenberg, M.S.; Tanev, K.; Marin, M.F.; Pitman, R.K. Stress, PTSD, and dementia. Alzheimer’s Dement. 2014, 10 (Suppl. 3), S155–S165. [Google Scholar] [CrossRef]
  181. Kuring, J.K.; Mathias, J.L.; Ward, L. Risk of Dementia in persons who have previously experienced clinically-significant Depression, Anxiety, or PTSD: A Systematic Review and Meta-Analysis. J. Affect. Disord. 2020, 274, 247–261. [Google Scholar] [CrossRef] [PubMed]
  182. Catania, C.; Sotiropoulos, I.; Silva, R.; Onofri, C.; Breen, K.C.; Sousa, N.; Almeida, O.F.X. The amyloidogenic potential and behavioral correlates of stress. Mol. Psychiatry 2007, 14, 95–105. [Google Scholar] [CrossRef] [PubMed]
  183. Wilson, R.S.; Evans, D.A.; Bienias, J.L.; Mendes De Leon, C.F.; Schneider, J.A.; Bennett, D.A. Proneness to psychological distress is associated with risk of Alzheimer’s disease. Neurology 2003, 61, 1479–1485. [Google Scholar] [CrossRef] [PubMed]
  184. Delic, V.; Ratliff, W.A.; Citron, B.A. Sleep Deprivation, a Link Between Post-Traumatic Stress Disorder and Alzheimer’s Disease. J. Alzheimer’s Dis. 2021, 79, 1443–1449. [Google Scholar] [CrossRef]
  185. Weiner, M.W.; Harvey, D.; Landau, S.M.; Veitch, D.P.; Neylan, T.C.; Grafman, J.H.; Aisen, P.S.; Petersen, R.C.; Jack, C.R., Jr.; Tosun, D.; et al. Traumatic brain injury and post-traumatic stress disorder are not associated with Alzheimer’s disease pathology measured with biomarkers. Alzheimer’s Dement. 2022, 19, 884–895. [Google Scholar] [CrossRef]
  186. Justice, N.J.; Huang, L.; Tian, J.B.; Cole, A.; Pruski, M.; Hunt, A.J.; Flores, R.; Zhu, M.X.; Arenkiel, B.R.; Zheng, H. Posttraumatic Stress Disorder-Like Induction Elevates β-Amyloid Levels, Which Directly Activates Corticotropin-Releasing Factor Neurons to Exacerbate Stress Responses. J. Neurosci. 2015, 35, 2612–2623. [Google Scholar] [CrossRef]
  187. Raber, J.; Huang, Y.; Ashford, J.W. ApoE genotype accounts for the vast majority of AD risk and AD pathology. Neurobiol. Aging 2004, 25, 641–650. [Google Scholar] [CrossRef]
  188. Ferguson, A.C.; Tank, R.; Lyall, L.M.; Ward, J.; Celis-Morales, C.; Strawbridge, R.; Ho, F.; Whelan, C.D.; Gill, J.; Welsh, P.; et al. Alzheimer’s Disease Susceptibility Gene Apolipoprotein E (APOE) and Blood Biomarkers in UK Biobank (N = 395,769). J. Alzheimer’s Dis. 2020, 76, 1541–1551. [Google Scholar] [CrossRef]
  189. Li, T.; Pappas, C.; Klinedinst, B.; Pollpeter, A.; Larsen, B.; Hoth, N.; Anton, F.; Wang, Q.; Willette, A.A. Associations Between Insulin-Like Growth Factor-1 and Resting-State Functional Connectivity in Cognitively Unimpaired Midlife Adults. J. Alzheimer’s Dis. 2023, 94, S309–S318. [Google Scholar] [CrossRef]
  190. Wang, W.; Yu, J.T.; Tan, L.; Liu, Q.Y.; Wang, H.F.; Ma, X.Y. Insulin-like growth factor 1 (IGF1) polymorphism is associated with Alzheimer’s disease in Han Chinese. Neurosci. Lett. 2012, 531, 20–23. [Google Scholar] [CrossRef]
  191. Traversy, M.T.; Vandal, M.; Tremblay, C.; Tournissac, M.; Giguère-Rancourt, A.; Bennett, A.D.; Calon, F. Altered cerebral insulin response in transgenic mice expressing the epsilon-4 allele of the human apolipoprotein E gene. Psychoneuroendocrinology 2017, 77, 203–210. [Google Scholar] [CrossRef] [PubMed]
  192. Keeney, J.T.; Ibrahimi, S.; Zhao, L. Human ApoE Isoforms Differentially Modulate Glucose and Amyloid Metabolic Pathways in Female Brain: Evidence of the Mechanism of Neuroprotection by ApoE2 and Implications for Alzheimer’s Disease Prevention and Early Intervention. J. Alzheimer’s Dis. 2015, 48, 411–424. [Google Scholar] [CrossRef] [PubMed]
  193. Gu, D.; Ou, S.; Liu, G. Traumatic Brain Injury and Risk of Dementia and Alzheimer’s Disease: A Systematic Review and Meta-Analysis. Neuroepidemiology 2022, 56, 4–16. [Google Scholar] [CrossRef] [PubMed]
  194. Dams-O’Connor, K.; Guetta, G.; Hahn-Ketter, A.E.; Fedor, A. Traumatic brain injury as a risk factor for Alzheimer’s disease: Current knowledge and future directions. Neurodegener. Dis. Manag. 2016, 6, 417–429. [Google Scholar] [CrossRef]
  195. Johnson, V.E.; Stewart, W.; Smith, D.H. Traumatic brain injury and amyloid-β pathology: A link to Alzheimer’s disease? Nat. Rev. Neurosci. 2010, 11, 361–370. [Google Scholar] [CrossRef]
  196. Mohamed, A.Z.; Nestor, P.J.; Cumming, P.; Nasrallah, F.A. Traumatic brain injury fast-forwards Alzheimer’s pathology: Evidence from amyloid positron emission tomorgraphy imaging. J. Neurol. 2022, 269, 873–884. [Google Scholar] [CrossRef]
  197. Zheng, P.; Tong, W. IGF-1: An endogenous link between traumatic brain injury and Alzheimer disease? J. Neurosurg. Sci. 2017, 61, 416–421. [Google Scholar] [CrossRef] [PubMed]
  198. Ramos-Cejudo, J.; Wisniewski, T.; Marmar, C.; Zetterberg, H.; Blennow, K.; de Leon, M.J.; Fossati, S. Traumatic Brain Injury and Alzheimer’s Disease: The Cerebrovascular Link. EBioMedicine 2018, 28, 21–30. [Google Scholar] [CrossRef]
  199. Brett, B.L.; Gardner, R.C.; Godbout, J.; Dams-O’Connor, K.; Keene, C.D. Traumatic Brain Injury and Risk of Neurodegenerative Disorder. Biol. Psychiatry 2022, 91, 498–507. [Google Scholar] [CrossRef]
  200. Kempuraj, D.; Ahmed, M.E.; Selvakumar, G.P.; Thangavel, R.; Dhaliwal, A.S.; Dubova, I.; Mentor, S.; Premkumar, K.; Saeed, D.; Zahoor, H.; et al. Brain Injury-Mediated Neuroinflammatory Response and Alzheimer’s Disease. Neuroscientist 2020, 26, 134–155. [Google Scholar] [CrossRef]
  201. Shin, M.K.; Vázquez-Rosa, E.; Koh, Y.; Dhar, M.; Chaubey, K.; Cintrón-Pérez, C.J.; Barker, S.; Miller, E.; Franke, K.; Noterman, M.F.; et al. Reducing acetylated tau is neuroprotective in brain injury. Cell 2021, 184, 2715–2732.e23. [Google Scholar] [CrossRef] [PubMed]
  202. Gao, F.; Hu, M.; Zhang, J.; Hashem, J.; Chen, C. TDP-43 drives synaptic and cognitive deterioration following traumatic brain injury. Acta Neuropathol. 2022, 144, 187–210. [Google Scholar] [CrossRef] [PubMed]
  203. Hook, V.; Yoon, M.; Mosier, C.; Ito, G.; Podvin, S.; Head, B.P.; Rissman, R.; O’Donoghue, A.J.; Hook, G. Cathepsin B in neurodegeneration of Alzheimer’s disease, traumatic brain injury, and related brain disorders. Biochim. Biophys. Acta Proteins Proteom. 2020, 1868, 140428. [Google Scholar] [CrossRef]
  204. Wu, Z.; Wang, Z.H.; Liu, X.; Zhang, Z.; Gu, X.; Yu, S.P.; Keene, C.D.; Cheng, L.; Ye, K. Traumatic brain injury triggers APP and Tau cleavage by delta-secretase, mediating Alzheimer’s disease pathology. Prog. Neurobiol. 2020, 185, 101730. [Google Scholar] [CrossRef] [PubMed]
  205. Graham, N.S.N.; Jolly, A.; Zimmerman, K.; Bourke, N.J.; Scott, G.; Cole, J.H.; Schott, J.M.; Sharp, D.J. Diffuse axonal injury predicts neurodegeneration after moderate-severe traumatic brain injury. Brain 2020, 143, 3685–3698. [Google Scholar] [CrossRef]
  206. Brabant, G.; Wallaschofski, H. Normal levels of serum IGF-I: Determinants and validity of current reference ranges. Pituitary 2007, 10, 129–133. [Google Scholar] [CrossRef]
  207. Harris, T.G.; Strickler, H.D.; Yu, H.; Pollak, M.N.; Monrad, E.S.; Travin, M.I.; Xue, X.; Rohan, T.E.; Kaplan, R.C. Specimen processing time and measurement of total insulin-like growth factor-I (IGF-I), free IGF-I, and IGF binding protein-3 (IGFBP-3). Growth Horm. IGF Res. 2006, 16, 86–92. [Google Scholar] [CrossRef]
  208. Simstich, S.; Züllig, T.; D’Aurizio, F.; Biasotto, A.; Colao, A.; Isidori, A.M.; Lenzi, A.; Fauler, G.; Köfeler, H.C.; Curcio, F.; et al. The impact of different calibration matrices on the determination of insulin-like growth factor 1 by high-resolution-LC-MS in acromegalic and growth hormone deficient patients. Clin. Biochem. 2023, 114, 95–102. [Google Scholar] [CrossRef]
  209. Frystyk, J. Utility of Free IGF-I Measurements. Pituitary 2007, 10, 181–187. [Google Scholar] [CrossRef]
  210. Galle, S.A.; van der Spek, A.; Drent, M.L.; Brugts, M.P.; Scherder, E.J.A.; Janssen, J.; Ikram, M.A.; van Duijn, C.M. Revisiting the Role of Insulin-Like Growth Factor-I Receptor Stimulating Activity and the Apolipoprotein E in Alzheimer’s Disease. Front. Aging Neurosci. 2019, 11, 20. [Google Scholar] [CrossRef]
  211. Miki Stein, A.; Munive, V.; Fernandez, A.M.; Nuñez, A.; Torres Aleman, I. Acute exercise does not modify brain activity and memory performance in APP/PS1 mice. PLoS ONE 2017, 12, e0178247. [Google Scholar] [CrossRef] [PubMed]
  212. Trueba-Saiz, A.; Cavada, C.; Fernandez, A.M.; Leon, T.; Gonzalez, D.A.; Fortea, O.J.; Lleo, A.; Del, S.T.; Nunez, A.; Torres-Aleman, I. Loss of serum IGF-I input to the brain as an early biomarker of disease onset in Alzheimer mice. Transl. Psychiatry 2013, 3, e330. [Google Scholar] [CrossRef] [PubMed]
  213. Rollo, J.; Crawford, J.; Hardy, J. A dynamical systems approach for multiscale synthesis of Alzheimer’s pathogenesis. Neuron 2023, 111, 2126–2139. [Google Scholar] [CrossRef] [PubMed]
  214. Frere, S.; Slutsky, I. Alzheimer’s Disease: From Firing Instability to Homeostasis Network Collapse. Neuron 2018, 97, 32–58. [Google Scholar] [CrossRef]
Figure 1. Preservation of brain IGF-I activity through behavior. There are four main behavioral approaches to preserving brain IGF-I activity. Three of them, balanced emotional life and active physical and mental life, are already well established to reduce sAD risk. Reduction in stress associated with current lifestyle through different approaches (i.e., meditation), regular (moderate) exercise, and engagement in cognitively demanding tasks, including social intercourse, are becoming common knowledge in the prevention of sAD. In the case of balanced diets, numerous studies have not yet reached a firm consensus for any one in particular, although the Mediterranean diet is probably the most favored at present [50]. Modified from [16].
Figure 1. Preservation of brain IGF-I activity through behavior. There are four main behavioral approaches to preserving brain IGF-I activity. Three of them, balanced emotional life and active physical and mental life, are already well established to reduce sAD risk. Reduction in stress associated with current lifestyle through different approaches (i.e., meditation), regular (moderate) exercise, and engagement in cognitively demanding tasks, including social intercourse, are becoming common knowledge in the prevention of sAD. In the case of balanced diets, numerous studies have not yet reached a firm consensus for any one in particular, although the Mediterranean diet is probably the most favored at present [50]. Modified from [16].
Ijms 24 16440 g001
Figure 2. Indirect determination of IGF-I activity in the brain using electro-encephalographic (EEG) responses to exercise as a surrogate. The proposed test is based on three observations in mice: (A) Exercise induces the entrance of circulating IGF-I into the brain [106]. (B) Circulating IGF-I modulates EEG patterns [212]. (C) Absence of exercise-induced changes in EEG patterns in mildly cognitively impaired APP/PS1 mice [211]. Right panel: the proposed test for measuring brain IGF-I activity in humans consists of EEG recordings at rest, followed by a bout of moderate exercise and subsequent EEG recordings. The prediction is that subjects showing no EEG changes after exercise are at risk of developing cognitive disturbances. The purported explanation is that IGF-I activity is lower in the brains of these subjects.
Figure 2. Indirect determination of IGF-I activity in the brain using electro-encephalographic (EEG) responses to exercise as a surrogate. The proposed test is based on three observations in mice: (A) Exercise induces the entrance of circulating IGF-I into the brain [106]. (B) Circulating IGF-I modulates EEG patterns [212]. (C) Absence of exercise-induced changes in EEG patterns in mildly cognitively impaired APP/PS1 mice [211]. Right panel: the proposed test for measuring brain IGF-I activity in humans consists of EEG recordings at rest, followed by a bout of moderate exercise and subsequent EEG recordings. The prediction is that subjects showing no EEG changes after exercise are at risk of developing cognitive disturbances. The purported explanation is that IGF-I activity is lower in the brains of these subjects.
Ijms 24 16440 g002
Table 1. Reported IGF-I activities related to mechanisms known to be altered in AD.
Table 1. Reported IGF-I activities related to mechanisms known to be altered in AD.
ActivityMain References
Neurogenesis[22,34]
Re-innervation[23]
Regulation of inflammation[24,35]
Regulation of oxidative stress[25,36]
Neuronal plasticity[28,37]
Cognition[29]
Mood[30]
Energy allocation[26,31,38]
Sleep/wake cycle[32]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Zegarra-Valdivia, J.A.; Pignatelli, J.; Nuñez, A.; Torres Aleman, I. The Role of Insulin-like Growth Factor I in Mechanisms of Resilience and Vulnerability to Sporadic Alzheimer’s Disease. Int. J. Mol. Sci. 2023, 24, 16440. https://doi.org/10.3390/ijms242216440

AMA Style

Zegarra-Valdivia JA, Pignatelli J, Nuñez A, Torres Aleman I. The Role of Insulin-like Growth Factor I in Mechanisms of Resilience and Vulnerability to Sporadic Alzheimer’s Disease. International Journal of Molecular Sciences. 2023; 24(22):16440. https://doi.org/10.3390/ijms242216440

Chicago/Turabian Style

Zegarra-Valdivia, Jonathan A., Jaime Pignatelli, Angel Nuñez, and Ignacio Torres Aleman. 2023. "The Role of Insulin-like Growth Factor I in Mechanisms of Resilience and Vulnerability to Sporadic Alzheimer’s Disease" International Journal of Molecular Sciences 24, no. 22: 16440. https://doi.org/10.3390/ijms242216440

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop