Insulin-like Growth Factor I in Sporadic Alzheimer’s Disease: History
Please note this is an old version of this entry, which may differ significantly from the current revision.

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. Insulin-like growth factor I (IGF-I) activity in the brain provides a common substrate for the mechanisms of resilience and vulnerability to AD. 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. 

  • Alzheimer’s disease
  • insulin-like growth factor I
  • AD

1. Introduction

Mutations in amyloid precursor protein (APP) and preselinins are associated with familial Alzheimer’s disease (fAD), which constitutes around 1% of Alzheimer’s disease (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.

2. Insulin-Like Growth Factor I (IGF-I) and AD Resilience

The concept of AD resilience has been coined to explain the presence of AD pathology in cognitively intact individuals [14]. 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 [15], and this is not surprising considering the heavy genetic make-up of AD risk [16]. This genetic component may help uncover novel targets of resilience, such as the recently reported reelin, a protein functionally related to ApoE [17], 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 [18], aerobic glycolysis [19], efficient microglial phagocytosis [20], and dendritic spine plasticity [21] 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 [22], 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 [23][24].

Mechanisms of IGF-I-Dependent AD Resilience

Potentiation of neurotrophic activity, most often BDNF [25], has already been invoked as a mechanism of AD resilience [26], but specific mechanisms and factors need to be defined. Since the neuroprotective actions of IGF-I are pleiotropic [22][27], 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 [28]. 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 [29]. 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, scholars first focus on cell-based processes that affect all types of brain cells [30]. Among them, synapse loss is considered a major structural disturbance associated with cognitive deterioration in sAD [31]. Thus, IGF-I is involved in physiological synaptogenesis during development [32], in adult brains [33], and in synapse repletion after an insult [34]. Importantly, dendritic spines, a major site of cortical synapses, provide AD resilience [21], while IGF-I promotes dendritogenesis [35] and is intricately involved in synaptic physiology [36][37].
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 [38][39]. 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 [40][41], leading to the alteration of structural and functional brain networks seen in AD, as recently reported [42]. This “double-edge sword” process [43] is also modulated by IGF-I acting through a calcineurin-NFκB pathway in astrocytes that reversibly drives AD pathology in AD mice [44]. Naturally, neuro-inflammation also impacts on many other cellular activities, such as astrocyte phagocytosis [45], microglial reactivity [46] and proliferation [47], and activity of brain resident macrophages [48], and it also interacts with the brain angiotensin anti-inflammatory pathway [49][50]. 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 [49].
Other cell-associated processes in AD pathology, such as excess oxidative stress [51], which is probably directly involved in AD-related cell demise [52], are also counteracted by IGF-I [53]. Since an efficient mechanism of prevention of oxidative stress has been suggested to work in the brain of individuals showing AD resilience [54], 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 [55].
At the system level, dysregulated neural circuit activity [56][57] and an altered astrocytic network [58][59], or both disturbances interacting with each other [60][61], 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 [62][63], tau accumulation [64], or early loss of inhibitory tone [65], 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 [36] and excitatory/inhibitory balance [66][67], which also includes its actions through astrocytes [68], a type of glial cell known to modulate neuronal circuits.

This entry is adapted from the peer-reviewed paper 10.3390/ijms242216440

References

  1. Mayeux, R. Epidemiology of neurodegeneration. Annu. Rev. Neurosci. 2003, 26, 81–104.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  10. Arenaza-Urquijo, E.M.; Vemuri, P. Resistance vs resilience to Alzheimer disease: Clarifying terminology for preclinical studies. Neurology 2018, 90, 695–703.
  11. Blennow, K.; de Leon, M.J.; Zetterberg, H. Alzheimer’s disease. Lancet 2006, 368, 387–403.
  12. Kosik, K.S. PErsonalized medicine for effective alzheimer disease treatment. JAMA Neurol. 2015, 72, 497–498.
  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.
  14. Gómez-Isla, T.; Frosch, M.P. Lesions without symptoms: Understanding resilience to Alzheimer disease neuropathological changes. Nat. Rev. Neurol. 2022, 18, 323–332.
  15. 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.
  16. 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.
  17. 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.
  18. 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.
  19. 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.
  20. 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.
  21. 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.
  22. 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.
  23. 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.
  24. 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.
  25. 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.
  26. 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.
  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.
  28. 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.
  29. 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.
  30. De Strooper, B.; Karran, E. The Cellular Phase of Alzheimer’s Disease. Cell 2016, 164, 603–615.
  31. 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.
  32. 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.
  33. 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.
  34. 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.
  35. 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.
  36. 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.
  37. 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.
  38. Patani, R.; Hardingham, G.E.; Liddelow, S.A. Functional roles of reactive astrocytes in neuroinflammation and neurodegeneration. Nat. Rev. Neurol. 2023, 19, 395–409.
  39. Aguzzi, A.; Barres, B.A.; Bennett, M.L. Microglia: Scapegoat, saboteur, or something else? Science 2013, 339, 156–161.
  40. Heppner, F.L.; Ransohoff, R.M.; Becher, B. Immune attack: The role of inflammation in Alzheimer disease. Nat. Rev. Neurosci. 2015, 16, 358–372.
  41. 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.
  42. 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.
  43. Wyss-Coray, T.; Mucke, L. Inflammation in neurodegenerative disease-a double-edged sword. Neuron 2002, 35, 419.
  44. 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.
  45. 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.
  46. 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.
  47. 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.
  48. 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.
  49. 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.
  50. 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.
  51. 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.
  52. Markesbery, W.R.; Carney, J.M. Oxidative alterations in Alzheimer’s disease. Brain Pathol. 1999, 9, 133–146.
  53. 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.
  54. 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.
  55. 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.
  56. 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.
  57. 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.
  58. 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.
  59. 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.
  60. 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.
  61. 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.
  62. 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.
  63. 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.
  64. 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.
  65. 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.
  66. 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.
  67. 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.
  68. 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.
More
This entry is offline, you can click here to edit this entry!
ScholarVision Creations