Actions of Insulin Expressed in the CNS: Comparison
Please note this is a comparison between Version 3 by Beatrix Zheng and Version 2 by Beatrix Zheng.

The expression of insulin receptors (IR) in the central nervous system (CNS) was first documented almost half a century ago. It is now known that both short (IR-A) and long (IR-B) isoform of this receptor is expressed in the hypothalamus, hippocampus, cerebral cortex, and cerebellum, the brain regions also associated with the production of insulin. In regard to the IR expression at the cellular level, it was shown that, unlike neurons that only express IR-A, astrocytes express both IR-A and IR-B. Ever since the transport of insulin across the BBB and the expression of functional IR in the CNS was documented, attempts have been made to learn more about the effects of insulin on the brain. In line with that, numerous actions of insulin in the CNS have been described so far. It has been known that insulin takes part in controlling food intake and body weight. In addition to that, insulin actions are also essential for proper neuronal development and survival, cognition, brain cholesterol synthesis, hepatic glucose production, lipolysis and lipogenesis, and even reproductive competence. Moreover, it was also shown that impairment in insulin signaling could trigger depression- and anxiety-like behaviors. Although new roles of insulin keep emerging, it is still mainly unknown which of them can be attributed to brain-derived and which to pancreatic insulin, or whether their actions in the CNS overlap and to what extent.

  • brain-derived insulin
  • hypothalamus
  • hippocampus
  • cerebellum
  • cerebral cortex

1. Brain-Derived Insulin Has Neuroprotective Effects

Some of the earliest studies concerning the role of insulin produced in neurons were performed on cultured cells. One of the first roles of neural insulin to be observed was that it promoted neurofilament distribution and axonal growth via mitogen-activated protein kinase (MAPK) phosphorylation in cultured rat fetal neurons [1][2]. Cultured neurons from fetal rat brains incubated in an insulin-free medium were first shown to contain Ins2 mRNA and preproinsulin, a biosynthetic precursor of insulin. Subsequently, neurofilament immunoreaction was detected in the somata, dendrites, and axons of these neurons. Lastly, the treatment with either insulin antibody or the inhibitor of insulin receptor tyrosine kinase activity was shown to result in neurite retraction rendering neurons hypertrophic and vacuolated. Inhibition of MAPK, but not phosphatidylinositol 3-kinase (PI3K), also resulted in shortening and/or complete retraction of neuritis and rounding of neurons, which suggested that insulin effects on neurofilament distribution to the axons were accomplished via MAPK activation.
A study with a similar design also showed that insulin of neural origin promoted neural differentiation and growth [3]. Moreover, it was demonstrated that in vivo interference with embryonic insulin signaling by blocking insulin receptors increased apoptosis during early neurulation and thus showed that proinsulin produced by neuroepithelial cells promoted cell survival during this stage of embryonic development [4]. In another study, proinsulin was shown to decrease the expression of neuroinflammation markers through the activation of protein kinase B (Akt) pathways and lower levels of tumor necrosis factor-α (TNF-α) and interleukin 1β in the mouse hippocampus [5]. Of note, these effects detected at the molecular level correlated with improved cognitive performance. Collectively, these findings indicate that centrally produced (pro)insulin exhibits neuroprotective and anti-inflammatory properties.

2. Fasting-Induced Increase in Hypothalamic Insulin Expression Appears Not to Be Associated with Facilitating Neuronal Glucose Uptake

The distinctiveness of mechanisms controlling peripheral and central insulin expression became evident in one of the researchers' studies which examined the effects of short-term fasting on insulin expression in the rat hypothalamus. The research showed that six-hour fasting increased both Ins2 mRNA expression and insulin content in the hypothalamic parenchyma, while the concentrations of circulating glucose and insulin were expectedly to decrease [6]. Furthermore, immunopositivity for preproinsulin was observed in the neurons of the hypothalamic periventricular nucleus (PEV) and the ependymal cells lining the roof of the third cerebral ventricle. These two cell types were already recognized as the sources of insulin within the central nervous system (CNS). The presence of Ins2 mRNA was first detected in the neurons of PEV almost four decades ago [7], while the epithelial cells, such as those of choroid plexus (EChP), were identified as the source of insulin more recently [8]. Namely, Mazucanti et al. [8] found that Ins2 mRNA, mature insulin, and C-peptide were all present in EChP. Furthermore, they demonstrated that insulin secretion from primary cultured mouse EChP was stimulated by serotonin (5-HT). More precisely, it was shown that activation of the 5HT2C receptor by serotonin treatment resulted in the opening of IP3-gated Ca2+ channels in the endoplasmic reticulum. This resulted in Ca2+ being mobilized from intracellular storage, which consequently led to insulin secretion. The role of serotonin in the secretion of insulin from EChP was indirectly corroborated in vivo by showing that serotonin depletion in the dorsal raphe nucleus downregulated insulin expression in ChP. The researchers' unpublished data showed that, such as insulin, serotonin content in the hypothalamus was also elevated following the six-hour fast. Additionally, hypothalamic insulin increment was observed following microdialysis administration of dexfenfluramine, a potent 5-HT stimulator [9]. This suggests that in addition to basal conditions, serotonin may take part in stimulating the secretion of brain insulin during metabolically challenging states such as fasting.
In regard to the ependymal cells in the researchers' study, strong granular proinsulin immunopositivity was observed in the apical portion of cuboidal ciliated cells in the region surrounding the lumen of the third ventricle. The intracellular location of proinsulin-containing granules implies that insulin produced in these cells was secreted into the CSF rather than being taken up from it. This conclusion was supported by the fact that, unlike circulating insulin which was lowered, the CSF insulin levels were at the control level despite the short-term food deprivation [6].
The researchers then sought to decipher the adaptive significance of the short-term fasting-induced increase in hypothalamic insulin expression. Considering that fasting represents metabolic strain for the organism as a whole, the researchers wanted to investigate whether this phenomenon was related to maintaining glucose homeostasis within the hypothalamus. It was found that short-term fasting markedly increased the amount of endothelial 55 kDa isoform of glucose transporter 1 (GLUT1) and neuronal GLUT3. The levels of GLUT2, whose presence was detected in neurons, ependymocytes, and tanycytes, were also elevated [10]. However, the absence of co-expression of these membrane transporters with the activated insulin receptor suggested that the actions of this locally produced insulin were unlikely associated with the regulation of glucose-facilitated diffusion in the hypothalamus during fasting.
A further attempt to determine the role of fasting-promoted increase in the hypothalamic insulin expression was made by looking into how short-term fasting affected signaling pathways typically activated by this hormone [11]. The researchers found that the hypothalamic content of total and activated insulin receptor substrates 1 and 2 (IRS1/2), PI3K, and the mammalian target of rapamycin (mTOR) was unaltered. However, the levels of phosphorylated Akt1/2/3 were decreased, unlike those of activated extracellular signal-regulated kinases (ERK1/2), which were increased. Moreover, activated ERK1/2 was co-expressed with activated insulin receptors in the nucleus arcuatus, which suggested that the ERK activation in the hypothalamus was at least partially initiated by the centrally produced insulin during short-term fasting.
Lastly, the researchers wanted to examine how fasting of the same duration would affect hypothalamic insulin expression in female rats during different phases of the estrus cycle [12]. Following the six-hour food deprivation, Ins2 mRNA expression and insulin content were not elevated as previously observed in male rats, but they were not reduced either. Both of these parameters remained unaltered in both proestrus and diestrus, despite circulating insulin being significantly lowered. Similar to findings observed in male rats, insulin immunopositivity was detected in the PEV neurons and the ependymal cells at the top of the third cerebral ventricle in females. When taken together, these data indicate that control of insulin expression in the hypothalamus during short-term fasting is a sex-specific process. However, hypothalamic insulin expression in both sexes appeared to be distinctly regulated during fasting than that in the pancreatic β cells.

3. Stimulation of Insulin-Producing Neurons in Dorsal Vagal Complex Results in Increased Appetite

Another recent study also shed light on the complexity of centrally-produced insulin expression patterns and actions [13]. First of all, this study showed that in addition to hypothalamic nuclei, such as nucleus arcuatus and paraventricular nucleus (PVN), the Ins2-promoter-containing cells were also found in the nucleus of the solitary tract and the dorsal vagal complex (DVC). Furthermore, this study showed that insulin expression in the brain was affected by dietary interventions in a region-specific manner. For example, eight-week exposure to a high-fat diet (HFD) decreased Ins2 mRNA expression in the hypothalamus while simultaneously increasing it in DVC. This finding suggests that the role of insulin produced in the hindbrain becomes more evident under metabolically stressful situations such as prolonged exposure to HFD. In contrast to the anorexic effects that insulin typically exhibits in the hypothalamus, this study revealed that stimulation of DVC insulin-producing neurons results in an acute increase in appetite. Namely, optogenetic stimulation of DVC insulin-producing neurons resulted in increased food intake, which peaked one hour after the onset of stimulation without affecting long-term appetite. Ventricular application of a highly specific insulin receptor antagonist blocked the aforementioned stimulating effects of DVC insulin-producing neurons on feeding.

4. Insulin Produced in the Paraventricular Nucleus Stimulates Growth Hormone Secretion

A major breakthrough in unveiling the physiological role of hypothalamus-derived insulin came from the study of Lee et al. [14]. Not only did the authors describe the properties of the hypothalamic insulin-producing neurons in mice, but they also showed that insulin synthesized in this region controls the secretion of one of the anterior pituitary hormones. In this study, Ins2 mRNA, proinsulin, and mature insulin were colocalized within the same subset of neurons in the PVN. Proinsulin was mainly found in the neuronal cell bodies. In addition to that, proinsulin was also detected in the ependymal cells lining the third ventricle and, to a lesser extent, in the ameboid microglia. Unlike proinsulin, C-peptide, which connects the A and B chains of proinsulin and is secreted together with insulin, was located in neurosecretory nerve terminals in the external zone of the median eminence (ME). This suggests that C-peptide and matured insulin are transported from the somata of PVN insulin-producing neurons through axonal projections to their axon terminals in the ME.
Most of the insulin-producing PVN neurons (90%) examined in this study were shown to also synthesize corticotropin-releasing hormone (CRH), while somatostatin was found in only about 20% of insulin-producing neurons. Moreover, CRH was extensively colocalized with C-peptide within the same large dense-core vesicles in the axon terminals in the ME. It is noteworthy that the expression patterns of insulin and CRH genes were reciprocal during the eight-hour exposure to restraint stress. While Crh mRNA expression proved to be biphasic, peaking half an hour and eight hours after the onset of restraint, hypothalamic Ins2 mRNA levels were continuously decreased during the exposure to restraint.
The proximity of PVN insulin-producing neuronal axon terminals to the ME led the authors of this study to examine whether PVN-derived insulin might be involved in the control of anterior pituitary hormone secretion. The only pituitary hormone affected by the knockdown of PVN insulin was growth hormone (GH). Namely, both pituitary Gh mRNA expression and the serum GH concentration were lowered after the insulin gene had been knocked down in the PVN. Considering that hypothalamic expression of growth hormone-releasing hormone (GHRH) and somatostatin was unaltered by PVN insulin knockdown, it was concluded that hypothalamic insulin-regulated GH secretion independently of these two hormones. Moreover, PVN insulin knockdown significantly suppressed the pituitary phosphorylation of Akt, the kinase known to be a major target of insulin signaling. Ultimately, the importance of PVN insulin in promoting GH secretion was confirmed in PVN insulin-knockdown young mice. Six weeks after hypothalamic Ins2 mRNA expression had been downregulated, a significant reduction in body length (without changes in food intake and body weight) of these mice was documented in comparison to the controls of the same age [14]. This finding is particularly relevant given that growth in children can be hampered by exposure to stressful stimuli [15].

5. In Utero Alcohol Exposure Decreases Insulin Expression in the Cerebellum and Thus Impairs Insulin-Mediated Neuronal Glucose Uptake

The significance of centrally produced insulin for normal brain development was examined in a study conducted by Monte et al. [16]. They studied the effect of in-utero alcohol exposure on insulin expression and signaling in the early postnatal rat cerebellum, one of the brain regions most susceptible to ethanol neurotoxicity. It was found that chronic exposure to ethanol during gestation caused cerebellar hypoplasia with loss of neurons due to increased apoptosis. On the molecular level, exposure to ethanol downregulated the expression of insulin mRNA both in the cerebellum as a whole and in the primary culture of granule neurons isolated from the cerebella of ethanol-exposed pups. In regard to its receptor, there were no significant differences in the levels of insulin receptor mRNA in either ethanol-exposed cerebella in general or neurons isolated from that brain region. However, the levels of the intrinsic insulin receptor tyrosine activity were reduced following the gestational ethanol exposure.
It had already been known from earlier studies that ethanol-induced inhibition of insulin-stimulated tyrosine phosphorylation of insulin receptor and IRS1 and the subsequent impairment of downstream PI3K/Akt signaling results in decreased neuronal survival [17]. Among other targets, Akt was shown to phosphorylate protein tyrosine phosphatase 1b (PTP1b) and thus decrease its ability to dephosphorylate insulin receptors [18]. Therefore, the reduction in the Akt-mediated negative regulation of PTP1b resulted in increased activity of this enzyme despite the fact that the levels of PTP1b mRNA levels were not changed within the cerebellum. Considering the aforementioned, decreased neuronal sensitivity to insulin following exposure to ethanol can be accounted for by PTP1b-mediated dephosphorylation of insulin receptor.
The implications of ethanol-induced decrease in insulin expression in the cerebellum were also analyzed within the context of glucose uptake and utilization. Chronic gestational exposure to ethanol reduced the levels of Glut4 mRNA [16] and the cerebellar amount of this glucose transporter, whose translocation to the cell membrane is known to be insulin-dependent [19]. Consequently, both basal and insulin-induced glucose uptake was decreased in neuronal cultures cultivated from ethanol-exposed cerebella. Reduced glucose uptake was reflected in the ATP levels being consistently lowered in these cells in comparison to the control values. These findings are in line with a previous finding that the expression of the insulin-responsive gene coding for glyceraldehydes-3- phosphate dehydrogenase (GAPDH) was significantly decreased [17]. Taken together, the data from various studies of gestational exposure to ethanol suggest that neuronal glucose uptake and utilization, and consequently neuronal survival in the developing cerebellum are, at least partially, mediated by insulin expressed within this brain region.

6. The Impairment in Insulin Expression and Signaling in the Brain Is Associated with Memory Decline

The effects of treatments mimicking the stress exposure on insulin and insulin receptor expression in the brain were described in the study of Osmanovic et al. [20]. The authors found that, following the chronic administration of exogenous corticosterone, the expressions of both insulin and insulin receptors were reduced in the cerebral cortex. However, the treatment which mimicked the exposure to chronic stress resulted in elevated tau protein mRNA expression within the same brain region. Of note, these molecular changes positively correlated with a decline in working and reference memory. All of these findings are in line with the fact that Alzheimer’s disease is often associated with insulin-deficient and/or insulin-resistant brain states [21][22]. Collectively, these data point to the contribution of brain-derived insulin in preserving cognition, the process which is also known to be compromised by impaired transport of pancreatic insulin across the BBB [23] and proper functionality of IR in the CNS [24].
As previously mentioned, studies have shown that insulin signaling impairment is associated with pathologies such as Alzheimer’s disease [25]. Moreover, insulin concentration is reduced in the CSF of patients with Alzheimer’s disease [26], while decreased insulin levels were found post-mortem in the brain parenchyma of patients who suffered from sporadic Alzheimer’s disease [27]. One of the first studies that provided insight into how hippocampal insulin expression and secretion was affected by amyloid-β1–42 (Aβ1–42) was that of Nemoto et al. [28]. They first reconfirmed that both Ins2 mRNA and proinsulin were present in the neurons of the hippocampus and cerebral cortex. They also established that insulin secretion from hippocampal neurons was regulated by Ca2+-dependent activator protein for secretion 2 (CAPS2). This means that, such as the myriad of other neuropeptides, insulin is secreted from neurons via exocytosis from dense-core vesicles. The treatment of hippocampal neurons with Aβ1–42 significantly decreased the amount of proinsulin detected in these cells. Moreover, the Aβ1–42 treatment decreased Ser9-phosphorylation of glycogen synthase kinase-3β (GSK-3β). Namely, the binding of insulin to its receptor activates PI3K/Akt cascade, which results in GSK-3β inactivation through Ser9-phosphorylation. When taken together, these data suggest that downregulation of hippocampal Ins2 expression in Aβ1–42-induced model of Alzheimer’s disease occurs via activation of GSK-3β. This conclusion was backed by the fact that the effect of Aβ1–42 treatment was reduced by both GSK-3β siRNA and lithium, a mood stabilizer is known to inhibit GSK-3β activity.
In addition to the hippocampus, the significance of insulin expression in the CNS in the context of Alzheimer’s disease was documented in the cerebral cortex as well [29]. This electrophysiological study revealed that adding glucose to the external solution of neocortical brain slices resulted in GABAergic NGFC reversibly decreasing the frequency and amplitude of spontaneous excitatory postsynaptic potentials (EPSP) in neighboring pyramidal neurons via a mechanism that included activation of the insulin receptor. The same effect was achieved by adding exogenous insulin to the brain slices, while the application of insulin receptor antagonists resulted in the prevention of these excitation-suppressing effects on local microcircuits detected after the addition of hyperglycemic solution. Considering that lower cerebral levels of insulin have been associated with neurodegenerative diseases such as Alzheimer’s [21], a potential adaptive significance of insulin expression in NGFC could be a modulation of neighboring neuronal circuits involved in learning and memory.

References

  1. Schechter, R.; Yanovitch, T.; Abboud, M.; Johnson, G.; Gaskins, J. Effects of Brain Endogenous Insulin on Neurofilament and MAPK in Fetal Rat Neuron Cell Cultures. Brain Res. 1998, 808, 270–278.
  2. Schechter, R.; Abboud, M.; Johnson, G. Brain Endogenous Insulin Effects on Neurite Growth within Fetal Rat Neuron Cell Cultures. Dev. Brain Res. 1999, 116, 159–167.
  3. Schechter, R.; Abboud, M. Neuronal Synthesized Insulin Roles on Neural Differentiation within Fetal Rat Neuron Cell Cultures. Dev. Brain Res. 2001, 127, 41–49.
  4. Hernández-Sánchez, C.; Rubio, E.; Serna, J.; de la Rosa, E.J.; de Pablo, F. Unprocessed Proinsulin Promotes Cell Survival during Neurulation in the Chick Embryo. Diabetes 2002, 51, 770–777.
  5. Corpas, R.; Hernández-Pinto, A.M.; Porquet, D.; Hernández-Sánchez, C.; Bosch, F.; Ortega-Aznar, A.; Comellas, F.; de la Rosa, E.J.; Sanfeliu, C. Proinsulin Protects against Age-Related Cognitive Loss through Anti-Inflammatory Convergent Pathways. Neuropharmacology 2017, 123, 221–232.
  6. Dakic, T.B.; Jevdjovic, T.V.; Peric, M.I.; Bjelobaba, I.M.; Markelic, M.B.; Milutinovic, B.S.; Lakic, I.V.; Jasnic, N.I.; Djordjevic, J.D.; Vujovic, P.Z. Short-Term Fasting Promotes Insulin Expression in Rat Hypothalamus. Eur. J. Neurosci. 2017, 46, 1730–1737.
  7. Young, W.S. Periventricular Hypothalamic Cells in the Rat Brain Contain Insulin MRNA. Neuropeptides 1986, 8, 93–97.
  8. Mazucanti, C.H.; Liu, Q.-R.; Lang, D.; Huang, N.; O’Connell, J.F.; Camandola, S.; Egan, J.M. Release of Insulin Produced by the Choroid Plexis Is Regulated by Serotonergic Signaling. JCI Insight 2019, 4, e131682.
  9. Orosco, M.; Rouch, C.; Gerozissis, K. Activation of Hypothalamic Insulin by Serotonin Is the Primary Event of the Insulin–Serotonin Interaction Involved in the Control of Feeding. Brain Res. 2000, 872, 64–70.
  10. Dakic, T.; Jevdjovic, T.; Lakic, I.; Djurasevic, S.F.; Djordjevic, J.; Vujovic, P. Food for Thought: Short-Term Fasting Upregulates Glucose Transporters in Neurons and Endothelial Cells, But Not in Astrocytes. Neurochem. Res. 2019, 44, 388–399.
  11. Dakic, T.; Jevdjovic, T.; Djordjevic, J.; Vujovic, P. Short-Term Fasting Differentially Regulates PI3K/AkT/MTOR and ERK Signalling in the Rat Hypothalamus. Mech. Ageing Dev. 2020, 192, 111358.
  12. Dakic, T.B.; Markelic, M.B.; Ruzicic, A.A.; Jevdjovic, T.V.; Lakic, I.V.; Djordjevic, J.D.; Vujovic, P.Z. Hypothalamic Insulin Expression Remains Unaltered after Short-Term Fasting in Female Rats. Endocrine 2022, 78, 476–483.
  13. Eerola, K.; Longo, F.; Reinbothe, T.M.; Richard, J.E.; Shevchouk, O.T.; López-Ferreras, L.; Mishra, D.; Asker, M.; Tolö, J.; Miranda, C.; et al. Hindbrain Insulin Controls Feeding Behavior. Mol. Metab. 2022, 66, 101614.
  14. Lee, J.; Kim, K.; Cho, J.H.; Bae, J.Y.; O’Leary, T.P.; Johnson, J.D.; Bae, Y.C.; Kim, E.-K. Insulin Synthesized in the Paraventricular Nucleus of the Hypothalamus Regulates Pituitary Growth Hormone Production. JCI Insight 2020, 5, e135412.
  15. Zhong, D.; Reid, B.M.; Donzella, B.; Miller, B.S.; Gunnar, M.R. Early-life Stress and Current Stress Predict BMI and Height Growth Trajectories in Puberty. Dev. Psychobiol. 2022, 64, e22342.
  16. de la Monte, S.M.; Xu, X.J.; Wands, J.R. Ethanol Inhibits Insulin Expression and Actions in the Developing Brain. CMLS Cell. Mol. Life Sci. 2005, 62, 1131–1145.
  17. Xu, J.; Yeon, J.E.; Chang, H.; Tison, G.; Chen, G.J.; Wands, J.; de la Monte, S. Ethanol Impairs Insulin-Stimulated Neuronal Survival in the Developing Brain: Role of PTEN Phosphatase. J. Biol. Chem. 2003, 278, 26929–26937.
  18. Ravichandran, L.V.; Chen, H.; Li, Y.; Quon, M.J. Phosphorylation of PTP1B at Ser(50) by Akt Impairs Its Ability to Dephosphorylate the Insulin Receptor. Mol. Endocrinol. 2001, 15, 1768–1780.
  19. Olson, A.L. Regulation of GLUT4 and Insulin-Dependent Glucose Flux. ISRN Mol. Biol. 2012, 2012, 856987.
  20. Osmanovic, J.; Plaschke, K.; Salkovic-Petrisic, M.; Grünblatt, E.; Riederer, P.; Hoyer, S. Chronic Exogenous Corticosterone Administration Generates an Insulin-Resistant Brain State in Rats. Stress 2010, 13, 123–131.
  21. Kellar, D.; Craft, S. Brain Insulin Resistance in Alzheimer’s Disease and Related Disorders: Mechanisms and Therapeutic Approaches. Lancet Neurol. 2020, 19, 758–766.
  22. Yoon, J.H.; Hwang, J.; Son, S.U.; Choi, J.; You, S.-W.; Park, H.; Cha, S.-Y.; Maeng, S. How Can Insulin Resistance Cause Alzheimer’s Disease? Int. J. Mol. Sci. 2023, 24, 3506.
  23. Leclerc, M.; Bourassa, P.; Tremblay, C.; Caron, V.; Sugère, C.; Emond, V.; Bennett, D.A.; Calon, F. Cerebrovascular Insulin Receptors Are Defective in Alzheimer’s Disease. Brain 2023, 146, 75–90.
  24. Folch, J.; Ettcheto, M.; Busquets, O.; Sánchez-López, E.; Castro-Torres, R.; Verdaguer, E.; Manzine, P.; Poor, S.; García, M.; Olloquequi, J.; et al. The Implication of the Brain Insulin Receptor in Late Onset Alzheimer’s Disease Dementia. Pharmaceuticals 2018, 11, 11.
  25. Sędzikowska, A.; Szablewski, L. Insulin and Insulin Resistance in Alzheimer’s Disease. Int. J. Mol. Sci. 2021, 22, 9987.
  26. Gil-Bea, F.J.; Solas, M.; Solomon, A.; Mugueta, C.; Winblad, B.; Kivipelto, M.; Ramirez, M.J.; Cedazo-Mínguez, A. Insulin Levels Are Decreased in the Cerebrospinal Fluid of Women with Prodomal Alzheimer’s Disease. J. Alzheimer’s Dis. 2010, 22, 405–413.
  27. Steen, E.; Terry, B.M.; Rivera, E.J.; Cannon, J.L.; Neely, T.R.; Tavares, R.; Xu, X.J.; Wands, J.R.; de la Monte, S.M. Impaired Insulin and Insulin-like Growth Factor Expression and Signaling Mechanisms in Alzheimer’s Disease—Is This Type 3 Diabetes? JAD 2005, 7, 63–80.
  28. Nemoto, T.; Toyoshima-Aoyama, F.; Yanagita, T.; Maruta, T.; Fujita, H.; Koshida, T.; Yonaha, T.; Wada, A.; Sawaguchi, A.; Murakami, M. New Insights Concerning Insulin Synthesis and Its Secretion in Rat Hippocampus and Cerebral Cortex: Amyloid-Β1–42-Induced Reduction of Proinsulin Level via Glycogen Synthase Kinase-3β. Cell Signal. 2014, 26, 253–259.
  29. Molnár, G.; Faragó, N.; Kocsis, Á.K.; Rózsa, M.; Lovas, S.; Boldog, E.; Báldi, R.; Csajbók, É.; Gardi, J.; Puskás, L.G.; et al. GABAergic Neurogliaform Cells Represent Local Sources of Insulin in the Cerebral Cortex. J. Neurosci. 2014, 34, 1133–1137.
More
Video Production Service