51
|
Servizi S, Corrigan RR, Casadesus G. The Importance of Understanding Amylin Signaling Mechanisms for Therapeutic Development in the Treatment of Alzheimer's Disease. Curr Pharm Des 2020; 26:1345-1355. [PMID: 32188374 PMCID: PMC10088426 DOI: 10.2174/1381612826666200318151146] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 03/04/2020] [Indexed: 12/12/2022]
Abstract
Type II Diabetes (T2D) is a major risk factor for Alzheimer's Disease (AD). These two diseases share several pathological features, including amyloid accumulation, inflammation, oxidative stress, cell death and cognitive decline. The metabolic hormone amylin and amyloid-beta are both amyloids known to self-aggregate in T2D and AD, respectively, and are thought to be the main pathogenic entities in their respective diseases. Furthermore, studies suggest amylin's ability to seed amyloid-beta aggregation, the activation of common signaling cascades in the pancreas and the brain, and the ability of amyloid beta to signal through amylin receptors (AMYR), at least in vitro. However, paradoxically, non-aggregating forms of amylin such as pramlintide are given to treat T2D and functional and neuroprotective benefits of amylin and pramlintide administration have been reported in AD transgenic mice. These paradoxical results beget a deeper study of the complex nature of amylin's signaling through the several AMYR subtypes and other receptors associated with amylin effects to be able to fully understand its potential role in mediating AD development and/or prevention. The goal of this review is to provide such critical insight to begin to elucidate how the complex nature of this hormone's signaling may explain its equally complex relationship with T2D and mechanisms of AD pathogenesis.
Collapse
Affiliation(s)
- Spencer Servizi
- School of Biomedical Sciences, Kent State University, Ohio, United States
| | - Rachel R Corrigan
- School of Biomedical Sciences, Kent State University, Ohio, United States
| | - Gemma Casadesus
- School of Biomedical Sciences, Kent State University, Ohio, United States.,Department of Biological Sciences, Kent State University, Ohio, United States
| |
Collapse
|
52
|
Xu Y, Chen F. Factors and Molecular Mechanisms Influencing the Protein Synthesis, Degradation and Membrane Trafficking of ASIC1a. Front Cell Dev Biol 2020; 8:596304. [PMID: 33195276 PMCID: PMC7644914 DOI: 10.3389/fcell.2020.596304] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 10/05/2020] [Indexed: 12/31/2022] Open
Abstract
Acid-sensing ion channels (ASICs) are members of the degenerin/epithelial sodium channel superfamily. They are extracellular pH sensors that are activated by protons. Among all ASICs, ASIC1a is one of the most intensively studied isoforms because of its unique ability to be permeable to Ca2+. In addition, it is considered to contribute to various pathophysiological conditions. As a membrane proton receptor, the number of ASIC1a present on the cell surface determines its physiological and pathological functions, and this number partially depends on protein synthesis, degradation, and membrane trafficking processes. Recently, several studies have shown that various factors affect these processes. Therefore, this review elucidated the major factors and underlying molecular mechanisms affecting ASIC1a protein expression and membrane trafficking.
Collapse
Affiliation(s)
- Yayun Xu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China.,The Key Laboratory of Major Autoimmune Diseases of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China
| | - Feihu Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China.,The Key Laboratory of Major Autoimmune Diseases of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China
| |
Collapse
|
53
|
Clinical Evidence of Antidepressant Effects of Insulin and Anti-Hyperglycemic Agents and Implications for the Pathophysiology of Depression-A Literature Review. Int J Mol Sci 2020; 21:ijms21186969. [PMID: 32971941 PMCID: PMC7554794 DOI: 10.3390/ijms21186969] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 08/21/2020] [Accepted: 09/15/2020] [Indexed: 02/07/2023] Open
Abstract
Close connections between depression and type 2 diabetes (T2DM) have been suggested by many epidemiological and experimental studies. Disturbances in insulin sensitivity due to the disruption of various molecular pathways cause insulin resistance, which underpins many metabolic disorders, including diabetes, as well as depression. Several anti-hyperglycemic agents have demonstrated antidepressant properties in clinical trials, probably due to their action on brain targets based on the shared pathophysiology of depression and T2DM. In this article, we review reports of clinical trials examining the antidepressant effect of these medications, including insulin, metformin, glucagon like peptide-1 receptor agonists (GLP-1RA), and peroxisome proliferator-activated receptor (PPAR)-γ agonists, and briefly consider possible molecular mechanisms underlying the associations between amelioration of insulin resistance and improvement of depressive symptoms. In doing so, we intend to suggest an integrative perspective for understanding the pathophysiology of depression.
Collapse
|
54
|
Spinelli M, Fusco S, Grassi C. Brain insulin resistance impairs hippocampal plasticity. VITAMINS AND HORMONES 2020; 114:281-306. [PMID: 32723548 DOI: 10.1016/bs.vh.2020.04.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Nutrient-related signals have been demonstrated to influence brain development and cognitive functions. In particular, insulin signaling has been shown to impact on molecular cascades underlying hippocampal plasticity, learning and memory. Alteration of brain insulin signaling interferes with the maintenance of neural stem cell niche and neuronal activity in the hippocampus. Brain insulin resistance is also emerging as key factor causing the cognitive impairment observed in metabolic and neurodegenerative diseases. Here, we review the molecular mechanisms involved in the insulin modulation of both adult neurogenesis and synaptic activity in the hippocampus. We also summarize the effects of altered insulin sensitivity on hippocampal plasticity. Finally, we reassume the experimental and epidemiological evidence highlighting the critical role of brain insulin resistance at the crossroad between type 2 diabetes and Alzheimer's disease.
Collapse
Affiliation(s)
- Matteo Spinelli
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Salvatore Fusco
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.
| | - Claudio Grassi
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.
| |
Collapse
|
55
|
Madhusudhanan J, Suresh G, Devanathan V. Neurodegeneration in type 2 diabetes: Alzheimer's as a case study. Brain Behav 2020; 10:e01577. [PMID: 32170854 PMCID: PMC7218246 DOI: 10.1002/brb3.1577] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 01/04/2020] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION Rigorous research in the last few years has shown that in addition to the classical mechanism of neurodegeneration, certain unconventional mechanisms may also lead to neurodegenerative disease. One of them is a widely studied metabolic disorder: type 2 diabetes mellitus (T2DM). We now have a clear understanding of glucose-mediated neurodegeneration, mostly from studies in Alzheimer's disease (AD) models. AD is recognized to be significantly associated with hyperglycemia, even earning the term "type 3 diabetes." Here, we review first the pathophysiology of AD, both from the perspective of classical protein accumulation, as well as the newer T2DM-dependent mechanisms supported by findings from patients with T2DM. Secondly, we review the different pathways through which neurodegeneration is aggravated in hyperglycemic conditions taking AD as a case study. Finally, some of the current advances in AD management as a result of recent research developments in metabolic disorders-driven neurodegeneration are also discussed. METHODS Relevant literatures found from PubMed search were reviewed. RESULTS Apart from the known causes of AD, type 2 diabetes opens a new window to the AD pathology in several ways. It is a bidirectional interaction, of which, the molecular and signaling mechanisms are recently studied. This is our attempt to connect all of them to draw a complete mechanistic explanation for the neurodegeneration in T2DM. Refer to Figure 3. CONCLUSION The perspective of AD as a classical neurodegenerative disease is changing, and it is now being looked at from a zoomed-out perspective. The correlation between T2DM and AD is something observed and studied extensively. It is promising to know that there are certain advances in AD management following these studies.
Collapse
Affiliation(s)
- Jalaja Madhusudhanan
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati, India
| | - Gowthaman Suresh
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati, India
| | - Vasudharani Devanathan
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati, India
| |
Collapse
|
56
|
Induction of LTM following an Insulin Injection. eNeuro 2020; 7:ENEURO.0088-20.2020. [PMID: 32291265 PMCID: PMC7218004 DOI: 10.1523/eneuro.0088-20.2020] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 03/21/2020] [Accepted: 03/26/2020] [Indexed: 02/06/2023] Open
Abstract
The pond snail Lymnaea stagnalis learns conditioned taste aversion (CTA) and consolidates it into long-term memory (LTM). One-day food-deprived snails (day 1 snails) show the best CTA learning and memory, whereas more severely food-deprived snails (5 d) do not express good memory. However, previous studies showed that CTA-LTM was indeed formed in 5-d food-deprived snails (day 5 snails), but its recall was prevented by the effects of food deprivation. CTA-LTM recall in day 5 snails was expressed following 7 d of feeding and then 1 d of food deprivation (day 13 snails). In the present study, we thus hypothesized that memory recall occurs because day 13 snails are in an optimal internal state. One day of food deprivation before the memory test in day 13 snails increased the mRNA level of molluscan insulin-related peptide (MIP) in the CNS. Thus, we further hypothesized that an injection of insulin into day 5 snails following seven additional days with access to food (day 12 snails) activates CTA neurons and mimics the food deprivation state before the memory test in day 13 snails. Day 12 snails injected with insulin could recall the memory. In addition, the simultaneous injection of an anti-insulin receptor antibody and insulin into day 12 snails did not allow memory recall. Insulin injection also decreased the hemolymph glucose concentration. Together, the results suggest that an optimal internal state (i.e., a spike in insulin release and specific glucose levels) are necessary for LTM recall following CTA training in snails.
Collapse
|
57
|
Abstract
Tau protein which was discovered in 1975 [310] became of great interest when it was identified as the main component of neurofibrillary tangles (NFT), a pathological feature in the brain of patients with Alzheimer's disease (AD) [39, 110, 232]. Tau protein is expressed mainly in the brain as six isoforms generated by alternative splicing [46, 97]. Tau is a microtubule associated proteins (MAPs) and plays a role in microtubules assembly and stability, as well as diverse cellular processes such as cell morphogenesis, cell division, and intracellular trafficking [49]. Additionally, Tau is involved in much larger neuronal functions particularly at the level of synapses and nuclei [11, 133, 280]. Tau is also physiologically released by neurons [233] even if the natural function of extracellular Tau remains to be uncovered (see other chapters of the present book).
Collapse
|
58
|
Hayashi T. Post-translational palmitoylation of ionotropic glutamate receptors in excitatory synaptic functions. Br J Pharmacol 2020; 178:784-797. [PMID: 32159240 DOI: 10.1111/bph.15050] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 02/07/2020] [Accepted: 03/05/2020] [Indexed: 12/17/2022] Open
Abstract
In the mammalian CNS, glutamate is the major excitatory neurotransmitter. Ionotropic glutamate receptors (iGluRs) are responsible for the glutamate-mediated postsynaptic excitation of neurons. Regulation of glutamatergic synapses is critical for higher brain functions including neural communication, memory formation, learning, emotion, and behaviour. Many previous studies have shown that post-translational protein S-palmitoylation, the only reversible covalent attachment of lipid to protein, regulates synaptic expression, intracellular localization, and membrane trafficking of iGluRs and their scaffolding proteins in neurons. This modification mechanism is extremely conserved in the vertebrate lineages. The failure of appropriate palmitoylation-dependent regulation of iGluRs leads to hyperexcitability that reduces the maintenance of network stability, resulting in brain disorders, such as epileptic seizures. This review summarizes advances in the study of palmitoylation of iGluRs, especially AMPA receptors and NMDA receptors, and describes the current understanding of palmitoylation-dependent regulation of excitatory glutamatergic synapses. LINKED ARTICLES: This article is part of a themed issue on Neurochemistry in Japan. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.4/issuetoc.
Collapse
Affiliation(s)
- Takashi Hayashi
- Section of Cellular Biochemistry, Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Kodaira, Tokyo, Japan
| |
Collapse
|
59
|
McNay EC, Pearson-Leary J. GluT4: A central player in hippocampal memory and brain insulin resistance. Exp Neurol 2020; 323:113076. [PMID: 31614121 PMCID: PMC6936336 DOI: 10.1016/j.expneurol.2019.113076] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 09/19/2019] [Accepted: 10/01/2019] [Indexed: 12/24/2022]
Abstract
Insulin is now well-established as playing multiple roles within the brain, and specifically as regulating hippocampal cognitive processes and metabolism. Impairments to insulin signaling, such as those seen in type 2 diabetes and Alzheimer's disease, are associated with brain hypometabolism and cognitive impairment, but the mechanisms of insulin's central effects are not determined. Several lines of research converge to suggest that the insulin-responsive glucose transporter GluT4 plays a central role in hippocampal memory processes, and that reduced activation of this transporter may underpin the cognitive impairments seen as a consequence of insulin resistance.
Collapse
Affiliation(s)
- Ewan C McNay
- Behavioral Neuroscience, University at Albany, Albany, NY, USA.
| | - Jiah Pearson-Leary
- Department of Anesthesiology, Abramson Research Center, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| |
Collapse
|
60
|
Krasil'nikova I, Surin A, Sorokina E, Fisenko A, Boyarkin D, Balyasin M, Demchenko A, Pomytkin I, Pinelis V. Insulin Protects Cortical Neurons Against Glutamate Excitotoxicity. Front Neurosci 2019; 13:1027. [PMID: 31611766 PMCID: PMC6769071 DOI: 10.3389/fnins.2019.01027] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 09/10/2019] [Indexed: 12/31/2022] Open
Abstract
Glutamate excitotoxicity is implicated in the pathogenesis of numerous diseases, such as stroke, traumatic brain injury, and Alzheimer's disease, for which insulin resistance is a concomitant condition, and intranasal insulin treatment is believed to be a promising therapy. Excitotoxicity is initiated primarily by the sustained stimulation of ionotropic glutamate receptors and leads to a rise in intracellular Ca2+ ([Ca2+] i ), followed by a cascade of intracellular events, such as delayed calcium deregulation (DCD), mitochondrial depolarization, adenosine triphosphate (ATP) depletion that collectively end in cell death. Therefore, cross-talk between insulin and glutamate signaling in excitotoxicity is of particular interest for research. In the present study, we investigated the effects of short-term insulin exposure on the dynamics of [Ca2+] i and mitochondrial potential in cultured rat cortical neurons during glutamate excitotoxicity. We found that insulin ameliorated the glutamate-evoked rise of [Ca2+] i and prevented the onset of DCD, the postulated point-of-no-return in excitotoxicity. Additionally, insulin significantly improved the glutamate-induced drop in mitochondrial potential, ATP depletion, and depletion of brain-derived neurotrophic factor (BDNF), which is a critical neuroprotector in excitotoxicity. Also, insulin improved oxygen consumption rates, maximal respiration, and spare respiratory capacity in neurons exposed to glutamate, as well as the viability of cells in the MTT assay. In conclusion, the short-term insulin exposure in our experiments was evidently a protective treatment against excitotoxicity, in a sharp contrast to chronic insulin exposure causal to neuronal insulin resistance, the adverse factor in excitotoxicity.
Collapse
Affiliation(s)
| | - Alexander Surin
- National Medical Research Center for Children's Health, Moscow, Russia.,Institute of General Pathology and Pathophysiology, Russian Academy of Sciences, Moscow, Russia
| | - Elena Sorokina
- National Medical Research Center for Children's Health, Moscow, Russia
| | - Andrei Fisenko
- National Medical Research Center for Children's Health, Moscow, Russia
| | - Dmitry Boyarkin
- National Medical Research Center for Children's Health, Moscow, Russia
| | - Maxim Balyasin
- Department of Advanced Cell Technologies, Institute of Regenerative Medicine, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Anna Demchenko
- Department of Advanced Cell Technologies, Institute of Regenerative Medicine, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Igor Pomytkin
- Department of Advanced Cell Technologies, Institute of Regenerative Medicine, I.M. Sechenov First Moscow State Medical University, Moscow, Russia.,Scientific Center for Biomedical Technologies, Federal Medical and Biological Agency, Svetlye Gory, Moscow, Russia
| | - Vsevolod Pinelis
- National Medical Research Center for Children's Health, Moscow, Russia
| |
Collapse
|
61
|
Spinelli M, Fusco S, Grassi C. Brain Insulin Resistance and Hippocampal Plasticity: Mechanisms and Biomarkers of Cognitive Decline. Front Neurosci 2019; 13:788. [PMID: 31417349 PMCID: PMC6685093 DOI: 10.3389/fnins.2019.00788] [Citation(s) in RCA: 159] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 07/15/2019] [Indexed: 12/27/2022] Open
Abstract
In the last decade, much attention has been devoted to the effects of nutrient-related signals on brain development and cognitive functions. A turning point was the discovery that brain areas other than the hypothalamus expressed receptors for hormones related to metabolism. In particular, insulin signaling has been demonstrated to impact on molecular cascades underlying hippocampal plasticity, learning and memory. Here, we summarize the molecular evidence linking alteration of hippocampal insulin sensitivity with changes of both adult neurogenesis and synaptic plasticity. We also review the epidemiological studies and experimental models emphasizing the critical role of brain insulin resistance at the crossroad between metabolic and neurodegenerative disease. Finally, we brief novel findings suggesting how biomarkers of brain insulin resistance, involving the study of brain-derived extracellular vesicles and brain glucose metabolism, may predict the onset and/or the progression of cognitive decline.
Collapse
Affiliation(s)
- Matteo Spinelli
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Salvatore Fusco
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Claudio Grassi
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| |
Collapse
|
62
|
Zhao F, Siu JJ, Huang W, Askwith C, Cao L. Insulin Modulates Excitatory Synaptic Transmission and Synaptic Plasticity in the Mouse Hippocampus. Neuroscience 2019; 411:237-254. [PMID: 31146008 PMCID: PMC6612444 DOI: 10.1016/j.neuroscience.2019.05.033] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 05/14/2019] [Accepted: 05/16/2019] [Indexed: 12/17/2022]
Abstract
The administration of exogenous insulin into the hippocampus has the potential to enhance cognitive function and exert other beneficial effects. Elucidating the neurobiological substrates of insulin action and its underlying physiological mechanisms may further improve treatment efficacy. Previous work has shown that insulin affects synaptic plasticity, however there are discrepancies and contradictory conclusions between studies. Here, we used extracellular field recordings in mouse hippocampal slices to investigate how insulin acutely modulates synaptic transmission and synaptic plasticity, both of which are correlated with learning and memory processes. Our data demonstrate that insulin application inhibited basal excitatory synaptic transmission and promoted long-term potentiation (LTP) induction at hippocampal Schaffer collateral-CA1 synapses. Under similar conditions, insulin strongly activated the PI3K/AKT pathway, but had only a weak effect on the MAPK/ERK pathway. Although insulin-induced inhibition of field excitatory post-synaptic potentials (fEPSPs) was previously termed insulin-long-term depression (insulin-LTD), insulin application potentiated recovery from classically induced LTD. Further analysis suggests suppression of presynaptic neurotransmitter release contributed to the insulin-LTD. At low concentrations, insulin primarily inhibited fEPSPs; however, at high concentration, its effects were of mixed inhibition and enhancement in different recordings. Moreover, a broad spectrum protein kinase C blocker, cannabinoid receptor type 1 activator, or a high glucose concentration inhibited fEPSPs per se, and disturbed insulin's effect on fEPSP. Insulin also caused depotentiation during LTP expression and triggered depression during LTD recovery. Given the essential roles of dynamic synaptic transmission and plasticity in learning and memory, our data provide more evidence that insulin application may actively modulate hippocampal-dependent cognitive events.
Collapse
Affiliation(s)
- Fangli Zhao
- College of Medicine, The Ohio State University
| | - Jason J Siu
- College of Medicine, The Ohio State University
| | - Wei Huang
- College of Medicine, The Ohio State University
| | | | - Lei Cao
- College of Medicine, The Ohio State University.
| |
Collapse
|
63
|
Frangou S, Shirali M, Adams MJ, Howard DM, Gibson J, Hall LS, Smith BH, Padmanabhan S, Murray AD, Porteous DJ, Haley CS, Deary IJ, Clarke TK, McIntosh AM. Insulin resistance: Genetic associations with depression and cognition in population based cohorts. Exp Neurol 2019; 316:20-26. [PMID: 30965038 PMCID: PMC6503941 DOI: 10.1016/j.expneurol.2019.04.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 03/26/2019] [Accepted: 04/03/2019] [Indexed: 01/07/2023]
Abstract
Insulin resistance, broadly defined as the reduced ability of insulin to exert its biological action, has been associated with depression and cognitive dysfunction in observational studies. However, it is unclear whether these associations are causal and whether they might be underpinned by other shared factors. To address this knowledge gap, we capitalized on the stability of genetic biomarkers through the lifetime, and on their unidirectional relationship with depression and cognition. Specifically, we determined the association between quantitative measures of cognitive function and depression and genetic instruments of insulin resistance traits in two large-scale population samples, the Generation Scotland: Scottish Family Health Study (GS: SFHS; N = 19,994) and in the UK Biobank (N = 331,374). In the GS:SFHS, the polygenic risk score (PRS) for fasting insulin was associated with verbal intelligence and depression while the PRS for the homeostasis model assessment of insulin resistance was associated with verbal intelligence. Despite this overlap in genetic architecture, Mendelian randomization analyses in the GS:SFHS and in the UK Biobank samples did not yield evidence for causal associations from insulin resistance traits to either depression or cognition. These findings may be due to weak genetic instruments, limited cognitive measures and insufficient power but they may also indicate the need to identify other biological mechanisms that may mediate the relationship from insulin resistance to depression and cognition.
Collapse
Affiliation(s)
- Sophia Frangou
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Masoud Shirali
- Division of Psychiatry, Royal Edinburgh Hospital, University of Edinburgh, Edinburgh, UK
| | - Mark J Adams
- Division of Psychiatry, Royal Edinburgh Hospital, University of Edinburgh, Edinburgh, UK
| | - David M Howard
- Division of Psychiatry, Royal Edinburgh Hospital, University of Edinburgh, Edinburgh, UK
| | - Jude Gibson
- Division of Psychiatry, Royal Edinburgh Hospital, University of Edinburgh, Edinburgh, UK
| | - Lynsey S Hall
- Division of Psychiatry, Royal Edinburgh Hospital, University of Edinburgh, Edinburgh, UK
| | - Blair H Smith
- Division of Population Health Sciences, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Sandosh Padmanabhan
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Alison D Murray
- Aberdeen Biomedical Imaging Centre, University of Aberdeen, Aberdeen, UK
| | - David J Porteous
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK; Generation Scotland, Medical Genetics Section, Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Chris S Haley
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Ian J Deary
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK; Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - Toni-Kim Clarke
- Division of Psychiatry, Royal Edinburgh Hospital, University of Edinburgh, Edinburgh, UK
| | - Andrew M McIntosh
- Division of Psychiatry, Royal Edinburgh Hospital, University of Edinburgh, Edinburgh, UK; Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK; Department of Psychology, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
64
|
de Mello NP, Orellana AM, Mazucanti CH, de Morais Lima G, Scavone C, Kawamoto EM. Insulin and Autophagy in Neurodegeneration. Front Neurosci 2019; 13:491. [PMID: 31231176 PMCID: PMC6558407 DOI: 10.3389/fnins.2019.00491] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 04/29/2019] [Indexed: 12/12/2022] Open
Abstract
Crosstalk in the pathophysiological processes underpinning metabolic diseases and neurodegenerative disorders have been the subject of extensive investigation, in which insulin signaling and autophagy impairment demonstrate to be a common factor in both conditions. Although it is still somewhat conflicting, pharmacological and genetic strategies that regulate these pathways may be a promising approach for aggregate protein clearancing and consequently the delaying of onset or progression of the disease. However, as the response due to this modulation seems to be time-dependent, finding the right regulation of autophagy may be a potential target for drug development for neurodegenerative diseases. In this way, this review focuses on the role of insulin signaling/resistance and autophagy in some neurodegenerative diseases, discussing pharmacological and non-pharmacological interventions in these diseases.
Collapse
Affiliation(s)
- Natália Prudente de Mello
- Laboratory of Molecular and Functional Neurobiology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Ana Maria Orellana
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Caio Henrique Mazucanti
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Geovanni de Morais Lima
- Laboratory of Molecular and Functional Neurobiology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Cristoforo Scavone
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Elisa Mitiko Kawamoto
- Laboratory of Molecular and Functional Neurobiology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
65
|
Jahanmahin A, Abbasnejad Z, Haghparast A, Ahmadiani A, Ghasemi R. The Effect of Intrahippocampal Insulin Injection on Scopolamine-induced Spatial Memory Impairment and Extracellular Signal-regulated Kinases Alteration. Basic Clin Neurosci 2019; 10:23-36. [PMID: 31031891 PMCID: PMC6484185 DOI: 10.32598/bcn.9.10.165] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 01/10/2018] [Accepted: 03/06/2018] [Indexed: 12/27/2022] Open
Abstract
Introduction It is well documented that insulin has neuroprotective and neuromodulator effects and can protect against different models of memory loss. Furthermore, cholinergic activity plays a significant role in memory, and scopolamine-induced memory loss is widely used as an experimental model of dementia. The current study aimed at investigating the possible effects of insulin against scopolamine-induced memory impairment in Wistar rat and its underlying molecular mechanisms. Methods Accordingly, animals were bilaterally cannulated in CA1, hippocampus. Intrahippocampal administration of insulin 6 MU and 12 MU in CA1 per day was performed during first 6 days after surgery. During next four days, the animal's spatial learning and memory were assessed in Morris water maze test (three days of learning and one day of retention test). The animals received scopolamine (1 mg/kg) Intraperitoneally (IP) 30 minutes before the onset of behavioral tests in each day. In the last day, the hippocampi were dissected and the levels of MAPK (mitogen-activated protein kinases) and caspase-3 activation were analyzed by Western blot technique. Results The behavioral results showed that scopolamine impaired spatial learning and memory without activating casapase-3, P38, and JNK, but chronic pretreatment by both doses of insulin was unable to restore this spatial memory impairment. In addition, scopolamine significantly reduced Extracellular signal-Regulated Kinases (ERKs) activity and insulin was unable to restore this reduction. Results revealed that scopolamine-mediated memory loss was not associated with hippocampal damage. Conclusion Insulin as a neuroprotective agent cannot restore memory when there is no hippocampal damage. In addition, the neuromodulator effect of insulin is not potent enough to overwhelm scopolamine-mediated disruptions of synaptic neurotransmission.
Collapse
Affiliation(s)
- Ahmad Jahanmahin
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Abbasnejad
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Haghparast
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abolhassan Ahmadiani
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Rasoul Ghasemi
- Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
66
|
Santiago JCP, Hallschmid M. Outcomes and clinical implications of intranasal insulin administration to the central nervous system. Exp Neurol 2019; 317:180-190. [PMID: 30885653 DOI: 10.1016/j.expneurol.2019.03.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 02/12/2019] [Accepted: 03/13/2019] [Indexed: 12/20/2022]
Abstract
Insulin signaling in the brain plays a critical role in metabolic control and cognitive function. Targeting insulinergic pathways in the central nervous system via peripheral insulin administration is feasible, but associated with systemic effects that necessitate tight supervision or countermeasures. The intranasal route of insulin administration, which largely bypasses the circulation and thereby greatly reduces these obstacles, has now been repeatedly tested in proof-of-concept studies in humans as well as animals. It is routinely used in experimental settings to investigate the impact on eating behavior, peripheral metabolism, memory function and brain activation of acute or long-term enhancements in central nervous system insulin signaling. Epidemiological and experimental evidence linking deteriorations in metabolic control such as diabetes with neurodegenerative diseases imply pathophysiological relevance of dysfunctional brain insulin signaling or brain insulin resistance, and suggest that targeting insulin in the brain holds some promise as a therapy or adjunct therapy. This short narrative review gives an overview over recent findings on brain insulin signaling as derived from human studies deploying intranasal insulin, and evaluates the potential of therapeutic interventions that target brain insulin resistance.
Collapse
Affiliation(s)
- João C P Santiago
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, 72076 Tübingen, Germany; German Center for Diabetes Research (DZD), 72076 Tübingen, Germany; Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, 72076 Tübingen, Germany
| | - Manfred Hallschmid
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, 72076 Tübingen, Germany; German Center for Diabetes Research (DZD), 72076 Tübingen, Germany; Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, 72076 Tübingen, Germany.
| |
Collapse
|
67
|
Insulin signaling in the hippocampus and amygdala regulates metabolism and neurobehavior. Proc Natl Acad Sci U S A 2019; 116:6379-6384. [PMID: 30765523 DOI: 10.1073/pnas.1817391116] [Citation(s) in RCA: 154] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Previous studies have shown that insulin and IGF-1 signaling in the brain, especially the hypothalamus, is important for regulation of systemic metabolism. Here, we develop mice in which we have specifically inactivated both insulin receptors (IRs) and IGF-1 receptors (IGF1Rs) in the hippocampus (Hippo-DKO) or central amygdala (CeA-DKO) by stereotaxic delivery of AAV-Cre into IRlox/lox/IGF1Rlox/lox mice. Consequently, both Hippo-DKO and CeA-DKO mice have decreased levels of the GluA1 subunit of glutamate AMPA receptor and display increased anxiety-like behavior, impaired cognition, and metabolic abnormalities, including glucose intolerance. Hippo-DKO mice also display abnormal spatial learning and memory whereas CeA-DKO mice have impaired cold-induced thermogenesis. Thus, insulin/IGF-1 signaling has common roles in the hippocampus and central amygdala, affecting synaptic function, systemic glucose homeostasis, behavior, and cognition. In addition, in the hippocampus, insulin/IGF-1 signaling is important for spatial learning and memory whereas insulin/IGF-1 signaling in the central amygdala controls thermogenesis via regulation of neural circuits innervating interscapular brown adipose tissue.
Collapse
|
68
|
Forny-Germano L, De Felice FG, Vieira MNDN. The Role of Leptin and Adiponectin in Obesity-Associated Cognitive Decline and Alzheimer's Disease. Front Neurosci 2019; 12:1027. [PMID: 30692905 PMCID: PMC6340072 DOI: 10.3389/fnins.2018.01027] [Citation(s) in RCA: 165] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 12/19/2019] [Indexed: 12/14/2022] Open
Abstract
Cross-talk between adipose tissue and central nervous system (CNS) underlies the increased risk of obese people to develop brain diseases such as cognitive and mood disorders. Detailed mechanisms for how peripheral changes caused by adipose tissue accumulation in obesity impact the CNS to cause brain dysfunction are poorly understood. Adipokines are a large group of substances secreted by the white adipose tissue to regulate a wide range of homeostatic processes including, but not limited to, energy metabolism and immunity. Obesity is characterized by a generalized change in the levels of circulating adipokines due to abnormal accumulation and dysfunction of adipose tissue. Altered adipokine levels underlie complications of obesity as well as the increased risk for the development of obesity-related comorbidities such as type 2 diabetes, cardiovascular and neurodegenerative diseases. Here, we review the literature for the role of adipokines as key mediators of the communication between periphery and CNS in health and disease. We will focus on the actions of leptin and adiponectin, two of the most abundant and well studied adipokines, in the brain, with particular emphasis on how altered signaling of these adipokines in obesity may lead to cognitive dysfunction and augmented risk for Alzheimer's disease. A better understanding of adipokine biology in brain disorders may prove of major relevance to diagnostic, prevention and therapy.
Collapse
Affiliation(s)
- Leticia Forny-Germano
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fernanda G. De Felice
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Centre for Neuroscience Studies, Department of Psychiatry, Queen’s University, Kingston, ON, Canada
| | | |
Collapse
|
69
|
Carvalho C, Cardoso SM, Correia SC, Moreira PI. Tortuous Paths of Insulin Signaling and Mitochondria in Alzheimer's Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1128:161-183. [PMID: 31062330 DOI: 10.1007/978-981-13-3540-2_9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Due to the exponential growth of aging population worldwide, neurodegenerative diseases became a major public health concern. Among them, Alzheimer's disease (AD) prevails as the most common in the elderly, rendering it a research priority. After several decades considering the brain as an insulin-insensitive organ, recent advances proved a central role for this hormone in learning and memory processes and showed that AD shares a high number of features with systemic conditions characterized by insulin resistance. Mitochondrial dysfunction has also been widely demonstrated to play a major role in AD development supporting the idea that this neurodegenerative disease is characterized by a pronounced metabolic dysregulation. This chapter is intended to discuss evidence demonstrating the key role of insulin signaling and mitochondrial anomalies in AD.
Collapse
Affiliation(s)
- Cristina Carvalho
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Susana M Cardoso
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Sónia C Correia
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Paula I Moreira
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal. .,Laboratory of Physiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
70
|
Molecular Connection Between Diabetes and Dementia. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1128:103-131. [DOI: 10.1007/978-981-13-3540-2_6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
71
|
Hersom M, Helms HC, Schmalz C, Pedersen TÅ, Buckley ST, Brodin B. The insulin receptor is expressed and functional in cultured blood-brain barrier endothelial cells but does not mediate insulin entry from blood to brain. Am J Physiol Endocrinol Metab 2018; 315:E531-E542. [PMID: 29584446 DOI: 10.1152/ajpendo.00350.2016] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Insulin and its receptor are known to be present and functional in the brain. Insulin cerebrospinal fluid concentrations have been shown to correlate with plasma levels of insulin in a nonlinear fashion, indicative of a saturable transport pathway from the blood to the brain interstitial fluid. The aim of the present study was to investigate whether insulin was transported across brain endothelial cells in vitro via an insulin receptor-dependent pathway. The study showed that the insulin receptor was expressed at both the mRNA and protein levels in bovine brain endothelial cells. Luminally applied radiolabeled insulin showed insulin receptor-mediated binding to the endothelial cells. This caused a dose-dependent increase in Akt-phosphorylation, which was inhibited by coapplication of an insulin receptor inhibitor, s961, demonstrating activation of insulin receptor signaling pathways. Transport of insulin across the blood-brain barrier in vitro was low and comparable to that of a similarly sized paracellular marker. Furthermore, insulin transport was not inhibited by coapplication of an excess of unlabeled insulin or an insulin receptor inhibitor. The insulin transport and uptake studies were repeated in mouse brain endothelial cells demonstrating similar results. Although it cannot be ruled out that culture-induced changes in the cell model could have impaired a potential insulin transport mechanism, these in vitro data indicate that peripheral insulin must reach the brain parenchyma through alternative pathways rather than crossing the blood-brain barrier via receptor mediated transcytosis.
Collapse
Affiliation(s)
- Maria Hersom
- Department of Pharmacy, University of Copenhagen , Copenhagen , Denmark
| | - Hans C Helms
- Department of Pharmacy, University of Copenhagen , Copenhagen , Denmark
- Discovery ADME, Global Research, Novo Nordisk, Måløv, Denmark
| | | | - Thomas Å Pedersen
- Insulin Metabolism and Safety Biology, Global Research, Novo Nordisk, Måløv, Denmark
| | | | - Birger Brodin
- Department of Pharmacy, University of Copenhagen , Copenhagen , Denmark
| |
Collapse
|
72
|
Biliverdin Reductase-A Mediates the Beneficial Effects of Intranasal Insulin in Alzheimer Disease. Mol Neurobiol 2018; 56:2922-2943. [PMID: 30073505 DOI: 10.1007/s12035-018-1231-5] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 07/10/2018] [Indexed: 12/19/2022]
Abstract
Impairment of biliverdin reductase-A (BVR-A) is an early event leading to brain insulin resistance in AD. Intranasal insulin (INI) administration is under evaluation as a strategy to alleviate brain insulin resistance; however, the molecular mechanisms underlying INI beneficial effects are still unclear. We show that INI improves insulin signaling activation in the hippocampus and cortex of adult and aged 3×Tg-AD mice by ameliorating BVR-A activation. These changes were associated with a reduction of nitrosative stress, Tau phosphorylation, and Aβ oligomers in brain, along with improved cognitive functions. The role of BVR-A was strengthened by showing that cells lacking BVR-A: (i) develop insulin resistance if treated with insulin and (ii) can be recovered from insulin resistance only if treated with a BVR-A-mimetic peptide. These novel findings shed light on the mechanisms underlying INI treatment effects and suggest BVR-A as potential therapeutic target to prevent brain insulin resistance in AD.
Collapse
|
73
|
Loprinzi PD, Frith E. Obesity and episodic memory function. J Physiol Sci 2018; 68:321-331. [PMID: 29667132 PMCID: PMC10717800 DOI: 10.1007/s12576-018-0612-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 04/10/2018] [Indexed: 12/30/2022]
Abstract
Obesity-related lifestyle factors, such as physical activity behavior and dietary intake, have been shown to be associated with episodic memory function. From animal work, there is considerable biological plausibility linking obesity with worse memory function. There are no published systematic reviews evaluating the effects of obesity on episodic memory function among humans, and examining whether physical activity and diet influences this obesity-memory link. Thus, the purpose of this systematic review was to evaluate the totality of research examining whether obesity is associated with episodic memory function, and whether physical activity and dietary behavior confounds this relationship. A review approach was employed, using PubMed, PsychInfo, and Sports Discus databases. Fourteen studies met our criteria. Among these 14 reviewed studies, eight were cross-sectional, four were prospective, and two employed a randomized controlled experimental design. Twelve of the 14 studies did not take into consideration dietary behavior in their analysis, and similarly, nine of the 14 studies did not take into consideration participant physical activity behavior. Among the 14 studies, ten found an inverse association of weight status on memory function, but for one of these studies, this association was attenuated after controlling for physical activity. Among the 14 evaluated studies, four did not find a direct effect of weight status on memory. Among the four null studies, one, however, found an indirect effect of BMI on episodic memory and another found a moderation effect of BMI and age on memory function. It appears that obesity may be associated with worse memory function, with the underlying mechanisms discussed herein. At this point, it is uncertain whether adiposity, itself, is influencing memory changes, or rather, whether adiposity-related lifestyle behaviors (e.g., physical inactivity and diet) are driving the obesity-memory relationship.
Collapse
Affiliation(s)
- Paul D Loprinzi
- Exercise Psychology Laboratory, Physical Activity Epidemiology Laboratory, Department of Health, Exercise Science and Recreation Management, The University of Mississippi, University, MS, 38677, USA.
| | - Emily Frith
- Exercise Psychology Laboratory, Physical Activity Epidemiology Laboratory, Department of Health, Exercise Science and Recreation Management, The University of Mississippi, University, MS, 38677, USA
| |
Collapse
|
74
|
Nguyen TTL, Chan LC, Borreginne K, Kale RP, Hu C, Tye SJ. A review of brain insulin signaling in mood disorders: From biomarker to clinical target. Neurosci Biobehav Rev 2018; 92:7-15. [PMID: 29758232 DOI: 10.1016/j.neubiorev.2018.05.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 04/08/2018] [Accepted: 05/08/2018] [Indexed: 12/16/2022]
Abstract
Patients with mood disorders are at increased risk for metabolic dysfunction. Co-occurrence of the two conditions is typically associated with a more severe disease course and poorer treatment outcomes. The specific pathophysiological mechanisms underlying this bidirectional relationship between mood and metabolic dysfunction remains poorly understood. However, it is likely that impairment of metabolic processes within the brain play a critical role. The insulin signaling pathway mediates metabolic homeostasis and is important in the regulation of neurotrophic and synaptic plasticity processes, including those involved in neurodegenerative diseases like Alzheimer's. Thus, insulin signaling in the brain may serve to link metabolic function and mood. Central insulin signaling is mediated through locally secreted insulin and widespread insulin receptor expression. Here we review the preclinical and clinical data addressing the relationships between central insulin signaling, cellular metabolism, neurotrophic processes, and mood regulation, including key points of mechanistic overlap. These relationships have important implications for developing biomarker-based diagnostics and precision medicine approaches to treat severe mood disorders.
Collapse
Affiliation(s)
- Thanh Thanh L Nguyen
- Department of Psychiatry and Psychology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, United States; Department of Biology and Psychology, Green Mountain College, 1 Brennan Cir, Poultney, VT, 05764, United States
| | - Lily C Chan
- Department of Psychiatry and Psychology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, United States
| | - Kristin Borreginne
- Department of Psychiatry and Psychology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, United States
| | - Rajas P Kale
- Department of Psychiatry and Psychology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, United States; School of Engineering, Deakin University, Waurn Ponds, VIC, 3216, Australia
| | - Chunling Hu
- Department of Psychiatry and Psychology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, United States
| | - Susannah J Tye
- Department of Psychiatry and Psychology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, United States; Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, United States; Department of Psychiatry, University of Minnesota, 3 Morrill Hall, 100 Church Street SE, Minneapolis, MN, 55454, United States; School of Psychology, Deakin University, Burwood, VIC, 3125, Australia; Queensland Brain Institute, The University of Queensland, St Lucia, QLD, 4072, Australia.
| |
Collapse
|
75
|
Mandal A, Prabhavalkar KS, Bhatt LK. Gastrointestinal hormones in regulation of memory. Peptides 2018; 102:16-25. [PMID: 29466709 DOI: 10.1016/j.peptides.2018.02.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 02/13/2018] [Accepted: 02/14/2018] [Indexed: 12/16/2022]
Abstract
The connection between the gastrointestinal hormones and the brain has been established many years ago. This relation is termed the gut-brain axis (GBA). The GBA is a bidirectional communication which not only regulates gastrointestinal homeostasis but is also linked with higher emotional and cognitive functions. Hypothalamus plays a critical role in the regulation of energy metabolism, nutrient partitioning and control of feeding behaviors. Various gut hormones are released inside the gastrointestinal tract on food intake. These hormones act peripherally and influence the different responses of the tissues to the food intake, but do also have effects on the brain. The hypothalamus, in turn, integrates visceral function with limbic system structures such as hippocampus, amygdala, and cerebral cortex. The hippocampus has been known for its involvement in the cognitive function and the modulation of synaptic plasticity. This review aims to establish the role of various gut hormones in learning and memory, through the interaction of various receptors in the hippocampus. Understanding their role in memory can also aid in finding novel therapeutic strategies for the treatment of the neurological disorders associated with memory dysfunctions.
Collapse
Affiliation(s)
- Anwesha Mandal
- Department of Pharmacology, SVKM's Dr Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai, India
| | - Kedar S Prabhavalkar
- Department of Pharmacology, SVKM's Dr Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai, India.
| | - Lokesh K Bhatt
- Department of Pharmacology, SVKM's Dr Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai, India
| |
Collapse
|
76
|
Aulston BD, Schapansky J, Huang Y, Odero GL, Glazner GW. Secreted amyloid precursor protein alpha activates neuronal insulin receptors and prevents diabetes-induced encephalopathy. Exp Neurol 2018; 303:29-37. [PMID: 29410317 DOI: 10.1016/j.expneurol.2018.01.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 12/20/2017] [Accepted: 01/17/2018] [Indexed: 01/06/2023]
Abstract
Secreted amyloid precursor protein alpha (sAPPα) is a potent neurotrophin in the CNS but a dedicated receptor has not been found. However, protein interactions involving amyloid beta (Aβ), a peptide cleaved from the same parent peptide as sAPPα, indicate that insulin receptors (IRs) could be a target of amyloid peptides. In this study, in vitro analysis of cortical neuronal cultures revealed that exogenous sAPPα increased IR phosphorylation in the absence of insulin. Furthermore, in an APP overexpressing mouse model, sAPPα bound IRs in the cortex with significantly greater binding in hypoinsulinemic animals. To further examine the effects of sAPPα on the diabetic brain, we next rendered sAPPα overexpressing mice insulin depleted and found that sAPPα blocked aberrant tau phosphorylation (T231) in cortical tissue after 16 weeks diabetes. sAPPα overexpression also prevented hyperphosphorylation of AKT/GSK3 and activation of the unfolded protein response (UPR). In total, these data show sAPPα binds and activates neuronal IRs and that sAPPα has a protective effect on diabetic brain tissue.
Collapse
Affiliation(s)
- Brent D Aulston
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, Manitoba, Canada; St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, Manitoba, Canada
| | - Jason Schapansky
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, Manitoba, Canada; St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, Manitoba, Canada
| | - YaWen Huang
- St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, Manitoba, Canada
| | - Gary L Odero
- St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, Manitoba, Canada
| | - Gordon W Glazner
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, Manitoba, Canada; St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, Manitoba, Canada.
| |
Collapse
|
77
|
Nday CM, Eleftheriadou D, Jackson G. Shared pathological pathways of Alzheimer's disease with specific comorbidities: current perspectives and interventions. J Neurochem 2018; 144:360-389. [PMID: 29164610 DOI: 10.1111/jnc.14256] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 11/10/2017] [Accepted: 11/10/2017] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD) belongs to one of the most multifactorial, complex and heterogeneous morbidity-leading disorders. Despite the extensive research in the field, AD pathogenesis is still at some extend obscure. Mechanisms linking AD with certain comorbidities, namely diabetes mellitus, obesity and dyslipidemia, are increasingly gaining importance, mainly because of their potential role in promoting AD development and exacerbation. Their exact cognitive impairment trajectories, however, remain to be fully elucidated. The current review aims to offer a clear and comprehensive description of the state-of-the-art approaches focused on generating in-depth knowledge regarding the overlapping pathology of AD and its concomitant ailments. Thorough understanding of associated alterations on a number of molecular, metabolic and hormonal pathways, will contribute to the further development of novel and integrated theranostics, as well as targeted interventions that may be beneficial for individuals with age-related cognitive decline.
Collapse
Affiliation(s)
- Christiane M Nday
- Department of Chemical Engineering, Laboratory of Inorganic Chemistry, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Despoina Eleftheriadou
- Department of Chemical Engineering, Laboratory of Inorganic Chemistry, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Graham Jackson
- Department of Chemistry, University of Cape Town, Rondebosch, Cape Town, South Africa
| |
Collapse
|
78
|
Griffith CM, Eid T, Rose GM, Patrylo PR. Evidence for altered insulin receptor signaling in Alzheimer's disease. Neuropharmacology 2018; 136:202-215. [PMID: 29353052 DOI: 10.1016/j.neuropharm.2018.01.008] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 12/19/2017] [Accepted: 01/05/2018] [Indexed: 12/11/2022]
Abstract
Epidemiological data have shown that metabolic disease can increase the propensity for developing cognitive decline and dementia, particularly Alzheimer's disease (AD). While this interaction is not completely understood, clinical studies suggest that both hyper- and hypoinsulinemia are associated with an increased risk for developing AD. Indeed, insulin signaling is altered in post-mortem brain tissue from AD patients and treatments known to enhance insulin signaling can improve cognitive function. Further, clinical evidence has shown that AD patients and mouse models of AD often display alterations in peripheral metabolism. Since insulin is primarily derived from the periphery, it is likely that changes in peripheral insulin levels lead to alterations in central nervous system (CNS) insulin signaling and could contribute to cognitive decline and pathogenesis. Developing a better understanding of the relationship between alterations in peripheral metabolism and cognitive function might provide a foundation for the development of better treatment options for patients with AD. In this article we will begin to piece together the present data defining this relationship by briefly discussing insulin signaling in the periphery and CNS, its role in cognitive function, insulin's relationship to AD, peripheral metabolic alterations in mouse models of AD and how information from these models helps understand the mechanisms through which these changes potentially lead to impairments in insulin signaling in the CNS, and potential ways to target insulin signaling that could improve cognitive function in AD. This article is part of the Special Issue entitled 'Metabolic Impairment as Risk Factors for Neurodegenerative Disorders.'
Collapse
Affiliation(s)
- Chelsea M Griffith
- Department of Physiology, Southern Illinois University School of Medicine, Carbondale, IL 62901, USA; Center for Integrated Research in Cognitive and Neural Sciences, Southern Illinois University Carbondale, IL 62901, USA
| | - Tore Eid
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06510, USA; Department of Neurosurgery, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Gregory M Rose
- Department of Physiology, Southern Illinois University School of Medicine, Carbondale, IL 62901, USA; Department of Anatomy, Southern Illinois University School of Medicine, Carbondale, IL 62901, USA; Center for Integrated Research in Cognitive and Neural Sciences, Southern Illinois University Carbondale, IL 62901, USA
| | - Peter R Patrylo
- Department of Physiology, Southern Illinois University School of Medicine, Carbondale, IL 62901, USA; Department of Anatomy, Southern Illinois University School of Medicine, Carbondale, IL 62901, USA; Center for Integrated Research in Cognitive and Neural Sciences, Southern Illinois University Carbondale, IL 62901, USA.
| |
Collapse
|
79
|
Navidhamidi M, Ghasemi M, Mehranfard N. Epilepsy-associated alterations in hippocampal excitability. Rev Neurosci 2018; 28:307-334. [PMID: 28099137 DOI: 10.1515/revneuro-2016-0059] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 11/03/2016] [Indexed: 11/15/2022]
Abstract
The hippocampus exhibits a wide range of epilepsy-related abnormalities and is situated in the mesial temporal lobe, where limbic seizures begin. These abnormalities could affect membrane excitability and lead to overstimulation of neurons. Multiple overlapping processes refer to neural homeostatic responses develop in neurons that work together to restore neuronal firing rates to control levels. Nevertheless, homeostatic mechanisms are unable to restore normal neuronal excitability, and the epileptic hippocampus becomes hyperexcitable or hypoexcitable. Studies show that there is hyperexcitability even before starting recurrent spontaneous seizures, suggesting although hippocampal hyperexcitability may contribute to epileptogenesis, it alone is insufficient to produce epileptic seizures. This supports the concept that the hippocampus is not the only substrate for limbic seizure onset, and a broader hyperexcitable limbic structure may contribute to temporal lobe epilepsy (TLE) seizures. Nevertheless, seizures also occur in conditions where the hippocampus shows a hypoexcitable phenotype. Since TLE seizures most often originate in the hippocampus, it could therefore be assumed that both hippocampal hypoexcitability and hyperexcitability are undesirable states that make the epileptic hippocampal network less stable and may, under certain conditions, trigger seizures.
Collapse
|
80
|
Gratuze M, Joly-Amado A, Vieau D, Buée L, Blum D. Mutual Relationship between Tau and Central Insulin Signalling: Consequences for AD and Tauopathies? Neuroendocrinology 2018; 107:181-195. [PMID: 29439247 DOI: 10.1159/000487641] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 02/13/2018] [Indexed: 12/30/2022]
Abstract
Alzheimer disease (AD) is a progressive neurodegenerative disorder mainly characterized by cognitive deficits and neuropathological changes such as Tau lesions and amyloid plaques, but also associated with non-cognitive symptomatology. Metabolic and neuroendocrine abnormalities, such as alterations in body weight, brain insulin impairments, and lower brain glucose metabolism, which often precede clinical diagnosis, have been extensively reported in AD patients. However, the origin of these symptoms and their relation to pathology and cognitive impairments remain misunderstood. Insulin is a hormone involved in the control of energy homeostasis both peripherally and centrally, and insulin-resistant state has been linked to increased risk of dementia. It is now well established that insulin resistance can exacerbate Tau lesions, mainly by disrupting the balance between Tau kinases and phosphatases. On the other hand, the emerging literature indicates that Tau protein can also modulate insulin signalling in the brain, thus creating a detrimental vicious circle. The following review will highlight our current understanding of the role of insulin in the brain and its relation to Tau protein in the context of AD and tauopathies. Considering that insulin signalling is prone to be pharmacologically targeted at multiple levels, it constitutes an appealing approach to improve both insulin brain sensitivity and mitigate brain pathology with expected positive outcome in terms of cognition.
Collapse
Affiliation(s)
- Maud Gratuze
- Centre de Recherche du Centre Hospitalier de l'Université Laval de Québec, Axe Neurosciences, Université Laval, Québec, Québec, Canada
| | - Aurélie Joly-Amado
- Byrd Alzheimer's Institute, Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida, USA
| | - Didier Vieau
- Université de Lille, Inserm, CHU Lille, UMR-S 1172 - JPArc, "Alzheimer and Tauopathies,", Lille, France
| | - Luc Buée
- Université de Lille, Inserm, CHU Lille, UMR-S 1172 - JPArc, "Alzheimer and Tauopathies,", Lille, France
| | - David Blum
- Université de Lille, Inserm, CHU Lille, UMR-S 1172 - JPArc, "Alzheimer and Tauopathies,", Lille, France
| |
Collapse
|
81
|
Rodriguez-Rodriguez P, Sandebring-Matton A, Merino-Serrais P, Parrado-Fernandez C, Rabano A, Winblad B, Ávila J, Ferrer I, Cedazo-Minguez A. Tau hyperphosphorylation induces oligomeric insulin accumulation and insulin resistance in neurons. Brain 2017; 140:3269-3285. [PMID: 29053786 DOI: 10.1093/brain/awx256] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 08/11/2017] [Indexed: 11/13/2022] Open
Abstract
Insulin signalling deficiencies and insulin resistance have been directly linked to the progression of neurodegenerative disorders like Alzheimer's disease. However, to date little is known about the underlying molecular mechanisms or insulin state and distribution in the brain under pathological conditions. Here, we report that insulin is accumulated and retained as oligomers in hyperphosphorylated tau-bearing neurons in Alzheimer's disease and in several of the most prevalent human tauopathies. The intraneuronal accumulation of insulin is directly dependent on tau hyperphosphorylation, and follows the tauopathy progression. Furthermore, cells accumulating insulin show signs of insulin resistance and decreased insulin receptor levels. These results suggest that insulin retention in hyperphosphorylated tau-bearing neurons is a causative factor for the insulin resistance observed in tauopathies, and describe a novel neuropathological concept with important therapeutic implications.
Collapse
Affiliation(s)
- Patricia Rodriguez-Rodriguez
- Karolinska Institutet, Center for Alzheimer Research, Department of Neurobiology Care Sciences and Society, Division of Neurogeriatrics, Stockholm, Sweden
| | - Anna Sandebring-Matton
- Karolinska Institutet, Center for Alzheimer Research, Department of Neurobiology Care Sciences and Society, Division of Neurogeriatrics, Stockholm, Sweden
| | - Paula Merino-Serrais
- Karolinska Institutet, Center for Alzheimer Research, Department of Neurobiology Care Sciences and Society, Division of Neurogeriatrics, Stockholm, Sweden
| | - Cristina Parrado-Fernandez
- Karolinska Institutet, Center for Alzheimer Research, Department of Neurobiology Care Sciences and Society, Division of Neurogeriatrics, Stockholm, Sweden
| | - Alberto Rabano
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Spain.,Fundación CIEN, Madrid, Spain
| | - Bengt Winblad
- Karolinska Institutet, Center for Alzheimer Research, Department of Neurobiology Care Sciences and Society, Division of Neurogeriatrics, Stockholm, Sweden
| | - Jesús Ávila
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Spain.,Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
| | - Isidre Ferrer
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Spain.,Institut de Neuropatologia, Servei Anatomia Patologica, IDIBELL-Hospital Universitari de Bellvitge, Universitat de Barcelona, Hospitalet de Llobregat, Barcelona, Spain
| | - Angel Cedazo-Minguez
- Karolinska Institutet, Center for Alzheimer Research, Department of Neurobiology Care Sciences and Society, Division of Neurogeriatrics, Stockholm, Sweden
| |
Collapse
|
82
|
Insulin-mediated synaptic plasticity in the CNS: Anatomical, functional and temporal contexts. Neuropharmacology 2017; 136:182-191. [PMID: 29217283 PMCID: PMC5988909 DOI: 10.1016/j.neuropharm.2017.12.001] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 12/01/2017] [Accepted: 12/03/2017] [Indexed: 12/17/2022]
Abstract
For decades the brain was erroneously considered an insulin insensitive organ. Although gaps in our knowledge base remain, conceptual frameworks are starting to emerge to provide insight into the mechanisms through which insulin facilitates critical brain functions like metabolism, cognition, and motivated behaviors. These diverse physiological and behavioral activities highlight the region-specific activities of insulin in the CNS; that is, there is an anatomical context to the activities of insulin in the CNS. Similarly, there is also a temporal context to the activities of insulin in the CNS. Indeed, brain insulin receptor activity can be conceptualized as a continuum in which insulin promotes neuroplasticity from development into adulthood where it is an integral part of healthy brain function. Unfortunately, brain insulin resistance likely contributes to neuroplasticity deficits in obesity and type 2 diabetes mellitus (T2DM). This neuroplasticity continuum can be conceptualized by the mechanisms through which insulin promotes cognitive function through its actions in brain regions like the hippocampus, as well as the ability of insulin to modulate motivated behaviors through actions in brain regions like the nucleus accumbens and the ventral tegmental area. Thus, the goals of this review are to highlight these anatomical, temporal, and functional contexts of insulin activity in these brain regions, and to identify potentially critical time points along this continuum where the transition from enhancement of neuroplasticity to impairment may take place.
Collapse
|
83
|
Neth BJ, Craft S. Insulin Resistance and Alzheimer's Disease: Bioenergetic Linkages. Front Aging Neurosci 2017; 9:345. [PMID: 29163128 PMCID: PMC5671587 DOI: 10.3389/fnagi.2017.00345] [Citation(s) in RCA: 202] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 10/13/2017] [Indexed: 12/14/2022] Open
Abstract
Metabolic dysfunction is a well-established feature of Alzheimer's disease (AD), evidenced by brain glucose hypometabolism that can be observed potentially decades prior to the development of AD symptoms. Furthermore, there is mounting support for an association between metabolic disease and the development of AD and related dementias. Individuals with insulin resistance, type 2 diabetes mellitus (T2D), hyperlipidemia, obesity, or other metabolic disease may have increased risk for the development of AD and similar conditions, such as vascular dementia. This association may in part be due to the systemic mitochondrial dysfunction that is common to these pathologies. Accumulating evidence suggests that mitochondrial dysfunction is a significant feature of AD and may play a fundamental role in its pathogenesis. In fact, aging itself presents a unique challenge due to inherent mitochondrial dysfunction and prevalence of chronic metabolic disease. Despite the progress made in understanding the pathogenesis of AD and in the development of potential therapies, at present we remain without a disease-modifying treatment. In this review, we will discuss insulin resistance as a contributing factor to the pathogenesis of AD, as well as the metabolic and bioenergetic disruptions linking insulin resistance and AD. We will also focus on potential neuroimaging tools for the study of the metabolic dysfunction commonly seen in AD with hopes of developing therapeutic and preventative targets.
Collapse
Affiliation(s)
- Bryan J Neth
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Suzanne Craft
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
84
|
Engin AB, Engin ED, Karakus R, Aral A, Gulbahar O, Engin A. N-Methyl-D aspartate receptor-mediated effect on glucose transporter-3 levels of high glucose exposed-SH-SY5Y dopaminergic neurons. Food Chem Toxicol 2017; 109:465-471. [PMID: 28951307 DOI: 10.1016/j.fct.2017.09.038] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 09/16/2017] [Accepted: 09/20/2017] [Indexed: 11/29/2022]
Abstract
High glucose and insulin lead to neuronal insulin resistance. Glucose transport into the neurons is achieved by regulatory induction of surface glucose transporter-3 (GLUT3) instead of the insulin. N-methyl-D aspartate (NMDA) receptor activity increases GLUT3 expression. This study explored whether an endogenous NMDA receptor antagonist, kynurenic acid (KynA) affects the neuronal cell viability at high glucose concentrations. SH-SY5Y neuroblastoma cells were exposed to 150-250 mg/dL glucose and 40 μU/mL insulin. In KynA and N-nitro-l-arginine methyl ester (L-NAME) supplemented cultures, oxidative stress, mitochondrial metabolic activity (MTT), nitric oxide as nitrite+nitrate (NOx) and GLUT3 were determined at the end of 24 and 48-h incubation periods. Viable cells were counted by trypan blue dye. High glucose-exposed SH-SY5Y cells showed two-times more GLUT3 expression at second 24-h period. While GLUT3-stimulated glucose transport and oxidative stress was increased, total mitochondrial metabolic activity was significantly reduced. Insulin supplementation to high glucose decreased NOx synthesis and GLUT3 levels, in contrast oxidative stress increased three-fold. KynA significantly reduced oxidative stress, and increased MTT by regulating NOx production and GLUT3 expression. KynA is a noteworthy compound, as an endogenous, specific NMDA receptor antagonist; it significantly reduces oxidative stress, while increasing cell viability at high glucose and insulin concentrations.
Collapse
Affiliation(s)
- Ayse Basak Engin
- Gazi University, Faculty of Pharmacy, Department of Toxicology, Hipodrom, Ankara, Turkey.
| | - Evren Doruk Engin
- Ankara University, Biotechnology Institute, Tandogan, Ankara, Turkey
| | - Resul Karakus
- Gazi University, Faculty of Medicine, Department of Immunology, Besevler, Ankara, Turkey
| | - Arzu Aral
- Gazi University, Faculty of Medicine, Department of Immunology, Besevler, Ankara, Turkey
| | - Ozlem Gulbahar
- Gazi University, Faculty of Medicine, Department of Biochemistry, Besevler, Ankara, Turkey
| | - Atilla Engin
- Gazi University, Faculty of Medicine, Department of General Surgery, Besevler, Ankara, Turkey
| |
Collapse
|
85
|
Maimaiti S, Frazier HN, Anderson KL, Ghoweri AO, Brewer LD, Porter NM, Thibault O. Novel calcium-related targets of insulin in hippocampal neurons. Neuroscience 2017; 364:130-142. [PMID: 28939258 DOI: 10.1016/j.neuroscience.2017.09.019] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 09/07/2017] [Accepted: 09/08/2017] [Indexed: 01/28/2023]
Abstract
Both insulin signaling disruption and Ca2+ dysregulation are closely related to memory loss during aging and increase the vulnerability to Alzheimer's disease (AD). In hippocampal neurons, aging-related changes in calcium regulatory pathways have been shown to lead to higher intracellular calcium levels and an increase in the Ca2+-dependent afterhyperpolarization (AHP), which is associated with cognitive decline. Recent studies suggest that insulin reduces the Ca2+-dependent AHP. Given the sensitivity of neurons to insulin and evidence that brain insulin signaling is reduced with age, insulin-mediated alterations in calcium homeostasis may underlie the beneficial actions of insulin in the brain. Indeed, increasing insulin signaling in the brain via intranasal delivery has yielded promising results such as improving memory in both clinical and animal studies. However, while several mechanisms have been proposed, few have focused on regulation on intracellular Ca2+. In the present study, we further examined the effects of acute insulin on calcium pathways in primary hippocampal neurons in culture. Using the whole-cell patch-clamp technique, we found that acute insulin delivery reduced voltage-gated calcium currents. Fura-2 imaging was used to also address acute insulin effects on spontaneous and depolarization-mediated Ca2+ transients. Results indicate that insulin reduced Ca2+ transients, which appears to have involved a reduction in ryanodine receptor function. Together, these results suggest insulin regulates pathways that control intracellular Ca2+ which may reduce the AHP and improve memory. This may be one mechanism contributing to improved memory recall in response to intranasal insulin therapy in the clinic.
Collapse
Affiliation(s)
- Shaniya Maimaiti
- Department of Pharmacology and Nutritional Sciences, University of Kentucky Medical Center, UKMC, MS-310; 800 Rose Street, Lexington, KY 40536, United States
| | - Hilaree N Frazier
- Department of Pharmacology and Nutritional Sciences, University of Kentucky Medical Center, UKMC, MS-310; 800 Rose Street, Lexington, KY 40536, United States
| | - Katie L Anderson
- Department of Pharmacology and Nutritional Sciences, University of Kentucky Medical Center, UKMC, MS-310; 800 Rose Street, Lexington, KY 40536, United States
| | - Adam O Ghoweri
- Department of Pharmacology and Nutritional Sciences, University of Kentucky Medical Center, UKMC, MS-310; 800 Rose Street, Lexington, KY 40536, United States
| | - Lawrence D Brewer
- Department of Pharmacology and Nutritional Sciences, University of Kentucky Medical Center, UKMC, MS-310; 800 Rose Street, Lexington, KY 40536, United States
| | - Nada M Porter
- Department of Pharmacology and Nutritional Sciences, University of Kentucky Medical Center, UKMC, MS-310; 800 Rose Street, Lexington, KY 40536, United States
| | - Olivier Thibault
- Department of Pharmacology and Nutritional Sciences, University of Kentucky Medical Center, UKMC, MS-310; 800 Rose Street, Lexington, KY 40536, United States.
| |
Collapse
|
86
|
Tundo GR, Sbardella D, Ciaccio C, Grasso G, Gioia M, Coletta A, Polticelli F, Di Pierro D, Milardi D, Van Endert P, Marini S, Coletta M. Multiple functions of insulin-degrading enzyme: a metabolic crosslight? Crit Rev Biochem Mol Biol 2017. [PMID: 28635330 DOI: 10.1080/10409238.2017.1337707] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Insulin-degrading enzyme (IDE) is a ubiquitous zinc peptidase of the inverzincin family, which has been initially discovered as the enzyme responsible for insulin catabolism; therefore, its involvement in the onset of diabetes has been largely investigated. However, further studies on IDE unraveled its ability to degrade several other polypeptides, such as β-amyloid, amylin, and glucagon, envisaging the possible implication of IDE dys-regulation in the "aggregopathies" and, in particular, in neurodegenerative diseases. Over the last decade, a novel scenario on IDE biology has emerged, pointing out a multi-functional role of this enzyme in several basic cellular processes. In particular, latest advances indicate that IDE behaves as a heat shock protein and modulates the ubiquitin-proteasome system, suggesting a major implication in proteins turnover and cell homeostasis. In addition, recent observations have highlighted that the regulation of glucose metabolism by IDE is not merely based on its largely proposed role in the degradation of insulin in vivo. There is increasing evidence that improper IDE function, regulation, or trafficking might contribute to the etiology of metabolic diseases. In addition, the enzymatic activity of IDE is affected by metals levels, thus suggesting a role also in the metal homeostasis (metallostasis), which is thought to be tightly linked to the malfunction of the "quality control" machinery of the cell. Focusing on the physiological role of IDE, we will address a comprehensive vision of the very complex scenario in which IDE takes part, outlining its crucial role in interconnecting several relevant cellular processes.
Collapse
Affiliation(s)
- Grazia R Tundo
- a Department of Clinical Sciences and Translation Medicine , University of Roma Tor Vergata , Roma , Italy.,b CIRCMSB , Bari , Italy
| | - Diego Sbardella
- a Department of Clinical Sciences and Translation Medicine , University of Roma Tor Vergata , Roma , Italy.,b CIRCMSB , Bari , Italy.,c Center for TeleInfrastructures, University of Roma Tor Vergata , Roma , Italy
| | - Chiara Ciaccio
- a Department of Clinical Sciences and Translation Medicine , University of Roma Tor Vergata , Roma , Italy.,b CIRCMSB , Bari , Italy
| | - Giuseppe Grasso
- d Department of Chemistry , University of Catania , Catania , Italy.,e CNR IBB , Catania , Italy
| | - Magda Gioia
- a Department of Clinical Sciences and Translation Medicine , University of Roma Tor Vergata , Roma , Italy.,b CIRCMSB , Bari , Italy
| | - Andrea Coletta
- f Department of Chemistry , University of Aarhus , Aarhus , Denmark
| | | | - Donato Di Pierro
- a Department of Clinical Sciences and Translation Medicine , University of Roma Tor Vergata , Roma , Italy.,b CIRCMSB , Bari , Italy
| | | | - Peter Van Endert
- h Université Paris Descartes, INSERM, U1151, CNRS , Paris , France
| | - Stefano Marini
- a Department of Clinical Sciences and Translation Medicine , University of Roma Tor Vergata , Roma , Italy.,b CIRCMSB , Bari , Italy.,c Center for TeleInfrastructures, University of Roma Tor Vergata , Roma , Italy
| | - Massimo Coletta
- a Department of Clinical Sciences and Translation Medicine , University of Roma Tor Vergata , Roma , Italy.,b CIRCMSB , Bari , Italy.,c Center for TeleInfrastructures, University of Roma Tor Vergata , Roma , Italy
| |
Collapse
|
87
|
Hippocampal insulin resistance and altered food decision-making as players on obesity risk. Neurosci Biobehav Rev 2017; 77:165-176. [DOI: 10.1016/j.neubiorev.2017.03.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 03/17/2017] [Accepted: 03/19/2017] [Indexed: 12/17/2022]
|
88
|
Murine model and mechanisms of treatment-induced painful diabetic neuropathy. Neuroscience 2017; 354:136-145. [PMID: 28476321 DOI: 10.1016/j.neuroscience.2017.04.036] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 04/21/2017] [Accepted: 04/23/2017] [Indexed: 11/21/2022]
Abstract
Diabetes mellitus represents a group of metabolic diseases that are characterized by hyperglycemia caused by either lack of insulin production or a reduced ability to respond to insulin. It is estimated that there were 347 million people worldwide who suffered from diabetes in 2008 and incidence is predicted to double by 2050. Neuropathy is the most common complication of long-term diabetes and approximately 30% of these subjects develop chronic neuropathic pain. A distinct acute, severe form of neuropathic pain, called insulin neuritis or treatment-induced painful neuropathy of diabetes (TIND), may also occur shortly after initiation of intensive glycemic control, with an incidence rate of up to 10.9%. The pathological mechanisms leading to TIND, which is mostly unresponsive to analgesics, are not yet understood, impeding the development of therapies. Studies to date have been clinical and with limited cohorts of patients. In the current study, we developed chronic and acute insulin-induced neuropathic pain in mice with type 2 insulin-resistant diabetes. Furthermore, we determined that insulin-induced acute allodynia is independent of glycemia levels, can also be induced with Insulin-like Growth Factor 1 (IGF1) and be prevented by inhibition of AKT, providing evidence of an insulin/IGF1 signaling pathway-based mechanism for TIND. This mouse model is useful for the elucidation of mechanisms contributing to TIND and for the testing of new therapeutic approaches to treat TIND.
Collapse
|
89
|
Vauzour D, Camprubi-Robles M, Miquel-Kergoat S, Andres-Lacueva C, Bánáti D, Barberger-Gateau P, Bowman GL, Caberlotto L, Clarke R, Hogervorst E, Kiliaan AJ, Lucca U, Manach C, Minihane AM, Mitchell ES, Perneczky R, Perry H, Roussel AM, Schuermans J, Sijben J, Spencer JPE, Thuret S, van de Rest O, Vandewoude M, Wesnes K, Williams RJ, Williams RSB, Ramirez M. Nutrition for the ageing brain: Towards evidence for an optimal diet. Ageing Res Rev 2017; 35:222-240. [PMID: 27713095 DOI: 10.1016/j.arr.2016.09.010] [Citation(s) in RCA: 140] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 09/05/2016] [Accepted: 09/29/2016] [Indexed: 02/07/2023]
Abstract
As people age they become increasingly susceptible to chronic and extremely debilitating brain diseases. The precise cause of the neuronal degeneration underlying these disorders, and indeed normal brain ageing remains however elusive. Considering the limits of existing preventive methods, there is a desire to develop effective and safe strategies. Growing preclinical and clinical research in healthy individuals or at the early stage of cognitive decline has demonstrated the beneficial impact of nutrition on cognitive functions. The present review is the most recent in a series produced by the Nutrition and Mental Performance Task Force under the auspice of the International Life Sciences Institute Europe (ILSI Europe). The latest scientific advances specific to how dietary nutrients and non-nutrient may affect cognitive ageing are presented. Furthermore, several key points related to mechanisms contributing to brain ageing, pathological conditions affecting brain function, and brain biomarkers are also discussed. Overall, findings are inconsistent and fragmented and more research is warranted to determine the underlying mechanisms and to establish dose-response relationships for optimal brain maintenance in different population subgroups. Such approaches are likely to provide the necessary evidence to develop research portfolios that will inform about new dietary recommendations on how to prevent cognitive decline.
Collapse
Affiliation(s)
- David Vauzour
- University of East Anglia, Norwich Medical School, Norwich NR4 7UQ, United Kingdom
| | - Maria Camprubi-Robles
- Abbott Nutrition R&D, Abbott Laboratories, Camino de Purchil 68, 18004 Granada, Spain
| | | | | | - Diána Bánáti
- International Life Sciences Institute, Europe (ILSI Europe), Av E. Mounier 83, Box 6, 1200 Brussels, Belgium
| | | | - Gene L Bowman
- Nestlé Institute of Health Sciences, EPFL Innovation Park, 1015 Lausanne, Switzerland
| | - Laura Caberlotto
- The Microsoft Research-University of Trento, Centre for Computational and Systems Biology (COSBI), Piazza Manifattura 1, 38068 Rovereto, TN, Italy
| | - Robert Clarke
- Oxford University, Richard Doll Building, Old Road Campus, Roosevelt Drive, OX3 7LF Oxford, United Kingdom
| | - Eef Hogervorst
- Loughborough University, Brockington Building, Asby Road, LE11 3TU Loughborough, United Kingdom
| | - Amanda J Kiliaan
- Radboud University Medical Center, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Ugo Lucca
- IRCCS-Instituto di Richerche Farmacologiche Mario Negri, Via G. La Masa 19, 20156 Milan, Italy
| | - Claudine Manach
- INRA, UMR 1019, Human Nutrition Unit, CRNH Auvergne, 63000 Clermont-Ferrand, France
| | - Anne-Marie Minihane
- University of East Anglia, Norwich Medical School, Norwich NR4 7UQ, United Kingdom
| | | | - Robert Perneczky
- Imperial College London, South Kensington Campus, SW7 2AZ London, United Kingdom
| | - Hugh Perry
- University of Southampton, Tremona Road, SO16 6YD Southampton, United Kingdom
| | - Anne-Marie Roussel
- Joseph Fourier University, Domaine de la Merci, 38706 La Tronche, France
| | - Jeroen Schuermans
- International Life Sciences Institute, Europe (ILSI Europe), Av E. Mounier 83, Box 6, 1200 Brussels, Belgium.
| | - John Sijben
- Nutricia Research, Nutricia Advances Medical Nutrition, P.O. Box 80141, 3508TC Utrecht, The Netherlands
| | - Jeremy P E Spencer
- University of Reading, Whiteknights, P.O. Box 217, RG6 6AH Reading, Berkshire, United Kingdom
| | - Sandrine Thuret
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, The Maurice Wohl Clinical Neuroscience Institute, 125 Coldharbour Lane, SE5 9NU London, United Kingdom
| | - Ondine van de Rest
- Wageningen University, P.O. Box 8129, 6700 EV Wageningen, The Netherlands
| | | | - Keith Wesnes
- Wesnes Cognition Ltd., Little Paddock, Streatley on Thames RG8 9RD, United Kingdom; Department of Psychology, Northumbria University, Newcastle, United Kingdom; Centre for Human Psychopharmacology, Swinburne University, Melbourne, Australia; Medicinal Plant Research Group, Newcastle University, United Kingdom
| | | | - Robin S B Williams
- Royal Holloway, University of London, Egham, TW20 0EX Surrey, United Kingdom
| | - Maria Ramirez
- Abbott Nutrition R&D, Abbott Laboratories, Camino de Purchil 68, 18004 Granada, Spain
| |
Collapse
|
90
|
Ogoh S. Relationship between cognitive function and regulation of cerebral blood flow. J Physiol Sci 2017; 67:345-351. [PMID: 28155036 PMCID: PMC10717011 DOI: 10.1007/s12576-017-0525-0] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 01/23/2017] [Indexed: 12/11/2022]
Abstract
Ageing is the primary risk factor for cognitive deterioration. Given that the cerebral blood flow (CBF) or regulation of cerebral circulation is attenuated in the elderly, it could be expected that ageing-induced cognitive deterioration may be affected by a decrease in CBF as a result of brain ischemia and energy depletion. CBF regulation associated with cerebral metabolism thus likely plays an important role in the preservation of cognitive function. However, in some specific conditions (e.g. during exercise), change in CBF does not synchronize with that of cerebral metabolism. Our recent study demonstrated that cognitive function was more strongly affected by changes in cerebral metabolism than by changes in CBF during exercise. Therefore, it remains unclear how an alteration in CBF or its regulation affects cognitive function. In this review, I summarize current knowledge on previous investigations providing the possibility of an interaction between regulation of CBF or cerebral metabolism and cognitive function.
Collapse
Affiliation(s)
- Shigehiko Ogoh
- Department of Biomedical Engineering, Toyo University, 2100 Kujirai, Kawagoe-Shi, Saitama, 350-8585, Japan.
| |
Collapse
|
91
|
Cheke LG, Bonnici HM, Clayton NS, Simons JS. Obesity and insulin resistance are associated with reduced activity in core memory regions of the brain. Neuropsychologia 2017; 96:137-149. [PMID: 28093279 PMCID: PMC5317178 DOI: 10.1016/j.neuropsychologia.2017.01.013] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 12/20/2016] [Accepted: 01/13/2017] [Indexed: 01/14/2023]
Abstract
Increasing research in animals and humans suggests that obesity may be associated with learning and memory deficits, and in particular with reductions in episodic memory. Rodent models have implicated the hippocampus in obesity-related memory impairments, but the neural mechanisms underlying episodic memory deficits in obese humans remain undetermined. In the present study, lean and obese human participants were scanned using fMRI while completing a What-Where-When episodic memory test (the “Treasure-Hunt Task”) that assessed the ability to remember integrated item, spatial, and temporal details of previously encoded complex events. In lean participants, the Treasure-Hunt task elicited significant activity in regions of the brain known to be important for recollecting episodic memories, such as the hippocampus, angular gyrus, and dorsolateral prefrontal cortex. Both obesity and insulin resistance were associated with significantly reduced functional activity throughout the core recollection network. These findings indicate that obesity is associated with reduced functional activity in core brain areas supporting episodic memory and that insulin resistance may be a key player in this association. Obesity associated with reduced activity in core recollection network during episodic memory. Insulin resistance associated with reduced activity in core recollection network during episodic memory. Insulin resistance, but not obesity, associated with poorer memory performance.
Collapse
Affiliation(s)
- Lucy G Cheke
- Department of Psychology, University of Cambrigde, UK.
| | | | | | - Jon S Simons
- Department of Psychology, University of Cambrigde, UK
| |
Collapse
|
92
|
Cardoso S, Seiça R, Moreira PI. Diabesity and Brain Energy Metabolism: The Case of Alzheimer's Disease. ADVANCES IN NEUROBIOLOGY 2017; 19:117-150. [PMID: 28933063 DOI: 10.1007/978-3-319-63260-5_5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/21/2023]
Abstract
It is widely accepted that high calorie diets and a sedentary lifestyle sturdily influence the incidence and outcome of type 2 diabetes and obesity, which can occur simultaneously, a situation called diabesity. Tightly linked with metabolic and energy regulation, a close association between diabetes and Alzheimer's disease (AD) has been proposed. Among the common pathogenic mechanisms that underpin both conditions, insulin resistance, brain glucose hypometabolism, and metabolic dyshomeostasis appear to have a pivotal role. This century is an unprecedented diabetogenic period in human history, so therapeutic strategies and/or approaches to control and/or revert this evolving epidemic is of utmost importance. This chapter will make a brief contextualization about the impact that diabetes and obesity can exert in brain structure and function alongside with a brief survey about the role of insulin in normal brain function, exploring its roles in cognition and brain glucose metabolism. Later, attention will be given to the intricate relation of diabesity, insulin resistance, and AD. Finally, both pharmacological and lifestyle interventions will also be reviewed as strategies aimed at fighting diabesity and/or AD-related metabolic effects.
Collapse
Affiliation(s)
- Susana Cardoso
- Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal.
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.
| | - Raquel Seiça
- Institute of Physiology, Institute for Biomedical Imaging and Life Sciences-IBILI, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Paula I Moreira
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Institute of Physiology, Institute for Biomedical Imaging and Life Sciences-IBILI, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
93
|
Bryan MR, Bowman AB. Manganese and the Insulin-IGF Signaling Network in Huntington's Disease and Other Neurodegenerative Disorders. ADVANCES IN NEUROBIOLOGY 2017; 18:113-142. [PMID: 28889265 PMCID: PMC6559248 DOI: 10.1007/978-3-319-60189-2_6] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Huntington's disease (HD) is an autosomal dominant neurodegenerative disease resulting in motor impairment and death in patients. Recently, several studies have demonstrated insulin or insulin-like growth factor (IGF) treatment in models of HD, resulting in potent amelioration of HD phenotypes via modulation of the PI3K/AKT/mTOR pathways. Administration of IGF and insulin can rescue microtubule transport, metabolic function, and autophagy defects, resulting in clearance of Huntingtin (HTT) aggregates, restoration of mitochondrial function, amelioration of motor abnormalities, and enhanced survival. Manganese (Mn) is an essential metal to all biological systems but, in excess, can be toxic. Interestingly, several studies have revealed the insulin-mimetic effects of Mn-demonstrating Mn can activate several of the same metabolic kinases and increase peripheral and neuronal insulin and IGF-1 levels in rodent models. Separate studies have shown mouse and human striatal neuroprogenitor cell (NPC) models exhibit a deficit in cellular Mn uptake, indicative of a Mn deficiency. Furthermore, evidence from the literature reveals a striking overlap between cellular consequences of Mn deficiency (i.e., impaired function of Mn-dependent enzymes) and known HD endophenotypes including excitotoxicity, increased reactive oxygen species (ROS) accumulation, and decreased mitochondrial function. Here we review published evidence supporting a hypothesis that (1) the potent effect of IGF or insulin treatment on HD models, (2) the insulin-mimetic effects of Mn, and (3) the newly discovered Mn-dependent perturbations in HD may all be functionally related. Together, this review will present the intriguing possibility that intricate regulatory cross-talk exists between Mn biology and/or toxicology and the insulin/IGF signaling pathways which may be deeply connected to HD pathology and, perhaps, other neurodegenerative diseases (NDDs) and other neuropathological conditions.
Collapse
Affiliation(s)
- Miles R Bryan
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
- Vanderbilt Brain Institute, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
| | - Aaron B Bowman
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Vanderbilt Brain Institute, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Vanderbilt Center in Molecular Toxicology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| |
Collapse
|
94
|
Gralle M. The neuronal insulin receptor in its environment. J Neurochem 2016; 140:359-367. [PMID: 27889917 DOI: 10.1111/jnc.13909] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 10/31/2016] [Accepted: 11/21/2016] [Indexed: 01/01/2023]
Abstract
Insulin is known mainly for its effects in peripheral tissues, such as the liver, skeletal muscles and adipose tissue, where the activation of the insulin receptor (IR) has both short-term and long-term effects. Insulin and the IR are also present in the brain, and since there is evidence that neuronal insulin signaling regulates synaptic plasticity and that it is impaired in disease, this pathway might be the key to protection or reversal of symptoms, especially in Alzheimer's disease. However, there are controversies about the importance of the neuronal IR, partly because biophysical data on its activation and signaling are much less complete than for the peripheral IR. This review briefly summarizes the neuronal IR signaling in health and disease, and then focuses on known differences between the neuronal and peripheral IR with regard to alternative splicing and glycosylation, and lack of data with respect to phosphorylation and membrane subdomain localization. Particularities in the neuronal IR itself and its environment may have consequences for downstream signaling and impact synaptic plasticity. Furthermore, establishing the relative importance of insulin signaling through IR or through hybrids with its homolog, the insulin-like growth factor 1 receptor, is crucial for evaluating the consequences of brain IR activation. An improved biophysical understanding of the neuronal IR may help predict the consequences of insulin-targeted interventions.
Collapse
Affiliation(s)
- Matthias Gralle
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
95
|
Collison KS, Inglis A, Shibin S, Andres B, Ubungen R, Thiam J, Mata P, Al-Mohanna FA. Differential effects of early-life NMDA receptor antagonism on aspartame-impaired insulin tolerance and behavior. Physiol Behav 2016; 167:209-221. [DOI: 10.1016/j.physbeh.2016.09.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 08/09/2016] [Accepted: 09/13/2016] [Indexed: 01/15/2023]
|
96
|
Ribe EM, Lovestone S. Insulin signalling in Alzheimer's disease and diabetes: from epidemiology to molecular links. J Intern Med 2016; 280:430-442. [PMID: 27739227 DOI: 10.1111/joim.12534] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
As populations across the world both age and become more obese, the numbers of individuals with Alzheimer's disease and diabetes are increasing; posing enormous challenges for society and consequently becoming priorities for governments and global organizations. These issues, an ageing population at risk of neurodegenerative diseases such as Alzheimer's disease and an increasingly obese population at risk of metabolic alterations such as type 2 diabetes, are usually considered as independent conditions, but increasing evidence from both epidemiological and molecular studies link these disorders. The aim of this review was to highlight these multifactorial links. We will discuss the impact of direct links between insulin and IGF-1 signalling and the Alzheimer's disease-associated pathological events as well as the impact of other processes such as inflammation, oxidative stress and mitochondrial dysfunction either common to both conditions or perhaps responsible for a mechanistic link between metabolic and neurodegenerative disease. An understanding of such associations might be of importance not only in the understanding of disease mechanisms but also in the search for novel therapeutic options.
Collapse
Affiliation(s)
- E M Ribe
- Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, UK
| | - S Lovestone
- Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, UK.
| |
Collapse
|
97
|
Abstract
Both type 1 (T1DM) and type 2 diabetes mellitus (T2DM) have been associated with reduced performance on multiple domains of cognitive function and with evidence of abnormal structural and functional brain magnetic resonance imaging (MRI). Cognitive deficits may occur at the very earliest stages of diabetes and are further exacerbated by the metabolic syndrome. The duration of diabetes and glycemic control may have an impact on the type and severity of cognitive impairment, but as yet we cannot predict who is at greatest risk of developing cognitive impairment. The pathophysiology of cognitive impairment is multifactorial, although dysfunction in each interconnecting pathway ultimately leads to discordance in metabolic signaling. The pathophysiology includes defects in insulin signaling, autonomic function, neuroinflammatory pathways, mitochondrial (Mt) metabolism, the sirtuin-peroxisome proliferator-activated receptor-gamma co-activator 1α (SIRT-PGC-1α) axis, and Tau signaling. Several promising therapies have been identified in pre-clinical studies, but remain to be validated in clinical trials.
Collapse
Affiliation(s)
- Lindsay A Zilliox
- Department of Neurology, Maryland VA Healthcare System and University of Maryland, 110 South Paca Street, Baltimore, MD, 21201, USA
| | - Krish Chadrasekaran
- Department of Neurology, Maryland VA Healthcare System and University of Maryland, 110 South Paca Street, Baltimore, MD, 21201, USA
| | - Justin Y Kwan
- Department of Neurology, Maryland VA Healthcare System and University of Maryland, 110 South Paca Street, Baltimore, MD, 21201, USA
| | - James W Russell
- Department of Neurology, Maryland VA Healthcare System and University of Maryland, 110 South Paca Street, Baltimore, MD, 21201, USA.
- School of Medicine, Department of Neurology, University of Maryland, 3S-129, 110 South Paca Street, Baltimore, MD, 21201-1595, USA.
| |
Collapse
|
98
|
Ribarič S. The Rationale for Insulin Therapy in Alzheimer's Disease. Molecules 2016; 21:molecules21060689. [PMID: 27240327 PMCID: PMC6273626 DOI: 10.3390/molecules21060689] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 05/14/2016] [Accepted: 05/19/2016] [Indexed: 12/30/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia, with a prevalence that increases with age. By 2050, the worldwide number of patients with AD is projected to reach more than 140 million. The prominent signs of AD are progressive memory loss, accompanied by a gradual decline in cognitive function and premature death. AD is the clinical manifestation of altered proteostasis. The initiating step of altered proteostasis in most AD patients is not known. The progression of AD is accelerated by several chronic disorders, among which the contribution of diabetes to AD is well understood at the cell biology level. The pathological mechanisms of AD and diabetes interact and tend to reinforce each other, thus accelerating cognitive impairment. At present, only symptomatic interventions are available for treating AD. To optimise symptomatic treatment, a personalised therapy approach has been suggested. Intranasal insulin administration seems to open the possibility for a safe, and at least in the short term, effective symptomatic intervention that delays loss of cognition in AD patients. This review summarizes the interactions of AD and diabetes from the cell biology to the patient level and the clinical results of intranasal insulin treatment of cognitive decline in AD.
Collapse
Affiliation(s)
- Samo Ribarič
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, SI-1000 Ljubljana, Slovenia.
| |
Collapse
|
99
|
Central Nervous Insulin Signaling in Sleep-Associated Memory Formation and Neuroendocrine Regulation. Neuropsychopharmacology 2016; 41:1540-50. [PMID: 26448203 PMCID: PMC4832015 DOI: 10.1038/npp.2015.312] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 09/16/2015] [Accepted: 10/03/2015] [Indexed: 01/30/2023]
Abstract
The neurochemical underpinnings of sleep's contribution to the establishment and maintenance of memory traces are largely unexplored. Considering that intranasal insulin administration to the CNS improves memory functions in healthy and memory-impaired humans, we tested whether brain insulin signaling and sleep interact to enhance memory consolidation in healthy participants. We investigated the effect of intranasal insulin on sleep-associated neurophysiological and neuroendocrine parameters and memory consolidation in 16 men and 16 women (aged 18-30 years), who learned a declarative word-pair task and a procedural finger sequence tapping task in the evening before intranasal insulin (160 IU) or placebo administration and 8 h of nocturnal sleep. On the subsequent evening, they learned interfering word-pairs and a new finger sequence before retrieving the original memories. Insulin increased growth hormone concentrations in the first night-half and EEG delta power during the second 90 min of non-rapid-eye-movement sleep. Insulin treatment impaired the acquisition of new contents in both the declarative and procedural memory systems on the next day, whereas retrieval of original memories was unchanged. Results indicate that sleep-associated memory consolidation is not a primary mediator of insulin's acute memory-improving effect, but that the peptide acts on mechanisms that diminish the subsequent encoding of novel information. Thus, by inhibiting processes of active forgetting during sleep, central nervous insulin might reduce the interfering influence of encoding new information.
Collapse
|
100
|
McGregor G, Malekizadeh Y, Harvey J. Minireview: Food for thought: regulation of synaptic function by metabolic hormones. Mol Endocrinol 2016; 29:3-13. [PMID: 25470238 DOI: 10.1210/me.2014-1328] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The peripheral actions of the metabolic hormones, leptin and insulin, are well documented. However, the functions of these hormones are not restricted to the periphery because evidence is growing that both leptin and insulin can readily cross the blood-brain barrier and have widespread central actions. The hippocampus in particular expresses high levels of both insulin and leptin receptors as well as key components of their associated signaling cascades. Moreover, recent studies indicate that both hormones are potential cognitive enhancers. Indeed, it has been demonstrated that both leptin and insulin markedly influence key cellular events that underlie hippocampal learning and memory including activity-dependent synaptic plasticity and the trafficking of glutamate receptors to and away from hippocampal synapses. The hippocampal formation is also a prime site for the neurodegenerative processes that occur during Alzheimer's disease, and impairments in either leptin or insulin function have been linked to central nervous system-driven diseases like Alzheimer's disease. Thus, the capacity of the metabolic hormones, leptin and insulin, to regulate hippocampal synaptic function has significant implications for normal brain function and also central nervous system-driven disease.
Collapse
Affiliation(s)
- Gemma McGregor
- Division of Neuroscience, Medical Research Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, United Kingdom
| | | | | |
Collapse
|