1
|
Černe U, Horvat A, Sanjković E, Kozoderc N, Kreft M, Zorec R, Scholz N, Vardjan N. Ca 2+ excitability of glia to neuromodulator octopamine in Drosophila living brain is greater than that of neurons. Acta Physiol (Oxf) 2025; 241:e14270. [PMID: 39801347 PMCID: PMC11726276 DOI: 10.1111/apha.14270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 11/13/2024] [Accepted: 01/01/2025] [Indexed: 01/16/2025]
Abstract
AIM Octopamine in the Drosophila brain has a neuromodulatory role similar to that of noradrenaline in mammals. After release from Tdc2 neurons, octopamine/tyramine may trigger intracellular Ca2+ signaling via adrenoceptor-like receptors on neural cells, modulating neurotransmission. Octopamine/tyramine receptors are expressed in neurons and glia, but how each of these cell types responds to octopamine remains elusive. This study aimed to characterize Ca2+ responses of neurons and astrocytes to neuromodulatory octopamine signals. METHODS We expressed Ca2+ indicator jGCaMP7b in specific cell type in adult Drosophila brains and performed intracellular Ca2+ imaging in the brain optic lobes upon bath application of octopamine by confocal microscopy. RESULTS Octopamine-stimulated Ca2+ responses in neurons were different from those of glial cells. The amplitude of octopamine-mediated Ca2+ signals in neurons was 3.4-fold greater than in astrocytes. However, astrocytes were more sensitive to octopamine; the median effective concentration that triggered Ca2+ responses was nearly 6-fold lower in astrocytes than in neurons. In both cell types, Ca2+ transients are shaped by Gq and Gs protein-coupled octopamine/tyramine receptors. Our snRNA-seq database screening uncovered differential expression patterns of these receptors between brain cell types, which may explain the difference in Ca2+ signaling. CONCLUSION In the brain optic lobes, astrocytes, not neurons, appear to be the sole responders to low concentration octopamine signals, and therefore likely drive synaptic plasticity and visual processing. Given the interconnectivity of the optic lobes with other brain regions, octopaminergic signals acting through the optic lobe astrocytes may also influence higher-order brain functions including learning and memory.
Collapse
Grants
- P40 OD018537 NIH HHS
- Deutsche Forschungsgemeinschaft (FOR 2149, 265903901/P01; CRC 1423, 421152132/B06)
- Slovenian Research and Innovation Agency (P3-0310, J3-2523, J3-50104, MR+ 2019, I0-0034, I0-0022: MRIC-Carl Zeiss Reference Centre for Laser Confocal Microscopy)
- European Cooperation in Science and Technology (COST) action CA18133 (European Research Network on Signal Transduction (ERNEST))
- European Cooperation in Science and Technology (COST) action CA18133 (European Research Network on Signal Transduction (ERNEST))
- Slovenian Research and Innovation Agency (P3‐0310, J3‐2523, J3‐50104, MR+ 2019, I0‐0034, I0‐0022: MRIC‐Carl Zeiss Reference Centre for Laser Confocal Microscopy)
- Deutsche Forschungsgemeinschaft (FOR 2149, 265903901/P01; CRC 1423, 421152132/B06)
Collapse
Affiliation(s)
- Urška Černe
- Laboratory of Neuroendocrinology‐Molecular Cell Physiology, Faculty of Medicine, Institute of PathophysiologyUniversity of LjubljanaLjubljanaSlovenia
- Laboratory of Cell Engineering, Celica BiomedicalLjubljanaSlovenia
| | - Anemari Horvat
- Laboratory of Neuroendocrinology‐Molecular Cell Physiology, Faculty of Medicine, Institute of PathophysiologyUniversity of LjubljanaLjubljanaSlovenia
- Laboratory of Cell Engineering, Celica BiomedicalLjubljanaSlovenia
| | - Ena Sanjković
- Laboratory of Neuroendocrinology‐Molecular Cell Physiology, Faculty of Medicine, Institute of PathophysiologyUniversity of LjubljanaLjubljanaSlovenia
| | - Nika Kozoderc
- Laboratory of Neuroendocrinology‐Molecular Cell Physiology, Faculty of Medicine, Institute of PathophysiologyUniversity of LjubljanaLjubljanaSlovenia
| | - Marko Kreft
- Laboratory of Neuroendocrinology‐Molecular Cell Physiology, Faculty of Medicine, Institute of PathophysiologyUniversity of LjubljanaLjubljanaSlovenia
- Laboratory of Cell Engineering, Celica BiomedicalLjubljanaSlovenia
- Department of Biology, Biotechnical FacultyUniversity of LjubljanaLjubljanaSlovenia
| | - Robert Zorec
- Laboratory of Neuroendocrinology‐Molecular Cell Physiology, Faculty of Medicine, Institute of PathophysiologyUniversity of LjubljanaLjubljanaSlovenia
- Laboratory of Cell Engineering, Celica BiomedicalLjubljanaSlovenia
| | - Nicole Scholz
- Division of General Biochemistry, Medical Faculty, Rudolf Schönheimer Institute of BiochemistryLeipzig UniversityLeipzigGermany
| | - Nina Vardjan
- Laboratory of Neuroendocrinology‐Molecular Cell Physiology, Faculty of Medicine, Institute of PathophysiologyUniversity of LjubljanaLjubljanaSlovenia
- Laboratory of Cell Engineering, Celica BiomedicalLjubljanaSlovenia
| |
Collapse
|
2
|
Verkhratsky A, Zorec R. Neuroglia in cognitive reserve. Mol Psychiatry 2024; 29:3962-3967. [PMID: 38956370 PMCID: PMC11609093 DOI: 10.1038/s41380-024-02644-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 06/18/2024] [Accepted: 06/21/2024] [Indexed: 07/04/2024]
Abstract
The concept of cognitive reserve was born to account for the disjunction between the objective extent of brain damage in pathology and its clinical and intellectual outcome. The cognitive reserve comprises structural (brain reserve) and functional (brain maintenance, resilience, compensation) aspects of the nervous tissue reflecting exposome-driven life-long plasticity, which defines the ability of the brain to withstand aging and pathology. The mechanistic background of this concept was primarily focused on adaptive changes in neurones and neuronal networks. We present arguments favoring the more inclusive view, positing that neuroglia are fundamental for defining the cognitive reserve through homeostatic, neuroprotective, and neurodegenerative mechanisms. Neuroglia are critical for the life-long shaping of synaptically connected neuronal circuits as well as the brain connectome thus defining cognitive reserve. Neuroglial homeostatic and protective physiological responses define brain maintenance and resilience, while neuroglia regenerative capabilities are critical for brain compensation in pathology. Targeting neuroglia may represent an untrodden path for prolonging cognitive longevity.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK.
- Department of Neurosciences, University of the Basque Country, 48940, Leioa, Bizkaia, Spain.
- IKERBASQUE Basque Foundation for Science, Bilbao, Spain.
- University of Ljubljana, Institute of Pathophysiology, Laboratory of Neuroendocrinology and Molecular Cell Physiology, Zaloška cesta 4, SI-1000, Ljubljana, Slovenia.
- Celica, BIOMEDICAL, Technology Park 24, 1000, Ljubljana, Slovenia.
| | - Robert Zorec
- University of Ljubljana, Institute of Pathophysiology, Laboratory of Neuroendocrinology and Molecular Cell Physiology, Zaloška cesta 4, SI-1000, Ljubljana, Slovenia.
- Celica, BIOMEDICAL, Technology Park 24, 1000, Ljubljana, Slovenia.
| |
Collapse
|
3
|
Tao Y, Li X, Dong Q, Kong L, Petersen AJ, Yan Y, Xu K, Zima S, Li Y, Schmidt DK, Ayala M, Mathivanan S, Sousa AMM, Chang Q, Zhang SC. Generation of locus coeruleus norepinephrine neurons from human pluripotent stem cells. Nat Biotechnol 2024; 42:1404-1416. [PMID: 37974010 PMCID: PMC11392812 DOI: 10.1038/s41587-023-01977-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 08/30/2023] [Indexed: 11/19/2023]
Abstract
Central norepinephrine (NE) neurons, located mainly in the locus coeruleus (LC), are implicated in diverse psychiatric and neurodegenerative diseases and are an emerging target for drug discovery. To facilitate their study, we developed a method to generate 40-60% human LC-NE neurons from human pluripotent stem cells. The approach depends on our identification of ACTIVIN A in regulating LC-NE transcription factors in dorsal rhombomere 1 (r1) progenitors. In vitro generated human LC-NE neurons display extensive axonal arborization; release and uptake NE; and exhibit pacemaker activity, calcium oscillation and chemoreceptor activity in response to CO2. Single-nucleus RNA sequencing (snRNA-seq) analysis at multiple timepoints confirmed NE cell identity and revealed the differentiation trajectory from hindbrain progenitors to NE neurons via an ASCL1-expressing precursor stage. LC-NE neurons engineered with an NE sensor reliably reported extracellular levels of NE. The availability of functional human LC-NE neurons enables investigation of their roles in psychiatric and neurodegenerative diseases and provides a tool for therapeutics development.
Collapse
Affiliation(s)
- Yunlong Tao
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA.
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, China.
| | - Xueyan Li
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Qiping Dong
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Linghai Kong
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | | | - Yuanwei Yan
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Ke Xu
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Seth Zima
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Yanru Li
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | | | - Melvin Ayala
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | | | - Andre M M Sousa
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Neuroscience, Department of Neurology, University of Wisconsin-Madison, Madison, WI, USA
| | - Qiang Chang
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Neuroscience, Department of Neurology, University of Wisconsin-Madison, Madison, WI, USA
| | - Su-Chun Zhang
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA.
- Department of Neuroscience, Department of Neurology, University of Wisconsin-Madison, Madison, WI, USA.
- Program in Neuroscience and Behavioral Disorders, Duke-NUS Medical School, Singapore, Singapore.
| |
Collapse
|
4
|
Rae CD, Baur JA, Borges K, Dienel G, Díaz-García CM, Douglass SR, Drew K, Duarte JMN, Duran J, Kann O, Kristian T, Lee-Liu D, Lindquist BE, McNay EC, Robinson MB, Rothman DL, Rowlands BD, Ryan TA, Scafidi J, Scafidi S, Shuttleworth CW, Swanson RA, Uruk G, Vardjan N, Zorec R, McKenna MC. Brain energy metabolism: A roadmap for future research. J Neurochem 2024; 168:910-954. [PMID: 38183680 PMCID: PMC11102343 DOI: 10.1111/jnc.16032] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 11/29/2023] [Accepted: 12/05/2023] [Indexed: 01/08/2024]
Abstract
Although we have learned much about how the brain fuels its functions over the last decades, there remains much still to discover in an organ that is so complex. This article lays out major gaps in our knowledge of interrelationships between brain metabolism and brain function, including biochemical, cellular, and subcellular aspects of functional metabolism and its imaging in adult brain, as well as during development, aging, and disease. The focus is on unknowns in metabolism of major brain substrates and associated transporters, the roles of insulin and of lipid droplets, the emerging role of metabolism in microglia, mysteries about the major brain cofactor and signaling molecule NAD+, as well as unsolved problems underlying brain metabolism in pathologies such as traumatic brain injury, epilepsy, and metabolic downregulation during hibernation. It describes our current level of understanding of these facets of brain energy metabolism as well as a roadmap for future research.
Collapse
Affiliation(s)
- Caroline D. Rae
- School of Psychology, The University of New South Wales, NSW 2052 & Neuroscience Research Australia, Randwick, New South Wales, Australia
| | - Joseph A. Baur
- Department of Physiology and Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Karin Borges
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, QLD, Australia
| | - Gerald Dienel
- Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Department of Cell Biology and Physiology, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| | - Carlos Manlio Díaz-García
- Department of Biochemistry and Molecular Biology, Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | | | - Kelly Drew
- Center for Transformative Research in Metabolism, Institute of Arctic Biology, University of Alaska Fairbanks, Fairbanks, Alaska, USA
| | - João M. N. Duarte
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, Lund, & Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - Jordi Duran
- Institut Químic de Sarrià (IQS), Universitat Ramon Llull (URL), Barcelona, Spain
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Oliver Kann
- Institute of Physiology and Pathophysiology, University of Heidelberg, D-69120; Interdisciplinary Center for Neurosciences (IZN), University of Heidelberg, Heidelberg, Germany
| | - Tibor Kristian
- Veterans Affairs Maryland Health Center System, Baltimore, Maryland, USA
- Department of Anesthesiology and the Center for Shock, Trauma, and Anesthesiology Research (S.T.A.R.), University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Dasfne Lee-Liu
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Región Metropolitana, Chile
| | - Britta E. Lindquist
- Department of Neurology, Division of Neurocritical Care, Gladstone Institute of Neurological Disease, University of California at San Francisco, San Francisco, California, USA
| | - Ewan C. McNay
- Behavioral Neuroscience, University at Albany, Albany, New York, USA
| | - Michael B. Robinson
- Departments of Pediatrics and System Pharmacology & Translational Therapeutics, Children’s Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Douglas L. Rothman
- Magnetic Resonance Research Center and Departments of Radiology and Biomedical Engineering, Yale University, New Haven, Connecticut, USA
| | - Benjamin D. Rowlands
- School of Chemistry, Faculty of Science, The University of Sydney, Sydney, New South Wales, Australia
| | - Timothy A. Ryan
- Department of Biochemistry, Weill Cornell Medicine, New York, New York, USA
| | - Joseph Scafidi
- Department of Neurology, Kennedy Krieger Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Susanna Scafidi
- Anesthesiology & Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - C. William Shuttleworth
- Department of Neurosciences, University of New Mexico School of Medicine Albuquerque, Albuquerque, New Mexico, USA
| | - Raymond A. Swanson
- Department of Neurology, University of California, San Francisco, and San Francisco Veterans Affairs Medical Center, San Francisco, California, USA
| | - Gökhan Uruk
- Department of Neurology, University of California, San Francisco, and San Francisco Veterans Affairs Medical Center, San Francisco, California, USA
| | - Nina Vardjan
- Laboratory of Cell Engineering, Celica Biomedical, Ljubljana, Slovenia
- Laboratory of Neuroendocrinology—Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Robert Zorec
- Laboratory of Cell Engineering, Celica Biomedical, Ljubljana, Slovenia
- Laboratory of Neuroendocrinology—Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Mary C. McKenna
- Department of Pediatrics and Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
5
|
Matt RA, Martin RS, Evans AK, Gever JR, Vargas GA, Shamloo M, Ford AP. Locus Coeruleus and Noradrenergic Pharmacology in Neurodegenerative Disease. Handb Exp Pharmacol 2024; 285:555-616. [PMID: 37495851 DOI: 10.1007/164_2023_677] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
Adrenoceptors (ARs) throughout the brain are stimulated by noradrenaline originating mostly from neurons of the locus coeruleus, a brainstem nucleus that is ostensibly the earliest to show detectable pathology in neurodegenerative diseases such as Alzheimer's and Parkinson's diseases. The α1-AR, α2-AR, and β-AR subtypes expressed in target brain regions and on a range of cell populations define the physiological responses to noradrenaline, which includes activation of cognitive function in addition to modulation of neurometabolism, cerebral blood flow, and neuroinflammation. As these heterocellular functions are critical for maintaining brain homeostasis and neuronal health, combating the loss of noradrenergic tone from locus coeruleus degeneration may therefore be an effective treatment for both cognitive symptoms and disease modification in neurodegenerative indications. Two pharmacologic approaches are receiving attention in recent clinical studies: preserving noradrenaline levels (e.g., via reuptake inhibition) and direct activation of target adrenoceptors. Here, we review the expression and role of adrenoceptors in the brain, the preclinical studies which demonstrate that adrenergic stimulation can support cognitive function and cerebral health by reversing the effects of noradrenaline depletion, and the human data provided by pharmacoepidemiologic analyses and clinical trials which together identify adrenoceptors as promising targets for the treatment of neurodegenerative disease.
Collapse
Affiliation(s)
| | | | - Andrew K Evans
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, CA, USA
| | | | | | - Mehrdad Shamloo
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, CA, USA
| | | |
Collapse
|
6
|
Wang D, Zheng J, Sun X, Xie L, Yang Y. Study on the Pharmacological Mechanism of Icariin for the Treatment of Alzheimer's Disease Based on Network Pharmacology and Molecular Docking Techniques. Metabolites 2023; 14:1. [PMID: 38276291 PMCID: PMC10820555 DOI: 10.3390/metabo14010001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 12/12/2023] [Accepted: 12/14/2023] [Indexed: 01/27/2024] Open
Abstract
The purpose of this study is to explore the pharmacological mechanism of icariin (ICA) in the treatment of Alzheimer's disease (AD) based on network pharmacology and network molecular docking technology. In order to investigate the regulatory effect of ICA on the expression level of AD pathological phosphorylation regulatory proteins, this study further explored the possible molecular mechanism of ICA regulating AD autophagy through network pharmacology. Macromolecular docking network was verified by Autodock Vina 1.1.2 software. The main active ingredients of ICA, the physicochemical properties, and pharmacokinetic information of ICA were predicted using online databases and relevant information. The results showed that the targets of MAPK3, AKT1, HSP90AA1, ESR1, and HSP90AA1 were more critical in the treatment of AD. Autophagy, apoptosis, senescence factors, phosphatidylinositide 3-kinase/protein kinase B (P13K/AKT) signaling pathway, MAKP, mTOR, and other pathways were significantly associated with AD. Docking of ICA with HIF-1, BNIP3, PINK1, and Parkin pathway molecules showed that the key targets of the signaling pathway were more stably bound to ICA, which may provide a better pathway for ICA to regulate autophagy by providing a better pathway. ICA can improve AD, and its mechanism may be related to the P13K/AKT, MAKP, and mTOR signaling pathways, thereby regulating autophagy-related proteins.
Collapse
Affiliation(s)
- Dongwei Wang
- College of Police Dog Technology, Criminal Investigation Police University of China, Shenyang 110854, China; (D.W.); (J.Z.); (X.S.); (L.X.)
| | - Jilong Zheng
- College of Police Dog Technology, Criminal Investigation Police University of China, Shenyang 110854, China; (D.W.); (J.Z.); (X.S.); (L.X.)
| | - Xingsheng Sun
- College of Police Dog Technology, Criminal Investigation Police University of China, Shenyang 110854, China; (D.W.); (J.Z.); (X.S.); (L.X.)
| | - Liuwei Xie
- College of Police Dog Technology, Criminal Investigation Police University of China, Shenyang 110854, China; (D.W.); (J.Z.); (X.S.); (L.X.)
- The Second Affiliated Hospital of Shenyang Medical College, Shenyang 110031, China
| | - Yang Yang
- The Second Affiliated Hospital of Shenyang Medical College, Shenyang 110031, China
| |
Collapse
|
7
|
Matt RA, Westhorpe FG, Romuar RF, Rana P, Gever JR, Ford AP. Fingerprinting heterocellular β-adrenoceptor functional expression in the brain using agonist activity profiles. Front Mol Biosci 2023; 10:1214102. [PMID: 37664183 PMCID: PMC10471193 DOI: 10.3389/fmolb.2023.1214102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 07/05/2023] [Indexed: 09/05/2023] Open
Abstract
Noradrenergic projections from the brainstem locus coeruleus drive arousal, attentiveness, mood, and memory, but specific adrenoceptor (AR) function across the varied brain cell types has not been extensively characterized, especially with agonists. This study reports a pharmacological analysis of brain AR function, offering insights for innovative therapeutic interventions that might serve to compensate for locus coeruleus decline, known to develop in the earliest phases of neurodegenerative diseases. First, β-AR agonist activities were measured in recombinant cell systems and compared with those of isoprenaline to generate Δlog(Emax/EC50) values, system-independent metrics of agonist activity, that, in turn, provide receptor subtype fingerprints. These fingerprints were then used to assess receptor subtype expression across human brain cell systems and compared with Δlog(Emax/EC50) values arising from β-arrestin activation or measurements of cAMP response desensitization to assess the possibility of ligand bias among β-AR agonists. Agonist activity profiles were confirmed to be system-independent and, in particular, revealed β2-AR functional expression across several human brain cell types. Broad β2-AR function observed is consistent with noradrenergic tone arising from the locus coeruleus exerting heterocellular neuroexcitatory and homeostatic influence. Notably, Δlog(Emax/EC50) measurements suggest that tested β-AR agonists do not show ligand bias as it pertains to homologous receptor desensitization in the system examined. Δlog(Emax/EC50) agonist fingerprinting is a powerful means of assessing receptor subtype expression regardless of receptor expression levels or assay readout, and the method may be applicable to future use for novel ligands and tissues expressing any receptor with available reference agonists.
Collapse
|
8
|
Zorec R, Vardjan N. Adrenergic regulation of astroglial aerobic glycolysis and lipid metabolism: Towards a noradrenergic hypothesis of neurodegeneration. Neurobiol Dis 2023; 182:106132. [PMID: 37094775 DOI: 10.1016/j.nbd.2023.106132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 04/18/2023] [Accepted: 04/19/2023] [Indexed: 04/26/2023] Open
Abstract
Ageing is a key factor in the development of cognitive decline and dementia, an increasing and challenging problem of the modern world. The most commonly diagnosed cognitive decline is related to Alzheimer's disease (AD), the pathophysiology of which is poorly understood. Several hypotheses have been proposed. The cholinergic hypothesis is the oldest, however, recently the noradrenergic system has been considered to have a role as well. The aim of this review is to provide evidence that supports the view that an impaired noradrenergic system is causally linked to AD. Although dementia is associated with neurodegeneration and loss of neurons, this likely develops due to a primary failure of homeostatic cells, astrocytes, abundant and heterogeneous neuroglial cells in the central nervous system (CNS). The many functions that astrocytes provide to maintain the viability of neural networks include the control of ionic balance, neurotransmitter turnover, synaptic connectivity and energy balance. This latter function is regulated by noradrenaline, released from the axon varicosities of neurons arising from the locus coeruleus (LC), the primary site of noradrenaline release in the CNS. The demise of the LC is linked to AD, whereby a hypometabolic CNS state is observed clinically. This is likely due to impaired release of noradrenaline in the AD brain during states of arousal, attention and awareness. These functions controlled by the LC are needed for learning and memory formation and require activation of the energy metabolism. In this review, we address first the process of neurodegeneration and cognitive decline, highlighting the function of astrocytes. Cholinergic and/or noradrenergic deficits lead to impaired astroglial function. Then, we focus on adrenergic control of astroglial aerobic glycolysis and lipid droplet metabolism, which play a protective role but also promote neurodegeneration under some circumstances, supporting the noradrenergic hypothesis of cognitive decline. We conclude that targeting astroglial metabolism, glycolysis and/or mitochondrial processes may lead to important new developments in the future when searching for medicines to prevent or even halt cognitive decline.
Collapse
Affiliation(s)
- Robert Zorec
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia; Laboratory of Cell Engineering, Celica Biomedical, Ljubljana, Slovenia.
| | - Nina Vardjan
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia; Laboratory of Cell Engineering, Celica Biomedical, Ljubljana, Slovenia.
| |
Collapse
|
9
|
Hippocampal Noradrenaline Is a Positive Regulator of Spatial Working Memory and Neurogenesis in the Rat. Int J Mol Sci 2023; 24:ijms24065613. [PMID: 36982688 PMCID: PMC10052298 DOI: 10.3390/ijms24065613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 02/22/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023] Open
Abstract
Loss of noradrenaline (NA)-rich afferents from the Locus Coeruleus (LC) ascending to the hippocampal formation has been reported to dramatically affect distinct aspects of cognitive function, in addition to reducing the proliferation of neural progenitors in the dentate gyrus. Here, the hypothesis that reinstating hippocampal noradrenergic neurotransmission with transplanted LC-derived neuroblasts would concurrently normalize both cognitive performance and adult hippocampal neurogenesis was investigated. Post-natal day (PD) 4 rats underwent selective immunolesioning of hippocampal noradrenergic afferents followed, 4 days later, by the bilateral intrahippocampal implantation of LC noradrenergic-rich or control cerebellar (CBL) neuroblasts. Starting from 4 weeks and up to about 9 months post-surgery, sensory-motor and spatial navigation abilities were evaluated, followed by post-mortem semiquantitative tissue analyses. All animals in the Control, Lesion, Noradrenergic Transplant and Control CBL Transplant groups exhibited normal sensory-motor function and were equally efficient in the reference memory version of the water maze task. By contrast, working memory abilities were seen to be consistently impaired in the Lesion-only and Control CBL-Transplanted rats, which also exhibited a virtually complete noradrenergic fiber depletion and a significant 62–65% reduction in proliferating 5-bromo-2′deoxyuridine (BrdU)-positive progenitors in the dentate gyrus. Notably, the noradrenergic reinnervation promoted by the grafted LC, but not cerebellar neuroblasts, significantly ameliorated working memory performance and reinstated a fairly normal density of proliferating progenitors. Thus, LC-derived noradrenergic inputs may act as positive regulators of hippocampus-dependent spatial working memory possibly via the concurrent maintenance of normal progenitor proliferation in the dentate gyrus.
Collapse
|
10
|
Leanza G, Zorec R. Towards Astroglia-based Noradrenergic Hypothesis of Alzheimer's Disease. FUNCTION (OXFORD, ENGLAND) 2022; 4:zqac060. [PMID: 36590326 PMCID: PMC9789502 DOI: 10.1093/function/zqac060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 12/01/2022] [Indexed: 12/25/2022]
Affiliation(s)
- Giampiero Leanza
- Dept. of Drug and Health Sciences, University of Catania, Piazza Università, 2, 95131 Catania, Italy
| | | |
Collapse
|
11
|
Infantino R, Boccella S, Scuteri D, Perrone M, Ricciardi F, Vitale R, Bonsale R, Parente A, Allocca I, Virtuoso A, De Luca C, Belardo C, Amodeo P, Gentile V, Cirillo G, Bagetta G, Luongo L, Maione S, Guida F. 2-pentadecyl-2-oxazoline prevents cognitive and social behaviour impairments in the Amyloid β-induced Alzheimer-like mice model: Bring the α2 adrenergic receptor back into play. Biomed Pharmacother 2022; 156:113844. [DOI: 10.1016/j.biopha.2022.113844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/03/2022] [Accepted: 10/06/2022] [Indexed: 11/15/2022] Open
|
12
|
Bolton CJ, Tam JW. Differential Involvement of the Locus Coeruleus in Early- and Late-Onset Alzheimer's Disease: A Potential Mechanism of Clinical Differences? J Geriatr Psychiatry Neurol 2022; 35:733-739. [PMID: 34496652 PMCID: PMC12023724 DOI: 10.1177/08919887211044755] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Sporadic early-onset Alzheimer's disease (sEOAD) is often associated with atypical clinical features, yet the cause of this heterogeneity remains unclear. This study investigated post-mortem atrophy of the locus coeruleus (LC) in sEOAD and late-onset Alzheimer's disease (LOAD). Levels of LC atrophy, as estimated by pathologist-rating of hypopigmentation, were compared between sEOAD (n = 115) and LOAD (n = 672) participants while controlling for other measures of pathological progression. Subsequent analyses compared low vs. high LC atrophy sEOAD subgroups on neuropsychological test performance. Results show nearly 4 times greater likelihood of higher LC atrophy in sEOAD as compared to LOAD (p < .005). sEOAD participants with greater LC atrophy displayed significantly worse performance on various baseline measures of attentional functioning (p < .05), despite similar global cognition (p = .25). These findings suggest the LC is an important potential driver of clinical and pathological heterogeneity in sEOAD.
Collapse
Affiliation(s)
- Corey J. Bolton
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Joyce W. Tam
- Department of Psychiatry and Behavioral Sciences, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
13
|
Navarro-Hortal MD, Romero-Márquez JM, Osta S, Jiménez-Trigo V, Muñoz-Ollero P, Varela-López A. Natural Bioactive Products and Alzheimer’s Disease Pathology: Lessons from Caenorhabditis elegans Transgenic Models. Diseases 2022; 10:diseases10020028. [PMID: 35645249 PMCID: PMC9149938 DOI: 10.3390/diseases10020028] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 04/28/2022] [Accepted: 05/04/2022] [Indexed: 02/04/2023] Open
Abstract
Alzheimer’s disease (AD) is an age-dependent, progressive disorder affecting millions of people. Currently, the therapeutics for AD only treat the symptoms. Although they have been used to discover new products of interest for this disease, mammalian models used to investigate the molecular determinants of this disease are often prohibitively expensive, time-consuming and very complex. On the other hand, cell cultures lack the organism complexity involved in AD. Given the highly conserved neurological pathways between mammals and invertebrates, Caenorhabditis elegans has emerged as a powerful tool for the investigation of the pathophysiology of human AD. Numerous models of both Tau- and Aβ-induced toxicity, the two prime components observed to correlate with AD pathology and the ease of performing RNA interference for any gene in the C. elegans genome, allow for the identification of multiple therapeutic targets. The effects of many natural products in main AD hallmarks using these models suggest promising health-promoting effects. However, the way in which they exert such effects is not entirely clear. One of the reasons is that various possible therapeutic targets have not been evaluated in many studies. The present review aims to explore shared therapeutical targets and the potential of each of them for AD treatment or prevention.
Collapse
|
14
|
Saporin as a Commercial Reagent: Its Uses and Unexpected Impacts in the Biological Sciences—Tools from the Plant Kingdom. Toxins (Basel) 2022; 14:toxins14030184. [PMID: 35324681 PMCID: PMC8952126 DOI: 10.3390/toxins14030184] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/23/2022] [Accepted: 02/23/2022] [Indexed: 02/02/2023] Open
Abstract
Saporin is a ribosome-inactivating protein that can cause inhibition of protein synthesis and causes cell death when delivered inside a cell. Development of commercial Saporin results in a technology termed ‘molecular surgery’, with Saporin as the scalpel. Its low toxicity (it has no efficient method of cell entry) and sturdy structure make Saporin a safe and simple molecule for many purposes. The most popular applications use experimental molecules that deliver Saporin via an add-on targeting molecule. These add-ons come in several forms: peptides, protein ligands, antibodies, even DNA fragments that mimic cell-binding ligands. Cells that do not express the targeted cell surface marker will not be affected. This review will highlight some newer efforts and discuss significant and unexpected impacts on science that molecular surgery has yielded over the last almost four decades. There are remarkable changes in fields such as the Neurosciences with models for Alzheimer’s Disease and epilepsy, and game-changing effects in the study of pain and itch. Many other uses are also discussed to record the wide-reaching impact of Saporin in research and drug development.
Collapse
|
15
|
Gray SR, Ye L, Ye JY, Paukert M. Noradrenergic terminal short-term potentiation enables modality-selective integration of sensory input and vigilance state. SCIENCE ADVANCES 2021; 7:eabk1378. [PMID: 34919424 PMCID: PMC8682997 DOI: 10.1126/sciadv.abk1378] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 10/28/2021] [Indexed: 06/14/2023]
Abstract
Recent years have seen compelling demonstrations of the importance of behavioral state on sensory processing and attention. Arousal plays a dominant role in controlling brain-wide neural activity patterns, particularly through modulation by norepinephrine. Noradrenergic brainstem nuclei, including locus coeruleus, can be activated by stimuli of multiple sensory modalities and broadcast modulatory signals via axonal projections throughout the brain. This organization might suggest proportional brain-wide norepinephrine release during states of heightened vigilance. Here, however, we have found that low-intensity, nonarousing visual stimuli enhanced vigilance-dependent noradrenergic signaling locally in visual cortex, revealed using dual-site fiber photometry to monitor noradrenergic Ca2+ responses of astroglia simultaneously in cerebellum and visual cortex and two-photon microscopy to monitor noradrenergic axonal terminal Ca2+ dynamics. Nitric oxide, following N-methyl-d-aspartate receptor activation in neuronal nitric oxide synthase-positive interneurons, mediated transient acceleration of norepinephrine-dependent astroglia Ca2+ activation. These findings reveal a candidate cortical microcircuit for sensory modality-selective modulation of attention.
Collapse
Affiliation(s)
- Shawn R. Gray
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Joint UTSA/UTHSCSA Graduate Program in Biomedical Engineering, San Antonio, TX, USA
| | - Liang Ye
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Jing Yong Ye
- Joint UTSA/UTHSCSA Graduate Program in Biomedical Engineering, San Antonio, TX, USA
- Department of Biomedical Engineering, University of Texas at San Antonio, San Antonio, TX, USA
| | - Martin Paukert
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Joint UTSA/UTHSCSA Graduate Program in Biomedical Engineering, San Antonio, TX, USA
- Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| |
Collapse
|
16
|
Fernandes C, Macedo I, Barbosa F, Marques-Teixeira J. Economic decision-making in the continuum between healthy aging and Alzheimer's Disease: A systematic review of 20 years of research. Neurosci Biobehav Rev 2021; 131:1243-1263. [PMID: 34715151 DOI: 10.1016/j.neubiorev.2021.10.030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 09/23/2021] [Accepted: 10/23/2021] [Indexed: 01/21/2023]
Abstract
The effect of pathological aging on economic decision-making is a topic of major relevance as impairments in this domain place older adults at increased risk for financial abuse. This review aims to characterize decision-making across the continuum that goes from healthy aging to Alzheimer's Dementia. We included 42 studies comparing patients with Mild Cognitive Impairment (MCI) and healthy older adults, patients with Alzheimer's Disease (AD) and healthy older adults, and patients with MCI and patients with AD. Substantial evidence emerged suggesting that both MCI as AD affect economic decision-making. However, a non-negligible number of behavioural tasks failed to find significant differences between patients and controls, and no differences were reported between patients with MCI and AD. On the contrary, measures of financial capacity reached more robust findings, showing that healthy older adults had better performance than patients, while MCI patients showed better performance than AD patients. This review presents the main conclusions that may be drawn from significant findings, as well as the hypotheses and recommendations for future research.
Collapse
Affiliation(s)
- Carina Fernandes
- Laboratory of Neuropsychophysiology, Faculty of Psychology and Education Sciences, University of Porto, Portugal.
| | - Inês Macedo
- Laboratory of Neuropsychophysiology, Faculty of Psychology and Education Sciences, University of Porto, Portugal
| | - Fernando Barbosa
- Laboratory of Neuropsychophysiology, Faculty of Psychology and Education Sciences, University of Porto, Portugal
| | - João Marques-Teixeira
- Laboratory of Neuropsychophysiology, Faculty of Psychology and Education Sciences, University of Porto, Portugal
| |
Collapse
|
17
|
Lalo U, Pankratov Y. Astrocytes as Perspective Targets of Exercise- and Caloric Restriction-Mimetics. Neurochem Res 2021; 46:2746-2759. [PMID: 33677759 PMCID: PMC8437875 DOI: 10.1007/s11064-021-03277-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 02/16/2021] [Accepted: 02/17/2021] [Indexed: 12/20/2022]
Abstract
Enhanced mental and physical activity can have positive effects on the function of aging brain, both in the experimental animals and human patients, although cellular mechanisms underlying these effects are currently unclear. There is a growing evidence that pre-clinical stage of many neurodegenerative diseases involves changes in interactions between astrocytes and neurons. Conversely, astrocytes are strategically positioned to mediate the positive influence of physical activity and diet on neuronal function. Thus, development of therapeutic agents which could improve the astroglia-neuron communications in ageing brain is of crucial importance. Recent advances in studies of cellular mechanisms of brain longevity suggest that astrocyte-neuron communications have a vital role in the beneficial effects of caloric restriction, physical exercise and their pharmacological mimetics on synaptic homeostasis and cognitive function. In particular, our recent data indicate that noradrenaline uptake inhibitor atomoxetine can enhance astrocytic Ca2+-signaling and astroglia-driven modulation of synaptic plasticity. Similar effects were exhibited by caloric restriction-mimetics metformin and resveratrol. The emerged data also suggest that astrocytes could be involved in the modulatory action of caloric restriction and its mimetics on neuronal autophagy. Still, the efficiency of astrocyte-targeting compounds in preventing age-related cognitive decline is yet to be fully explored, in particular in the animal models of neurodegenerative diseases and autophagy impairment.
Collapse
Affiliation(s)
- Ulyana Lalo
- School of Life Sciences, Immanuel Kant Baltic Federal University, Kaliningrad, Russia
- School of Life Sciences, University of Warwick, Coventry, UK
| | - Yuriy Pankratov
- School of Life Sciences, Immanuel Kant Baltic Federal University, Kaliningrad, Russia.
- School of Life Sciences, University of Warwick, Coventry, UK.
| |
Collapse
|
18
|
Gallo A, Pillet LE, Verpillot R. New frontiers in Alzheimer's disease diagnostic: Monoamines and their derivatives in biological fluids. Exp Gerontol 2021; 152:111452. [PMID: 34182050 DOI: 10.1016/j.exger.2021.111452] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 04/29/2021] [Accepted: 06/08/2021] [Indexed: 10/21/2022]
Abstract
Current diagnosis of Alzheimer's disease (AD) relies on a combination of neuropsychological evaluations, biomarker measurements and brain imaging. Nevertheless, these approaches are either expensive, invasive or lack sensitivity to early AD stages. The main challenge of ongoing research is therefore to identify early non-invasive biomarkers to diagnose AD at preclinical stage. Accumulating evidence support the hypothesis that initial degeneration of profound monoaminergic nuclei may trigger a transneuronal spread of AD pathology towards hippocampus and cortex. These studies aroused great interest on monoamines, i.e. noradrenaline (NA), dopamine (D) ad serotonin (5-HT), as early hallmarks of AD pathology. The present work reviews current literature on the potential role of monoamines and related metabolites as biomarkers of AD. First, morphological changes in the monoaminergic systems during AD are briefly described. Second, we focus on concentration changes of these molecules and their derivatives in biological fluids, including cerebrospinal fluid, obtained by lumbar puncture, and blood or urine, sampled via less invasive procedures. Starting from initial observations, we then discuss recent insights on metabolomics-based analysis, highlighting the promising clinical utility of monoamines for the identification of a molecular AD signature, aimed at improving early diagnosis and discrimination from other dementia.
Collapse
|
19
|
Rivera AD, Chacon-De-La-Rocha I, Pieropan F, Papanikolau M, Azim K, Butt AM. Keeping the ageing brain wired: a role for purine signalling in regulating cellular metabolism in oligodendrocyte progenitors. Pflugers Arch 2021; 473:775-783. [PMID: 33712969 PMCID: PMC8076121 DOI: 10.1007/s00424-021-02544-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 02/04/2021] [Accepted: 02/19/2021] [Indexed: 02/07/2023]
Abstract
White matter (WM) is a highly prominent feature in the human cerebrum and is comprised of bundles of myelinated axons that form the connectome of the brain. Myelin is formed by oligodendrocytes and is essential for rapid neuronal electrical communication that underlies the massive computing power of the human brain. Oligodendrocytes are generated throughout life by oligodendrocyte precursor cells (OPCs), which are identified by expression of the chondroitin sulphate proteoglycan NG2 (Cspg4), and are often termed NG2-glia. Adult NG2+ OPCs are slowly proliferating cells that have the stem cell-like property of self-renewal and differentiation into a pool of 'late OPCs' or 'differentiation committed' OPCs(COPs) identified by specific expression of the G-protein-coupled receptor GPR17, which are capable of differentiation into myelinating oligodendrocytes. In the adult brain, these reservoirs of OPCs and COPs ensure rapid myelination of new neuronal connections formed in response to neuronal signalling, which underpins learning and cognitive function. However, there is an age-related decline in myelination that is associated with a loss of neuronal function and cognitive decline. The underlying causes of myelin loss in ageing are manifold, but a key factor is the decay in OPC 'stemness' and a decline in their replenishment of COPs, which results in the ultimate failure of myelin regeneration. These changes in ageing OPCs are underpinned by dysregulation of neuronal signalling and OPC metabolic function. Here, we highlight the role of purine signalling in regulating OPC self-renewal and the potential importance of GPR17 and the P2X7 receptor subtype in age-related changes in OPC metabolism. Moreover, age is the main factor in the failure of myelination in chronic multiple sclerosis and myelin loss in Alzheimer's disease, hence understanding the importance of purine signalling in OPC regeneration and myelination is critical for developing new strategies for promoting repair in age-dependent neuropathology.
Collapse
Affiliation(s)
- Andrea D Rivera
- School of Pharmacy and Biomedical Science, University of Portsmouth, Portsmouth, UK
- Department of Neuroscience, Institute of Human Anatomy, University of Padua, Padua, Italy
| | | | - Francesca Pieropan
- School of Pharmacy and Biomedical Science, University of Portsmouth, Portsmouth, UK
| | - Maria Papanikolau
- School of Pharmacy and Biomedical Science, University of Portsmouth, Portsmouth, UK
| | - Kasum Azim
- Department of Neurology, Neuroregeneration, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Arthur M Butt
- School of Pharmacy and Biomedical Science, University of Portsmouth, Portsmouth, UK.
| |
Collapse
|
20
|
Horvat A, Muhič M, Smolič T, Begić E, Zorec R, Kreft M, Vardjan N. Ca 2+ as the prime trigger of aerobic glycolysis in astrocytes. Cell Calcium 2021; 95:102368. [PMID: 33621899 DOI: 10.1016/j.ceca.2021.102368] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 02/05/2021] [Accepted: 02/08/2021] [Indexed: 12/17/2022]
Abstract
Astroglial aerobic glycolysis, a process during which d-glucose is converted to l-lactate, a brain fuel and signal, is regulated by the plasmalemmal receptors, including adrenergic receptors (ARs) and purinergic receptors (PRs), modulating intracellular Ca2+ and cAMP signals. However, the extent to which the two signals regulate astroglial aerobic glycolysis is poorly understood. By using agonists to stimulate intracellular α1-/β-AR-mediated Ca2+/cAMP signals, β-AR-mediated cAMP and P2R-mediated Ca2+ signals and genetically encoded fluorescence resonance energy transfer-based glucose and lactate nanosensors in combination with real-time microscopy, we show that intracellular Ca2+, but not cAMP, initiates a robust increase in the concentration of intracellular free d-glucose ([glc]i) and l-lactate ([lac]i), both depending on extracellular d-glucose, suggesting Ca2+-triggered glucose uptake and aerobic glycolysis in astrocytes. When the glycogen shunt, a process of glycogen remodelling, was inhibited, the α1-/β-AR-mediated increases in [glc]i and [lac]i were reduced by ∼65 % and ∼30 %, respectively, indicating that at least ∼30 % of the utilization of d-glucose is linked to glycogen remodelling and aerobic glycolysis. Additional activation of β-AR/cAMP signals aided to α1-/β-AR-triggered [lac]i increase, whereas the [glc]i increase was unaltered. Taken together, an increase in intracellular Ca2+ is the prime mechanism of augmented aerobic glycolysis in astrocytes, while cAMP has only a moderate role. The results provide novel information on the signals regulating brain metabolism and open new avenues to explore whether astroglial Ca2+ signals are dysregulated and contribute to neuropathologies with impaired brain metabolism.
Collapse
Affiliation(s)
- Anemari Horvat
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia; Laboratory of Cell Engineering, Celica Biomedical, Ljubljana, Slovenia
| | - Marko Muhič
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Tina Smolič
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Ena Begić
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Robert Zorec
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia; Laboratory of Cell Engineering, Celica Biomedical, Ljubljana, Slovenia
| | - Marko Kreft
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia; Laboratory of Cell Engineering, Celica Biomedical, Ljubljana, Slovenia; Department of Biology, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Nina Vardjan
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia; Laboratory of Cell Engineering, Celica Biomedical, Ljubljana, Slovenia.
| |
Collapse
|
21
|
Bekdash RA. The Cholinergic System, the Adrenergic System and the Neuropathology of Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms22031273. [PMID: 33525357 PMCID: PMC7865740 DOI: 10.3390/ijms22031273] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 01/23/2021] [Accepted: 01/25/2021] [Indexed: 12/16/2022] Open
Abstract
Neurodegenerative diseases are a major public health problem worldwide with a wide spectrum of symptoms and physiological effects. It has been long reported that the dysregulation of the cholinergic system and the adrenergic system are linked to the etiology of Alzheimer’s disease. Cholinergic neurons are widely distributed in brain regions that play a role in cognitive functions and normal cholinergic signaling related to learning and memory is dependent on acetylcholine. The Locus Coeruleus norepinephrine (LC-NE) is the main noradrenergic nucleus that projects and supplies norepinephrine to different brain regions. Norepinephrine has been shown to be neuroprotective against neurodegeneration and plays a role in behavior and cognition. Cholinergic and adrenergic signaling are dysregulated in Alzheimer’s disease. The degeneration of cholinergic neurons in nucleus basalis of Meynert in the basal forebrain and the degeneration of LC-NE neurons were reported in Alzheimer’s disease. The aim of this review is to describe current literature on the role of the cholinergic system and the adrenergic system (LC-NE) in the pathology of Alzheimer’s disease and potential therapeutic implications.
Collapse
Affiliation(s)
- Rola A Bekdash
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA
| |
Collapse
|
22
|
Clark I, Vissel B. Broader Insights into Understanding Tumor Necrosis Factor and Neurodegenerative Disease Pathogenesis Infer New Therapeutic Approaches. J Alzheimers Dis 2021; 79:931-948. [PMID: 33459706 PMCID: PMC7990436 DOI: 10.3233/jad-201186] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/11/2020] [Indexed: 12/12/2022]
Abstract
Proinflammatory cytokines such as tumor necrosis factor (TNF), with its now appreciated key roles in neurophysiology as well as neuropathophysiology, are sufficiently well-documented to be useful tools for enquiry into the natural history of neurodegenerative diseases. We review the broader literature on TNF to rationalize why abruptly-acquired neurodegenerative states do not exhibit the remorseless clinical progression seen in those states with gradual onsets. We propose that the three typically non-worsening neurodegenerative syndromes, post-stroke, post-traumatic brain injury (TBI), and post cardiac arrest, usually become and remain static because of excess cerebral TNF induced by the initial dramatic peak keeping microglia chronically activated through an autocrine loop of microglial activation through excess cerebral TNF. The existence of this autocrine loop rationalizes post-damage repair with perispinal etanercept and proposes a treatment for cerebral aspects of COVID-19 chronicity. Another insufficiently considered aspect of cerebral proinflammatory cytokines is the fitness of the endogenous cerebral anti-TNF system provided by norepinephrine (NE), generated and distributed throughout the brain from the locus coeruleus (LC). We propose that an intact LC, and therefore an intact NE-mediated endogenous anti-cerebral TNF system, plus the DAMP (damage or danger-associated molecular pattern) input having diminished, is what allows post-stroke, post-TBI, and post cardiac arrest patients a strong long-term survival advantage over Alzheimer's disease and Parkinson's disease sufferers. In contrast, Alzheimer's disease and Parkinson's disease patients remorselessly worsen, being handicapped by sustained, accumulating, DAMP and PAMP (pathogen-associated molecular patterns) input, as well as loss of the LC-origin, NE-mediated, endogenous anti-cerebral TNF system. Adrenergic receptor agonists may counter this.
Collapse
Affiliation(s)
- I.A. Clark
- Research School of Biology, Australian National University, Canberra, Australia
| | - B. Vissel
- Centre for Neuroscience and Regenerative Medicine, Faculty of Science, University of Technology, Sydney, Australia
- St. Vincent’s Centre for Applied Medical Research, Sydney, Australia
| |
Collapse
|
23
|
Novel PET Biomarkers to Disentangle Molecular Pathways across Age-Related Neurodegenerative Diseases. Cells 2020; 9:cells9122581. [PMID: 33276490 PMCID: PMC7761606 DOI: 10.3390/cells9122581] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/25/2020] [Accepted: 11/28/2020] [Indexed: 12/11/2022] Open
Abstract
There is a need to disentangle the etiological puzzle of age-related neurodegenerative diseases, whose clinical phenotypes arise from known, and as yet unknown, pathways that can act distinctly or in concert. Enhanced sub-phenotyping and the identification of in vivo biomarker-driven signature profiles could improve the stratification of patients into clinical trials and, potentially, help to drive the treatment landscape towards the precision medicine paradigm. The rapidly growing field of neuroimaging offers valuable tools to investigate disease pathophysiology and molecular pathways in humans, with the potential to capture the whole disease course starting from preclinical stages. Positron emission tomography (PET) combines the advantages of a versatile imaging technique with the ability to quantify, to nanomolar sensitivity, molecular targets in vivo. This review will discuss current research and available imaging biomarkers evaluating dysregulation of the main molecular pathways across age-related neurodegenerative diseases. The molecular pathways focused on in this review involve mitochondrial dysfunction and energy dysregulation; neuroinflammation; protein misfolding; aggregation and the concepts of pathobiology, synaptic dysfunction, neurotransmitter dysregulation and dysfunction of the glymphatic system. The use of PET imaging to dissect these molecular pathways and the potential to aid sub-phenotyping will be discussed, with a focus on novel PET biomarkers.
Collapse
|
24
|
Bolton CJ, Tam JW. Differential Involvement of the Locus Coeruleus in Early- and Late-Onset Alzheimer's Disease: A Potential Mechanism of Clinical Differences? MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2020:2020.11.01.20224139. [PMID: 33173930 PMCID: PMC7654926 DOI: 10.1101/2020.11.01.20224139] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Early-onset Alzheimer's disease (EOAD) has been associated with an increased likelihood of atypical clinical manifestations such as attentional impairment, yet the cause of this heterogeneity remains unclear. The locus coeruleus (LC) is implicated early in Alzheimer's disease pathology and is associated with attentional functioning. This study investigated post-mortem atrophy of the LC in EOAD and late-onset Alzheimer's disease (LOAD) in a large, well-characterized sample. Results show nearly four times greater likelihood of higher LC atrophy in EOAD as compared to LOAD after controlling for other measures of pathological progression ( p < .005). Follow-up analyses within the EOAD group revealed that compared to those who displayed mild or no LC atrophy at autopsy, those with moderate-severe atrophy of the LC displayed significantly worse performance on various baseline measures of attentional functioning ( p < .05), despite similar overall cognition ( p = .25). These findings suggest the LC is an important potential driver of clinical and pathological heterogeneity in EOAD.
Collapse
Affiliation(s)
- Corey J. Bolton
- Vanderbilt Memory & Alzheimer’s Center, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
- Deparment of Psychiatry and Behavioral Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Joyce W. Tam
- Deparment of Psychiatry and Behavioral Sciences, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
25
|
Shao YR, Kahali P, Houle TT, Deng H, Colvin C, Dickerson BC, Brown EN, Purdon PL. Low Frontal Alpha Power Is Associated With the Propensity for Burst Suppression: An Electroencephalogram Phenotype for a "Vulnerable Brain". Anesth Analg 2020; 131:1529-1539. [PMID: 33079876 PMCID: PMC7553194 DOI: 10.1213/ane.0000000000004781] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/24/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND A number of recent studies have reported an association between intraoperative burst suppression and postoperative delirium. These studies suggest that anesthesia-induced burst suppression may be an indicator of underlying brain vulnerability. A prominent feature of electroencephalogram (EEG) under propofol and sevoflurane anesthesia is the frontal alpha oscillation. This frontal alpha oscillation is known to decline significantly during aging and is generated by prefrontal brain regions that are particularly prone to age-related neurodegeneration. Given that burst suppression and frontal alpha oscillations are both associated with brain vulnerability, we hypothesized that anesthesia-induced frontal alpha power could also be associated with burst suppression. METHODS We analyzed EEG data from a previously reported cohort in which 155 patients received propofol (n = 60) or sevoflurane (n = 95) as the primary anesthetic. We computed the EEG spectrum during stable anesthetic maintenance and identified whether or not burst suppression occurred during the anesthetic. We characterized the relationship between burst suppression and alpha power using logistic regression. We proposed 5 different models consisting of different combinations of potential contributing factors associated with burst suppression: (1) a Base Model consisting of alpha power; (2) an Extended Mechanistic Model consisting of alpha power, age, and drug dosing information; (3) a Clinical Confounding Factors Model consisting of alpha power, hypotension, and other confounds; (4) a Simplified Model consisting only of alpha power and propofol bolus administration; and (5) a Full Model consisting of all of these variables to control for as much confounding as possible. RESULTS All models show a consistent significant association between alpha power and burst suppression while adjusting for different sets of covariates, all with consistent effect size estimates. Using the Simplified Model, we found that for each decibel decrease in alpha power, the odds of experiencing burst suppression increased by 1.33-fold. CONCLUSIONS In this study, we show how a decrease in anesthesia-induced frontal alpha power is associated with an increased propensity for burst suppression, in a manner that captures individualized information above and beyond a patient's chronological age. Lower frontal alpha band power is strongly associated with higher propensity for burst suppression and, therefore, potentially higher risk of postoperative neurocognitive disorders. We hypothesize that low frontal alpha power and increased propensity for burst suppression together characterize a "vulnerable brain" phenotype under anesthesia that could be mechanistically linked to brain metabolism, cognition, and brain aging.
Collapse
Affiliation(s)
- Yu Raymond Shao
- From the Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina
| | - Pegah Kahali
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Timothy T. Houle
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Hao Deng
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Christopher Colvin
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Bradford C. Dickerson
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Emery N. Brown
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, Massachusetts
- Institute for Medical Engineering and Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Patrick L. Purdon
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
26
|
Kelberman M, Keilholz S, Weinshenker D. What's That (Blue) Spot on my MRI? Multimodal Neuroimaging of the Locus Coeruleus in Neurodegenerative Disease. Front Neurosci 2020; 14:583421. [PMID: 33122996 PMCID: PMC7573566 DOI: 10.3389/fnins.2020.583421] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 09/16/2020] [Indexed: 01/04/2023] Open
Abstract
The locus coeruleus (LC) has long been underappreciated for its role in the pathophysiology of Alzheimer’s disease (AD), Parkinson’s disease (PD), and other neurodegenerative disorders. While AD and PD are distinct in clinical presentation, both are characterized by prodromal protein aggregation in the LC, late-stage degeneration of the LC, and comorbid conditions indicative of LC dysfunction. Many of these early studies were limited to post-mortem histological techniques due to the LC’s small size and location deep in the brainstem. Thus, there is a growing interest in utilizing in vivo imaging of the LC as a predictor of preclinical neurodegenerative processes and biomarker of disease progression. Simultaneously, neuroimaging in animal models of neurodegenerative disease holds promise for identifying early alterations to LC circuits, but has thus far been underutilized. While still in its infancy, a handful of studies have reported effects of single gene mutations and pathology on LC function in disease using various neuroimaging techniques. Furthermore, combining imaging and optogenetics or chemogenetics allows for interrogation of network connectivity in response to changes in LC activity. The purpose of this article is twofold: (1) to review what magnetic resonance imaging (MRI) and positron emission tomography (PET) have revealed about LC dysfunction in neurodegenerative disease and its potential as a biomarker in humans, and (2) to explore how animal models can be used to test hypotheses derived from clinical data and establish a mechanistic framework to inform LC-focused therapeutic interventions to alleviate symptoms and impede disease progression.
Collapse
Affiliation(s)
- Michael Kelberman
- Department of Human Genetics, Emory University, Atlanta, GA, United States
| | - Shella Keilholz
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Emory University, Atlanta, GA, United States
| | - David Weinshenker
- Department of Human Genetics, Emory University, Atlanta, GA, United States
| |
Collapse
|
27
|
Bolshakov AP, Stepanichev MY, Dobryakova YV, Spivak YS, Markevich VA. Saporin from Saponaria officinalis as a Tool for Experimental Research, Modeling, and Therapy in Neuroscience. Toxins (Basel) 2020; 12:toxins12090546. [PMID: 32854372 PMCID: PMC7551693 DOI: 10.3390/toxins12090546] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 08/17/2020] [Accepted: 08/21/2020] [Indexed: 01/06/2023] Open
Abstract
Saporin, which is extracted from Saponaria officinalis, is a protein toxin that inactivates ribosomes. Saporin itself is non-selective toxin but acquires high specificity after conjugation with different ligands such as signaling peptides or antibodies to some surface proteins expressed in a chosen cell subpopulation. The saporin-based conjugated toxins were widely adopted in neuroscience as a convenient tool to induce highly selective degeneration of desired cell subpopulation. Induction of selective cell death is one of approaches used to model neurodegenerative diseases, study functions of certain cell subpopulations in the brain, and therapy. Here, we review studies where saporin-based conjugates were used to analyze cell mechanisms of sleep, general anesthesia, epilepsy, pain, and development of Parkinson’s and Alzheimer’s diseases. Limitations and future perspectives of use of saporin-based toxins in neuroscience are discussed.
Collapse
Affiliation(s)
- Alexey P. Bolshakov
- Laboratory of Molecular Neurobiology, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, 119991 Moscow, Russia;
- Correspondence:
| | - Mikhail Yu. Stepanichev
- Laboratory of Functional Biochemistry of the Nervous System, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, 119991 Moscow, Russia;
| | - Yulia V. Dobryakova
- Laboratory of Neurophysiology of Learning, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, 119991 Moscow, Russia; (Y.V.D.); (V.A.M.)
| | - Yulia S. Spivak
- Laboratory of Molecular Neurobiology, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, 119991 Moscow, Russia;
| | - Vladimir A. Markevich
- Laboratory of Neurophysiology of Learning, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, 119991 Moscow, Russia; (Y.V.D.); (V.A.M.)
| |
Collapse
|
28
|
Belbin O, Morgan K, Medway C, Warden D, Cortina-Borja M, van Duijn CM, Adams HHH, Frank-Garcia A, Brookes K, Sánchez-Juan P, Alvarez V, Heun R, Kölsch H, Coto E, Kehoe PG, Rodriguez-Rodriguez E, Bullido MJ, Ikram MA, Smith AD, Lehmann DJ. The Epistasis Project: A Multi-Cohort Study of the Effects of BDNF, DBH, and SORT1 Epistasis on Alzheimer's Disease Risk. J Alzheimers Dis 2020; 68:1535-1547. [PMID: 30909233 DOI: 10.3233/jad-181116] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Pre-synaptic secretion of brain-derived neurotrophic factor (BDNF) from noradrenergic neurons may protect the Alzheimer's disease (AD) brain from amyloid pathology. While the BDNF polymorphism (rs6265) is associated with faster cognitive decline and increased hippocampal atrophy, a replicable genetic association of BDNF with AD risk has yet to be demonstrated. This could be due to masking by underlying epistatic interactions between BDNF and other loci that encode proteins involved in moderating BDNF secretion (DBH and Sortilin). We performed a multi-cohort case-control association study of the BDNF, DBH, and SORT1 loci comprising 5,682 controls and 2,454 AD patients from Northern Europe (87% of samples) and Spain (13%). The BDNF locus was associated with increased AD risk (odds ratios; OR = 1.1-1.2, p = 0.005-0.3), an effect size that was consistent in the Northern European (OR = 1.1-1.2, p = 0.002-0.8) but not the smaller Spanish (OR = 0.8-1.6, p = 0.4-1.0) subset. A synergistic interaction between BDNF and sex (synergy factor; SF = 1.3-1.5 p = 0.002-0.02) translated to a greater risk of AD associated with BDNF in women (OR = 1.2-1.3, p = 0.007-0.00008) than men (OR = 0.9-1.0, p = 0.3-0.6). While the DBH polymorphism (rs1611115) was also associated with increased AD risk (OR = 1.1, p = 0.04) the synergistic interaction (SF = 2.2, p = 0.007) between BDNF (rs6265) and DBH (rs1611115) contributed greater AD risk than either gene alone, an effect that was greater in women (SF = 2.4, p = 0.04) than men (SF = 2.0, p = 0.2). These data support a complex genetic interaction at loci encoding proteins implicated in the DBH-BDNF inflammatory pathway that modifies AD risk, particularly in women.
Collapse
Affiliation(s)
- Olivia Belbin
- Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Kevin Morgan
- Human Genetics School of Life Sciences, University of Nottingham, UK
| | - Chris Medway
- Institute of Medical Genetics, University Hospital Wales, Cardiff, UK
| | - Donald Warden
- Oxford Project to Investigate Memory and Ageing (OPTIMA), University Department of Pharmacology, Oxford, UK
| | - Mario Cortina-Borja
- Clinical Epidemiology, Nutrition and Biostatistics, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Cornelia M van Duijn
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Hieab H H Adams
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Ana Frank-Garcia
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain.,Centro de Biología Molecular Severo Ochoa (UAM-CSIC), Madrid, Spain
| | - Keeley Brookes
- Human Genetics School of Life Sciences, University of Nottingham, UK
| | - Pascual Sánchez-Juan
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain.,Neurology Service, Marqués de Valdecilla University Hospital (University of Cantabria and IDIVAL), Santander, Spain
| | - Victoria Alvarez
- Laboratorio de Genética, AGC Laboratorio de Medicina, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Reinhard Heun
- Department of Psychiatry, University of Bonn, Bonn, Germany
| | - Heike Kölsch
- Department of Psychiatry, University of Bonn, Bonn, Germany
| | - Eliecer Coto
- Laboratorio de Genética, AGC Laboratorio de Medicina, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Patrick G Kehoe
- Dementia Research Group, Bristol Medical School Translational Health Sciences, University of Bristol, Southmead Hospital, Bristol, UK
| | - Eloy Rodriguez-Rodriguez
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain.,Neurology Service, Marqués de Valdecilla University Hospital (University of Cantabria and IDIVAL), Santander, Spain
| | - Maria J Bullido
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain.,Centro de Biología Molecular Severo Ochoa (UAM-CSIC), Madrid, Spain
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - A David Smith
- Oxford Project to Investigate Memory and Ageing (OPTIMA), University Department of Pharmacology, Oxford, UK
| | - Donald J Lehmann
- Oxford Project to Investigate Memory and Ageing (OPTIMA), University Department of Pharmacology, Oxford, UK
| |
Collapse
|
29
|
Umek N. Cyclization step of noradrenaline and adrenaline autoxidation: a quantum chemical study. RSC Adv 2020; 10:16650-16658. [PMID: 35498869 PMCID: PMC9053094 DOI: 10.1039/d0ra02713h] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 04/17/2020] [Indexed: 12/14/2022] Open
Abstract
Catecholamine autoxidation has been recognized as one of the potential trigger factors for catecholaminergic neuron loss characteristics of neurodegenerative diseases. The cyclization step with intramolecular Michael addition of catecholamine o-quinones has been shown to be the irreversible and rate limiting step of the autoxidation reaction across a broad pH range and has a complex pH dependence that has not yet been fully understood. Using quantum chemical calculations, we demonstrated that in the case of noradrenaline and adrenaline two catecholamine o-quinone species, one with an unprotonated and one with a protonated quinone group can participate in the cyclization reaction and that the mechanisms of these reactions significantly differ, emphasizing the importance of quinone group protonation states in the reaction mechanism. With a thorough exploration of the reaction kinetics, we further showed that at acidic pH the cyclization reaction rate is pH independent, while at alkaline pH the pH dependence is marked, explaining the experimentally observed complex pH dependence. The quinone group protonation state determines the reaction mechanism of noradrenaline and adrenaline o-quinone cyclization.![]()
Collapse
Affiliation(s)
- Nejc Umek
- Institute of Anatomy, Faculty of Medicine, University of Ljubljana Korytkova ulica 2 1000 Ljubljana Slovenia +386 15437314
| |
Collapse
|
30
|
Beta-blocker therapy and risk of vascular dementia: A population-based prospective study. Vascul Pharmacol 2020; 125-126:106649. [PMID: 31958512 DOI: 10.1016/j.vph.2020.106649] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 12/29/2019] [Accepted: 01/16/2020] [Indexed: 02/07/2023]
Abstract
There are a few studies that report cognitive impairment as a complication of treatment with beta- blockers. We aimed to evaluate the longitudinal association between use of beta-blockers, as a class, and incident risk of all-cause dementia, vascular dementia, Alzheimer's and mixed dementia in the prospective population-based Malmö Preventive Project. We included 18,063 individuals (mean age 68.2, males 63.4%) followed up for 84,506 person-years. Dementia cases were retrieved from the Swedish National Patient Register and validated by review of medical records and neuroimaging data. We performed propensity score matching analysis, resulting in 3720 matched pairs of beta-blocker users and non-users at baseline, and multivariable Cox proportional-hazards regression. Overall, 122 study participants (1.6%) were diagnosed with dementia during the follow-up. Beta-blocker therapy was independently associated with increased risk of developing vascular dementia, regardless of confounding factors (HR: 1.72, 95%CI 1.01-3.78; p = .048). Conversely, treatment with beta-blockers was not associated with increased risk of all-cause, Alzheimer's and mixed dementia (HR:1.15; 95%CI 0.80-1.66; p = .44; HR:0.85; 95%CI 0.48-1.54; P = .59 and HR:1.35; 95%CI 0.56-3.27; p = .50, respectively). We observed that use of beta-blockers, as a class, is associated with increased longitudinal risk of vascular dementia in the general elderly population, regardless of cardiovascular risk factors, prevalent or incident history of atrial fibrillation, stroke, coronary events and heart failure. Further studies are needed to confirm our findings in the general population and to explore the mechanisms underlying the relationship between use of beta- blockers and increased risk of vascular dementia.
Collapse
|
31
|
Marcinkowska M, Śniecikowska J, Fajkis N, Paśko P, Franczyk W, Kołaczkowski M. Management of Dementia-Related Psychosis, Agitation and Aggression: A Review of the Pharmacology and Clinical Effects of Potential Drug Candidates. CNS Drugs 2020; 34:243-268. [PMID: 32052375 PMCID: PMC7048860 DOI: 10.1007/s40263-020-00707-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Along with cognitive decline, 90% of patients with dementia experience behavioral and psychological symptoms of dementia, such as psychosis, aggression, agitation, and depression. Atypical antipsychotics are commonly prescribed off-label to manage certain symptoms, despite warnings from the regulatory agencies regarding the increased risk of mortality associated with their use in elderly patients. Moreover, these compounds display a limited clinical efficacy, mostly owing to the fact that they were developed to treat schizophrenia, a disease characterized by neurobiological deficits. Thus, to improve clinical efficacy, it has been suggested that patients with dementia should be treated with exclusively designed and developed drugs that interact with pharmacologically relevant targets. Within this context, numerous studies have suggested druggable targets that might achieve therapeutically acceptable pharmacological profiles. Based on this, several different drug candidates have been proposed that are being investigated in clinical trials for behavioral and psychological symptoms of dementia. We highlight the recent advances toward the development of therapeutic agents for dementia-related psychosis and agitation/aggression and discuss the relationship between the relevant biological targets and their etiology. In addition, we review the compounds that are in the early stage of development (discovery or preclinical phase) and those that are currently being investigated in clinical trials for dementia-related psychosis and agitation/aggression. We also discuss the mechanism of action of these compounds and their pharmacological utility in patients with dementia.
Collapse
Affiliation(s)
- Monika Marcinkowska
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna Street, Kraków, 30-688, Poland.
| | - Joanna Śniecikowska
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna Street, Kraków, 30-688 Poland ,Adamed Pharma S.A., Czosnow, Poland
| | - Nikola Fajkis
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna Street, Kraków, 30-688 Poland
| | - Paweł Paśko
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna Street, Kraków, 30-688 Poland
| | - Weronika Franczyk
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna Street, Kraków, 30-688 Poland
| | - Marcin Kołaczkowski
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna Street, Kraków, 30-688 Poland ,Adamed Pharma S.A., Czosnow, Poland
| |
Collapse
|
32
|
Khan S, Barve KH, Kumar MS. Recent Advancements in Pathogenesis, Diagnostics and Treatment of Alzheimer's Disease. Curr Neuropharmacol 2020; 18:1106-1125. [PMID: 32484110 PMCID: PMC7709159 DOI: 10.2174/1570159x18666200528142429] [Citation(s) in RCA: 369] [Impact Index Per Article: 73.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 05/06/2020] [Accepted: 05/25/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The only conclusive way to diagnose Alzheimer's is to carry out brain autopsy of the patient's brain tissue and ascertain whether the subject had Alzheimer's or any other form of dementia. However, due to the non-feasibility of such methods, to diagnose and conclude the conditions, medical practitioners use tests that examine a patient's mental ability. OBJECTIVE Accurate diagnosis at an early stage is the need of the hour for initiation of therapy. The cause for most Alzheimer's cases still remains unknown except where genetic distinctions have been observed. Thus, a standard drug regimen ensues in every Alzheimer's patient, irrespective of the cause, which may not always be beneficial in halting or reversing the disease progression. To provide a better life to such patients by suppressing existing symptoms, early diagnosis, curative therapy, site-specific delivery of drugs, and application of hyphenated methods like artificial intelligence need to be brought into the main field of Alzheimer's therapeutics. METHODS In this review, we have compiled existing hypotheses to explain the cause of the disease, and highlighted gene therapy, immunotherapy, peptidomimetics, metal chelators, probiotics and quantum dots as advancements in the existing strategies to manage Alzheimer's. CONCLUSION Biomarkers, brain-imaging, and theranostics, along with artificial intelligence, are understood to be the future of the management of Alzheimer's.
Collapse
Affiliation(s)
- Sahil Khan
- SVKM’S NMIMS, Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, V.L. Mehta Road, Vile Parle West, Mumbai-400056, India
| | - Kalyani H. Barve
- SVKM’S NMIMS, Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, V.L. Mehta Road, Vile Parle West, Mumbai-400056, India
| | - Maushmi S. Kumar
- SVKM’S NMIMS, Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, V.L. Mehta Road, Vile Parle West, Mumbai-400056, India
| |
Collapse
|
33
|
Gonzalez‐Lopez E, Vrana KE. Dopamine beta‐hydroxylase and its genetic variants in human health and disease. J Neurochem 2019; 152:157-181. [DOI: 10.1111/jnc.14893] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 09/18/2019] [Accepted: 09/26/2019] [Indexed: 12/12/2022]
Affiliation(s)
| | - Kent E. Vrana
- Department of Pharmacology Penn State College of Medicine Hershey PA USA
| |
Collapse
|
34
|
Abstract
Noradrenergic system of brain supplies the neurotransmitter noradrenalin throughout the brain through widespread efferent projections and play pivotal role in cognitive activities and could be involve in motor and non-motor symptoms of Parkinson's disease (PD) pathology. Profound loss of noradrenergic pathways has been reported in both Parkinson's and Alzheimer's disease (AD) pathology however their employment in therapeutics is still scarce. Therefore the present review is providing the various aspects for involvement on noradrenergic pathways in PD and AD pathology as well as the imaging of locus coeruleus as indicative diagnostic marker for disease. The present review is describing about the role of tiny nucleus locus coeruleus located noradrenergic pathways in said pathologies and discussing the past research as well as lacunas in this regard.
Collapse
Affiliation(s)
- Sarika Singh
- Toxicology and Experimental Medicine Division, CDRI-CSIR, Lucknow, UP, India
| |
Collapse
|
35
|
Slezak M, Kandler S, Van Veldhoven PP, Van den Haute C, Bonin V, Holt MG. Distinct Mechanisms for Visual and Motor-Related Astrocyte Responses in Mouse Visual Cortex. Curr Biol 2019; 29:3120-3127.e5. [PMID: 31495587 PMCID: PMC6859477 DOI: 10.1016/j.cub.2019.07.078] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 06/19/2019] [Accepted: 07/26/2019] [Indexed: 02/02/2023]
Abstract
Astrocytes are a major cell type in the mammalian nervous system, are in close proximity to neurons, and show rich Ca2+ activity thought to mediate cellular outputs. Astrocytes show activity linked to sensory [1, 2] and motor [3, 4] events, reflecting local neural activity and brain-wide neuromodulatory inputs. Sensory responses are highly variable [5, 6, 7, 8, 9, 10], which may reflect interactions between distinct input types [6, 7, 9]. However, the diversity of inputs generating astrocyte activity, particularly during sensory stimulation and behavior, is not fully understood [11, 12]. Using a combination of Ca2+ imaging, a treadmill assay, and visual stimulation, we examined the properties of astrocyte activity in mouse visual cortex associated with motor or sensory events. Consistent with previous work, motor activity activated astrocytes across the cortex with little specificity, reflecting a diffuse neuromodulatory mechanism. In contrast, moving visual stimuli generated specific activity patterns that reflected the stimulus' trajectory within the visual field, precisely as one would predict if astrocytes reported local neural activity. Visual responses depended strongly on behavioral state, with astrocytes showing high amplitude Ca2+ transients during locomotion and little activity during stillness. Furthermore, the amplitudes of visual responses were highly correlated with pupil size, suggesting a role of arousal. Interestingly, while depletion of cortical noradrenaline abolished locomotor responses, visual responses were only reduced in amplitude and their spatiotemporal organization remained intact, suggesting two distinct types of inputs underlie visual responses. We conclude that cortical astrocytes integrate local sensory information and behavioral state, suggesting a role in information processing. Astrocytes of moving mice display robust retinotopic responses to visual stimuli Sensory responses are distinguishable from responses to locomotion Vision-driven responses are correlated to arousal Vision- and arousal-driven responses are differentially regulated by noradrenaline
Collapse
Affiliation(s)
- Michal Slezak
- Neuro-Electronics Research Flanders (NERF), Kapeldreef 75, 3001 Leuven, Belgium; VIB Center for Brain and Disease Research, Herestraat 49-Box 602, 3000 Leuven, Belgium
| | - Steffen Kandler
- Neuro-Electronics Research Flanders (NERF), Kapeldreef 75, 3001 Leuven, Belgium; IMEC, Kapeldreef 75, 3001 Leuven, Belgium
| | - Paul P Van Veldhoven
- KU Leuven, Department of Cellular and Molecular Medicine, LIPIT, Herestraat 49-Box 601, 3000 Leuven, Belgium
| | - Chris Van den Haute
- KU Leuven, Laboratory for Neurobiology and Gene Therapy and Leuven Brain Institute, RK-Herestraat 49-Box 1023, 3000 Leuven, Belgium; KU Leuven, Leuven Viral Vector Core, RK-Herestraat 49, 3000 Leuven, Belgium
| | - Vincent Bonin
- Neuro-Electronics Research Flanders (NERF), Kapeldreef 75, 3001 Leuven, Belgium; IMEC, Kapeldreef 75, 3001 Leuven, Belgium; KU Leuven, Department of Biology and Leuven Brain Institute, Naamsestraat 59-Box 2465, 3001 Leuven, Belgium.
| | - Matthew G Holt
- VIB Center for Brain and Disease Research, Herestraat 49-Box 602, 3000 Leuven, Belgium; KU Leuven, Department of Neuroscience and Leuven Brain Institute, Herestraat 49-Box 1021, 3000 Leuven, Belgium.
| |
Collapse
|
36
|
Mauricio R, Benn C, Davis J, Dawson G, Dawson LA, Evans A, Fox N, Gallacher J, Hutton M, Isaac J, Jones DN, Jones L, Lalli G, Libri V, Lovestone S, Moody C, Noble W, Perry H, Pickett J, Reynolds D, Ritchie C, Rohrer JD, Routledge C, Rowe J, Snyder H, Spires-Jones T, Swartz J, Truyen L, Whiting P. Tackling gaps in developing life-changing treatments for dementia. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2019; 5:241-253. [PMID: 31297438 PMCID: PMC6597931 DOI: 10.1016/j.trci.2019.05.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Since the G8 dementia summit in 2013, a number of initiatives have been established with the aim of facilitating the discovery of a disease-modifying treatment for dementia by 2025. This report is a summary of the findings and recommendations of a meeting titled "Tackling gaps in developing life-changing treatments for dementia", hosted by Alzheimer's Research UK in May 2018. The aim of the meeting was to identify, review, and highlight the areas in dementia research that are not currently being addressed by existing initiatives. It reflects the views of leading experts in the field of neurodegeneration research challenged with developing a strategic action plan to address these gaps and make recommendations on how to achieve the G8 dementia summit goals. The plan calls for significant advances in (1) translating newly identified genetic risk factors into a better understanding of the impacted biological processes; (2) enhanced understanding of selective neuronal resilience to inform novel drug targets; (3) facilitating robust and reproducible drug-target validation; (4) appropriate and evidence-based selection of appropriate subjects for proof-of-concept clinical trials; (5) improving approaches to assess drug-target engagement in humans; and (6) innovative approaches in conducting clinical trials if we are able to detect disease 10-15 years earlier than we currently do today.
Collapse
Affiliation(s)
| | | | - John Davis
- Alzheimer's Research UK Oxford Drug Discovery Institute, University of Oxford, Oxford, UK
| | - Gerry Dawson
- P1 Vital, Howbery Business Park, Wallingford, Oxfordshire, UK
| | - Lee A. Dawson
- Cerevance Ltd, Cambridge Science Park, Cambridge, UK
| | | | - Nick Fox
- Dementia Research Centre, Institute of Neurology, University College London, London, UK
| | - John Gallacher
- Department of Psychiatry, University of Oxford, Oxford, UK
| | | | - John Isaac
- Neuroscience External Innovation, Neuroscience Therapeutic Area, Johnson & Johnson Innovation, London, UK
| | - Declan N.C. Jones
- Neuroscience External Innovation, Neuroscience Therapeutic Area, Johnson & Johnson Innovation, London, UK
| | - Lesley Jones
- MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, Wales, UK
| | | | - Vincenzo Libri
- Institute of Neurology, University College London, London, UK
| | | | | | - Wendy Noble
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Hugh Perry
- Faculty of Natural and Environmental Sciences, University of Southampton, Southampton, UK
| | | | | | - Craig Ritchie
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Jonathan D. Rohrer
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | | | - James Rowe
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | | | - Tara Spires-Jones
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Jina Swartz
- European Innovation Hub, Merck Sharp and Dohme, London, UK
| | - Luc Truyen
- Janssen Research & Development LLC, Titusville, NJ, USA
| | - Paul Whiting
- Dementia Research Institute, UCL, London, UK
- ARUK Drug Discovery Institute, Institute of Neurology, University College London, London, UK
| | | |
Collapse
|
37
|
Morita M, Ikeshima-Kataoka H, Kreft M, Vardjan N, Zorec R, Noda M. Metabolic Plasticity of Astrocytes and Aging of the Brain. Int J Mol Sci 2019; 20:ijms20040941. [PMID: 30795555 PMCID: PMC6413111 DOI: 10.3390/ijms20040941] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 02/17/2019] [Accepted: 02/18/2019] [Indexed: 01/03/2023] Open
Abstract
As part of the blood-brain-barrier, astrocytes are ideally positioned between cerebral vasculature and neuronal synapses to mediate nutrient uptake from the systemic circulation. In addition, astrocytes have a robust enzymatic capacity of glycolysis, glycogenesis and lipid metabolism, managing nutrient support in the brain parenchyma for neuronal consumption. Here, we review the plasticity of astrocyte energy metabolism under physiologic and pathologic conditions, highlighting age-dependent brain dysfunctions. In astrocytes, glycolysis and glycogenesis are regulated by noradrenaline and insulin, respectively, while mitochondrial ATP production and fatty acid oxidation are influenced by the thyroid hormone. These regulations are essential for maintaining normal brain activities, and impairments of these processes may lead to neurodegeneration and cognitive decline. Metabolic plasticity is also associated with (re)activation of astrocytes, a process associated with pathologic events. It is likely that the recently described neurodegenerative and neuroprotective subpopulations of reactive astrocytes metabolize distinct energy substrates, and that this preference is supposed to explain some of their impacts on pathologic processes. Importantly, physiologic and pathologic properties of astrocytic metabolic plasticity bear translational potential in defining new potential diagnostic biomarkers and novel therapeutic targets to mitigate neurodegeneration and age-related brain dysfunctions.
Collapse
Affiliation(s)
- Mitsuhiro Morita
- Department of Biology, Graduate School of Sciences, Kobe University, 657-8501 Kobe, Japan.
| | - Hiroko Ikeshima-Kataoka
- Faculty of Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo 169-8555, Japan.
| | - Marko Kreft
- Laboratory of Cell Engineering, Celica Biomedical, 1000 Ljubljana, Slovenia.
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia.
- Department of Biology, Biotechnical Faculty University of Ljubljana, 1000 Ljubljana, Slovenia.
| | - Nina Vardjan
- Laboratory of Cell Engineering, Celica Biomedical, 1000 Ljubljana, Slovenia.
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia.
| | - Robert Zorec
- Laboratory of Cell Engineering, Celica Biomedical, 1000 Ljubljana, Slovenia.
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia.
| | - Mami Noda
- Laboratory of Pathophysiology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan.
| |
Collapse
|
38
|
Farfán-García ED, Márquez-Gómez R, Barrón-González M, Pérez-Capistran T, Rosales-Hernández MC, Pinto-Almazán R, Soriano-Ursúa MA. Monoamines and their Derivatives on GPCRs: Potential Therapy for Alzheimer's Disease. Curr Alzheimer Res 2019; 16:871-894. [PMID: 30963972 DOI: 10.2174/1570159x17666190409144558] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 03/18/2019] [Accepted: 04/01/2019] [Indexed: 02/07/2023]
Abstract
Albeit cholinergic depletion remains the key event in Alzheimer's Disease (AD), recent information describes stronger links between monoamines (trace amines, catecholamines, histamine, serotonin, and melatonin) and AD than those known in the past century. Therefore, new drug design strategies focus efforts to translate the scope on these topics and to offer new drugs which can be applied as therapeutic tools in AD. In the present work, we reviewed the state-of-art regarding genetic, neuropathology and neurochemistry of AD involving monoamine systems. Then, we compiled the effects of monoamines found in the brain of mammals as well as the reported effects of their derivatives and some structure-activity relationships. Recent derivatives have triggered exciting effects and pharmacokinetic properties in both murine models and humans. In some cases, the mechanism of action is clear, essentially through the interaction on G-protein-coupled receptors as revised in this manuscript. Additional mechanisms are inhibition of enzymes for their biotransformation, regulation of free-radicals in the central nervous system and others for the effects on Tau phosphorylation or amyloid-beta accumulation. All these data make the monoamines and their derivatives attractive potential elements for AD therapy.
Collapse
Affiliation(s)
- Eunice D Farfán-García
- Departamento de Fisiologia y Bioquimica. Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Plan de San Luis y Diaz Miron s/n, 11340, Mexico City, Mexico
| | - Ricardo Márquez-Gómez
- MRC Anatomical Neuropharmacology Unit, Department of Pharmacology, University of Oxford, OX1 3TH, Oxford, United Kingdom
| | - Mónica Barrón-González
- Departamento de Fisiologia y Bioquimica. Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Plan de San Luis y Diaz Miron s/n, 11340, Mexico City, Mexico
| | - Teresa Pérez-Capistran
- Departamento de Fisiologia y Bioquimica. Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Plan de San Luis y Diaz Miron s/n, 11340, Mexico City, Mexico
| | - Martha C Rosales-Hernández
- Laboratorio de Biofisica y Biocatalisis, Seccion de Estudios de Posgrado e Investigacion Escuela Superior de Medicina, Instituto Politecnico Nacional, Plan de San Luis y Diaz Miron s/n, 11340, Mexico City, Mexico
| | - Rodolfo Pinto-Almazán
- Unidad de Investigacion Hospital Regional de Alta Especialidad Ixtapaluca, Carretera Federal Mexico-Puebla km 34.5, C.P. 56530. Ixtapaluca, State of Mexico, Mexico
| | - Marvin A Soriano-Ursúa
- Departamento de Fisiologia y Bioquimica. Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Plan de San Luis y Diaz Miron s/n, 11340, Mexico City, Mexico
| |
Collapse
|
39
|
Verkhratsky A, Parpura V, Rodriguez-Arellano JJ, Zorec R. Astroglia in Alzheimer's Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1175:273-324. [PMID: 31583592 DOI: 10.1007/978-981-13-9913-8_11] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease is the most common cause of dementia. Cellular changes in the brains of the patients suffering from Alzheimer's disease occur well in advance of the clinical symptoms. At the cellular level, the most dramatic is a demise of neurones. As astroglial cells carry out homeostatic functions of the brain, it is certain that these cells are at least in part a cause of Alzheimer's disease. Historically, Alois Alzheimer himself has recognised this at the dawn of the disease description. However, the role of astroglia in this disease has been understudied. In this chapter, we summarise the various aspects of glial contribution to this disease and outline the potential of using these cells in prevention (exercise and environmental enrichment) and intervention of this devastating disease.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK. .,Faculty of Health and Medical Sciences, Center for Basic and Translational Neuroscience, University of Copenhagen, 2200, Copenhagen, Denmark. .,Achucarro Center for Neuroscience, IKERBASQUE, Basque Foundation for Science, 48011, Bilbao, Spain.
| | - Vladimir Parpura
- Department of Neurobiology, The University of Alabama at Birmingham, Birmingham, AL, USA.,University of Rijeka, Rijeka, Croatia
| | - Jose Julio Rodriguez-Arellano
- BioCruces Health Research Institute, IKERBASQUE, Basque Foundation for Science, Bilbao, Spain.,Department of Neuroscience, The University of the Basque Country UPV/EHU, Plaza de Cruces 12, 48903, Barakaldo, Bizkaia, Spain
| | - Robert Zorec
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Faculty of Medicine, Institute of Pathophysiology, University of Ljubljana, Ljubljana, Slovenia.,Celica BIOMEDICAL, Ljubljana, Slovenia
| |
Collapse
|
40
|
Age-dependent behavioral and biochemical characterization of single APP knock-in mouse (APP NL-G-F/NL-G-F) model of Alzheimer's disease. Neurobiol Aging 2018; 75:25-37. [PMID: 30508733 DOI: 10.1016/j.neurobiolaging.2018.10.026] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 10/22/2018] [Accepted: 10/28/2018] [Indexed: 12/23/2022]
Abstract
Saito et al developed a novel amyloid precursor protein (APP) knock-in mouse model (APPNL-G-F) for Alzheimer's disease (AD) to overcome the problem of overexpression of APP in available transgenic mouse models. However, this new mouse model for AD is not fully characterized age-dependently with respect to behavioral and biochemical changes. Therefore, in the present study, we performed an age-dependent behavioral and biochemical characterization of this newly developed mouse model. Here, we used 3-, 6-, 9-, and 12-month-old APPNL-G-F and C57BL/6J mice. We used a separate cohort of animals at each age point. Morris water maze, object recognition, and fear-conditioning tests were used for the assessment of learning and memory functions and open-field test to measure the general locomotor activity of mice. After each testing point, we perfused the mice and collected the brain for immunostaining. We performed the immunostaining for amyloid burden (4G8), glial fibrillary acidic protein, choline acetyltransferase, and tyrosine hydroxylase. The results of the present study indicate that APPNL-G-F mice showed age-dependent memory impairments with maximum impairment at the age of 12 months. These mice showed memory impairment in Morris water maze and fear conditioning tests when they were 6 months old, whereas, in object recognition test, memory deficit was found in 9-month-old mice. APPNL-G-F mice age dependently showed an increase in amyloid load in different brain regions. However, no amyloid pathology was found in 3-month-old APPNL-G-F mice. Choline acetyltransferase neurons in medial septum-diagonal band complex and tyrosine hydroxylase neurons in locus coeruleus were decreased significantly in APPNL-G-F mice. This mouse model also indicated an age-dependent increase in glial fibrillary acidic protein load. It can be concluded from the results that the APPNL-G-F mouse model may be used to explore the Aβ hypothesis, molecular, and cellular mechanisms involved in AD pathology and to screen the therapeutic potential compounds for the treatment of AD.
Collapse
|