1
|
Benita BA, Koss KM. Peptide discovery across the spectrum of neuroinflammation; microglia and astrocyte phenotypical targeting, mediation, and mechanistic understanding. Front Mol Neurosci 2024; 17:1443985. [PMID: 39634607 PMCID: PMC11616451 DOI: 10.3389/fnmol.2024.1443985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 07/24/2024] [Indexed: 12/07/2024] Open
Abstract
Uncontrolled and chronic inflammatory states in the Central Nervous System (CNS) are the hallmark of neurodegenerative pathology and every injury or stroke-related insult. The key mediators of these neuroinflammatory states are glial cells known as microglia, the resident immune cell at the core of the inflammatory event, and astroglia, which encapsulate inflammatory insults in proteoglycan-rich scar tissue. Since the majority of neuroinflammation is exclusively based on the responses of said glia, their phenotypes have been identified to be on an inflammatory spectrum encompassing developmental, homeostatic, and reparative behaviors as opposed to their ability to affect devastating cell death cascades and scar tissue formation. Recently, research groups have focused on peptide discovery to identify these phenotypes, find novel mechanisms, and mediate or re-engineer their actions. Peptides retain the diverse function of proteins but significantly reduce the activity dependence on delicate 3D structures. Several peptides targeting unique phenotypes of microglia and astroglia have been identified, along with several capable of mediating deleterious behaviors or promoting beneficial outcomes in the context of neuroinflammation. A comprehensive review of the peptides unique to microglia and astroglia will be provided along with their primary discovery methodologies, including top-down approaches using known biomolecules and naïve strategies using peptide and phage libraries.
Collapse
Affiliation(s)
| | - Kyle M. Koss
- Department of Surgery, University of Arizona, Tucson, AZ, United States
- Department of Neurobiology, University of Texas Medical Branch (UTMB) at Galvestion, Galvestion, TX, United States
- Sealy Institute for Drug Discovery (SIDD), University of Texas Medical Branch (UTMB) at Galvestion, Galvestion, TX, United States
| |
Collapse
|
2
|
Lemche E, Killick R, Mitchell J, Caton PW, Choudhary P, Howard JK. Molecular mechanisms linking type 2 diabetes mellitus and late-onset Alzheimer's disease: A systematic review and qualitative meta-analysis. Neurobiol Dis 2024; 196:106485. [PMID: 38643861 DOI: 10.1016/j.nbd.2024.106485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 03/18/2024] [Accepted: 03/23/2024] [Indexed: 04/23/2024] Open
Abstract
Research evidence indicating common metabolic mechanisms through which type 2 diabetes mellitus (T2DM) increases risk of late-onset Alzheimer's dementia (LOAD) has accumulated over recent decades. The aim of this systematic review is to provide a comprehensive review of common mechanisms, which have hitherto been discussed in separate perspectives, and to assemble and evaluate candidate loci and epigenetic modifications contributing to polygenic risk linkages between T2DM and LOAD. For the systematic review on pathophysiological mechanisms, both human and animal studies up to December 2023 are included. For the qualitative meta-analysis of genomic bases, human association studies were examined; for epigenetic mechanisms, data from human studies and animal models were accepted. Papers describing pathophysiological studies were identified in databases, and further literature gathered from cited work. For genomic and epigenomic studies, literature mining was conducted by formalised search codes using Boolean operators in search engines, and augmented by GeneRif citations in Entrez Gene, and other sources (WikiGenes, etc.). For the systematic review of pathophysiological mechanisms, 923 publications were evaluated, and 138 gene loci extracted for testing candidate risk linkages. 3 57 publications were evaluated for genomic association and descriptions of epigenomic modifications. Overall accumulated results highlight insulin signalling, inflammation and inflammasome pathways, proteolysis, gluconeogenesis and glycolysis, glycosylation, lipoprotein metabolism and oxidation, cell cycle regulation or survival, autophagic-lysosomal pathways, and energy. Documented findings suggest interplay between brain insulin resistance, neuroinflammation, insult compensatory mechanisms, and peripheral metabolic dysregulation in T2DM and LOAD linkage. The results allow for more streamlined longitudinal studies of T2DM-LOAD risk linkages.
Collapse
Affiliation(s)
- Erwin Lemche
- Section of Cognitive Neuropsychiatry and Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, United Kingdom.
| | - Richard Killick
- Section of Old Age Psychiatry, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, United Kingdom
| | - Jackie Mitchell
- Department of Basic and Clinical Neurosciences, Maurice Wohl CIinical Neurosciences Institute, Institute of Psychiatry, Psychology & Neuroscience, King's College London, 125 Coldharbour Lane, London SE5 9NU, United Kingdom
| | - Paul W Caton
- Diabetes Research Group, School of Life Course Sciences, King's College London, Hodgkin Building, Guy's Campus, London SE1 1UL, United Kingdom
| | - Pratik Choudhary
- Diabetes Research Group, Weston Education Centre, King's College London, 10 Cutcombe Road, London SE5 9RJ, United Kingdom
| | - Jane K Howard
- School of Cardiovascular and Metabolic Medicine & Sciences, Hodgkin Building, Guy's Campus, King's College London, Great Maze Pond, London SE1 1UL, United Kingdom
| |
Collapse
|
3
|
Moțățăianu A, Mănescu IB, Șerban G, Bărcuțean L, Ion V, Bălașa R, Andone S. Exploring the Role of Metabolic Hormones in Amyotrophic Lateral Sclerosis. Int J Mol Sci 2024; 25:5059. [PMID: 38791099 PMCID: PMC11121721 DOI: 10.3390/ijms25105059] [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: 03/26/2024] [Revised: 04/27/2024] [Accepted: 05/04/2024] [Indexed: 05/26/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease characterized by progressive loss of motor neurons. Emerging evidence suggests a potential link between metabolic dysregulation and ALS pathogenesis. This study aimed to investigate the relationship between metabolic hormones and disease progression in ALS patients. A cross-sectional study was conducted involving 44 ALS patients recruited from a tertiary care center. Serum levels of insulin, total amylin, C-peptide, active ghrelin, GIP (gastric inhibitory peptide), GLP-1 active (glucagon-like peptide-1), glucagon, PYY (peptide YY), PP (pancreatic polypeptide), leptin, interleukin-6, MCP-1 (monocyte chemoattractant protein-1), and TNFα (tumor necrosis factor alpha) were measured, and correlations with ALSFRS-R, evolution scores, and biomarkers were analyzed using Spearman correlation coefficients. Subgroup analyses based on ALS subtypes, progression pattern of disease, and disease progression rate patterns were performed. Significant correlations were observed between metabolic hormones and ALS evolution scores. Insulin and amylin exhibited strong correlations with disease progression and clinical functional outcomes, with insulin showing particularly robust associations. Other hormones such as C-peptide, leptin, and GLP-1 also showed correlations with ALS progression and functional status. Subgroup analyses revealed differences in hormone levels based on sex and disease evolution patterns, with male patients showing higher amylin and glucagon levels. ALS patients with slower disease progression exhibited elevated levels of amylin and insulin. Our findings suggest a potential role for metabolic hormones in modulating ALS progression and functional outcomes. Further research is needed to elucidate the underlying mechanisms and explore the therapeutic implications of targeting metabolic pathways in ALS management.
Collapse
Affiliation(s)
- Anca Moțățăianu
- Department of Neurology, University of Medicine, Pharmacy, Science and Technology of Târgu Mureș ‘George Emil Palade’, 540142 Târgu Mureș, Romania
- 1st Neurology Clinic, Mures County Clinical Emergency Hospital, 540136 Târgu Mureș, Romania
| | - Ion Bogdan Mănescu
- Department of Laboratory Medicine, University of Medicine, Pharmacy, Science and Technology of Târgu Mureș ‘George Emil Palade’, 540142 Târgu Mureș, Romania
| | - Georgiana Șerban
- Doctoral School, University of Medicine, Pharmacy, Science and Technology of Târgu Mureș ‘George Emil Palade’, 540142 Târgu Mureș, Romania
| | - Laura Bărcuțean
- Department of Neurology, University of Medicine, Pharmacy, Science and Technology of Târgu Mureș ‘George Emil Palade’, 540142 Târgu Mureș, Romania
- 1st Neurology Clinic, Mures County Clinical Emergency Hospital, 540136 Târgu Mureș, Romania
| | - Valentin Ion
- Faculty of Pharmacy, Department of Analytical Chemistry and Drug Analysis, University of Medicine, Pharmacy, Science and Technology of Târgu Mureș ‘George Emil Palade’, 540142 Târgu Mureș, Romania
- Drug Testing Laboratory, University of Medicine, Pharmacy, Science and Technology of Târgu Mureș ‘George Emil Palade’, 540142 Târgu Mureș, Romania
| | - Rodica Bălașa
- Department of Neurology, University of Medicine, Pharmacy, Science and Technology of Târgu Mureș ‘George Emil Palade’, 540142 Târgu Mureș, Romania
- 1st Neurology Clinic, Mures County Clinical Emergency Hospital, 540136 Târgu Mureș, Romania
| | - Sebastian Andone
- Department of Neurology, University of Medicine, Pharmacy, Science and Technology of Târgu Mureș ‘George Emil Palade’, 540142 Târgu Mureș, Romania
- 1st Neurology Clinic, Mures County Clinical Emergency Hospital, 540136 Târgu Mureș, Romania
| |
Collapse
|
4
|
Widjaya MA, Liu CH, Lee SD, Cheng WC. Transcriptomics Meta-Analysis Reveals Phagosome and Innate Immune System Dysfunction as Potential Mechanisms in the Cortex of Alzheimer's Disease Mouse Strains. J Mol Neurosci 2023; 73:773-786. [PMID: 37733230 DOI: 10.1007/s12031-023-02152-9] [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: 07/05/2023] [Accepted: 08/30/2023] [Indexed: 09/22/2023]
Abstract
Immune-related pathways can affect the immune system directly, such as the chemokine signaling pathway, or indirectly, such as the phagosome pathway. Alzheimer's disease (AD) is reportedly associated with several immune-related pathways. However, exploring its underlying mechanism is challenging in animal studies because AD mouse strains differentially express immune-related pathway characteristics. To overcome this problem, we performed a meta-analysis to identify significant and consistent immune-related AD pathways that are expressed in different AD mouse strains. Next-generation RNA sequencing (RNA-seq) and microarray datasets for the cortex of AD mice from different strains such as APP/PSEN1, APP/PS2, 3xTg, TREM, and 5xFAD were collected from the NCBI GEO database. Each dataset's quality control and normalization were already processed from each original study source using various methods depending on the high-throughput analysis platform (FastQC, median of ratios, RMA, between array normalization). Datasets were analyzed using DESeq2 for RNA-seq and GEO2R for microarray to identify differentially expressed (DE) genes. Significantly DE genes were meta-analyzed using Stouffer's method, with significant genes further analyzed for functional enrichment. Ten datasets representing 20 conditions were obtained from the NCBI GEO database, comprising 116 control and 120 AD samples. The DE analysis identified 284 significant DE genes. The meta-analysis identified three significantly enriched immune-related AD pathways: phagosome, the complement and coagulation cascade, and chemokine signaling. Phagosomes-related genes correlated with complement and immune system. Meanwhile, phagosomes and chemokine signaling genes overlapped with B cells receptors pathway genes indicating potential correlation between phagosome, chemokines, and adaptive immune system as well. The transcriptomic meta-analysis showed that AD is associated with immune-related pathways in the brain's cortex through the phagosome, complement and coagulation cascade, and chemokine signaling pathways. Interestingly, phagosome and chemokine signaling pathways had potential correlation with B cells receptors pathway.
Collapse
Affiliation(s)
- Michael Anekson Widjaya
- Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung, 40402, Taiwan
| | - Chia-Hsin Liu
- Cancer Biology and Precision Therapeutics Center, China Medical University and Academia Sinica China Medical University, Taichung, 40403, Taiwan
| | - Shin-Da Lee
- Department of Physical Therapy, PhD program in Healthcare Science, China Medical University, Taichung, 406040, Taiwan.
| | - Wei-Chung Cheng
- Cancer Biology and Precision Therapeutics Center, China Medical University and Academia Sinica China Medical University, Taichung, 40403, Taiwan.
- Ph.D. Program for Cancer Biology and Drug Discovery, China Medical University and Academia Sinica, Taichung, Taiwan.
| |
Collapse
|
5
|
Peng G, Li M, Meng Z. Polysaccharides: potential bioactive macromolecules for Alzheimer's disease. Front Nutr 2023; 10:1249018. [PMID: 37781122 PMCID: PMC10540640 DOI: 10.3389/fnut.2023.1249018] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 09/01/2023] [Indexed: 10/03/2023] Open
Abstract
Alzheimer's disease (AD) is one of the leading causes of death and disability. AD is a devastating disease that has caused an overwhelming burden. However, no disease-modified treatment was discovered. The approval of sodium oligomannate (GV-971) in mild-moderate AD patients has attracted great attention to investigate the role of saccharides in AD. Therefore, summarizing and explaining the role of saccharides in AD is urgent and promising. Recent studies showed that polysaccharides (PSs) potentially benefit AD in vitro and in vivo. PSs could alleviate the pathological damage and improve cognitive symptoms via (1) antagonizing the toxicity of abnormal amyloid-beta and tau proteins; (2) attenuating oxidative stress and proinflammation; (3) rebuilding neuroplasticity. PSs exhibit one-multiple pathological hits of AD. However, a thorough chemical investigation is needed for further study.
Collapse
Affiliation(s)
- Gong Peng
- Laboratory of Tumor Immunology, The First Hospital of Jilin University, Changchun, China
| | - Ming Li
- Department of Neurology, The Second Hospital of Nanchang University, Nanchang, China
| | - Zhaoli Meng
- Laboratory of Tumor Immunology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
6
|
Papiri G, D’Andreamatteo G, Cacchiò G, Alia S, Silvestrini M, Paci C, Luzzi S, Vignini A. Multiple Sclerosis: Inflammatory and Neuroglial Aspects. Curr Issues Mol Biol 2023; 45:1443-1470. [PMID: 36826039 PMCID: PMC9954863 DOI: 10.3390/cimb45020094] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/28/2023] [Accepted: 02/02/2023] [Indexed: 02/11/2023] Open
Abstract
Multiple sclerosis (MS) represents the most common acquired demyelinating disorder of the central nervous system (CNS). Its pathogenesis, in parallel with the well-established role of mechanisms pertaining to autoimmunity, involves several key functions of immune, glial and nerve cells. The disease's natural history is complex, heterogeneous and may evolve over a relapsing-remitting (RRMS) or progressive (PPMS/SPMS) course. Acute inflammation, driven by infiltration of peripheral cells in the CNS, is thought to be the most relevant process during the earliest phases and in RRMS, while disruption in glial and neural cells of pathways pertaining to energy metabolism, survival cascades, synaptic and ionic homeostasis are thought to be mostly relevant in long-standing disease, such as in progressive forms. In this complex scenario, many mechanisms originally thought to be distinctive of neurodegenerative disorders are being increasingly recognized as crucial from the beginning of the disease. The present review aims at highlighting mechanisms in common between MS, autoimmune diseases and biology of neurodegenerative disorders. In fact, there is an unmet need to explore new targets that might be involved as master regulators of autoimmunity, inflammation and survival of nerve cells.
Collapse
Affiliation(s)
- Giulio Papiri
- Neurology Unit, Ospedale Provinciale “Madonna del Soccorso”, 63074 San Benedetto del Tronto, Italy
| | - Giordano D’Andreamatteo
- Neurology Unit, Ospedale Provinciale “Madonna del Soccorso”, 63074 San Benedetto del Tronto, Italy
| | - Gabriella Cacchiò
- Neurology Unit, Ospedale Provinciale “Madonna del Soccorso”, 63074 San Benedetto del Tronto, Italy
| | - Sonila Alia
- Section of Biochemistry, Biology and Physics, Department of Clinical Sciences, Università Politecnica delle Marche, 60100 Ancona, Italy
| | - Mauro Silvestrini
- Neurology Unit, Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60100 Ancona, Italy
| | - Cristina Paci
- Neurology Unit, Ospedale Provinciale “Madonna del Soccorso”, 63074 San Benedetto del Tronto, Italy
| | - Simona Luzzi
- Neurology Unit, Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60100 Ancona, Italy
| | - Arianna Vignini
- Section of Biochemistry, Biology and Physics, Department of Clinical Sciences, Università Politecnica delle Marche, 60100 Ancona, Italy
- Correspondence:
| |
Collapse
|
7
|
Piras IS, Brokaw D, Kong Y, Weisenberger DJ, Krate J, Delvaux E, Mahurkar S, Blattler A, Siegmund KD, Sue L, Serrano GE, Beach TG, Laird PW, Huentelman MJ, Coleman PD. Integrated DNA Methylation/RNA Profiling in Middle Temporal Gyrus of Alzheimer's Disease. Cell Mol Neurobiol 2023:10.1007/s10571-022-01307-3. [PMID: 36596913 DOI: 10.1007/s10571-022-01307-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 11/08/2022] [Indexed: 01/05/2023]
Abstract
Alzheimer's disease is a neurodegenerative disorder clinically defined by gradual cognitive impairment and alteration in executive function. We conducted an epigenome-wide association study (EWAS) of a clinically and neuropathologically characterized cohort of 296 brains, including Alzheimer's disease (AD) and non-demented controls (ND), exploring the relationship with the RNA expression from matched donors. We detected 5246 CpGs and 832 regions differentially methylated, finding overlap with previous EWAS but also new associations. CpGs previously identified in ANK1, MYOC, and RHBDF2 were differentially methylated, and one of our top hits (GPR56) was not previously detected. ANK1 was differentially methylated at the region level, along with APOE and RHBDF2. Only a small number of genes showed a correlation between DNA methylation and RNA expression statistically significant. Multiblock partial least-squares discriminant analysis showed several CpG sites and RNAs discriminating AD and ND (AUC = 0.908) and strongly correlated with each other. Furthermore, the CpG site cg25038311 was negatively correlated with the expression of 22 genes. Finally, with the functional epigenetic module analysis, we identified a protein-protein network characterized by inverse RNA/DNA methylation correlation and enriched for "Regulation of insulin-like growth factor transport", with IGF1 as the hub gene. Our results confirm and extend the previous EWAS, providing new information about a brain region not previously explored in AD DNA methylation studies. The relationship between DNA methylation and gene expression is not significant for most of the genes in our sample, consistently with the complexities in the gene expression regulation.
Collapse
Affiliation(s)
- Ignazio S Piras
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ, 85004, USA
| | - Danielle Brokaw
- Biodesign Institute, Neurodegenerative Disease Research Center, Arizona State University, Tempe, AZ, 85287, USA
| | - Yinfei Kong
- Department of Information Systems and Decision Sciences, California State University Fullerton, Fullerton, CA, 92831, USA
| | - Daniel J Weisenberger
- Department of Biochemistry and Molecular Biology, University of South California, Los Angeles, CA, 90033, USA
| | - Jonida Krate
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ, 85004, USA
- UnityPoint Clinic, Waterloo, IA, USA
| | - Elaine Delvaux
- Biodesign Institute, Neurodegenerative Disease Research Center, Arizona State University, Tempe, AZ, USA
| | - Swapna Mahurkar
- UCLA Division of Digestive Diseases, University of California, Los Angeles, CA, 90024, USA
| | - Adam Blattler
- Department of Biochemistry and Molecular Biology, University of South California, Los Angeles, CA, 90033, USA
- Genetics Graduate Group, University of California, Davis, CA, 95616, USA
| | - Kimberly D Siegmund
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA, 90089-9175, USA
| | - Lucia Sue
- Civin Laboratory of Neuropathology, Banner Sun Health Research Institute, Sun City, AZ, 85351, USA
| | - Geidy E Serrano
- Civin Laboratory of Neuropathology, Banner Sun Health Research Institute, Sun City, AZ, 85351, USA
| | - Thomas G Beach
- Civin Laboratory of Neuropathology, Banner Sun Health Research Institute, Sun City, AZ, 85351, USA
| | - Peter W Laird
- Center for Epigenetics, Van Andel Institute, Grand Rapids, MI, 49503, USA
| | - Matthew J Huentelman
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ, 85004, USA
| | - Paul D Coleman
- Biodesign Institute, Neurodegenerative Disease Research Center, Arizona State University, Tempe, AZ, 85287, USA.
| |
Collapse
|
8
|
Garcia-Segura ME, Durainayagam BR, Liggi S, Graça G, Jimenez B, Dehghan A, Tzoulaki I, Karaman I, Elliott P, Griffin JL. Pathway-based integration of multi-omics data reveals lipidomics alterations validated in an Alzheimer's disease mouse model and risk loci carriers. J Neurochem 2023; 164:57-76. [PMID: 36326588 PMCID: PMC10107183 DOI: 10.1111/jnc.15719] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 09/14/2022] [Accepted: 09/16/2022] [Indexed: 11/06/2022]
Abstract
Alzheimer's disease (AD) is a highly prevalent neurodegenerative disorder. Despite increasing evidence of the importance of metabolic dysregulation in AD, the underlying metabolic changes that may impact amyloid plaque formation are not understood, particularly for late-onset AD. This study analyzed genome-wide association studies (GWAS), transcriptomics, and proteomics data obtained from several data repositories to obtain differentially expressed (DE) multi-omics elements in mouse models of AD. We characterized the metabolic modulation in these data sets using gene ontology, transcription factor, pathway, and cell-type enrichment analyses. A predicted lipid signature was extracted from genome-scale metabolic networks (GSMN) and subsequently validated in a lipidomic data set derived from cortical tissue of ABCA-7 null mice, a mouse model of one of the genes associated with late-onset AD. Moreover, a metabolome-wide association study (MWAS) was performed to further characterize the association between dysregulated lipid metabolism in human blood serum and genes associated with AD risk. We found 203 DE transcripts, 164 DE proteins, and 58 DE GWAS-derived mouse orthologs associated with significantly enriched metabolic biological processes. Lipid and bioenergetic metabolic pathways were significantly over-represented across the AD multi-omics data sets. Microglia and astrocytes were significantly enriched in the lipid-predominant AD-metabolic transcriptome. We also extracted a predicted lipid signature that was validated and robustly modeled class separation in the ABCA7 mice cortical lipidome, with 11 of these lipid species exhibiting statistically significant modulations. MWAS revealed 298 AD single nucleotide polymorphisms-metabolite associations, of which 70% corresponded to lipid classes. These results support the importance of lipid metabolism dysregulation in AD and highlight the suitability of mapping AD multi-omics data into GSMNs to identify metabolic alterations.
Collapse
Affiliation(s)
- Monica Emili Garcia-Segura
- Department of Brain Sciences, Imperial College London, London, UK.,Section of Biomolecular Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Brenan R Durainayagam
- Section of Biomolecular Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.,UK-Dementia Research Institute (UK-DRI) at Imperial College London, London, UK
| | - Sonia Liggi
- Section of Biomolecular Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Gonçalo Graça
- Section of Bioinformatics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Beatriz Jimenez
- Section of Bioanalytical Chemistry and the National Phenome Centre, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Abbas Dehghan
- UK-Dementia Research Institute (UK-DRI) at Imperial College London, London, UK.,Department of Epidemiology and Biostatistics, Imperial College London, London, UK.,MRC Centre for Environment and Health, Imperial College London, London, UK
| | - Ioanna Tzoulaki
- UK-Dementia Research Institute (UK-DRI) at Imperial College London, London, UK.,Department of Epidemiology and Biostatistics, Imperial College London, London, UK.,National Institute for Health Research Imperial Biomedical Research Centre, Imperial College London, UK.,Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | - Ibrahim Karaman
- Section of Bioinformatics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.,Department of Epidemiology and Biostatistics, Imperial College London, London, UK
| | - Paul Elliott
- UK-Dementia Research Institute (UK-DRI) at Imperial College London, London, UK.,Department of Epidemiology and Biostatistics, Imperial College London, London, UK.,MRC Centre for Environment and Health, Imperial College London, London, UK.,National Institute for Health Research Imperial Biomedical Research Centre, Imperial College London, UK
| | - Julian L Griffin
- Section of Biomolecular Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.,UK-Dementia Research Institute (UK-DRI) at Imperial College London, London, UK.,Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge, UK.,The Rowett Institute, University of Aberdeen, Aberdeen, Scotland
| |
Collapse
|
9
|
Corrigan RR, Labrador L, Grizzanti J, Mey M, Piontkivska H, Casadesús G. Neuroprotective Mechanisms of Amylin Receptor Activation, Not Antagonism, in the APP/PS1 Mouse Model of Alzheimer's Disease. J Alzheimers Dis 2023; 91:1495-1514. [PMID: 36641678 DOI: 10.3233/jad-221057] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
BACKGROUND Amylin, a pancreatic amyloid peptide involved in energy homeostasis, is increasingly studied in the context of Alzheimer's disease (AD) etiology. To date, conflicting pathogenic and neuroprotective roles for this peptide and its analogs for AD pathogenesis have been described. OBJECTIVE Whether the benefits of amylin are associated with peripheral improvement of metabolic tone/function or directly through the activation of central amylin receptors is also unknown and downstream signaling mechanisms of amylin receptors are major objectives of this study. METHODS To address these questions more directly we delivered the amylin analog pramlintide systemically (IP), at previously identified therapeutic doses, while centrally (ICV) inhibiting the receptor using an amylin receptor antagonist (AC187), at doses known to impact CNS function. RESULTS Here we show that pramlintide improved cognitive function independently of CNS receptor activation and provide transcriptomic data that highlights potential mechanisms. Furthermore, we show than inhibition of the amylin receptor increased amyloid-beta pathology in female APP/PS1 mice, an effect than was mitigated by peripheral delivery of pramlintide. Through transcriptomic analysis of pramlintide therapy in AD-modeled mice we found sexual dimorphic modulation of neuroprotective mechanisms: oxidative stress protection in females and membrane stability and reduced neuronal excitability markers in males. CONCLUSION These data suggest an uncoupling of functional and pathology-related events and highlighting a more complex receptor system and pharmacological relationship that must be carefully studied to clarify the role of amylin in CNS function and AD.
Collapse
Affiliation(s)
| | - Luis Labrador
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA
| | - John Grizzanti
- School of Biomedical Sciences, Kent State University, Kent, OH, USA
| | - Megan Mey
- School of Biomedical Sciences, Kent State University, Kent, OH, USA
| | - Helen Piontkivska
- Department of Biological Sciences, Kent State University, Kent, OH, USA
| | - Gemma Casadesús
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA
| |
Collapse
|
10
|
Garcia-Segura ME, Durainayagam BR, Liggi S, Graça G, Jimenez B, Dehghan A, Tzoulaki I, Karaman I, Elliott P, Griffin JL. Pathway-based integration of multi-omics data reveals lipidomics alterations validated in an Alzheimer's disease mouse model and risk loci carriers. J Neurochem 2023. [PMID: 36326588 DOI: 10.1101/2021.05.10.21255052v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Alzheimer's disease (AD) is a highly prevalent neurodegenerative disorder. Despite increasing evidence of the importance of metabolic dysregulation in AD, the underlying metabolic changes that may impact amyloid plaque formation are not understood, particularly for late-onset AD. This study analyzed genome-wide association studies (GWAS), transcriptomics, and proteomics data obtained from several data repositories to obtain differentially expressed (DE) multi-omics elements in mouse models of AD. We characterized the metabolic modulation in these data sets using gene ontology, transcription factor, pathway, and cell-type enrichment analyses. A predicted lipid signature was extracted from genome-scale metabolic networks (GSMN) and subsequently validated in a lipidomic data set derived from cortical tissue of ABCA-7 null mice, a mouse model of one of the genes associated with late-onset AD. Moreover, a metabolome-wide association study (MWAS) was performed to further characterize the association between dysregulated lipid metabolism in human blood serum and genes associated with AD risk. We found 203 DE transcripts, 164 DE proteins, and 58 DE GWAS-derived mouse orthologs associated with significantly enriched metabolic biological processes. Lipid and bioenergetic metabolic pathways were significantly over-represented across the AD multi-omics data sets. Microglia and astrocytes were significantly enriched in the lipid-predominant AD-metabolic transcriptome. We also extracted a predicted lipid signature that was validated and robustly modeled class separation in the ABCA7 mice cortical lipidome, with 11 of these lipid species exhibiting statistically significant modulations. MWAS revealed 298 AD single nucleotide polymorphisms-metabolite associations, of which 70% corresponded to lipid classes. These results support the importance of lipid metabolism dysregulation in AD and highlight the suitability of mapping AD multi-omics data into GSMNs to identify metabolic alterations.
Collapse
Affiliation(s)
- Monica Emili Garcia-Segura
- Department of Brain Sciences, Imperial College London, London, UK
- Section of Biomolecular Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Brenan R Durainayagam
- Section of Biomolecular Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- UK-Dementia Research Institute (UK-DRI) at Imperial College London, London, UK
| | - Sonia Liggi
- Section of Biomolecular Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Gonçalo Graça
- Section of Bioinformatics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Beatriz Jimenez
- Section of Bioanalytical Chemistry and the National Phenome Centre, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Abbas Dehghan
- UK-Dementia Research Institute (UK-DRI) at Imperial College London, London, UK
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- MRC Centre for Environment and Health, Imperial College London, London, UK
| | - Ioanna Tzoulaki
- UK-Dementia Research Institute (UK-DRI) at Imperial College London, London, UK
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- National Institute for Health Research Imperial Biomedical Research Centre, Imperial College London, UK
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | - Ibrahim Karaman
- Section of Bioinformatics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
| | - Paul Elliott
- UK-Dementia Research Institute (UK-DRI) at Imperial College London, London, UK
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- MRC Centre for Environment and Health, Imperial College London, London, UK
- National Institute for Health Research Imperial Biomedical Research Centre, Imperial College London, UK
| | - Julian L Griffin
- Section of Biomolecular Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- UK-Dementia Research Institute (UK-DRI) at Imperial College London, London, UK
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge, UK
- The Rowett Institute, University of Aberdeen, Aberdeen, Scotland
| |
Collapse
|
11
|
Russo M, De Rosa MA, Calisi D, Consoli S, Evangelista G, Dono F, Santilli M, Granzotto A, Onofrj M, Sensi SL. Migraine Pharmacological Treatment and Cognitive Impairment: Risks and Benefits. Int J Mol Sci 2022; 23:11418. [PMID: 36232720 PMCID: PMC9569564 DOI: 10.3390/ijms231911418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/19/2022] [Accepted: 09/20/2022] [Indexed: 11/23/2022] Open
Abstract
Migraine is a common neurological disorder impairing the quality of life of patients. The condition requires, as an acute or prophylactic line of intervention, the frequent use of drugs acting on the central nervous system (CNS). The long-term impact of these medications on cognition and neurodegeneration has never been consistently assessed. The paper reviews pharmacological migraine treatments and discusses their biological and clinical effects on the CNS. The different anti-migraine drugs show distinct profiles concerning neurodegeneration and the risk of cognitive deficits. These features should be carefully evaluated when prescribing a pharmacological treatment as many migraineurs are of scholar or working age and their performances may be affected by drug misuse. Thus, a reconsideration of therapy guidelines is warranted. Furthermore, since conflicting results have emerged in the relationship between migraine and dementia, future studies must consider present and past pharmacological regimens as potential confounding factors.
Collapse
Affiliation(s)
- Mirella Russo
- Department of Neurosciences, Imaging and Clinical Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- CAST—Center for Advanced Studies and Technology, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Matteo A. De Rosa
- Department of Neurosciences, Imaging and Clinical Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Dario Calisi
- Department of Neurosciences, Imaging and Clinical Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Stefano Consoli
- Department of Neurosciences, Imaging and Clinical Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Giacomo Evangelista
- Department of Neurosciences, Imaging and Clinical Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Fedele Dono
- Department of Neurosciences, Imaging and Clinical Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- CAST—Center for Advanced Studies and Technology, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Matteo Santilli
- Department of Neurosciences, Imaging and Clinical Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Alberto Granzotto
- Department of Neurosciences, Imaging and Clinical Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- CAST—Center for Advanced Studies and Technology, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Marco Onofrj
- Department of Neurosciences, Imaging and Clinical Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- CAST—Center for Advanced Studies and Technology, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Stefano L. Sensi
- Department of Neurosciences, Imaging and Clinical Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- CAST—Center for Advanced Studies and Technology, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- Institute for Mind Impairments and Neurological Disorders-iMIND, University of California, Irvine, Irvine, CA 92697, USA
- ITAB—Institute of Advanced Biomedical Technology, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| |
Collapse
|
12
|
Wang Q, Lu M, Zhu X, Gu X, Zhang T, Xia C, Yang L, Xu Y, Zhou M. The role of microglia immunometabolism in neurodegeneration: Focus on molecular determinants and metabolic intermediates of metabolic reprogramming. Biomed Pharmacother 2022; 153:113412. [DOI: 10.1016/j.biopha.2022.113412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 07/10/2022] [Accepted: 07/11/2022] [Indexed: 11/16/2022] Open
|
13
|
Ma Z, Yang Z, Feng X, Deng J, He C, Li R, Zhao Y, Ge Y, Zhang Y, Song C, Zhong S. The Emerging Evidence for a Protective Role of Fucoidan from Laminaria japonica in Chronic Kidney Disease-Triggered Cognitive Dysfunction. Mar Drugs 2022; 20:258. [PMID: 35447931 PMCID: PMC9025131 DOI: 10.3390/md20040258] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 04/04/2022] [Accepted: 04/05/2022] [Indexed: 02/06/2023] Open
Abstract
This study aimed to explore the mechanism of fucoidan in chronic kidney disease (CKD)-triggered cognitive dysfunction. The adenine-induced ICR strain CKD mice model was applied, and RNA-Seq was performed for differential gene analysis between aged-CKD and normal mice. As a result, fucoidan (100 and 200 mg kg-1) significantly reversed adenine-induced high expression of urea, uric acid in urine, and creatinine in serum, as well as the novel object recognition memory and spatial memory deficits. RNA sequencing analysis indicated that oxidative and inflammatory signaling were involved in adenine-induced kidney injury and cognitive dysfunction; furthermore, fucoidan inhibited oxidative stress via GSK3β-Nrf2-HO-1 signaling and ameliorated inflammatory response through regulation of microglia/macrophage polarization in the kidney and hippocampus of CKD mice. Additionally, we clarified six hallmarks in the hippocampus and four in the kidney, which were correlated with CKD-triggered cognitive dysfunction. This study provides a theoretical basis for the application of fucoidan in the treatment of CKD-triggered memory deficits.
Collapse
Affiliation(s)
- Zhihui Ma
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (Z.M.); (X.F.); (J.D.); (C.H.); (R.L.); (Y.Z.); (Y.Z.); (C.S.); (S.Z.)
| | - Zhiyou Yang
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (Z.M.); (X.F.); (J.D.); (C.H.); (R.L.); (Y.Z.); (Y.Z.); (C.S.); (S.Z.)
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| | - Xinyue Feng
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (Z.M.); (X.F.); (J.D.); (C.H.); (R.L.); (Y.Z.); (Y.Z.); (C.S.); (S.Z.)
| | - Jiahang Deng
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (Z.M.); (X.F.); (J.D.); (C.H.); (R.L.); (Y.Z.); (Y.Z.); (C.S.); (S.Z.)
| | - Chuantong He
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (Z.M.); (X.F.); (J.D.); (C.H.); (R.L.); (Y.Z.); (Y.Z.); (C.S.); (S.Z.)
| | - Rui Li
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (Z.M.); (X.F.); (J.D.); (C.H.); (R.L.); (Y.Z.); (Y.Z.); (C.S.); (S.Z.)
| | - Yuntao Zhao
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (Z.M.); (X.F.); (J.D.); (C.H.); (R.L.); (Y.Z.); (Y.Z.); (C.S.); (S.Z.)
| | - Yuewei Ge
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, Guangdong Pharmaceutical University, Guangzhou 510006, China;
| | - Yongping Zhang
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (Z.M.); (X.F.); (J.D.); (C.H.); (R.L.); (Y.Z.); (Y.Z.); (C.S.); (S.Z.)
| | - Cai Song
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (Z.M.); (X.F.); (J.D.); (C.H.); (R.L.); (Y.Z.); (Y.Z.); (C.S.); (S.Z.)
| | - Saiyi Zhong
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (Z.M.); (X.F.); (J.D.); (C.H.); (R.L.); (Y.Z.); (Y.Z.); (C.S.); (S.Z.)
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| |
Collapse
|
14
|
Papiri G, Vignini A, Capriotti L, Verdenelli P, Alia S, Di Paolo A, Fiori C, Baldinelli S, Silvestrini M, Luzzi S. Cerebrospinal Fluid α-Calcitonin Gene-Related Peptide: A Comparison between Alzheimer's Disease and Multiple Sclerosis. Biomolecules 2022; 12:199. [PMID: 35204700 PMCID: PMC8961604 DOI: 10.3390/biom12020199] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/17/2022] [Accepted: 01/21/2022] [Indexed: 02/04/2023] Open
Abstract
Alzheimer's disease (AD) and Multiple Sclerosis (MS) represent an emerging health problem on a global scale, as they are responsible for a significant contribution to the burden of disability in Western countries. Limited numbers of cerebrospinal fluid (CSF) diagnostic markers are available for each disease (amyloid and tau deposition markers for AD and oligoclonal bands for MS) representing mostly state markers that provide few, if any, clues about the severity of the clinical phenotype. α-CGRP is a neuropeptide implied in nociception, vasodilation, synaptic plasticity and immune functions. This neuropeptide is expressed in encephalic regions connected to memory, attention, autonomic and behavioral functions and is also expressed by spinal motor neurons. The present work confronted α-CGRP levels between 19 AD, 27 MS and 17 control subjects using an ELISA/EIA assay. We measured higher CSF α-CGRP contents in control subjects with respect to AD, as shown in previous studies, as well as in MS patients in comparison to AD. The control subjects and MS patients did not significantly differ between each other. We did not observe a relationship between CSF protein content, albumin quotient and α-CGRP. We also describe, retrospectively, an association between higher CSF CGRP content and higher MRI overall lesion count in MS and between lower α-CGRP and worse attention and visuo-perceptual skills in AD. We speculate that α-CGRP could be differentially involved in both disabling diseases.
Collapse
Affiliation(s)
- Giulio Papiri
- Neurology Clinic, Azienda Ospedaliero Universitaria, Ospedali Riuniti di Ancona, 60126 Torrette di Ancona, Italy; (G.P.); (C.F.); (S.B.); (M.S.); (S.L.)
| | - Arianna Vignini
- Section of Biochemistry, Department of Clinical Sciences, Biology and Physics, Università Politecnica delle Marche, 60126 Ancona, Italy; (S.A.); (A.D.P.)
| | - Luigi Capriotti
- Anesthesiology and Intensive Care Unit, Ospedale Provinciale “Madonna del Soccorso”, 63074 San Benedetto del Tronto, Italy; (L.C.); (P.V.)
| | - Paola Verdenelli
- Anesthesiology and Intensive Care Unit, Ospedale Provinciale “Madonna del Soccorso”, 63074 San Benedetto del Tronto, Italy; (L.C.); (P.V.)
| | - Sonila Alia
- Section of Biochemistry, Department of Clinical Sciences, Biology and Physics, Università Politecnica delle Marche, 60126 Ancona, Italy; (S.A.); (A.D.P.)
| | - Alice Di Paolo
- Section of Biochemistry, Department of Clinical Sciences, Biology and Physics, Università Politecnica delle Marche, 60126 Ancona, Italy; (S.A.); (A.D.P.)
| | - Chiara Fiori
- Neurology Clinic, Azienda Ospedaliero Universitaria, Ospedali Riuniti di Ancona, 60126 Torrette di Ancona, Italy; (G.P.); (C.F.); (S.B.); (M.S.); (S.L.)
| | - Sara Baldinelli
- Neurology Clinic, Azienda Ospedaliero Universitaria, Ospedali Riuniti di Ancona, 60126 Torrette di Ancona, Italy; (G.P.); (C.F.); (S.B.); (M.S.); (S.L.)
| | - Mauro Silvestrini
- Neurology Clinic, Azienda Ospedaliero Universitaria, Ospedali Riuniti di Ancona, 60126 Torrette di Ancona, Italy; (G.P.); (C.F.); (S.B.); (M.S.); (S.L.)
| | - Simona Luzzi
- Neurology Clinic, Azienda Ospedaliero Universitaria, Ospedali Riuniti di Ancona, 60126 Torrette di Ancona, Italy; (G.P.); (C.F.); (S.B.); (M.S.); (S.L.)
| |
Collapse
|
15
|
Patel A, Kimura R, Fu W, Soudy R, MacTavish D, Westaway D, Yang J, Davey RA, Zajac JD, Jhamandas JH. Genetic Depletion of Amylin/Calcitonin Receptors Improves Memory and Learning in Transgenic Alzheimer's Disease Mouse Models. Mol Neurobiol 2021; 58:5369-5382. [PMID: 34312771 PMCID: PMC8497456 DOI: 10.1007/s12035-021-02490-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 07/08/2021] [Indexed: 01/05/2023]
Abstract
Based upon its interactions with amyloid β peptide (Aβ), the amylin receptor, a class B G protein-coupled receptor (GPCR), is a potential modulator of Alzheimer's disease (AD) pathogenesis. However, past pharmacological approaches have failed to resolve whether activation or blockade of this receptor would have greater therapeutic benefit. To address this issue, we generated compound mice expressing a human amyloid precursor protein gene with familial AD mutations in combination with deficiency of amylin receptors produced by hemizygosity for the critical calcitonin receptor subunit of this heterodimeric GPCR. These compound transgenic AD mice demonstrated attenuated responses to human amylin- and Aβ-induced depression of hippocampal long-term potentiation (LTP) in keeping with the genetic depletion of amylin receptors. Both the LTP responses and spatial memory (as measured with Morris water maze) in these mice were improved compared to AD mouse controls and, importantly, a reduction in both the amyloid plaque burden and markers of neuroinflammation was observed. Our data support the notion of further development of antagonists of the amylin receptor as AD-modifying therapies.
Collapse
Affiliation(s)
- Aarti Patel
- Department of Medicine (Neurology), Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, T6G 2S2, Canada
| | - Ryoichi Kimura
- Department of Medicine (Neurology), Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, T6G 2S2, Canada
- Center for Liberal Arts and Sciences, Sanyo-Onoda City University, Yamaguchi , 756-0884, Japan
| | - Wen Fu
- Department of Medicine (Neurology), Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, T6G 2S2, Canada
| | - Rania Soudy
- Department of Medicine (Neurology), Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, T6G 2S2, Canada
- Faculty of Pharmacy, Cairo University, Giza, Egypt
| | - David MacTavish
- Department of Medicine (Neurology), Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, T6G 2S2, Canada
| | - David Westaway
- Department of Medicine (Neurology), Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, T6G 2S2, Canada
- Department of Biochemistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB, T6G 2M8, Canada
| | - Jing Yang
- Department of Medicine (Neurology), Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, T6G 2S2, Canada
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB, T6G 2M8, Canada
| | - Rachel A Davey
- Department of Medicine, University of Melbourne, Austin HealthHeidelberg, VIC, 3074, Australia
| | - Jeffrey D Zajac
- Department of Medicine, University of Melbourne, Austin HealthHeidelberg, VIC, 3074, Australia
| | - Jack H Jhamandas
- Department of Medicine (Neurology), Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, T6G 2S2, Canada.
| |
Collapse
|
16
|
Li H, Zou L, Shi J, Han X. Bioinformatics analysis of differentially expressed genes and identification of an miRNA-mRNA network associated with entorhinal cortex and hippocampus in Alzheimer's disease. Hereditas 2021; 158:25. [PMID: 34243818 PMCID: PMC8272337 DOI: 10.1186/s41065-021-00190-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 06/28/2021] [Indexed: 01/09/2023] Open
Abstract
Background Alzheimer’s disease (AD) is a fatal neurodegenerative disorder, and the lesions originate in the entorhinal cortex (EC) and hippocampus (HIP) at the early stage of AD progression. Gaining insight into the molecular mechanisms underlying AD is critical for the diagnosis and treatment of this disorder. Recent discoveries have uncovered the essential roles of microRNAs (miRNAs) in aging and have identified the potential of miRNAs serving as biomarkers in AD diagnosis. Methods We sought to apply bioinformatics tools to investigate microarray profiles and characterize differentially expressed genes (DEGs) in both EC and HIP and identify specific candidate genes and pathways that might be implicated in AD for further analysis. Furthermore, we considered that DEGs might be dysregulated by miRNAs. Therefore, we investigated patients with AD and healthy controls by studying the gene profiling of their brain and blood samples to identify AD-related DEGs, differentially expressed miRNAs (DEmiRNAs), along with gene ontology (GO) analysis, KEGG pathway analysis, and construction of an AD-specific miRNA–mRNA interaction network. Results Our analysis identified 10 key hub genes in the EC and HIP of patients with AD, and these hub genes were focused on energy metabolism, suggesting that metabolic dyshomeostasis contributed to the progression of the early AD pathology. Moreover, after the construction of an miRNA–mRNA network, we identified 9 blood-related DEmiRNAs, which regulated 10 target genes in the KEGG pathway. Conclusions Our findings indicated these DEmiRNAs having the potential to act as diagnostic biomarkers at an early stage of AD. Supplementary Information The online version contains supplementary material available at 10.1186/s41065-021-00190-0.
Collapse
Affiliation(s)
- Haoming Li
- Department of Human Anatomy, Institute of Neurobiology, Medical School of Nantong University, 19 Qixiu Road, Nantong, 226001, Jiangsu, China.,Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center, Neuroregeneration of Nantong University, Nantong, 226001, Jiangsu, China
| | - Linqing Zou
- Department of Human Anatomy, Institute of Neurobiology, Medical School of Nantong University, 19 Qixiu Road, Nantong, 226001, Jiangsu, China
| | - Jinhong Shi
- Department of Human Anatomy, Institute of Neurobiology, Medical School of Nantong University, 19 Qixiu Road, Nantong, 226001, Jiangsu, China.
| | - Xiao Han
- Department of Human Anatomy, Institute of Neurobiology, Medical School of Nantong University, 19 Qixiu Road, Nantong, 226001, Jiangsu, China. .,Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center, Neuroregeneration of Nantong University, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
17
|
Yu W, Yu W, Yang Y, Lü Y. Exploring the Key Genes and Identification of Potential Diagnosis Biomarkers in Alzheimer's Disease Using Bioinformatics Analysis. Front Aging Neurosci 2021; 13:602781. [PMID: 34194312 PMCID: PMC8236887 DOI: 10.3389/fnagi.2021.602781] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 05/06/2021] [Indexed: 12/30/2022] Open
Abstract
Background Alzheimer’s disease (AD) is one of the major threats of the twenty-first century and lacks available therapy. Identification of novel molecular markers for diagnosis and treatment of AD is urgently demanded, and genetic biomarkers show potential prospects. Method We identify and intersected differentially expressed genes (DEGs) from five microarray datasets to detect consensus DEGs. Based on these DEGs, we conducted Gene Ontology (GO), performed the Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis, constructed a protein—protein interaction (PPI) network, and utilized Cytoscape to identify hub genes. The least absolute shrinkage and selection operator (LASSO) logistic regression was applied to identify potential diagnostic biomarkers. Gene set enrichment analysis (GSEA) was performed to investigate the biological functions of the key genes. Result We identified 608 consensus DEGs, several dysregulated pathways, and 18 hub genes. Sixteen hub genes dysregulated as AD progressed. The diagnostic model of 35 genes was constructed, which has a high area under the curve (AUC) value in both the validation dataset and combined dataset (AUC = 0.992 and AUC = 0.985, respectively). The model can also differentiate mild cognitive impairment and AD patients from controls in two blood datasets. Brain-derived neurotrophic factor (BDNF) and WW domain-containing transcription regulator protein 1 (WWTR1), which are associated with the Braak stage, Aβ 42 levels, and β-secretase activity, were identified as critical genes of AD. Conclusion Our study identified 16 hub genes correlated to the neuropathological stage and 35 potential biomarkers for the diagnosis of AD. WWTR1 were identified as candidate genes for future studies. This study deepens our understanding of the transcriptomic and functional features and provides new potential diagnostic biomarkers and therapeutic targets for AD.
Collapse
Affiliation(s)
- Wuhan Yu
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Weihua Yu
- Institutes of Neuroscience, Chongqing Medical University, Chongqing, China
| | - Yan Yang
- State Key Laboratory of Power Transmission Equipment and System Security and New Technology, College of Electrical Engineering, Chongqing University, Chongqing, China
| | - Yang Lü
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
18
|
Burillo J, Marqués P, Jiménez B, González-Blanco C, Benito M, Guillén C. Insulin Resistance and Diabetes Mellitus in Alzheimer's Disease. Cells 2021; 10:1236. [PMID: 34069890 PMCID: PMC8157600 DOI: 10.3390/cells10051236] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/11/2021] [Accepted: 05/13/2021] [Indexed: 12/12/2022] Open
Abstract
Type 2 diabetes mellitus is a progressive disease that is characterized by the appearance of insulin resistance. The term insulin resistance is very wide and could affect different proteins involved in insulin signaling, as well as other mechanisms. In this review, we have analyzed the main molecular mechanisms that could be involved in the connection between type 2 diabetes and neurodegeneration, in general, and more specifically with the appearance of Alzheimer's disease. We have studied, in more detail, the different processes involved, such as inflammation, endoplasmic reticulum stress, autophagy, and mitochondrial dysfunction.
Collapse
Affiliation(s)
- Jesús Burillo
- Department of Biochemistry, Complutense University, 28040 Madrid, Spain; (J.B.); (P.M.); (B.J.); (C.G.-B.); (M.B.)
- Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28040 Madrid, Spain
- Mechanisms of Insulin Resistance (MOIR2), General Direction of Universities and Investigation (CCMM), 28040 Madrid, Spain
| | - Patricia Marqués
- Department of Biochemistry, Complutense University, 28040 Madrid, Spain; (J.B.); (P.M.); (B.J.); (C.G.-B.); (M.B.)
- Mechanisms of Insulin Resistance (MOIR2), General Direction of Universities and Investigation (CCMM), 28040 Madrid, Spain
| | - Beatriz Jiménez
- Department of Biochemistry, Complutense University, 28040 Madrid, Spain; (J.B.); (P.M.); (B.J.); (C.G.-B.); (M.B.)
- Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28040 Madrid, Spain
- Mechanisms of Insulin Resistance (MOIR2), General Direction of Universities and Investigation (CCMM), 28040 Madrid, Spain
| | - Carlos González-Blanco
- Department of Biochemistry, Complutense University, 28040 Madrid, Spain; (J.B.); (P.M.); (B.J.); (C.G.-B.); (M.B.)
- Mechanisms of Insulin Resistance (MOIR2), General Direction of Universities and Investigation (CCMM), 28040 Madrid, Spain
| | - Manuel Benito
- Department of Biochemistry, Complutense University, 28040 Madrid, Spain; (J.B.); (P.M.); (B.J.); (C.G.-B.); (M.B.)
- Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28040 Madrid, Spain
- Mechanisms of Insulin Resistance (MOIR2), General Direction of Universities and Investigation (CCMM), 28040 Madrid, Spain
| | - Carlos Guillén
- Department of Biochemistry, Complutense University, 28040 Madrid, Spain; (J.B.); (P.M.); (B.J.); (C.G.-B.); (M.B.)
- Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28040 Madrid, Spain
- Mechanisms of Insulin Resistance (MOIR2), General Direction of Universities and Investigation (CCMM), 28040 Madrid, Spain
| |
Collapse
|
19
|
Na H, Tian H, Zhang Z, Li Q, Yang JB, Mcparland L, Gan Q, Qiu WQ. Oral Amylin Treatment Reduces the Pathological Cascade of Alzheimer's Disease in a Mouse Model. Am J Alzheimers Dis Other Demen 2021; 36:15333175211012867. [PMID: 34137273 PMCID: PMC10623958 DOI: 10.1177/15333175211012867] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 03/24/2021] [Accepted: 04/04/2021] [Indexed: 11/16/2022]
Abstract
Intraperitoneal injection of amylin or its analog reduces Alzheimer's disease (AD) pathology in the brains. However, self-injecting amylin analogs is difficult for patients due to cognitive deficits. This work aims to study the effects of amylin on the brain could be achieved by oral delivery as some study reported that amylin receptor may be present in the gastrointestinal tract. A 6-week course of oral amylin treatment reduced components of AD pathology, including the levels of amyloid-β, phosphorylated tau, and ionized calcium binding adaptor molecule 1. The treatment reduced active forms of cyclin-dependent kinase 5. Oral amylin treatment led to improvements in social deficit in AD mouse. Using immunofluorescence, we observed the amylin receptor complexed with the calcitonin receptor and receptor activity-modifying proteins in the enteric neurons. The study suggests the potential of the oral delivery of amylin analogs for the treatment of AD and other neurodegenerative diseases through enteric neurons.
Collapse
Affiliation(s)
- Hana Na
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Hua Tian
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
- Department of Pharmacology, Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | - Zhengrong Zhang
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Qiang Li
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
- Nursing School, Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | - Jack B. Yang
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Liam Mcparland
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Qini Gan
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Wei Qiao Qiu
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
- Alzheimer’s Disease Center, Boston University School of Medicine, Boston, MA, USA
- Department of Psychiatry, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
20
|
Servizi S, Corrigan RR, Casadesus G. The Importance of Understanding Amylin Signaling Mechanisms for Therapeutic Development in the Treatment of Alzheimer's Disease. Curr Pharm Des 2020; 26:1345-1355. [PMID: 32188374 PMCID: PMC10088426 DOI: 10.2174/1381612826666200318151146] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 03/04/2020] [Indexed: 12/12/2022]
Abstract
Type II Diabetes (T2D) is a major risk factor for Alzheimer's Disease (AD). These two diseases share several pathological features, including amyloid accumulation, inflammation, oxidative stress, cell death and cognitive decline. The metabolic hormone amylin and amyloid-beta are both amyloids known to self-aggregate in T2D and AD, respectively, and are thought to be the main pathogenic entities in their respective diseases. Furthermore, studies suggest amylin's ability to seed amyloid-beta aggregation, the activation of common signaling cascades in the pancreas and the brain, and the ability of amyloid beta to signal through amylin receptors (AMYR), at least in vitro. However, paradoxically, non-aggregating forms of amylin such as pramlintide are given to treat T2D and functional and neuroprotective benefits of amylin and pramlintide administration have been reported in AD transgenic mice. These paradoxical results beget a deeper study of the complex nature of amylin's signaling through the several AMYR subtypes and other receptors associated with amylin effects to be able to fully understand its potential role in mediating AD development and/or prevention. The goal of this review is to provide such critical insight to begin to elucidate how the complex nature of this hormone's signaling may explain its equally complex relationship with T2D and mechanisms of AD pathogenesis.
Collapse
Affiliation(s)
- Spencer Servizi
- School of Biomedical Sciences, Kent State University, Ohio, United States
| | - Rachel R Corrigan
- School of Biomedical Sciences, Kent State University, Ohio, United States
| | - Gemma Casadesus
- School of Biomedical Sciences, Kent State University, Ohio, United States.,Department of Biological Sciences, Kent State University, Ohio, United States
| |
Collapse
|
21
|
Nassar SZ, Badae NM, Issa YA. Effect of amylin on memory and central insulin resistance in a rat model of Alzheimer's disease. Arch Physiol Biochem 2020; 126:326-334. [PMID: 30449203 DOI: 10.1080/13813455.2018.1534244] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Context: Alzheimer's disease is strongly associated with brain insulin signalling.Objective: Investigating the effect of amylin as a novel treatment in streptozotocin (STZ) rat model of AD.Materials and methods: Alzheimer's disease (AD) was induced in albino rats by intracerebroventricular injection of STZ (3 mg/kg). Rats received either amylin analogue (Pramlintide 200 μg/kg/day) or Metformin (30 mg/kg/day) for 5 weeks.Results: Both Pramlintide and Metformin improve learning and memory through enhancing insulin signalling (p-IR and p-PI3K) which lead to lowering level of CSF glucose, phosphorylated tau proteins, and amyloid-β peptide (Aβ) in hippocampus.Conclusions: Insulin sensitisers as Metformin and Pramlintide can improve learning and memory and decrease the pathological changes in STZ induced rat model of AD. However, Pramlintide is superior to Metformin in some memory tests which related to its action as an amylin analogue. Amylin improves learning and memory through an independent effect other than insulin sensitisation.
Collapse
Affiliation(s)
- Seham Zakaria Nassar
- Department of Medical Physiology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Noha Mohamed Badae
- Department of Medical Physiology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Yasmine Amr Issa
- Department of Medical Biochemistry, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| |
Collapse
|
22
|
Patrick S, Corrigan R, Grizzanti J, Mey M, Blair J, Pallas M, Camins A, Lee HG, Casadesus G. Neuroprotective Effects of the Amylin Analog, Pramlintide, on Alzheimer's Disease Are Associated with Oxidative Stress Regulation Mechanisms. J Alzheimers Dis 2020; 69:157-168. [PMID: 30958347 DOI: 10.3233/jad-180421] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Administration of the recombinant analog of the pancreatic amyloid amylin, Pramlintide, has shown therapeutic benefits in aging and Alzheimer's disease (AD) models, both on cognition and amyloid-β (Aβ) pathology. However, the neuroprotective mechanisms underlying the benefits of Pramlintide remain unclear. Given the early and critical role of oxidative stress in AD pathogenesis and the known reactive oxygen species (ROS) modulating function of amyloids, we sought to determine whether Pramlintide's neuroprotective effects involve regulation of oxidative stress mechanisms. To address this, we treated APP/PS1 transgenic mice with Pramlintide for 3 months, starting at 5.5 months prior to widespread AD pathology onset, and measured cognition (Morris Water Maze), AD pathology, and oxidative stress-related markers and enzymes in vivo. In vitro, we determined the ability of Pramlintide to modulate H2O2-induced oxidative stress levels. Our data show that Pramlintide improved cognitive function, altered amyloid-processing enzymes, reduced plaque burden in the hippocampus, and regulated endogenous antioxidant enzymes (MnSOD and GPx1) and the stress marker HO-1 in a location specific manner. In vitro, Pramlintide treatment in neuronal models reduced H2O2-induced endogenous ROS production and lipid peroxidation in a dose-dependent manner. Together, these results indicate that Pramlintide's benefits on cognitive function and pathology may involve antioxidant-like properties of this compound.
Collapse
Affiliation(s)
- Sarah Patrick
- School of Biomedical Sciences, Kent State University, Kent, OH, USA
| | - Rachel Corrigan
- School of Biomedical Sciences, Kent State University, Kent, OH, USA
| | - John Grizzanti
- School of Biomedical Sciences, Kent State University, Kent, OH, USA
| | - Megan Mey
- School of Biomedical Sciences, Kent State University, Kent, OH, USA
| | - Jeff Blair
- School of Biomedical Sciences, Kent State University, Kent, OH, USA
| | - Merce Pallas
- Facultat de Farmacia, Universitat de Barcelona, Barcelona, Spain
| | - Antonio Camins
- Facultat de Farmacia, Universitat de Barcelona, Barcelona, Spain
| | - Hyoung-Gon Lee
- Department of Biology, The University of Texas San Antonio, San Antonio, TX, USA
| | - Gemma Casadesus
- School of Biomedical Sciences, Kent State University, Kent, OH, USA.,Department of Biological Sciences, Kent State University, Kent, OH, USA
| |
Collapse
|
23
|
Cunnane SC, Trushina E, Morland C, Prigione A, Casadesus G, Andrews ZB, Beal MF, Bergersen LH, Brinton RD, de la Monte S, Eckert A, Harvey J, Jeggo R, Jhamandas JH, Kann O, la Cour CM, Martin WF, Mithieux G, Moreira PI, Murphy MP, Nave KA, Nuriel T, Oliet SHR, Saudou F, Mattson MP, Swerdlow RH, Millan MJ. Brain energy rescue: an emerging therapeutic concept for neurodegenerative disorders of ageing. Nat Rev Drug Discov 2020; 19:609-633. [PMID: 32709961 PMCID: PMC7948516 DOI: 10.1038/s41573-020-0072-x] [Citation(s) in RCA: 523] [Impact Index Per Article: 104.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/03/2020] [Indexed: 12/11/2022]
Abstract
The brain requires a continuous supply of energy in the form of ATP, most of which is produced from glucose by oxidative phosphorylation in mitochondria, complemented by aerobic glycolysis in the cytoplasm. When glucose levels are limited, ketone bodies generated in the liver and lactate derived from exercising skeletal muscle can also become important energy substrates for the brain. In neurodegenerative disorders of ageing, brain glucose metabolism deteriorates in a progressive, region-specific and disease-specific manner - a problem that is best characterized in Alzheimer disease, where it begins presymptomatically. This Review discusses the status and prospects of therapeutic strategies for countering neurodegenerative disorders of ageing by improving, preserving or rescuing brain energetics. The approaches described include restoring oxidative phosphorylation and glycolysis, increasing insulin sensitivity, correcting mitochondrial dysfunction, ketone-based interventions, acting via hormones that modulate cerebral energetics, RNA therapeutics and complementary multimodal lifestyle changes.
Collapse
Affiliation(s)
- Stephen C Cunnane
- Department of Medicine, Université de Sherbrooke, Sherbrooke, QC, Canada.
- Research Center on Aging, Sherbrooke, QC, Canada.
| | | | - Cecilie Morland
- Department of Pharmaceutical Biosciences, Institute of Pharmacy, University of Oslo, Oslo, Norway
| | - Alessandro Prigione
- Department of General Pediatrics, Neonatology, and Pediatric Cardiology, University of Dusseldorf, Dusseldorf, Germany
| | - Gemma Casadesus
- Department of Biological Sciences, Kent State University, Kent, OH, USA
| | - Zane B Andrews
- Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
- Department of Physiology, Monash University, Clayton, VIC, Australia
| | - M Flint Beal
- Department of Neurology, Weill Cornell Medicine, New York, NY, USA
| | - Linda H Bergersen
- Department of Anatomy, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | | | | | | | - Jenni Harvey
- Ninewells Hospital, University of Dundee, Dundee, UK
- Medical School, University of Dundee, Dundee, UK
| | - Ross Jeggo
- Centre for Therapeutic Innovation in Neuropsychiatry, Institut de Recherche Servier, Croissy sur Seine, France
| | - Jack H Jhamandas
- Department of Medicine, University of Albeta, Edmonton, AB, Canada
- Neuroscience and Mental Health Institute, University of Albeta, Edmonton, AB, Canada
| | - Oliver Kann
- Institute of Physiology and Pathophysiology, University of Heidelberg, Heidelberg, Germany
| | - Clothide Mannoury la Cour
- Centre for Therapeutic Innovation in Neuropsychiatry, Institut de Recherche Servier, Croissy sur Seine, France
| | - William F Martin
- Institute of Molecular Evolution, University of Dusseldorf, Dusseldorf, Germany
| | | | - Paula I Moreira
- CNC Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Michael P Murphy
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| | - Klaus-Armin Nave
- Department of Biosciences, University of Heidelberg, Heidelberg, Germany
| | - Tal Nuriel
- Columbia University Medical Center, New York, NY, USA
| | - Stéphane H R Oliet
- Neurocentre Magendie, INSERM U1215, Bordeaux, France
- Université de Bordeaux, Bordeaux, France
| | - Frédéric Saudou
- University of Grenoble Alpes, Grenoble, France
- INSERM U1216, CHU Grenoble Alpes, Grenoble Institute Neurosciences, Grenoble, France
| | - Mark P Mattson
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Mark J Millan
- Centre for Therapeutic Innovation in Neuropsychiatry, Institut de Recherche Servier, Croissy sur Seine, France.
| |
Collapse
|
24
|
Watanabe-Nakayama T, Sahoo BR, Ramamoorthy A, Ono K. High-Speed Atomic Force Microscopy Reveals the Structural Dynamics of the Amyloid-β and Amylin Aggregation Pathways. Int J Mol Sci 2020; 21:E4287. [PMID: 32560229 PMCID: PMC7352471 DOI: 10.3390/ijms21124287] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 06/09/2020] [Accepted: 06/14/2020] [Indexed: 12/18/2022] Open
Abstract
Individual Alzheimer's disease (AD) patients have been shown to have structurally distinct amyloid-β (Aβ) aggregates, including fibrils, in their brain. These findings suggest the possibility of a relationship between AD progression and Aβ fibril structures. Thus, the characterization of the structural dynamics of Aβ could aid the development of novel therapeutic strategies and diagnosis. Protein structure and dynamics have typically been studied separately. Most of the commonly used biophysical approaches are limited in providing substantial details regarding the combination of both structure and dynamics. On the other hand, high-speed atomic force microscopy (HS-AFM), which simultaneously visualizes an individual protein structure and its dynamics in liquid in real time, can uniquely link the structure and the kinetic details, and it can also unveil novel insights. Although amyloidogenic proteins generate heterogeneously aggregated species, including transient unstable states during the aggregation process, HS-AFM elucidated the structural dynamics of individual aggregates in real time in liquid without purification and isolation. Here, we review and discuss the HS-AFM imaging of amyloid aggregation and strategies to optimize the experiments showing findings from Aβ and amylin, which is associated with type II diabetes, shares some common biological features with Aβ, and is reported to be involved in AD.
Collapse
Affiliation(s)
| | - Bikash R. Sahoo
- Biophysics Program, Department of Chemistry, Macromolecular Science and Engineering, and Biomedical Engineering, The University of Michigan, Ann Arbor, MI 48109-1055, USA;
| | - Ayyalusamy Ramamoorthy
- Biophysics and Department of Chemistry, University of Michigan, Ann Arbor, MI 48109-1055, USA;
| | - Kenjiro Ono
- Division of Neurology, Department of Internal Medicine, School of Medicine, Showa University, Hatanodai, Shinagawa district, Tokyo 142-8666, Japan;
| |
Collapse
|
25
|
Madhusudhanan J, Suresh G, Devanathan V. Neurodegeneration in type 2 diabetes: Alzheimer's as a case study. Brain Behav 2020; 10:e01577. [PMID: 32170854 PMCID: PMC7218246 DOI: 10.1002/brb3.1577] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 01/04/2020] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION Rigorous research in the last few years has shown that in addition to the classical mechanism of neurodegeneration, certain unconventional mechanisms may also lead to neurodegenerative disease. One of them is a widely studied metabolic disorder: type 2 diabetes mellitus (T2DM). We now have a clear understanding of glucose-mediated neurodegeneration, mostly from studies in Alzheimer's disease (AD) models. AD is recognized to be significantly associated with hyperglycemia, even earning the term "type 3 diabetes." Here, we review first the pathophysiology of AD, both from the perspective of classical protein accumulation, as well as the newer T2DM-dependent mechanisms supported by findings from patients with T2DM. Secondly, we review the different pathways through which neurodegeneration is aggravated in hyperglycemic conditions taking AD as a case study. Finally, some of the current advances in AD management as a result of recent research developments in metabolic disorders-driven neurodegeneration are also discussed. METHODS Relevant literatures found from PubMed search were reviewed. RESULTS Apart from the known causes of AD, type 2 diabetes opens a new window to the AD pathology in several ways. It is a bidirectional interaction, of which, the molecular and signaling mechanisms are recently studied. This is our attempt to connect all of them to draw a complete mechanistic explanation for the neurodegeneration in T2DM. Refer to Figure 3. CONCLUSION The perspective of AD as a classical neurodegenerative disease is changing, and it is now being looked at from a zoomed-out perspective. The correlation between T2DM and AD is something observed and studied extensively. It is promising to know that there are certain advances in AD management following these studies.
Collapse
Affiliation(s)
- Jalaja Madhusudhanan
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati, India
| | - Gowthaman Suresh
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati, India
| | - Vasudharani Devanathan
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati, India
| |
Collapse
|
26
|
Benlier N, Ozer G, Orhan N. Relation between serum amylin level and epilepsy. THE EGYPTIAN JOURNAL OF NEUROLOGY, PSYCHIATRY AND NEUROSURGERY 2020. [DOI: 10.1186/s41983-020-00164-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Abstract
Background
Epilepsy is a neurological disorder characterized by convulsions. Identification of biological pathways underlying epilepsy and novel genes may shed light on the pathogenesis of epilepsy as well as new targets for treatment.
Objectives
Amylin is cosecreted with insulin from the pancreatic β-cells in a pulsatile manner as a response to nutrient stimuli. In vitro studies have shown the neurotoxicity potential of amylin. We aimed to investigate serum amylin levels between epilepsy patients and a healthy control group.
Subjects and methods
For this study, 45 patients with epilepsy and 60 healthy controls were enrolled. Routine blood analysis and electroencephalography scan were performed for all participants. Five cc venous blood sample was collected from each participant. Sera were isolated and stored at − 80 °C until the time of amylin analysis with the enzyme-linked immunosorbent assay.
Results
Gender distribution of the two groups was as follows: 44.4% males and 55.6% females among epileptic patients and 53.3% males and 46.7% females for control subjects.
Body mass index was 23.09 ± 3.99 kg/m2 for epileptic patients and 26.29 ± 4.83 kg/m2 for controls, with a statistically significantly higher body mass index in control subjects (p ˂ 0.001). With regard to serum amylin levels, a statistically significant difference was observed between the two groups (p ˂ 0.001). The median serum amylin concentration was 226.62 ng/ml (69.49–6961.19 (min–max)) for epileptic patients and 103.66 ng/ml (37.42–607.11 (min–max)) for controls (p ˂ 0.001).
Conclusion
In the present study, a significant difference was observed between patient and control groups in serum amylin concentrations, which were considerably higher in epileptic patients.
Collapse
|
27
|
Mousa YM, Abdallah IM, Hwang M, Martin DR, Kaddoumi A. Amylin and pramlintide modulate γ-secretase level and APP processing in lipid rafts. Sci Rep 2020; 10:3751. [PMID: 32111883 PMCID: PMC7048857 DOI: 10.1038/s41598-020-60664-5] [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: 07/02/2019] [Accepted: 02/04/2020] [Indexed: 12/13/2022] Open
Abstract
A major characteristic of Alzheimer's disease (AD) is the accumulation of misfolded amyloid-β (Aβ) peptide. Several studies linked AD with type 2 diabetes due to similarities between Aβ and human amylin. This study investigates the effect of amylin and pramlintide on Aβ pathogenesis and the predisposing molecular mechanism(s) behind the observed effects in TgSwDI mouse, a cerebral amyloid angiopathy (CAA) and AD model. Our findings showed that thirty days of intraperitoneal injection with amylin or pramlintide increased Aβ burden in mice brains. Mechanistic studies revealed both peptides altered the amyloidogenic pathway and increased Aβ production by modulating amyloid precursor protein (APP) and γ-secretase levels in lipid rafts. In addition, both peptides increased levels of B4GALNT1 enzyme and GM1 ganglioside, and only pramlintide increased the level of GM2 ganglioside. Increased levels of GM1 and GM2 gangliosides play an important role in regulating amyloidogenic pathway proteins in lipid rafts. Increased brain Aβ burden by amylin and pramlintide was associated with synaptic loss, apoptosis, and microglia activation. In conclusion, our findings showed amylin or pramlintide increase Aβ levels and related pathology in TgSwDI mice brains, and suggest that increased amylin levels or the therapeutic use of pramlintide could increase the risk of AD.
Collapse
Affiliation(s)
- Youssef M Mousa
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, USA
| | - Ihab M Abdallah
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, USA
| | - Misako Hwang
- Scott-Ritchey Research Center, Auburn University, Auburn, USA
| | - Douglas R Martin
- Scott-Ritchey Research Center, Auburn University, Auburn, USA.,Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, USA.,Center for Neuroscience Initiative, Auburn University, Auburn, AL, USA
| | - Amal Kaddoumi
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, USA. .,Center for Neuroscience Initiative, Auburn University, Auburn, AL, USA.
| |
Collapse
|
28
|
Grizzanti J, Corrigan R, Casadesus G. Neuroprotective Effects of Amylin Analogues on Alzheimer's Disease Pathogenesis and Cognition. J Alzheimers Dis 2019; 66:11-23. [PMID: 30282360 DOI: 10.3233/jad-180433] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Type II diabetes (T2D) has been identified as a major risk factor for the development of Alzheimer's disease (AD). Interestingly, both AD and T2D have similar characteristics including amyloid peptide aggregation, decreased metabolism, and increased oxidative stress and inflammation. Despite their prevalence, therapies for these diseases are limited. To date, most therapies for AD have targeted amyloid-β or tau. Unfortunately, most of these clinical trials have been largely unsuccessful, creating a crucial need for novel therapies. A number of studies have shown that metabolic hormone therapies are effective at ameliorating high blood glucose levels in diabetics as well as improving cognitive function in AD and mild cognitive impairment patients. Pramlintide, a synthetic analogue of the pancreatic hormone amylin, has been developed and used for years now as a treatment for both type I diabetes and T2D due to the loss of β-islet cells responsible for producing amylin. Importantly, recent data demonstrates its potential therapeutic role for AD as well. This review aims at addressing parallels between T2D and AD at a pathological and functional level, focusing on amylin signaling as a key, overlapping mediator in both diseases. The potential therapeutic use of this hormone to treat AD will also be explored from a mechanistic viewpoint.
Collapse
Affiliation(s)
- John Grizzanti
- School of Biomedical Sciences, Kent State University, Kent, OH, USA
| | - Rachel Corrigan
- School of Biomedical Sciences, Kent State University, Kent, OH, USA
| | - Gemma Casadesus
- School of Biomedical Sciences, Kent State University, Kent, OH, USA.,Department of Biological Sciences, Kent State University, Kent, OH, USA
| |
Collapse
|
29
|
Ling W, Huang YM, Qiao YC, Zhang XX, Zhao HL. Human Amylin: From Pathology to Physiology and Pharmacology. Curr Protein Pept Sci 2019; 20:944-957. [DOI: 10.2174/1389203720666190328111833] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 03/12/2019] [Accepted: 03/15/2019] [Indexed: 12/18/2022]
Abstract
The histopathological hallmark of type 2 diabetes is islet amyloid implicated in the developing treatment options. The major component of human islet amyloid is 37 amino acid peptide known as amylin or islet amyloid polypeptide (IAPP). Amylin is an important hormone that is co-localized, copackaged, and co-secreted with insulin from islet β cells. Physiologically, amylin regulates glucose homeostasis by inhibiting insulin and glucagon secretion. Furthermore, amylin modulates satiety and inhibits gastric emptying via the central nervous system. Normally, human IAPP is soluble and natively unfolded in its monomeric state. Pathologically, human IAPP has a propensity to form oligomers and aggregate. The oligomers show misfolded α-helix conformation and can further convert themselves to β-sheet-rich fibrils as amyloid deposits. The pathological findings and physiological functions of amylin have led to the introduction of pramlintide, an amylin analog, for the treatment of diabetes. The history of amylin’s discovery is a representative example of how a pathological finding can translate into physiological exploration and lead to pharmacological intervention. Understanding the importance of transitioning from pathology to physiology and pharmacology can provide novel insight into diabetes mellitus and Alzheimer's disease.
Collapse
Affiliation(s)
- Wei Ling
- Center for Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin 541004, China
| | - Yan-Mei Huang
- Center for Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin 541004, China
| | - Yong-Chao Qiao
- Department of Laboratory, the Affiliated Hospital of Guilin Medical University, Guilin 541004, China
| | - Xiao-Xi Zhang
- Center for Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin 541004, China
| | - Hai-Lu Zhao
- Center for Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin 541004, China
| |
Collapse
|
30
|
Short amylin receptor antagonist peptides improve memory deficits in Alzheimer's disease mouse model. Sci Rep 2019; 9:10942. [PMID: 31358858 PMCID: PMC6662706 DOI: 10.1038/s41598-019-47255-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 07/12/2019] [Indexed: 12/12/2022] Open
Abstract
Recent evidence supports involvement of amylin and the amylin receptor in the pathogenesis of Alzheimer’s disease (AD). We have previously shown that amylin receptor antagonist, AC253, improves spatial memory in AD mouse models. Herein, we generated and screened a peptide library and identified two short sequence amylin peptides (12–14 aa) that are proteolytically stable, brain penetrant when administered intraperitoneally, neuroprotective against Aβ toxicity and restore diminished levels of hippocampal long term potentiation in AD mice. Systemic administration of the peptides for five weeks in aged 5XFAD mice improved spatial memory, reduced amyloid plaque burden, and neuroinflammation. The common residue SQELHRLQTY within the peptides is an essential sequence for preservation of the beneficial effects of the fragments that we report here and constitutes a new pharmacological target. These findings suggest that the amylin receptor antagonism may represent a novel therapy for AD.
Collapse
|
31
|
Grizzanti J, Corrigan R, Servizi S, Casadesus G. Amylin Signaling in Diabetes and Alzheimer's Disease: Therapy or Pathology? JOURNAL OF NEUROLOGY & NEUROMEDICINE 2019; 4:12-16. [PMID: 31511851 PMCID: PMC6738967 DOI: 10.29245/2572.942x/2019/1.1212] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Growing evidence highlights the intimate relationship between type II diabetes (T2D) and Alzheimer's disease (AD). Importantly, these two diseases share a number of pathological similarities, including amyloid accumulation, oxidative stress, inflammation, and cell death. To date, drug therapies for AD and T2D are lacking and there is a crucial need for the discovery and development of novel therapeutics for these diseases. A number of human and rodent studies have given evidence that metabolic hormone supplementation is highly valuable for improving cognitive function and overall metabolic health in both T2D and AD. The pancreatic hormone amylin has arisen as a crucial component of the disease etiology of both T2D and AD, though the exact role that amylin plays in these diseases is not yet well understood. Here, we critically review the current literature that utilizes human amylin or its synthetic analogue, pramlintide, as well as amylin receptor antagonists for the treatment of AD.
Collapse
Affiliation(s)
- John Grizzanti
- School of Biomedical Sciences, Kent State University, Ohio, USA
| | - Rachel Corrigan
- School of Biomedical Sciences, Kent State University, Ohio, USA
| | - Spencer Servizi
- School of Biomedical Sciences, Kent State University, Ohio, USA
| | - Gemma Casadesus
- School of Biomedical Sciences, Kent State University, Ohio, USA
- Department of Biological Sciences, Kent State University, Ohio, USA
| |
Collapse
|
32
|
Maletínská L, Popelová A, Železná B, Bencze M, Kuneš J. The impact of anorexigenic peptides in experimental models of Alzheimer's disease pathology. J Endocrinol 2019; 240:R47-R72. [PMID: 30475219 DOI: 10.1530/joe-18-0532] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 11/20/2018] [Indexed: 12/16/2022]
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disorder in the elderly population. Numerous epidemiological and experimental studies have demonstrated that patients who suffer from obesity or type 2 diabetes mellitus have a higher risk of cognitive dysfunction and AD. Several recent studies demonstrated that food intake-lowering (anorexigenic) peptides have the potential to improve metabolic disorders and that they may also potentially be useful in the treatment of neurodegenerative diseases. In this review, the neuroprotective effects of anorexigenic peptides of both peripheral and central origins are discussed. Moreover, the role of leptin as a key modulator of energy homeostasis is discussed in relation to its interaction with anorexigenic peptides and their analogs in AD-like pathology. Although there is no perfect experimental model of human AD pathology, animal studies have already proven that anorexigenic peptides exhibit neuroprotective properties. This phenomenon is extremely important for the potential development of new drugs in view of the aging of the human population and of the significantly increasing incidence of AD.
Collapse
Affiliation(s)
- Lenka Maletínská
- Institute of Organic Chemistry and Biochemistry AS CR, Prague, Czech Republic
| | - Andrea Popelová
- Institute of Organic Chemistry and Biochemistry AS CR, Prague, Czech Republic
| | - Blanka Železná
- Institute of Organic Chemistry and Biochemistry AS CR, Prague, Czech Republic
| | - Michal Bencze
- Institute of Organic Chemistry and Biochemistry AS CR, Prague, Czech Republic
- Institute of Physiology AS CR, Prague, Czech Republic
| | - Jaroslav Kuneš
- Institute of Organic Chemistry and Biochemistry AS CR, Prague, Czech Republic
- Institute of Physiology AS CR, Prague, Czech Republic
| |
Collapse
|
33
|
Mietlicki-Baase EG. Amylin in Alzheimer's disease: Pathological peptide or potential treatment? Neuropharmacology 2018; 136:287-297. [PMID: 29233636 PMCID: PMC5994175 DOI: 10.1016/j.neuropharm.2017.12.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 12/06/2017] [Accepted: 12/07/2017] [Indexed: 12/19/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease for which we currently lack effective treatments or a cure. The pancreatic peptide hormone amylin has recently garnered interest as a potential pharmacological target for the treatment of AD. A number of studies have demonstrated that amylin and amylin analogs like the FDA-approved diabetes drug pramlintide can reduce amyloid burden in the brain and improve cognitive symptoms of AD. However, other data suggest that amylin may have pathological effects in AD due to its propensity to misfold and aggregate under certain conditions. Here, the literature supporting a beneficial versus harmful role of amylin in AD is reviewed. Additionally, several critical gaps in the literature are discussed, such as our limited understanding of the amylin system during aging and in disease states, as well as complexities of amylin receptor signaling and of changing pathophysiology during AD progression that might underlie the seemingly conflicting or contradictory results in the amylin/AD literature. This article is part of the Special Issue entitled 'Metabolic Impairment as Risk Factors for Neurodegenerative Disorders.'
Collapse
Affiliation(s)
- Elizabeth G Mietlicki-Baase
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, State University of New York at Buffalo, Buffalo, NY 14214, USA.
| |
Collapse
|
34
|
Lau A, Bourkas M, Lu YQQ, Ostrowski LA, Weber-Adrian D, Figueiredo C, Arshad H, Shoaei SZS, Morrone CD, Matan-Lithwick S, Abraham KJ, Wang H, Schmitt-Ulms G. Functional Amyloids and their Possible Influence on Alzheimer Disease. Discoveries (Craiova) 2017; 5:e79. [PMID: 32309597 PMCID: PMC7159844 DOI: 10.15190/d.2017.9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 09/29/2017] [Accepted: 10/02/2017] [Indexed: 12/21/2022] Open
Abstract
Amyloids play critical roles in human diseases but have increasingly been recognized to also exist naturally. Shared physicochemical characteristics of amyloids and of their smaller oligomeric building blocks offer the prospect of molecular interactions and crosstalk amongst these assemblies, including the propensity to mutually influence aggregation. A case in point might be the recent discovery of an interaction between the amyloid β peptide (Aβ) and somatostatin (SST). Whereas Aβ is best known for its role in Alzheimer disease (AD) as the main constituent of amyloid plaques, SST is intermittently stored in amyloid-form in dense core granules before its regulated release into the synaptic cleft. This review was written to introduce to readers a large body of literature that surrounds these two peptides. After introducing general concepts and recent progress related to our understanding of amyloids and their aggregation, the review focuses separately on the biogenesis and interactions of Aβ and SST, before attempting to assess the likelihood of encounters of the two peptides in the brain, and summarizing key observations linking SST to the pathobiology of AD. While the review focuses on Aβ and SST, it is to be anticipated that crosstalk amongst functional and disease-associated amyloids will emerge as a general theme with much broader significance in the etiology of dementias and other amyloidosis.
Collapse
Affiliation(s)
- Angus Lau
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Medical Sciences Building, 6th Floor, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Centre, 6th Floor, 60 Leonard Avenue, Toronto, Ontario M5T 2S8, Canada
| | - Matthew Bourkas
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Medical Sciences Building, 6th Floor, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Centre, 6th Floor, 60 Leonard Avenue, Toronto, Ontario M5T 2S8, Canada
| | - Yang Qing Qin Lu
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Medical Sciences Building, 6th Floor, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| | - Lauren Anne Ostrowski
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Medical Sciences Building, 6th Floor, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| | - Danielle Weber-Adrian
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Medical Sciences Building, 6th Floor, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| | - Carlyn Figueiredo
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Medical Sciences Building, 6th Floor, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| | - Hamza Arshad
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Medical Sciences Building, 6th Floor, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Centre, 6th Floor, 60 Leonard Avenue, Toronto, Ontario M5T 2S8, Canada
| | - Seyedeh Zahra Shams Shoaei
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Medical Sciences Building, 6th Floor, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| | - Christopher Daniel Morrone
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Medical Sciences Building, 6th Floor, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| | - Stuart Matan-Lithwick
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Medical Sciences Building, 6th Floor, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| | - Karan Joshua Abraham
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Medical Sciences Building, 6th Floor, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| | - Hansen Wang
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Centre, 6th Floor, 60 Leonard Avenue, Toronto, Ontario M5T 2S8, Canada
| | - Gerold Schmitt-Ulms
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Medical Sciences Building, 6th Floor, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Centre, 6th Floor, 60 Leonard Avenue, Toronto, Ontario M5T 2S8, Canada
| |
Collapse
|
35
|
Fu W, Vukojevic V, Patel A, Soudy R, MacTavish D, Westaway D, Kaur K, Goncharuk V, Jhamandas J. Role of microglial amylin receptors in mediating beta amyloid (Aβ)-induced inflammation. J Neuroinflammation 2017; 14:199. [PMID: 28985759 PMCID: PMC5639602 DOI: 10.1186/s12974-017-0972-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 09/27/2017] [Indexed: 02/06/2023] Open
Abstract
Background Neuroinflammation in the brain consequent to activation of microglia is viewed as an important component of Alzheimer’s disease (AD) pathology. Amyloid beta (Aβ) protein is known to activate microglia and unleash an inflammatory cascade that eventually results in neuronal dysfunction and death. In this study, we sought to identify the presence of amylin receptors on human fetal and murine microglia and determine whether Aβ activation of the inflammasome complex and subsequent release of cytokines is mediated through these receptors. Methods The presence of dimeric components of the amylin receptor (calcitonin receptor and receptor activity modifying protein 3) were first immunohistochemically identified on microglia. Purified human fetal microglial (HFM) cultures were incubated with an in vivo microglial marker, DyLight 594-conjugated tomato lectin, and loaded with the membrane-permeant green fluorescent dye, Fluo-8L-AM for measurements of intracellular calcium [Ca2+]i. HFM and BV-2 cells were primed with lipopolysaccharide and then exposed to either human amylin or soluble oligomeric Aβ1–42 prior to treatment with and without the amylin receptor antagonist, AC253. Changes in the inflammasome complex, NLRP3 and caspase-1, were examined in treated cell cultures with Western blot and fluorometric assays. RT-PCR measurements were performed to assess cytokine release. Finally, in vivo studies were performed in transgenic mouse model of AD (5xFAD) to examine the effects of systemic administration of AC253 on markers of neuroinflammation in the brain. Results Acute applications of human amylin or Aβ1–42 resulted in an increase in [Ca2+]i that could be blocked by the amylin receptor antagonist, AC253. Activation of the NLRP3 and caspase-1 and subsequent release of cytokines, TNFα and IL-1β, was diminished by AC253 pretreatment of HFMs and BV2 cells. In vivo, intraperitoneal administration of AC253 resulted in a reduction in microglial markers (Iba-1 and CD68), caspase-1, TNFα, and IL-1β. These reductions in inflammatory markers were accompanied by reduction in amyloid plaque and size in the brains of 5xFAD mice compared to controls. Conclusion Microglial amylin receptors mediate Aβ-evoked inflammation, and amylin receptor antagonists therefore offer an attractive therapeutic target for intervention in AD. Electronic supplementary material The online version of this article (10.1186/s12974-017-0972-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wen Fu
- Department of Medicine (Neurology), Neuroscience and Mental Health Institute, University of Alberta, 530 Heritage Medical Research Centre, Edmonton, AB, T6G 2S2, Canada
| | - Vlatka Vukojevic
- Department of Medicine (Neurology), Neuroscience and Mental Health Institute, University of Alberta, 530 Heritage Medical Research Centre, Edmonton, AB, T6G 2S2, Canada
| | - Aarti Patel
- Department of Medicine (Neurology), Neuroscience and Mental Health Institute, University of Alberta, 530 Heritage Medical Research Centre, Edmonton, AB, T6G 2S2, Canada
| | - Rania Soudy
- Department of Medicine (Neurology), Neuroscience and Mental Health Institute, University of Alberta, 530 Heritage Medical Research Centre, Edmonton, AB, T6G 2S2, Canada.,Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - David MacTavish
- Department of Medicine (Neurology), Neuroscience and Mental Health Institute, University of Alberta, 530 Heritage Medical Research Centre, Edmonton, AB, T6G 2S2, Canada
| | - David Westaway
- Department of Medicine (Neurology), Neuroscience and Mental Health Institute, University of Alberta, 530 Heritage Medical Research Centre, Edmonton, AB, T6G 2S2, Canada.,Department of Biochemistry, University of Alberta, Edmonton, AB, Canada.,Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB, Canada
| | - Kamaljit Kaur
- Chapman University School of Pharmacy, Irvine, CA, USA
| | | | - Jack Jhamandas
- Department of Medicine (Neurology), Neuroscience and Mental Health Institute, University of Alberta, 530 Heritage Medical Research Centre, Edmonton, AB, T6G 2S2, Canada.
| |
Collapse
|
36
|
Amylin Receptor: A Potential Therapeutic Target for Alzheimer's Disease. Trends Mol Med 2017; 23:709-720. [PMID: 28694141 DOI: 10.1016/j.molmed.2017.06.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 06/08/2017] [Accepted: 06/14/2017] [Indexed: 01/29/2023]
Abstract
Alzheimer'sdisease (AD) is a progressive neurodegenerative disorder, characterized by senile plaques constituting extracellular deposits of β-amyloid (Aβ) fibrils. Since Aβ accumulation in the brain is considered an early event preceding, by decades, cognitive dysfunction, disease-modifying treatments are aimed at facilitating clearance of this protein from the brain or ameliorating its toxic effects. Recent studies have identified the amylin receptor as a capable mediator of the deleterious actions of Aβ and furthermore, administration of amylin receptor-based peptides has been shown to improve spatial memory and learning in transgenic mouse models of AD. Here, by discussing available evidence, we posit that the amylin receptor could be considered a potential therapeutic target for AD, and present the rationale for using amylin receptor antagonists to treat this debilitating condition.
Collapse
|
37
|
Amylin and its G-protein-coupled receptor: A probable pathological process and drug target for Alzheimer's disease. Neuroscience 2017; 356:44-51. [PMID: 28528968 DOI: 10.1016/j.neuroscience.2017.05.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 05/11/2017] [Accepted: 05/12/2017] [Indexed: 12/31/2022]
Abstract
G-protein-coupled receptors (GPCRs) are shown to be involved in Alzheimer's disease (AD) pathogenesis. However, because GPCRs include a large family of membrane receptors, it is unclear which specific GPCR or pathway with rational ligands can become effective therapeutic targets for AD. Amylin receptor (AmR) is a GPCR that mediates several activities, such as improving glucose metabolism, relaxing cerebrovascular structure, modulating inflammatory reactions and potentially enhancing neural regeneration. Recent studies show that peripheral treatments with amylin or its clinical analog, pramlintide, reduced several components of AD pathology, including amyloid plaques, tauopathy, neuroinflammation and other components in the brain, corresponding with improved learning and memory in AD mouse models. Because amylin shares a similar secondary structure with amyloid-β peptide (Aβ), I propose that the AmR/GPCR pathway is disturbed by a large amount of Aβ in the AD brain, leading to tau phosphorylation, neuroinflammation and neuronal death in the pathological cascade. Amylin-type peptides, readily crossing the blood-brain barrier (BBB), are the rational ligands to enhance this GPCR pathway and may exhibit utility as novel therapeutic agents for treating AD.
Collapse
|
38
|
Zhu H, Xue X, Wang E, Wallack M, Na H, Hooker JM, Kowall N, Tao Q, Stein TD, Wolozin B, Qiu WQ. Amylin receptor ligands reduce the pathological cascade of Alzheimer's disease. Neuropharmacology 2017; 119:170-181. [PMID: 28363773 DOI: 10.1016/j.neuropharm.2017.03.030] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 03/21/2017] [Accepted: 03/25/2017] [Indexed: 01/01/2023]
Abstract
Amylin is an important gut-brain axis hormone. Since amylin and amyloid-β peptide (Aβ) share similar β sheet secondary structure despite not having the same primary sequences, we hypothesized that the accumulation of Aβ in the brains of subjects with Alzheimer's disease (AD) might compete with amylin for binding to the amylin receptor (AmR). If true, adding exogenous amylin type peptides would compete with Aβ and reduce the AD pathological cascade, improving cognition. Here we report that a 10-week course of peripheral treatment with human amylin significantly reduced multiple different markers associated with AD pathology, including reducing levels of phospho-tau, insoluble tau, two inflammatory markers (Iba1 and CD68), as well as cerebral Aβ. Amylin treatment also led to improvements in learning and memory in two AD mouse models. Mechanistic studies showed that an amylin receptor antagonist successfully antagonized some protective effects of amylin in vivo, suggesting that the protective effects of amylin require interaction with its cognate receptor. Comparison of signaling cascades emanating from AmR suggest that amylin electively suppresses activation of the CDK5 pathway by Aβ. Treatment with amylin significantly reduced CDK5 signaling in a receptor dependent manner, dramatically decreasing the levels of p25, the active form of CDK5 with a corresponding reduction in tau phosphorylation. This is the first report documenting the ability of amylin treatment to reduce tauopathy and inflammation in animal models of AD. The data suggest that the clinical analog of amylin, pramlintide, might exhibit utility as a therapeutic agent for AD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Haihao Zhu
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA.
| | - Xiehua Xue
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA; Affiliated Rehabilitation Hospital of Fujian, TCM University, China
| | - Erming Wang
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Max Wallack
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA; Harvard Medical School, USA
| | - Hana Na
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Jacob M Hooker
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, USA; Harvard Medical School, USA
| | - Neil Kowall
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA; Alzheimer's Disease Center, Boston University School of Medicine, Boston, MA, USA
| | - Qiushan Tao
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Thor D Stein
- Department of Pathology, Boston University School of Medicine, Boston, MA, USA; Alzheimer's Disease Center, Boston University School of Medicine, Boston, MA, USA
| | - Benjamin Wolozin
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA; Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Wei Qiao Qiu
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA; Alzheimer's Disease Center, Boston University School of Medicine, Boston, MA, USA; Department of Psychiatry, Boston University School of Medicine, Boston, MA, USA.
| |
Collapse
|
39
|
Mohamed LA, Zhu H, Mousa YM, Wang E, Qiu WQ, Kaddoumi A. Amylin Enhances Amyloid-β Peptide Brain to Blood Efflux Across the Blood-Brain Barrier. J Alzheimers Dis 2017; 56:1087-1099. [PMID: 28059785 PMCID: PMC5466167 DOI: 10.3233/jad-160800] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Findings from Alzheimer's disease (AD) mouse models showed that amylin treatment improved AD pathology and enhanced amyloid-β (Aβ) brain to blood clearance; however, the mechanism was not investigated. Using the Tg2576 AD mouse model, a single intraperitoneal injection of amylin significantly increased Aβ serum levels, and the effect was abolished by AC253, an amylin receptor antagonist, suggesting that amylin effect could be mediated by its receptor. Subsequent mechanistic studies showed amylin enhanced Aβ transport across a cell-based model of the blood-brain barrier (BBB), an effect that was abolished when the amylin receptor was inhibited by two amylin antagonists and by siRNA knockdown of amylin receptor Ramp3. To explain this finding, amylin effect on Aβ transport proteins expressed at the BBB was evaluated. Findings indicated that cells treated with amylin induced LRP1 expression, a major receptor involved in brain Aβ efflux, in plasma membrane fraction, suggesting intracellular translocation of LRP1 from the cytoplasmic pool. Increased LRP1 in membrane fraction could explain, at least in part, the enhanced uptake and transport of Aβ across the BBB. Collectively, our findings indicated that amylin induced Aβ brain to blood clearance through amylin receptor by inducing LRP1 subcellular translocation to the plasma membrane of the BBB endothelium.
Collapse
Affiliation(s)
- Loqman A. Mohamed
- Department of Basic Pharmaceutical Science, School of Pharmacy, University of Louisiana at Monroe, Monroe, LA, USA
| | - Haihao Zhu
- Departments of Pharmacology & Experimental Therapeutics, Psychiatry, Boston University School of Medicine, Boston, MA, USA
| | - Youssef M. Mousa
- Department of Basic Pharmaceutical Science, School of Pharmacy, University of Louisiana at Monroe, Monroe, LA, USA
| | - Erming Wang
- Departments of Pharmacology & Experimental Therapeutics, Psychiatry, Boston University School of Medicine, Boston, MA, USA
| | - Wei Qiao Qiu
- Departments of Pharmacology & Experimental Therapeutics, Psychiatry, Boston University School of Medicine, Boston, MA, USA
| | - Amal Kaddoumi
- Department of Basic Pharmaceutical Science, School of Pharmacy, University of Louisiana at Monroe, Monroe, LA, USA
| |
Collapse
|