1
|
Janciauskiene S, Lechowicz U, Pelc M, Olejnicka B, Chorostowska-Wynimko J. Diagnostic and therapeutic value of human serpin family proteins. Biomed Pharmacother 2024; 175:116618. [PMID: 38678961 DOI: 10.1016/j.biopha.2024.116618] [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: 01/11/2024] [Revised: 04/15/2024] [Accepted: 04/17/2024] [Indexed: 05/01/2024] Open
Abstract
SERPIN (serine proteinase inhibitors) is an acronym for the superfamily of structurally similar proteins found in animals, plants, bacteria, viruses, and archaea. Over 1500 SERPINs are known in nature, while only 37 SERPINs are found in humans, which participate in inflammation, coagulation, angiogenesis, cell viability, and other pathophysiological processes. Both qualitative or quantitative deficiencies or overexpression and/or abnormal accumulation of SERPIN can lead to diseases commonly referred to as "serpinopathies". Hence, strategies involving SERPIN supplementation, elimination, or correction are utilized and/or under consideration. In this review, we discuss relationships between certain SERPINs and diseases as well as putative strategies for the clinical explorations of SERPINs.
Collapse
Affiliation(s)
- Sabina Janciauskiene
- Department of Pulmonary and Infectious Diseases and BREATH German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany; Department of Genetics and Clinical Immunology, National Institute of Tuberculosis and Lung Diseases, 26 Plocka St, Warsaw 01-138, Poland
| | - Urszula Lechowicz
- Department of Genetics and Clinical Immunology, National Institute of Tuberculosis and Lung Diseases, 26 Plocka St, Warsaw 01-138, Poland
| | - Magdalena Pelc
- Department of Genetics and Clinical Immunology, National Institute of Tuberculosis and Lung Diseases, 26 Plocka St, Warsaw 01-138, Poland
| | - Beata Olejnicka
- Department of Pulmonary and Infectious Diseases and BREATH German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
| | - Joanna Chorostowska-Wynimko
- Department of Genetics and Clinical Immunology, National Institute of Tuberculosis and Lung Diseases, 26 Plocka St, Warsaw 01-138, Poland.
| |
Collapse
|
2
|
Ali NH, Al-Kuraishy HM, Al-Gareeb AI, Alnaaim SA, Alexiou A, Papadakis M, Saad HM, Batiha GES. The probable role of tissue plasminogen activator/neuroserpin axis in Alzheimer's disease: a new perspective. Acta Neurol Belg 2024; 124:377-388. [PMID: 37917293 DOI: 10.1007/s13760-023-02403-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 10/09/2023] [Indexed: 11/04/2023]
Abstract
Alzheimer's disease (AD) is the most common type of dementia associated with amyloid beta (Aβ) deposition. Dysfunction of the neuronal clearance pathway promotes the accumulation of Aβ. The plasminogen-activating system (PAS) is controlled by various enzymes like tissue plasminogen activators (tPA). Neuronal tPA enhances the conversion of plasminogen to plasmin, which cleaves Aβ; this function is controlled by many inhibitors of PAS, including a plasminogen-activating inhibitor (PAI-1) and neuroserpin. Therefore, the objective of the present narrative review was to explore the potential role of tPA/neuroserpin in the pathogenesis of AD. PAI-1 activity is increased in AD, which is involved in accumulating Aβ. Progressive increase of Aβ level during AD neuropathology is correlated with the over-production of PAI-1 with subsequent reduction of plasmin and tPA activities. Reducing plasmin and tPA activities promote Aβ by reducing Aβ clearance. Neuroserpin plays a critical role in the pathogenesis of AD as it regulates the expression and accumulation of Aβ. Higher expression of neuroserpin inhibits the neuroprotective tPA and the generation of plasmin with subsequent reduction in the clearance of Aβ. These observations raise conflicting evidence on whether neuroserpin is neuroprotective or involved in AD progression. Thus, neuroserpin over-expression with subsequent reduction of tPA may propagate AD neuropathology.
Collapse
Affiliation(s)
- Naif H Ali
- Department of Internal Medicine, Medical College, Najran University, Najran, Saudi Arabia
| | - Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, ALmustansiriyia University, PO Box 14132, Baghdad, Iraq
| | - Ali I Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, ALmustansiriyia University, PO Box 14132, Baghdad, Iraq
| | - Saud A Alnaaim
- Clinical Neurosciences Department, College of Medicine, King Faisal University, Hofuf, Saudi Arabia
| | - Athanasios Alexiou
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, NSW, 2770, Australia
- AFNP Med, 1030, Vienna, Austria
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, Heusnerstrasse 40, University of Witten-Herdecke, 42283, Wuppertal, Germany.
| | - Hebatallah M Saad
- Department of Pathology, Faculty of Veterinary Medicine, Matrouh University, Matrouh, 51744, Matrouh, Egypt.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, AlBeheira, Egypt.
| |
Collapse
|
3
|
Li Z, Liu J. Thyroid dysfunction and Alzheimer's disease, a vicious circle. Front Endocrinol (Lausanne) 2024; 15:1354372. [PMID: 38419953 PMCID: PMC10899337 DOI: 10.3389/fendo.2024.1354372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 02/01/2024] [Indexed: 03/02/2024] Open
Abstract
Recently, research into the link between thyroid dysfunction and Alzheimer's disease (AD) remains a current topic of interest. Previous research has primarily concentrated on examining the impact of thyroid dysfunction on the risk of developing AD, or solely explored the mechanisms of interaction between hypothyroidism and AD, a comprehensive analysis of the mechanisms linking thyroid dysfunction, including hyperthyroidism and hypothyroidism, to Alzheimer's disease (AD) still require further elucidation. Therefore, the aim of this review is to offer a thorough and comprehensive explanation of the potential mechanisms underlying the causal relationship between thyroid dysfunction and AD, highlighting the existence of a vicious circle. The effect of thyroid dysfunction on AD includes neuron death, impaired synaptic plasticity and memory, misfolded protein deposition, oxidative stress, and diffuse and global neurochemical disturbances. Conversely, AD can also contribute to thyroid dysfunction by affecting the stress repair response and disrupting pathways involved in thyroid hormone (TH) production, transport, and activation. Furthermore, this review briefly discusses the role and significance of utilizing the thyroid as a therapeutic target for cognitive recovery in AD. By exploring potential mechanisms and therapeutic avenues, this research contributes to our understanding and management of this devastating neurodegenerative disease.
Collapse
Affiliation(s)
| | - Jia Liu
- Department of Thyroid Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
4
|
Ma W, Song X, Yuan GC, Wang P. RECCIPE: A new framework assessing localized cell-cell interaction on gene expression in multicellular ST data. Front Genet 2024; 15:1322886. [PMID: 38327830 PMCID: PMC10847567 DOI: 10.3389/fgene.2024.1322886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 01/10/2024] [Indexed: 02/09/2024] Open
Abstract
Cell-cell interaction (CCI) plays a pivotal role in cellular communication within the tissue microenvironment. The recent development of spatial transcriptomics (ST) technology and associated data analysis methods has empowered researchers to systematically investigate CCI. However, existing methods are tailored to single-cell resolution datasets, whereas the majority of ST platforms lack such resolution. Additionally, the detection of CCI through association screening based on ST data, which has complicated dependence structure, necessitates proper control of false discovery rates due to the multiple hypothesis testing issue in high dimensional spaces. To address these challenges, we introduce RECCIPE, a novel method designed for identifying cell signaling interactions across multiple cell types in spatial transcriptomic data. RECCIPE integrates gene expression data, spatial information and cell type composition in a multivariate regression framework, enabling genome-wide screening for changes in gene expression levels attributed to CCIs. We show that RECCIPE not only achieves high accuracy in simulated datasets but also provides new biological insights from real data obtained from a mouse model of Alzheimer's disease (AD). Overall, our framework provides a useful tool for studying impact of cell-cell interactions on gene expression in multicellular systems.
Collapse
Affiliation(s)
- Weiping Ma
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Xiaoyu Song
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Institute for Health Care Delivery Science, Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Guo-Cheng Yuan
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Pei Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
5
|
Ren B, Ma J, Tao M, Jing G, Han S, Zhou C, Wang X, Wang J. The disturbance of thyroid-associated hormone and its receptors in brain and blood circulation existed in the early stage of mouse model of Alzheimer's disease. Aging (Albany NY) 2023; 15:1591-1602. [PMID: 36897166 PMCID: PMC10042683 DOI: 10.18632/aging.204570] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 02/20/2023] [Indexed: 03/09/2023]
Abstract
BACKGROUND Studies showed that thyroid function plays an important role in the pathology of Alzheimer's disease (AD). However, changes in brain thyroid hormone and related receptors in the early stage of AD were rarely reported. The aim of this study was to explore the relationship between the early stage of AD and local thyroid hormone and its receptors in the brain. METHODS The animal model was established by stereotactic injection of okadaic acid (OA) into hippocampal region for the experiment, and 0.9% NS for the control. Blood sample from each mouse was collected and then the mice were sacrificed and the brain tissue was collected for detecting free triiodothyronine (FT3), free thyroid hormone (FT4), and thyroid-stimulating hormone (TSH), thyrotropin-releasing hormone (TRH) and phosphorylated tau, amyloid-β (Aβ) and thyroid hormone receptors (THRs) in the hippocampus of the mice were detected as well. RESULTS Enzyme-linked immunosorbent assay showed that compared with the control, FT3, FT4, TSH and TRH in brain were significantly increased in the experimental group; in the serum, FT4, TSH and TRH were increased, while FT3 had no change; western blot analysis indicated that the expression of THR α and β in the hippocampus of the experimental group was significantly higher than that of the control. CONCLUSION Based on the results of this study, a mouse AD model can be established successfully by injecting a small dose of OA into the hippocampus. We speculate that early AD brain and circulating thyroid dysfunction may be an early local and systemic stress repair response.
Collapse
Affiliation(s)
- Bingxiu Ren
- Department of Nuclear Medicine, The Third Affiliated Hospital of Zunyi Medical University (The First People’s Hospital of Zunyi), Zunyi, Guizhou 563000, China
| | - Jinxin Ma
- Department of Hospital Infection Management, The Third Affiliated Hospital of Zunyi Medical University (The First People’s Hospital of Zunyi), Zunyi, Guizhou 563000, China
| | - Min Tao
- Department of Nuclear Medicine, The Third Affiliated Hospital of Zunyi Medical University (The First People’s Hospital of Zunyi), Zunyi, Guizhou 563000, China
| | - Gongwei Jing
- Department of Nuclear Medicine, The Third Affiliated Hospital of Zunyi Medical University (The First People’s Hospital of Zunyi), Zunyi, Guizhou 563000, China
| | - Sheng Han
- Department of Nuclear Medicine, The Third Affiliated Hospital of Zunyi Medical University (The First People’s Hospital of Zunyi), Zunyi, Guizhou 563000, China
| | - Chengyi Zhou
- Department of Nuclear Medicine, The Third Affiliated Hospital of Zunyi Medical University (The First People’s Hospital of Zunyi), Zunyi, Guizhou 563000, China
| | - Xin Wang
- Department of Nuclear Medicine, The Third Affiliated Hospital of Zunyi Medical University (The First People’s Hospital of Zunyi), Zunyi, Guizhou 563000, China
| | - Jiaoya Wang
- Department of Nuclear Medicine, The Third Affiliated Hospital of Zunyi Medical University (The First People’s Hospital of Zunyi), Zunyi, Guizhou 563000, China
| |
Collapse
|
6
|
Silvestri B, Mochi M, Garone MG, Rosa A. Emerging Roles for the RNA-Binding Protein HuD (ELAVL4) in Nervous System Diseases. Int J Mol Sci 2022; 23:14606. [PMID: 36498933 PMCID: PMC9736382 DOI: 10.3390/ijms232314606] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/15/2022] [Accepted: 11/21/2022] [Indexed: 11/24/2022] Open
Abstract
The main goal of this review is to provide an updated overview of the involvement of the RNA-binding protein (RBP) HuD, encoded by the ELAVL4 gene, in nervous system development, maintenance, and function, and its emerging role in nervous system diseases. A particular focus is on recent studies reporting altered HuD levels, or activity, in disease models and patients. Substantial evidence suggests HuD involvement in Parkinson's disease (PD), Alzheimer's disease (AD), and amyotrophic lateral sclerosis (ALS). Interestingly, while possible disease-causing mutations in the ELAVL4 gene remain elusive, a common theme in these diseases seems to be the altered regulation of HuD at multiple steps, including post-transcriptional and post-translational levels. In turn, the changed activity of HuD can have profound implications for its target transcripts, which are overly stabilized in case of HuD gain of function (as proposed in PD and ALS) or reduced in case of decreased HuD binding (as suggested by some studies in AD). Moreover, the recent discovery that HuD is a component of pathological cytoplasmic inclusion in both familial and sporadic ALS patients might help uncover the common molecular mechanisms underlying such complex diseases. We believe that deepening our understanding of the involvement of HuD in neurodegeneration could help developing new diagnostic and therapeutic tools.
Collapse
Affiliation(s)
- Beatrice Silvestri
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, 00185 Rome, Italy
- Center for Life Nano & Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy
| | - Michela Mochi
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, 00185 Rome, Italy
| | - Maria Giovanna Garone
- Department of Stem Cell Biology, Murdoch Children’s Research Institute, The Royal Children’s Hospital, Melbourne, VIC 3052, Australia
| | - Alessandro Rosa
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, 00185 Rome, Italy
- Center for Life Nano & Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy
| |
Collapse
|
7
|
van der Linden RJ, Gerritsen JS, Liao M, Widomska J, Pearse RV, White FM, Franke B, Young-Pearse TL, Poelmans G. RNA-binding protein ELAVL4/HuD ameliorates Alzheimer's disease-related molecular changes in human iPSC-derived neurons. Prog Neurobiol 2022; 217:102316. [PMID: 35843356 PMCID: PMC9912016 DOI: 10.1016/j.pneurobio.2022.102316] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 05/18/2022] [Accepted: 07/12/2022] [Indexed: 11/26/2022]
Abstract
The RNA binding protein ELAVL4/HuD regulates the translation and splicing of multiple Alzheimer's disease (AD) candidate genes. We generated ELAVL4 knockout (KO) human induced pluripotent stem cell-derived neurons to study the effect that ELAVL4 has on AD-related cellular phenotypes. ELAVL4 KO significantly increased the levels of specific APP isoforms and intracellular phosphorylated tau, molecular changes that are related to the pathological hallmarks of AD. Overexpression of ELAVL4 in wild-type neurons and rescue experiments in ELAVL4 KO cells showed opposite effects and also led to a reduction of the extracellular amyloid-beta (Aβ)42/40 ratio. All these observations were made in familial AD (fAD) and fAD-corrected neurons. To gain insight into the molecular cascades involved in neuronal ELAVL4 signaling, we conducted pathway and upstream regulator analyses of transcriptomic and proteomic data from the generated neurons. These analyses revealed that ELAVL4 affects multiple biological pathways linked to AD, including those involved in synaptic function, as well as gene expression downstream of APP and tau signaling. The analyses also suggest that ELAVL4 expression is regulated by insulin receptor-FOXO1 signaling in neurons. Taken together, ELAVL4 expression ameliorates AD-related molecular changes in neurons and affects multiple synaptic pathways, making it a promising target for novel drug development.
Collapse
Affiliation(s)
- Robert J van der Linden
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands; Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Jacqueline S Gerritsen
- Koch Institute for Integrative Cancer Research; Center for Precision Cancer Medicine; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Meichen Liao
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Joanna Widomska
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Richard V Pearse
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Forest M White
- Koch Institute for Integrative Cancer Research; Center for Precision Cancer Medicine; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Barbara Franke
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands; Donders Institute for Brain, Cognition and Behaviour, Nijmegen, the Netherlands; Department of Psychiatry, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Tracy L Young-Pearse
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| | - Geert Poelmans
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands.
| |
Collapse
|
8
|
Andrews EJ, Martini AC, Head E. Exploring the role of sex differences in Alzheimer's disease pathogenesis in Down syndrome. Front Neurosci 2022; 16:954999. [PMID: 36033603 PMCID: PMC9411995 DOI: 10.3389/fnins.2022.954999] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 07/25/2022] [Indexed: 11/14/2022] Open
Abstract
Women are disproportionately affected by Alzheimer's disease (AD), yet little is known about sex-specific effects on the development of AD in the Down syndrome (DS) population. DS is caused by a full or partial triplication of chromosome 21, which harbors the amyloid precursor protein (APP) gene, among others. The majority of people with DS in their early- to mid-40s will accumulate sufficient amyloid-beta (Aβ) in their brains along with neurofibrillary tangles (NFT) for a neuropathological diagnosis of AD, and the triplication of the APP gene is regarded as the main cause. Studies addressing sex differences with age and impact on dementia in people with DS are inconsistent. However, women with DS experience earlier age of onset of menopause, marked by a drop in estrogen, than women without DS. This review focuses on key sex differences observed with age and AD in people with DS and a discussion of possible underlying mechanisms that could be driving or protecting from AD development in DS. Understanding how biological sex influences the brain will lead to development of dedicated therapeutics and interventions to improve the quality of life for people with DS and AD.
Collapse
Affiliation(s)
- Elizabeth J. Andrews
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA, United States
| | - Alessandra C. Martini
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA, United States
| | - Elizabeth Head
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA, United States
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
9
|
Loss of RNA binding protein HuD facilitates the production of the senescence-associated secretory phenotype. Cell Death Dis 2022; 13:329. [PMID: 35411051 PMCID: PMC9001635 DOI: 10.1038/s41419-022-04792-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 02/08/2022] [Accepted: 02/16/2022] [Indexed: 02/06/2023]
Abstract
HuD, an RNA binding protein, plays a role in the regulation of gene expression in certain types of cells, including neuronal cells and pancreatic β-cells, via RNA metabolism. Its aberrant expression is associated with the pathogenesis of several human diseases. To explore HuD-mediated gene regulation, stable cells expressing short hairpin RNA against HuD were established using mouse neuroblastoma Neuro2a (N2a) cells, which displayed enhanced phenotypic characteristics of cellular senescence. Two approaches, RNA immunoprecipitation (RNA IP)-NanoString profiling and cytokine array, were used to subsequently identify a subset of putative HuD targets that act as senescence-associated secretory phenotype (SASP), including C-C motif ligand 2 (CCL2), CCL20, C-X-C motif chemokine ligand 2 (CXCL2), and interleukin-6 (IL-6). Here, we further demonstrated that HuD regulates the expression of CCL2, a SASP candidate upregulated in cells following HuD knockdown, by binding to the 3′-untranslated region (UTR) of Ccl2 mRNA. Downregulation of HuD increased the level of CCL2 in N2a cells and the brain tissues of HuD knockout (KO) mice. Exposure to γ-irradiation induced cellular senescence in N2a cells and HuD knockdown facilitated stress-induced cellular senescence. Our results reveal that HuD acts as a novel regulator of CCL2 expression, and its aberrant expression may contribute to cellular senescence by regulating SASP production.
Collapse
|
10
|
Godinez A, Rajput R, Chitranshi N, Gupta V, Basavarajappa D, Sharma S, You Y, Pushpitha K, Dhiman K, Mirzaei M, Graham S, Gupta V. Neuroserpin, a crucial regulator for axogenesis, synaptic modelling and cell-cell interactions in the pathophysiology of neurological disease. Cell Mol Life Sci 2022; 79:172. [PMID: 35244780 PMCID: PMC8897380 DOI: 10.1007/s00018-022-04185-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 02/01/2022] [Accepted: 02/02/2022] [Indexed: 01/31/2023]
Abstract
Neuroserpin is an axonally secreted serpin that is involved in regulating plasminogen and its enzyme activators, such as tissue plasminogen activator (tPA). The protein has been increasingly shown to play key roles in neuronal development, plasticity, maturation and synaptic refinement. The proteinase inhibitor may function both independently and through tPA-dependent mechanisms. Herein, we discuss the recent evidence regarding the role of neuroserpin in healthy and diseased conditions and highlight the participation of the serpin in various cellular signalling pathways. Several polymorphisms and mutations have also been identified in the protein that may affect the serpin conformation, leading to polymer formation and its intracellular accumulation. The current understanding of the involvement of neuroserpin in Alzheimer's disease, cancer, glaucoma, stroke, neuropsychiatric disorders and familial encephalopathy with neuroserpin inclusion bodies (FENIB) is presented. To truly understand the detrimental consequences of neuroserpin dysfunction and the effective therapeutic targeting of this molecule in pathological conditions, a cross-disciplinary understanding of neuroserpin alterations and its cellular signaling networks is essential.
Collapse
Affiliation(s)
- Angela Godinez
- Faculty of Medicine, Health and Human Sciences, Macquarie University, F10A, 2 Technology Place, North Ryde, NSW, 2109, Australia
| | - Rashi Rajput
- Faculty of Medicine, Health and Human Sciences, Macquarie University, F10A, 2 Technology Place, North Ryde, NSW, 2109, Australia
| | - Nitin Chitranshi
- Faculty of Medicine, Health and Human Sciences, Macquarie University, F10A, 2 Technology Place, North Ryde, NSW, 2109, Australia.
| | - Veer Gupta
- School of Medicine, Deakin University, Melbourne, VIC, Australia
| | - Devaraj Basavarajappa
- Faculty of Medicine, Health and Human Sciences, Macquarie University, F10A, 2 Technology Place, North Ryde, NSW, 2109, Australia
| | - Samridhi Sharma
- Faculty of Medicine, Health and Human Sciences, Macquarie University, F10A, 2 Technology Place, North Ryde, NSW, 2109, Australia
| | - Yuyi You
- Faculty of Medicine, Health and Human Sciences, Macquarie University, F10A, 2 Technology Place, North Ryde, NSW, 2109, Australia
| | - Kanishka Pushpitha
- Faculty of Medicine, Health and Human Sciences, Macquarie University, F10A, 2 Technology Place, North Ryde, NSW, 2109, Australia
| | - Kunal Dhiman
- School of Medicine, Deakin University, Melbourne, VIC, Australia
| | - Mehdi Mirzaei
- Faculty of Medicine, Health and Human Sciences, Macquarie University, F10A, 2 Technology Place, North Ryde, NSW, 2109, Australia
| | - Stuart Graham
- Faculty of Medicine, Health and Human Sciences, Macquarie University, F10A, 2 Technology Place, North Ryde, NSW, 2109, Australia
- Save Sight Institute, University of Sydney, Sydney, NSW, Australia
| | - Vivek Gupta
- Faculty of Medicine, Health and Human Sciences, Macquarie University, F10A, 2 Technology Place, North Ryde, NSW, 2109, Australia.
| |
Collapse
|
11
|
Khaleghzadeh-Ahangar H, Talebi A, Mohseni-Moghaddam P. Thyroid Disorders and Development of Cognitive Impairment: A Review Study. Neuroendocrinology 2022; 112:835-844. [PMID: 34963121 DOI: 10.1159/000521650] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 12/22/2021] [Indexed: 11/19/2022]
Abstract
Dementia is a neurological disorder that is spreading with increasing human lifespan. In this neurological disorder, memory and cognition are declined and eventually impaired. Various factors can be considered as the background of this disorder, one of which is endocrine disorders. Thyroid hormones are involved in various physiological processes in the body; one of the most important of them is neuromodulation. Thyroid disorders, including hyperthyroidism or hypothyroidism, can affect the nervous system and play a role in the development of dementia. Despite decades of investigation, the nature of the association between thyroid disorders and cognition remains a mystery. Given the enhancing global burden of dementia, the principal purpose of this study was to elucidate the association between thyroid disturbances as a potentially modifiable risk factor of cognitive dysfunction. In this review study, we have tried to collect almost all of the reported mechanisms demonstrating the role of hypothyroidism and hyperthyroidism in the pathogenesis of dementia.
Collapse
Affiliation(s)
- Hossein Khaleghzadeh-Ahangar
- Department of Physiology, School of Medicine, Babol University of Medical Sciences, Babol, Iran
- Immunoregulation Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Anis Talebi
- Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Parvaneh Mohseni-Moghaddam
- Department of Physiology, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| |
Collapse
|
12
|
D'Acunto E, Fra A, Visentin C, Manno M, Ricagno S, Galliciotti G, Miranda E. Neuroserpin: structure, function, physiology and pathology. Cell Mol Life Sci 2021; 78:6409-6430. [PMID: 34405255 PMCID: PMC8558161 DOI: 10.1007/s00018-021-03907-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 07/23/2021] [Accepted: 07/27/2021] [Indexed: 12/24/2022]
Abstract
Neuroserpin is a serine protease inhibitor identified in a search for proteins implicated in neuronal axon growth and synapse formation. Since its discovery over 30 years ago, it has been the focus of active research. Many efforts have concentrated in elucidating its neuroprotective role in brain ischemic lesions, the structural bases of neuroserpin conformational change and the effects of neuroserpin polymers that underlie the neurodegenerative disease FENIB (familial encephalopathy with neuroserpin inclusion bodies), but the investigation of the physiological roles of neuroserpin has increased over the last years. In this review, we present an updated and critical revision of the current literature dealing with neuroserpin, covering all aspects of research including the expression and physiological roles of neuroserpin, both inside and outside the nervous system; its inhibitory and non-inhibitory mechanisms of action; the molecular structure of the monomeric and polymeric conformations of neuroserpin, including a detailed description of the polymerisation mechanism; and the involvement of neuroserpin in human disease, with particular emphasis on FENIB. Finally, we briefly discuss the identification by genome-wide screening of novel neuroserpin variants and their possible pathogenicity.
Collapse
Affiliation(s)
- Emanuela D'Acunto
- Department of Biology and Biotechnologies 'Charles Darwin', Sapienza University of Rome, Rome, Italy
| | - Annamaria Fra
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Cristina Visentin
- Department of Biosciences, University of Milan, Milan, Italy
- Institute of Molecular and Translational Cardiology, I.R.C.C.S. Policlinico San Donato, Milan, Italy
| | - Mauro Manno
- Institute of Biophysics, National Research Council of Italy, Palermo, Italy
| | - Stefano Ricagno
- Department of Biosciences, University of Milan, Milan, Italy
| | - Giovanna Galliciotti
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Elena Miranda
- Department of Biology and Biotechnologies 'Charles Darwin', Sapienza University of Rome, Rome, Italy.
- Pasteur Institute-Cenci Bolognetti Foundation, Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
13
|
Quertermous BP, Kavuri S, Walsh DW. An old disease with a new twist: CNS and thyroid sarcoidosis presenting as subacute dementia. Clin Case Rep 2021; 9:e04829. [PMID: 34631064 PMCID: PMC8493448 DOI: 10.1002/ccr3.4829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 08/17/2021] [Accepted: 08/23/2021] [Indexed: 12/03/2022] Open
Abstract
Dementia in the elderly is extremely common and is often irreversible. When a patient presents with rapid cognitive decline, uncommon reversible etiologies should be investigated with the goal of restoring cognitive function.
Collapse
Affiliation(s)
| | - Sravan Kavuri
- Medical College of Georgia Augusta University Augusta GA USA
| | - David W Walsh
- Medical College of Georgia Augusta University Augusta GA USA
| |
Collapse
|
14
|
Han S, Zhang D, Dong Q, Wang X, Wang L. Overexpression of neuroserpin in larval and adult zebrafish shows different behavioral phenotypes. Neurosci Lett 2021; 762:136175. [PMID: 34400286 DOI: 10.1016/j.neulet.2021.136175] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 08/09/2021] [Accepted: 08/11/2021] [Indexed: 01/08/2023]
Abstract
Serpini1, which encodes neuroserpin, has been implicated in the development and maintenance of the nervous system. In this study, an inducible neuroserpin overexpression transgenic zebrafish was generated to investigate its role in different developmental stages. Neuroserpin overexpression was induced by doxycycline in larval and adult zebrafish respectively. Locomotion and thigmotaxis were recorded and analyzed using the ZebraBox high-throughput monitoring equipment and the ZebraLab software system. We find that Tg (serpini1) (+DOX) zebrafish larvae are more hypoactive than their wild-type counterparts at 7 day-postfertilization and anxiety-like behavior is observed in Tg (serpini1) (+DOX) adult zebrafish at 3 month-postfertilization. Furthermore, RNA-sequencing analysis reveals that neuroserpin overexpression affects neurodegeneration-related gene expression. In summary, we report that neuroserpin overexpression in larval and adult zebrafish shows different behavioral phenotypes.
Collapse
Affiliation(s)
- Sha Han
- Department of Neurology, Huashan Hospital, Fudan University, 12 Wulumuqi Zhong Road, Shanghai 200040, People's Republic of China
| | - Dongyang Zhang
- Department of Neurology, Huashan Hospital, Fudan University, 12 Wulumuqi Zhong Road, Shanghai 200040, People's Republic of China
| | - Qiang Dong
- Department of Neurology, Huashan Hospital, Fudan University, 12 Wulumuqi Zhong Road, Shanghai 200040, People's Republic of China
| | - Xu Wang
- Cancer Metabolism Laboratory, Cancer Research Institute, Fudan University Shanghai Cancer Center, Shanghai 200032, People's Republic of China
| | - Liang Wang
- Department of Neurology, Huashan Hospital, Fudan University, 12 Wulumuqi Zhong Road, Shanghai 200040, People's Republic of China.
| |
Collapse
|
15
|
Ambrosio FA, Coricello A, Costa G, Lupia A, Micaelli M, Marchesi N, Sala F, Pascale A, Rossi D, Vasile F, Alcaro S, Collina S. Identification of Compounds Targeting HuD. Another Brick in the Wall of Neurodegenerative Disease Treatment. J Med Chem 2021; 64:9989-10000. [PMID: 34219450 DOI: 10.1021/acs.jmedchem.1c00191] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
ELAV-like (ELAVL) RNA-binding proteins play a pivotal role in post-transcriptional processes, and their dysregulation is involved in several pathologies. This work was focused on HuD (ELAVL4), which is specifically expressed in nervous tissues, and involved in differentiation and synaptic plasticity mechanisms. HuD represents a new, albeit unexplored, candidate target for the treatment of several relevant neurodegenerative diseases. The aim of this pioneering work was the identification of new molecules able to recognize and bind HuD, thus interfering with its activity. We combined virtual screening, molecular dynamics (MD), and STD-NMR techniques. Starting from around 51 000 compounds, four promising hits eventually provided experimental evidence of their ability to bind HuD. Among the selected best hits, folic acid was found to be the most interesting one, being able to well recognize the HuD binding site. Our results provide a basis for the identification of new HuD interfering compounds which may be useful against neurodegenerative syndromes.
Collapse
Affiliation(s)
- Francesca Alessandra Ambrosio
- Dipartimento di Scienze della Salute, Università "Magna Græcia" di Catanzaro, Campus "S. Venuta", 88100 Catanzaro, Italy
| | - Adriana Coricello
- Dipartimento di Scienze della Salute, Università "Magna Græcia" di Catanzaro, Campus "S. Venuta", 88100 Catanzaro, Italy
| | - Giosuè Costa
- Dipartimento di Scienze della Salute, Università "Magna Græcia" di Catanzaro, Campus "S. Venuta", 88100 Catanzaro, Italy.,Net4Science Academic Spin-Off, Università "Magna Græcia" di Catanzaro, Campus "S. Venuta", 88100 Catanzaro, Italy.,Associazione CRISEA-Centro di Ricerca e Servizi Avanzati per l'Innovazione Rurale, Località Condoleo, Belcastro, Catanzaro, Italy
| | - Antonio Lupia
- Net4Science Academic Spin-Off, Università "Magna Græcia" di Catanzaro, Campus "S. Venuta", 88100 Catanzaro, Italy.,Associazione CRISEA-Centro di Ricerca e Servizi Avanzati per l'Innovazione Rurale, Località Condoleo, Belcastro, Catanzaro, Italy
| | - Mariachiara Micaelli
- CIBIO-Department of Cellular, Computational and Integrative Biology, University of Trento, Via Sommarive 9, Povo, 38123 Trento, Italy
| | - Nicoletta Marchesi
- Department of Drug Sciences, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy
| | - Federico Sala
- Department of Drug Sciences, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy.,Department of Chemistry, University of Milan, Via Golgi 19, 20133 Milano, Italy
| | - Alessia Pascale
- Department of Drug Sciences, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy
| | - Daniela Rossi
- Department of Drug Sciences, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy
| | - Francesca Vasile
- Department of Chemistry, University of Milan, Via Golgi 19, 20133 Milano, Italy
| | - Stefano Alcaro
- Dipartimento di Scienze della Salute, Università "Magna Græcia" di Catanzaro, Campus "S. Venuta", 88100 Catanzaro, Italy.,Net4Science Academic Spin-Off, Università "Magna Græcia" di Catanzaro, Campus "S. Venuta", 88100 Catanzaro, Italy.,Associazione CRISEA-Centro di Ricerca e Servizi Avanzati per l'Innovazione Rurale, Località Condoleo, Belcastro, Catanzaro, Italy
| | - Simona Collina
- Department of Drug Sciences, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy
| |
Collapse
|
16
|
RNA-Binding Protein HuD as a Versatile Factor in Neuronal and Non-Neuronal Systems. BIOLOGY 2021; 10:biology10050361. [PMID: 33922479 PMCID: PMC8145660 DOI: 10.3390/biology10050361] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/16/2021] [Accepted: 04/20/2021] [Indexed: 12/12/2022]
Abstract
Simple Summary Tight regulation of gene expression is critical for various biological processes such as proliferation, development, differentiation, and death; its dysregulation is linked to the pathogenesis of diseases. Gene expression is dynamically regulated by numerous factors at DNA, RNA, and protein levels, and RNA binding proteins (RBPs) and non–coding RNAs play important roles in the regulation of RNA metabolisms. RBPs govern a diverse spectrum of RNA metabolism by recognizing and binding to the secondary structure or the certain sequence of target mRNAs, and their malfunctions caused by aberrant expression or mutation are implicated in disease pathology. HuD, an RBP in the human antigen (Hu) family, has been studied as a pivotal regulator of gene expression in neuronal systems; however, accumulating evidence reveals the significance of HuD in non–neuronal systems including certain types of cancer cells or endocrine cells in the lung, pancreas, and adrenal gland. In addition, the abnormal function of HuD suggests its pathological association with neurological disorders, cancers, and diabetes. Thus, this review discusses HuD–mediated gene regulation in neuronal and non–neuronal systems to address how it works to orchestrate gene expression and how its expression is controlled in the stress response of pathogenesis of diseases. Abstract HuD (also known as ELAVL4) is an RNA–binding protein belonging to the human antigen (Hu) family that regulates stability, translation, splicing, and adenylation of target mRNAs. Unlike ubiquitously distributed HuR, HuD is only expressed in certain types of tissues, mainly in neuronal systems. Numerous studies have shown that HuD plays essential roles in neuronal development, differentiation, neurogenesis, dendritic maturation, neural plasticity, and synaptic transmission by regulating the metabolism of target mRNAs. However, growing evidence suggests that HuD also functions as a pivotal regulator of gene expression in non–neuronal systems and its malfunction is implicated in disease pathogenesis. Comprehensive knowledge of HuD expression, abundance, molecular targets, and regulatory mechanisms will broaden our understanding of its role as a versatile regulator of gene expression, thus enabling novel treatments for diseases with aberrant HuD expression. This review focuses on recent advances investigating the emerging role of HuD, its molecular mechanisms of target gene regulation, and its disease relevance in both neuronal and non–neuronal systems.
Collapse
|
17
|
HuD regulates SOD1 expression during oxidative stress in differentiated neuroblastoma cells and sporadic ALS motor cortex. Neurobiol Dis 2020; 148:105211. [PMID: 33271327 DOI: 10.1016/j.nbd.2020.105211] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 10/09/2020] [Accepted: 11/25/2020] [Indexed: 02/07/2023] Open
Abstract
The neuronal RNA-binding protein (RBP) HuD plays an important role in brain development, synaptic plasticity and neurodegenerative diseases such as Parkinson's (PD) and Alzheimer's (AD). Bioinformatics analysis of the human SOD1 mRNA 3' untranslated region (3'UTR) demonstrated the presence of HuD binding adenine-uridine (AU)-rich instability-conferring elements (AREs). Using differentiated SH-SY5Y cells along with brain tissues from sporadic amyotrophic lateral sclerosis (sALS) patients, we assessed HuD-dependent regulation of SOD1 mRNA. In vitro binding and mRNA decay assays demonstrate that HuD specifically binds to SOD1 ARE motifs promoting mRNA stabilization. In SH-SY5Y cells, overexpression of full-length HuD increased SOD1 mRNA and protein levels while a dominant negative form of the RBP downregulated its expression. HuD regulation of SOD1 mRNA was also found to be oxidative stress (OS)-dependent, as shown by the increased HuD binding and upregulation of this mRNA after H2O2 exposure. This treatment also induced a shift in alternative polyadenylation (APA) site usage in SOD1 3'UTR, increasing the levels of a long variant bearing HuD binding sites. The requirement of HuD for SOD1 upregulation during oxidative damage was validated using a specific siRNA that downregulated HuD protein levels to 36% and prevented upregulation of SOD1 and 91 additional genes. In the motor cortex from sALS patients, we found increases in SOD1 and HuD mRNAs and proteins, accompanied by greater HuD binding to this mRNA as confirmed by RNA-immunoprecipitation (RIP) assays. Altogether, our results suggest a role of HuD in the post-transcriptional regulation of SOD1 expression during ALS pathogenesis.
Collapse
|
18
|
Cai L, Zhou Y, Wang Z, Zhu Y. Neuroserpin extends the time window of tPA thrombolysis in a rat model of middle cerebral artery occlusion. J Biochem Mol Toxicol 2020; 34:e22570. [PMID: 32659866 DOI: 10.1002/jbt.22570] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 05/20/2020] [Accepted: 06/23/2020] [Indexed: 12/13/2022]
Abstract
Tissue-type plasminogen activator (tPA) is characterized as an effective drug for early thrombolytic therapy in acute cerebral infarction (ACI). However, tPA will increase the risk of hemorrhage if it is used beyond the treatment time window. The study aims to explore the effects of neuroserpin (NSP) on the time window of tPA thrombolysis in ACI and the underlying mechanism. The middle cerebral artery occlusion (MCAO) model was constructed in rats, which were randomly divided into six groups: sham operation group, infarction group, 1-hour thrombolysis group, 1-hour thrombolytic + NSP intervention group, 4-hour thrombolytic group, and 4-hour thrombolysis + NSP intervention group. The neurological changes in rats were evaluated by modified neurological severity scores and rota-rod test. The brain edema and cerebral infarction area were evaluated by dry-wet method and triphenyl tetrazolium chloride staining. The blood-brain barrier (BBB) integrity was examined by Evans blue method. The expressions of malondialdehyde, superoxide dismutase, and glutathione peroxidase in brain were also investigated. The expression of caspase-3 and Bcl-2 in brain tissue and apoptosis of neurons were examined by Western blot analysis and toluidine blue staining. tPA thrombolysis significantly attenuated the neurological impairment in rats with MCAO at 1 hour. Conversely, the effect of tPA thrombolysis at 4 hours after MCAO did not significantly help the recovery of neurological function. However, a combination of tPA treatment and NSP treatment at 4 hours after MCAO markedly ameliorated the neurological impairment, cerebral edema, cerebral infarction volume, BBB injury, oxidative stress products, and neuron apoptosis. NSP can probably expand the time window for tPA treatment to reduce neurological impairment in ACI.
Collapse
Affiliation(s)
- Li Cai
- Department of Craniocerebral Surgery, Guangxi International Zhuang Medicine Hospital, Nanning, China
| | - Youdong Zhou
- Department of Neurosurgery, Xiangyang Central Hospital Affiliated to Hubei University of Arts and Science, Xiangyang, China
| | - Zhiyong Wang
- Department of Neurosurgery, Xiangyang Central Hospital Affiliated to Hubei University of Arts and Science, Xiangyang, China
| | - Yaozu Zhu
- Department of Neurosurgery, Xiangyang Central Hospital Affiliated to Hubei University of Arts and Science, Xiangyang, China
| |
Collapse
|
19
|
Joy Mathew C, Jose MT, Elshaikh AO, Shah L, Lee R, Cancarevic I. Is Hyperthyroidism a Possible Etiology of Early Onset Dementia? Cureus 2020; 12:e10603. [PMID: 33133806 PMCID: PMC7586400 DOI: 10.7759/cureus.10603] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Dementia, a disabling syndrome of the elderly characterized by the decline in memory and cognition, is increasing in incidence and affects not only the individual but also their family and close ones. Hyperthyroidism can mimic many other diseases and untreated hyperthyroidism can lead to adverse problems of various systems including the heart, bones, muscles, menstrual cycle, and fertility. In this article, we have tried to evaluate the association between hyperthyroidism and dementia, as well as the impact of hyperthyroidism management in the treatment and prevention of dementia. Studies available in the PubMed database have been used, excluding animal studies and including studies of adults above the age of 50. The analysis of studies reveals that thyroid dysfunction can lead to cognitive impairment. It has not been able to prove that hyperthyroidism can lead to an earlier onset of dementia. But subclinical hyperthyroidism, thyroid-stimulating hormone (TSH) levels below the normal range, and high free thyroxine (T4) levels increase the risk of dementia among the elderly. The possible mechanisms involved in this association have also been discussed. Thus, we concluded that it is essential to detect and manage hyperthyroidism at an earlier stage since hyperthyroidism increases the risk of dementia. The possibility of using antithyroid treatment in euthyroid dementia is yet to be studied extensively.
Collapse
Affiliation(s)
| | - Merin Tresa Jose
- Family Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Abeer O Elshaikh
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Lisa Shah
- Family and Community Medicine, Smt. Nathiba Hargovandas Lakhmichand Municipal Medical College, Ahmedabad, IND
| | - Robert Lee
- Surgery, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Ivan Cancarevic
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| |
Collapse
|
20
|
Bavarsad K, Hosseini M, Hadjzadeh MAR, Sahebkar A. The effects of thyroid hormones on memory impairment and Alzheimer's disease. J Cell Physiol 2019; 234:14633-14640. [PMID: 30680727 DOI: 10.1002/jcp.28198] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 01/16/2019] [Indexed: 01/24/2023]
Abstract
Thyroid hormones (THs) have a wide and important range of effects within the central nervous system beginning from fetal life and continuing throughout the adult life. Thyroid disorders are one of the major causes of cognitive impairment including Alzheimer's disease (AD). Several studies in recent years have indicated an association between hypothyroidism or hyperthyroidism and AD. Despite available evidence for this association, it remains unclear whether thyroid dysfunction results from or contributes to the progression of AD. This review discusses the role of THs in learning and memory and summarizes the studies that have linked thyroid function and AD. Eventually, we elaborate how THs may be effective in treating AD by putting forward potential mechanisms.
Collapse
Affiliation(s)
- Kowsar Bavarsad
- Department of Physiology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Hosseini
- Department of Physiology, Mashhad University of Medical Sciences, Mashhad, Iran.,Division of Neurocognitive Sciences, Psychiatry and Behavioral Sciences Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mousa-Al-Reza Hadjzadeh
- Department of Physiology, Mashhad University of Medical Sciences, Mashhad, Iran.,Division of Neurocognitive Sciences, Psychiatry and Behavioral Sciences Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
21
|
Bonnet J, Garcia C, Leger T, Couquet MP, Vignoles P, Vatunga G, Ndung'u J, Boudot C, Bisser S, Courtioux B. Proteome characterization in various biological fluids of Trypanosoma brucei gambiense-infected subjects. J Proteomics 2018; 196:150-161. [PMID: 30414516 DOI: 10.1016/j.jprot.2018.11.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 10/02/2018] [Accepted: 11/05/2018] [Indexed: 02/04/2023]
Abstract
Human African trypanosomiasis (HAT) is a neglected tropical disease that is endemic in sub-Saharan Africa. Control of the disease has been recently improved by better screening and treatment strategies, and the disease is on the WHO list of possible elimination. However, some physiopathological aspects of the disease transmission and progression remain unclear. We propose a new proteomic approach to identify new targets and thus possible new biomarkers of the disease. We also focused our attention on fluids classically associated with HAT (serum and cerebrospinal fluid (CSF)) and on the more easily accessible biological fluids urine and saliva. Liquid chromatography-tandem mass spectrometry (LC-MS/MS) established the proteomic profile of patients with early and late stage disease. The serum, CSF, urine and saliva of 3 uninfected controls, 3 early stage patients and 4 late stage patients were analyzed. Among proteins identified, in CSF, urine and saliva, respectively, 37, 8 and 24 proteins were differentially expressed and showed particular interest with regards to their function. The most promising proteins (Neogenin, Neuroserpin, secretogranin 2 in CSF; moesin in urine and intelectin 2 in saliva) were quantified by enzyme-linked immunosorbent assay in a confirmatory cohort of 14 uninfected controls, 23 patients with early stage disease and 43 patients with late stage disease. The potential of two proteins, neuroserpin and moesin, with the latter present in urine, were further characterized. Our results showed the potential of proteomic analysis to discover new biomarkers and provide the basis of the establishment of a new proteomic catalogue applied to HAT-infected subjects and controls. SIGNIFICANCE: Sleeping sickness, also called Human African Trypanosomiasis (HAT), is a parasitic infection caused by a parasitic protozoan, Trypanosoma brucei gambiense or T. b. rhodesiense which are transmitted via an infected tsetse fly: Glossina. For both, the haemolymphatic stage (or first stage) signs and symptoms are intermittent fever, lymphadenopathy, hepatosplenomegaly, headaches, pruritus, and for T. b. rhodesiense infection a chancre is often formed at the bite site. Meningoencephalitic stage (or second stage) occurs when parasites invade the CNS, it is characterised by neurological signs and symptoms such as altered gait, tremors, neuropathy, somnolence which can lead to coma and death if untreated. first stage of the disease is characterizing by fevers, headaches, itchiness, and joint pains and progressive lethargy corresponding to the second stage with confusion, poor coordination, numbness and trouble sleeping. Actually, diagnosing HAT requires specialized expertise and significant resources such as well-equipped health centers and qualified staff. Such resources are lacking in many endemic areas that are often in rural locales, so many individuals with HAT die before the diagnosis is established. In this study, we analysed by mass spectrometry the entire proteome of serum, CSF, urine and saliva samples from infected and non-infected Angolan individuals to define new biomarkers of the disease. This work of proteomics analysis is a preliminary stage to the characterization of the whole proteome, of these 4 biological fluids, of HAT patients. We have identified 69 new biomarkers. Five of them have been thoroughly investigated by ELISA quantification. Neuroserpine and Moesin are respectively promising new biomarkers in CSF and urine's patient for a better diagnosis.
Collapse
Affiliation(s)
- Julien Bonnet
- Institute of Neuroepidemiology and Tropical Neurology, School of Medicine, CNRS FR 3503 GEIST, University of Limoges, INSERM UMR 1094 Tropical Neuroepidemiology, Limoges, France.
| | - Camille Garcia
- Jacques Monod Institute, Proteomics Facility, University Paris Diderot Sorbonne Paris Cité, Paris, France..
| | - Thibaut Leger
- Jacques Monod Institute, Proteomics Facility, University Paris Diderot Sorbonne Paris Cité, Paris, France..
| | - Marie-Pauline Couquet
- Institute of Neuroepidemiology and Tropical Neurology, School of Medicine, CNRS FR 3503 GEIST, University of Limoges, INSERM UMR 1094 Tropical Neuroepidemiology, Limoges, France.
| | - Philippe Vignoles
- Institute of Neuroepidemiology and Tropical Neurology, School of Medicine, CNRS FR 3503 GEIST, University of Limoges, INSERM UMR 1094 Tropical Neuroepidemiology, Limoges, France.
| | - Gedeao Vatunga
- Instituto de Combate e controlo das Tripanossomiases (ICCT), Luanda, Angola.
| | - Joseph Ndung'u
- Foundation for Innovative New Diagnostics (FIND), Geneva, Switzerland.
| | - Clotilde Boudot
- Institute of Neuroepidemiology and Tropical Neurology, School of Medicine, CNRS FR 3503 GEIST, University of Limoges, INSERM UMR 1094 Tropical Neuroepidemiology, Limoges, France.
| | - Sylvie Bisser
- Institute of Neuroepidemiology and Tropical Neurology, School of Medicine, CNRS FR 3503 GEIST, University of Limoges, INSERM UMR 1094 Tropical Neuroepidemiology, Limoges, France; Pasteur Institute in French Guiana, 23 Boulevard Pasteur, 973006, Cayenne Cedex, French Guiana.
| | - Bertrand Courtioux
- Institute of Neuroepidemiology and Tropical Neurology, School of Medicine, CNRS FR 3503 GEIST, University of Limoges, INSERM UMR 1094 Tropical Neuroepidemiology, Limoges, France.
| |
Collapse
|
22
|
Armañanzas R. Revealing post-transcriptional microRNA-mRNA regulations in Alzheimer's disease through ensemble graphs. BMC Genomics 2018; 19:668. [PMID: 30255799 PMCID: PMC6157163 DOI: 10.1186/s12864-018-5025-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND In silico investigations on the integration of multiple datasets are in need of higher statistical power methods to unveil secondary findings that were hidden from the initial analyses. We present here a novel method for the network analysis of messenger RNA post-translational regulation by microRNA molecules. The method integrates expression data and sequence binding predictions through a set of sound machine learning techniques, forwarding all results to an ensemble graph of regulations. RESULTS Bayesian network classifiers are induced based on a pool of ensemble graphs with ascending order of complexity. Individual goodness-of-fit and classification performances are evaluated for each learned model. As a testbed, four Alzheimer's disease datasets are integrated using the new approach, achieving top values of 0.9794 ± 0.01 for the area under the receiver operating characteristic curve and 0.9439 ± 0.0234 for the prediction accuracy. CONCLUSIONS Post-transcriptional regulations found by the optimal network classifier concur with previous literature findings. Furthermore, additional network structures suggest previously unreported regulations in the state of the art of Alzheimer's research. The quantitative performance as well as sound biological findings provide confidence in the ensemble approach and encourage similar integrative analyses for other conditions.
Collapse
Affiliation(s)
- Rubén Armañanzas
- Department of Bioengineering, Krasnow Institute for Advanced Study, George Mason University, 4400 University Dr, MS2A1, Fairfax, 22030, VA, USA.
| |
Collapse
|
23
|
Bavarsad K, Hadjzadeh MAR, Hosseini M, Pakdel R, Beheshti F, Bafadam S, Ashaari Z. Effects of levothyroxine on learning and memory deficits in a rat model of Alzheimer’s disease: the role of BDNF and oxidative stress. Drug Chem Toxicol 2018; 43:57-63. [DOI: 10.1080/01480545.2018.1481085] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Kowsar Bavarsad
- Student Research Committee, Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mousa-Al-Reza Hadjzadeh
- Division of Neurocognitive Sciences, Psychiatry and Behavioral Sciences Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Hosseini
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Roghayeh Pakdel
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran
| | - Farimah Beheshti
- Department of Basic Sciences and Neuroscience Research Center, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Soleyman Bafadam
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zeinab Ashaari
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
24
|
Li H, Sun X, Yu F, Xu L, Miu J, Xiao P. In Silico Investigation of the Pharmacological Mechanisms of Beneficial Effects of Ginkgo biloba L. on Alzheimer's Disease. Nutrients 2018; 10:nu10050589. [PMID: 29747475 PMCID: PMC5986469 DOI: 10.3390/nu10050589] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Revised: 05/01/2018] [Accepted: 05/08/2018] [Indexed: 11/21/2022] Open
Abstract
Based on compelling experimental and clinical evidence, Ginkgo biloba L. exerts a beneficial effect in ameliorating mild to moderate dementia in patients with Alzheimer’s disease (AD) and other neurological disorders, although the pharmacological mechanisms remain unknown. In the present study, compounds, their putative target proteins identified using an inverse docking approach, and clinically tested AD-related target proteins were systematically integrated together with applicable bioinformatics methods in silico. The results suggested that the beneficial effects of G. biloba on AD may be contributed by the regulation of hormone sensitivity, improvements in endocrine homeostasis, maintenance of endothelial microvascular integrity, and proteolysis of tau proteins, particularly prior to amyloid β-protein (Aβ) plaque formation. Moreover, we identified six putative protein targets that are significantly related to AD, but have not been researched or have had only preliminary studies conducted on the anti-AD effects of G. biloba. These mechanisms and protein targets are very significant for future scientific research. In addition, the existing mechanisms were also verified, such as the reduction of oxidative stress, anti-apoptotic effects, and protective effects against amyloidogenesis and Aβ aggregation. The discoveries summarized here may provide a macroscopic perspective that will improve our understanding of the molecular mechanism of medicinal plants or dietary supplements, as well as new clues for the future development of therapeutic strategies for AD.
Collapse
Affiliation(s)
- Hongxiang Li
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, 151 Malianwa North Road, Beijing 100193, China.
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, 151 Malianwa North Road, Beijing 100193, China.
| | - Xiaoyuan Sun
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, 151 Malianwa North Road, Beijing 100193, China.
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, 151 Malianwa North Road, Beijing 100193, China.
| | - Fan Yu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, 151 Malianwa North Road, Beijing 100193, China.
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, 151 Malianwa North Road, Beijing 100193, China.
| | - Lijia Xu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, 151 Malianwa North Road, Beijing 100193, China.
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, 151 Malianwa North Road, Beijing 100193, China.
| | - Jianhua Miu
- Guangxi Institute of Medicinal Plant Development, Nanning, 189 Changgang Road, Nanning 520023, China.
| | - Peigen Xiao
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, 151 Malianwa North Road, Beijing 100193, China.
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, 151 Malianwa North Road, Beijing 100193, China.
| |
Collapse
|
25
|
Giampietro C, Lionetti MC, Costantini G, Mutti F, Zapperi S, La Porta CAM. Cholesterol impairment contributes to neuroserpin aggregation. Sci Rep 2017; 7:43669. [PMID: 28255164 PMCID: PMC5334643 DOI: 10.1038/srep43669] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 01/27/2017] [Indexed: 01/05/2023] Open
Abstract
Intraneural accumulation of misfolded proteins is a common feature of several neurodegenerative pathologies including Alzheimer's and Parkinson's diseases, and Familial Encephalopathy with Neuroserpin Inclusion Bodies (FENIB). FENIB is a rare disease due to a point mutation in neuroserpin which accelerates protein aggregation in the endoplasmic reticulum (ER). Here we show that cholesterol depletion induced either by prolonged exposure to statins or by inhibiting the sterol reg-ulatory binding-element protein (SREBP) pathway also enhances aggregation of neuroserpin proteins. These findings can be explained considering a computational model of protein aggregation under non-equilibrium conditions, where a decrease in the rate of protein clearance improves aggregation. Decreasing cholesterol in cell membranes affects their biophysical properties, including their ability to form the vesicles needed for protein clearance, as we illustrate by a simple mathematical model. Taken together, these results suggest that cholesterol reduction induces neuroserpin aggregation, even in absence of specific neuroserpin mutations. The new mechanism we uncover could be relevant also for other neurodegenerative diseases associated with protein aggregation.
Collapse
Affiliation(s)
| | - Maria Chiara Lionetti
- Center for Complexity and Biosystems, Department of Biosciences, University of Milano, via Celoria 26, 20133 Milano, Italy
| | - Giulio Costantini
- Center for Complexity and Biosystems, Department of Physics, University of Milano, via Celoria 16, 20133 Milano, Italy
| | - Federico Mutti
- Center for Complexity and Biosystems, Department of Biosciences, University of Milano, via Celoria 26, 20133 Milano, Italy
| | - Stefano Zapperi
- Center for Complexity and Biosystems, Department of Physics, University of Milano, via Celoria 16, 20133 Milano, Italy
- CNR - Consiglio Nazionale delle Ricerche, Istituto di Chimica della Materia Condensata e di Tecnologie per l’Energia, Via R. Cozzi 53, 20125 Milano, Italy
- ISI Foundation, Via Alassio 11C, Torino, Italy
- Department of Applied Physics, Aalto University, P.O. Box 14100, FIN-00076, Aalto, Finland
| | - Caterina A. M. La Porta
- Center for Complexity and Biosystems, Department of Biosciences, University of Milano, via Celoria 26, 20133 Milano, Italy
| |
Collapse
|
26
|
Lee TW, Tsang VWK, Loef EJ, Birch NP. Physiological and pathological functions of neuroserpin: Regulation of cellular responses through multiple mechanisms. Semin Cell Dev Biol 2017; 62:152-159. [PMID: 27639894 DOI: 10.1016/j.semcdb.2016.09.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 09/09/2016] [Accepted: 09/12/2016] [Indexed: 11/17/2022]
Abstract
It is 27 years since neuroserpin was first discovered in the nervous system and identified as a member of the serpin superfamily. Since that time potential roles for this serine protease inhibitor have been identified in neuronal and non-neuronal systems. Many are linked to inhibition of neuroserpin's principal enzyme target, tissue plasminogen activator (tPA), although some have been suggested to involve alternate non-inhibitory mechanisms. This review focuses mainly on the inhibitory roles of neuroserpin and discusses the evidence supporting tPA as the physiological target. While the major sites of neuroserpin expression are neural, endocrine and immune tissues, most progress on characterizing functional roles for neuroserpin have been in the brain. Roles in emotional behaviour, synaptic plasticity and neuroprotection in stroke and excitotoxicity models are discussed. Current knowledge on three neurological diseases associated with neuroserpin mutation or activity, Familial Encephalopathy with Neuroserpin Inclusion Bodies (FENIB), Alzheimer's disease and brain metastasis is presented. Finally, we consider mechanistic studies that have revealed a distinct inhibitory mechanism for neuroserpin and its possible implications for neuroserpin function.
Collapse
Affiliation(s)
- Tet Woo Lee
- School of Biological Sciences and Centre for Brain Research, University of Auckland, Auckland, New Zealand.
| | - Vicky W K Tsang
- School of Biological Sciences and Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Evert Jan Loef
- School of Biological Sciences and Centre for Brain Research, University of Auckland, Auckland, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| | - Nigel P Birch
- School of Biological Sciences and Centre for Brain Research, University of Auckland, Auckland, New Zealand; Brain Research New Zealand, Rangahau Roro Aotearoa, Auckland, New Zealand.
| |
Collapse
|
27
|
D Potdar P, U Shetti A. Molecular Biomarkers for Diagnosis & Therapies of Alzheimer’s Disease. AIMS Neurosci 2016. [DOI: 10.3934/neuroscience.2016.4.433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
28
|
The plasminogen activation system in neuroinflammation. Biochim Biophys Acta Mol Basis Dis 2015; 1862:395-402. [PMID: 26493446 DOI: 10.1016/j.bbadis.2015.10.011] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 10/08/2015] [Accepted: 10/15/2015] [Indexed: 01/30/2023]
Abstract
The plasminogen activation (PA) system consists in a group of proteases and protease inhibitors regulating the activation of the zymogen plasminogen into its proteolytically active form, plasmin. Here, we give an update of the current knowledge about the role of the PA system on different aspects of neuroinflammation. These include modification in blood-brain barrier integrity, leukocyte diapedesis, removal of fibrin deposits in nervous tissues, microglial activation and neutrophil functions. Furthermore, we focus on the molecular mechanisms (some of them independent of plasmin generation and even of proteolysis) and target receptors responsible for these effects. The description of these mechanisms of action may help designing new therapeutic strategies targeting the expression, activity and molecular mediators of the PA system in neurological disorders involving neuroinflammatory processes. This article is part of a Special Issue entitled: Neuro Inflammation edited by Helga E. de Vries and Markus Schwaninger.
Collapse
|
29
|
Lee YS, Lee JA, Kaang BK. Regulation of mRNA stability by ARE-binding proteins in synaptic plasticity and memory. Neurobiol Learn Mem 2015; 124:28-33. [PMID: 26291750 DOI: 10.1016/j.nlm.2015.08.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 08/01/2015] [Accepted: 08/03/2015] [Indexed: 11/27/2022]
Abstract
Formation of long-term memories requires coordinated gene expression, which can be regulated at transcriptional, post-transcriptional, and translational levels. Post-transcriptional stabilization and destabilization of mRNAs provides precise temporal and spatial regulation of gene expression, which is critical for consolidation of synaptic plasticity and memory. mRNA stability is regulated by interactions between the cis-acting elements of mRNAs, such as adenine-uridine-rich elements (AREs), and the trans-acting elements, ARE-binding proteins (AUBPs). There are several AUBPs in the nervous system. Among AUBPs, Hu/ELAV-like proteins and AUF1 are the most studied mRNA stabilizing and destabilizing factors, respectively. Here, we summarize compelling evidence for critical roles of these AUBPs in synaptic plasticity, as well as learning and memory, in both vertebrates and invertebrates. Furthermore, we also briefly review the deregulations of AUBPs in neurological disorders.
Collapse
Affiliation(s)
- Yong-Seok Lee
- Department of Life Science, College of Natural Sciences, Chung-Ang University, Seoul, South Korea.
| | - Jin-A Lee
- Department of Biotechnology and Biological Sciences, Hannam University, Daejeon, South Korea
| | - Bong-Kiun Kaang
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea.
| |
Collapse
|
30
|
|