51
|
Kishino A, Hayashi K, Hidai C, Masuda T, Nomura Y, Oshima T. XBP1-FoxO1 interaction regulates ER stress-induced autophagy in auditory cells. Sci Rep 2017; 7:4442. [PMID: 28667325 PMCID: PMC5493624 DOI: 10.1038/s41598-017-02960-1] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 04/21/2017] [Indexed: 01/07/2023] Open
Abstract
The purpose of this study was to clarify the relationship among X-box-binding protein 1 unspliced, spliced (XBP1u, s), Forkhead box O1 (FoxO1) and autophagy in the auditory cells under endoplasmic reticulum (ER) stress. In addition, the relationship between ER stress that causes unfolded protein response (UPR) and autophagy was also investigated. The present study reported ER stress induction by tunicamycin treatment that resulted in IRE1α-mediated XBP1 mRNA splicing and autophagy. XBP1 mRNA splicing and FoxO1 were found to be involved in ER stress-induced autophagy. This inference was based on the observation that the expression of LC3-II was suppressed by knockdown of IRE1α, XBP1 or FoxO1. In addition, XBP1u was found to interact with XBP1s in auditory cells under ER stress, functioning as a negative feedback regulator that was based on two important findings. Firstly, there was a significant inverse correlation between XBP1u and XBP1s expressions, and secondly, the expression of XBP1 protein showed different dynamics compared to the XBP1 mRNA level. Furthermore, our results regarding the relationship between XBP1 and FoxO1 by small interfering RNA (siRNA) paradoxically showed negative regulation of FoxO1 expression by XBP1. Our findings revealed that the XBP1-FoxO1 interaction regulated the ER stress-induced autophagy in auditory cells.
Collapse
Affiliation(s)
- Akihiro Kishino
- Department of Otolaryngology, School of Medicine, Nihon University, Tokyo, 173-8610, Japan
| | - Ken Hayashi
- Department of Otolaryngology, Kamio Memorial Hospital, Tokyo, 101-0063, Japan
| | - Chiaki Hidai
- Department of Physiology, School of Medicine, Nihon University, Tokyo, 173-8610, Japan
| | - Takeshi Masuda
- Department of Otolaryngology, School of Medicine, Nihon University, Tokyo, 173-8610, Japan
| | - Yasuyuki Nomura
- Department of Otolaryngology, School of Medicine, Nihon University, Tokyo, 173-8610, Japan
| | - Takeshi Oshima
- Department of Otolaryngology, School of Medicine, Nihon University, Tokyo, 173-8610, Japan.
| |
Collapse
|
52
|
Wetzel S, Seipold L, Saftig P. The metalloproteinase ADAM10: A useful therapeutic target? BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017. [PMID: 28624438 DOI: 10.1016/j.bbamcr.2017.06.005] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Proteolytic cleavage represents a unique and irreversible posttranslational event regulating the function and half-life of many intracellular and extracellular proteins. The metalloproteinase ADAM10 has raised attention since it cleaves an increasing number of protein substrates close to the extracellular membrane leaflet. This "ectodomain shedding" regulates the turnover of a number of transmembrane proteins involved in cell adhesion and receptor signaling. It can initiate intramembrane proteolysis followed by nuclear transport and signaling of the cytoplasmic domain. ADAM10 has also been implicated in human disorders ranging from neurodegeneration to dysfunction of the immune system and cancer. Targeting proteases for therapeutic purposes remains a challenge since these enzymes including ADAM10 have a wide range of substrates. Accelerating or inhibiting a specific protease activity is in most cases associated with unwanted side effects and a therapeutic useful window of application has to be carefully defined. A better understanding of the regulatory mechanisms controlling the expression, subcellular localization and activity of ADAM10 will likely uncover suitable drug targets which will allow a more specific and fine-tuned modulation of its proteolytic activity.
Collapse
Affiliation(s)
- Sebastian Wetzel
- Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, Olshausenstrasse 40, D-24098 Kiel, Germany
| | - Lisa Seipold
- Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, Olshausenstrasse 40, D-24098 Kiel, Germany
| | - Paul Saftig
- Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, Olshausenstrasse 40, D-24098 Kiel, Germany.
| |
Collapse
|
53
|
Wang L, Cheng W, Zhang Z. Respiratory syncytial virus infection accelerates lung fibrosis through the unfolded protein response in a bleomycin-induced pulmonary fibrosis animal model. Mol Med Rep 2017; 16:310-316. [DOI: 10.3892/mmr.2017.6558] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 02/16/2017] [Indexed: 11/06/2022] Open
|
54
|
Meng T, Cao Q, Lei P, Bush AI, Xiang Q, Su Z, He X, Rogers JT, Chiu IM, Zhang Q, Huang Y. Tat-haFGF 14-154 Upregulates ADAM10 to Attenuate the Alzheimer Phenotype of APP/PS1 Mice through the PI3K-CREB-IRE1α/XBP1 Pathway. MOLECULAR THERAPY-NUCLEIC ACIDS 2017. [PMID: 28624220 PMCID: PMC5443968 DOI: 10.1016/j.omtn.2017.05.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Acid fibroblast growth factor (aFGF) has shown neuroprotection in Alzheimer’s disease (AD) models in previous studies, yet its mechanism is still uncertain. Here we report that the efficacy of Tat-haFGF14–154 is markedly increased when loaded cationic liposomes for intranasal delivery are intranasally administered to APP/PS1 mice. Our results demonstrated that liposomal Tat-haFGF14–154 treatment significantly ameliorated behavioral deficits, relieved brain Aβ burden, and increased the expression and activity of disintegrin and metalloproteinase domain-containing protein 10 (ADAM10) in the brain. Tat-haFGF14–154 antagonized Aβ1–42-induced cell death and structural damage in rat primary neurons in an ADAM10-dependent manner, which, in turn, was promoted by the activation of XBP1 splicing and modulated by the PI3K-CREB pathway. Both knockdown of ADAM10 and inhibition of PI3K (LY294002) negated Tat-haFGF14–154 rescue. Thus, Tat-haFGF14–154 activates the IRE1α/XBP1 pathway of the unfolded protein response (UPR) against the endoplasmic reticulum (ER) stress induced by Aβ, and, subsequently, the nuclear translocation of spliced XBP1 (XBP1s) promotes transcription of ADAM10. These results highlight the important role of ADAM10 and its activation through the PI3K-CREB-IRE1α/XBP1 pathway as a key factor in the mechanism of neuroprotection for Tat-haFGF14–154.
Collapse
Affiliation(s)
- Tian Meng
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China
| | - Qin Cao
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China
| | - Peng Lei
- Oxidation Biology Unit, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Ashley I Bush
- Oxidation Biology Unit, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Qi Xiang
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China; Cell Biology Department and National Engineering Research Center of Genetic Medicine, Jinan University, Guangzhou 510632, China
| | - Zhijian Su
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China; Cell Biology Department and National Engineering Research Center of Genetic Medicine, Jinan University, Guangzhou 510632, China
| | - Xiang He
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China
| | - Jack T Rogers
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02114, USA
| | - Ing-Ming Chiu
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Qihao Zhang
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China; Cell Biology Department and National Engineering Research Center of Genetic Medicine, Jinan University, Guangzhou 510632, China.
| | - Yadong Huang
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China; Cell Biology Department and National Engineering Research Center of Genetic Medicine, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
55
|
Freitas-Rodríguez S, Folgueras AR, López-Otín C. The role of matrix metalloproteinases in aging: Tissue remodeling and beyond. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:2015-2025. [PMID: 28499917 DOI: 10.1016/j.bbamcr.2017.05.007] [Citation(s) in RCA: 171] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 05/03/2017] [Accepted: 05/04/2017] [Indexed: 12/28/2022]
Abstract
Proteases are a set of enzymes that have been involved in multiple biological processes throughout evolution. Among them, extracellular matrix (ECM) remodeling has emerged as one of the most relevant functions exerted by these proteins, being essential in the regulation of critical events such as embryonic development or tissue homeostasis. Hence, it is not surprising that dysregulation in any protease function that affects ECM homeostasis may contribute to the aging process. Matrix metalloproteinases (MMPs) are one of the most important families of proteases involved in the tight control of ECM remodeling over time. In this review, we will discuss how MMPs and other proteases alter ECM composition and mechanical properties in aging, thereby affecting stem cell niches and the development of senescent phenotypes. Finally, we will summarize recent findings that associate MMPs with the development of age-related diseases, such as neurodegenerative disorders.
Collapse
Affiliation(s)
- Sandra Freitas-Rodríguez
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de Oviedo, 33006 Oviedo, Spain
| | - Alicia R Folgueras
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de Oviedo, 33006 Oviedo, Spain
| | - Carlos López-Otín
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de Oviedo, 33006 Oviedo, Spain; Centro de Investigación Biomédica en Red de Cáncer, Spain.
| |
Collapse
|
56
|
Aivazidis S, Coughlan CM, Rauniyar AK, Jiang H, Liggett LA, Maclean KN, Roede JR. The burden of trisomy 21 disrupts the proteostasis network in Down syndrome. PLoS One 2017; 12:e0176307. [PMID: 28430800 PMCID: PMC5400264 DOI: 10.1371/journal.pone.0176307] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Accepted: 04/07/2017] [Indexed: 12/27/2022] Open
Abstract
Down syndrome (DS) is a genetic disorder caused by trisomy of chromosome 21. Abnormalities in chromosome number have the potential to lead to disruption of the proteostasis network (PN) and accumulation of misfolded proteins. DS individuals suffer from several comorbidities, and we hypothesized that disruption of proteostasis could contribute to the observed pathology and decreased cell viability in DS. Our results confirm the presence of a disrupted PN in DS, as several of its elements, including the unfolded protein response, chaperone system, and proteasomal degradation exhibited significant alterations compared to euploid controls in both cell and mouse models. Additionally, when cell models were treated with compounds that promote disrupted proteostasis, we observed diminished levels of cell viability in DS compared to controls. Collectively our findings provide a cellular-level characterization of PN dysfunction in DS and an improved understanding of the potential pathogenic mechanisms contributing to disrupted cellular physiology in DS. Lastly, this study highlights the future potential of designing therapeutic strategies that mitigate protein quality control dysfunction.
Collapse
Affiliation(s)
- Stefanos Aivazidis
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, United States of America
| | - Christina M. Coughlan
- Department of Neurology, University of Colorado School of Medicine, Aurora, CO, United States of America
- The Linda Crnic Institute for Down Syndrome, University of Colorado, Aurora, CO, United States of America
| | - Abhishek K. Rauniyar
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, United States of America
| | - Hua Jiang
- The Linda Crnic Institute for Down Syndrome, University of Colorado, Aurora, CO, United States of America
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, United States of America
| | - L. Alexander Liggett
- The Linda Crnic Institute for Down Syndrome, University of Colorado, Aurora, CO, United States of America
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, United States of America
| | - Kenneth N. Maclean
- The Linda Crnic Institute for Down Syndrome, University of Colorado, Aurora, CO, United States of America
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, United States of America
| | - James R. Roede
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, United States of America
- * E-mail:
| |
Collapse
|
57
|
Endres K, Deller T. Regulation of Alpha-Secretase ADAM10 In vitro and In vivo: Genetic, Epigenetic, and Protein-Based Mechanisms. Front Mol Neurosci 2017; 10:56. [PMID: 28367112 PMCID: PMC5355436 DOI: 10.3389/fnmol.2017.00056] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 02/20/2017] [Indexed: 12/21/2022] Open
Abstract
ADAM10 (A Disintegrin and Metalloproteinase 10) has been identified as the major physiological alpha-secretase in neurons, responsible for cleaving APP in a non-amyloidogenic manner. This cleavage results in the production of a neuroprotective APP-derived fragment, APPs-alpha, and an attenuated production of neurotoxic A-beta peptides. An increase in ADAM10 activity shifts the balance of APP processing toward APPs-alpha and protects the brain from amyloid deposition and disease. Thus, increasing ADAM10 activity has been proposed an attractive target for the treatment of neurodegenerative diseases and it appears to be timely to investigate the physiological mechanisms regulating ADAM10 expression. Therefore, in this article, we will (1) review reports on the physiological regulation of ADAM10 at the transcriptional level, by epigenetic factors, miRNAs and/or protein interactions, (2) describe conditions, which change ADAM10 expression in vitro and in vivo, (3) report how neuronal ADAM10 expression may be regulated in humans, and (4) discuss how this knowledge on the physiological and pathophysiological regulation of ADAM10 may help to preserve or restore brain function.
Collapse
Affiliation(s)
- Kristina Endres
- Clinic of Psychiatry and Psychotherapy, University Medical Center Johannes Gutenberg-University Mainz Mainz, Germany
| | - Thomas Deller
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University Frankfurt/Main, Germany
| |
Collapse
|
58
|
Cissé M, Duplan E, Checler F. The transcription factor XBP1 in memory and cognition: Implications in Alzheimer disease. Mol Med 2017; 22:905-917. [PMID: 28079229 DOI: 10.2119/molmed.2016.00229] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 12/23/2016] [Indexed: 12/21/2022] Open
Abstract
X-box binding protein 1 (XBP1) is a unique basic region leucine zipper transcription factor isolated two decades ago in a search for regulators of major histocompatibility complex class II gene expression. XBP1 is a very complex protein regulating many physiological functions, including immune system, inflammatory responses, and lipid metabolism. Evidence over the past few years suggests that XBP1 also plays important roles in pathological settings since its activity as transcription factor has profound effects on the prognosis and progression of diseases such as cancer, neurodegeneration, and diabetes. Here we provide an overview on recent advances in our understanding of this multifaceted molecule, particularly in regulating synaptic plasticity and memory function, and the implications in neurodegenerative diseases with emphasis on Alzheimer disease.
Collapse
Affiliation(s)
- Moustapha Cissé
- Université Côte d'Azur, INSERM, CNRS, IPMC, team labeled "Fondation pour la Recherche Médicale" and "Laboratory of Excellence (LABEX) Distalz", 660 route des Lucioles, 06560, Sophia-Antipolis, Valbonne, France
| | - Eric Duplan
- Université Côte d'Azur, INSERM, CNRS, IPMC, team labeled "Fondation pour la Recherche Médicale" and "Laboratory of Excellence (LABEX) Distalz", 660 route des Lucioles, 06560, Sophia-Antipolis, Valbonne, France
| | - Frédéric Checler
- Université Côte d'Azur, INSERM, CNRS, IPMC, team labeled "Fondation pour la Recherche Médicale" and "Laboratory of Excellence (LABEX) Distalz", 660 route des Lucioles, 06560, Sophia-Antipolis, Valbonne, France
| |
Collapse
|
59
|
Identification of Phlogacantholide C as a Novel ADAM10 Enhancer from Traditional Chinese Medicinal Plants. MEDICINES 2016; 3:medicines3040030. [PMID: 28930140 PMCID: PMC5456242 DOI: 10.3390/medicines3040030] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 11/14/2016] [Accepted: 11/28/2016] [Indexed: 12/13/2022]
Abstract
Background: Alzheimer's disease is one of the most prevalent dementias in the elderly population with increasing numbers of patients. One pivotal hallmark of this disorder is the deposition of protein aggregates stemming from neurotoxic amyloid-beta peptides. Synthesis of those peptides has been efficiently prevented in AD model mice by activation of an enzyme called alpha-secretase. Therefore, drugs with the capability to increase the expression of this enzyme, named ADAM10, have been suggested as a valuable therapeutic medication. Methods: We investigated 69 substances from a drug library derived from traditional Chinese medicine by luciferase reporter assay in human neuronal cells for their potential to selectively induce alpha-secretase expression. Western blot analysis was used to confirm results on the protein level. Results: Ten of the 69 investigated compounds led to induction of ADAM10 transcriptional activity while BACE-1 (beta-site APP cleaving enzyme 1) and APP (amyloid precursor protein) expression were not induced. Two of them-Norkurarinol and Phlogacantholide C-showed substantial elevation of ADAM10 protein levels and Phlogacantholide C also increased secretion of the ADAM10-derived cleavage product APPs-alpha. Conclusion: Phlogacantholide C represents a novel ADAM10 gene expression enhancer from traditional Chinese medicinal herbs that may lay the groundwork for evolving potential novel therapeutics in Alzheimer's disease.
Collapse
|
60
|
Gerakis Y, Dunys J, Bauer C, Checler F. Aβ42 oligomers modulate β-secretase through an XBP-1s-dependent pathway involving HRD1. Sci Rep 2016; 6:37436. [PMID: 27853315 PMCID: PMC5112606 DOI: 10.1038/srep37436] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 10/28/2016] [Indexed: 01/18/2023] Open
Abstract
The aspartyl protease β-site APP cleaving enzyme, BACE1, is the rate-limiting enzyme involved in the production of amyloid-β peptide, which accumulates in both sporadic and familial cases of Alzheimer’s disease and is at the center of gravity of the amyloid cascade hypothesis. In this context, unravelling the molecular mechanisms controlling BACE1 expression and activity in both physiological and pathological conditions remains of major importance. We previously demonstrated that Aβ controlled BACE1 transcription in an NFκB-dependent manner. Here, we delineate an additional cellular pathway by which natural and synthetic Aβ42 oligomers enhance active X-box binding protein XBP-1s. XBP-1s lowers BACE1 expression and activity indirectly, via the up-regulation of the ubiquitin-ligase HRD1 that acts as an endogenous down-regulator of BACE1. Thus, we delineate a novel pathway by which cells could compensate for Aβ42 oligomers production and thus, associated toxicity, by triggering a compensatory mechanism aimed at lowering BACE-1-mediated Aβ production by a molecular cascade involving XBP-1s and HRD1. It thus identifies HRD1 as a potential target for a novel Aβ-centered therapeutic strategy.
Collapse
Affiliation(s)
- Yannis Gerakis
- Université Côte d'Azur, INSERM, CNRS, IPMC, France, Laboratory of excellence DistALZ, 660 route des Lucioles, 06560, Sophia-Antipolis, Valbonne, France
| | - Julie Dunys
- Université Côte d'Azur, INSERM, CNRS, IPMC, France, Laboratory of excellence DistALZ, 660 route des Lucioles, 06560, Sophia-Antipolis, Valbonne, France
| | - Charlotte Bauer
- Université Côte d'Azur, INSERM, CNRS, IPMC, France, Laboratory of excellence DistALZ, 660 route des Lucioles, 06560, Sophia-Antipolis, Valbonne, France
| | - Fréderic Checler
- Université Côte d'Azur, INSERM, CNRS, IPMC, France, Laboratory of excellence DistALZ, 660 route des Lucioles, 06560, Sophia-Antipolis, Valbonne, France
| |
Collapse
|
61
|
Anspach L, Unger RE, Gibson MI, Klok HA, Kirkpatrick CJ, Freese C. Impact of polymer-modified gold nanoparticles on brain endothelial cells: exclusion of endoplasmic reticulum stress as a potential risk factor. Nanotoxicology 2016; 10:1341-50. [PMID: 27492761 PMCID: PMC5166978 DOI: 10.1080/17435390.2016.1214761] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 04/18/2016] [Accepted: 06/13/2016] [Indexed: 12/17/2022]
Abstract
A library of polymer-coated gold nanoparticles (AuNPs) differing in size and surface modifications was examined for uptake and induction of cellular stress responses in the endoplasmic reticulum (ER stress) in human brain endothelial cells (hCMEC/D3). ER stress is known to affect the physiology of endothelial cells (ECs) and may lead to inflammation or apoptosis. Thus, even if applied at non-cytotoxic concentrations ER stress caused by nanoparticles should be prevented to reduce the risk of vascular diseases and negative effects on the integrity of barriers (e.g. blood-brain barrier). We exposed hCMEC/D3 to twelve different AuNPs (three sizes: 18, 35, and 65 nm, each with four surface-modifications) for various times and evaluated their effects on cytotoxicity, proinflammatory mediators, barrier functions and factors involved in ER stress. We demonstrated a time-dependent uptake of all AuNPs and no cytotoxicity for up to 72 h of exposure. Exposure to certain AuNPs resulted in a time-dependent increase in the proinflammatory markers IL-8, MCP-1, sVCAM, sICAM. However, none of the AuNPs induced an increase in expression of the chaperones and stress sensor proteins BiP and GRP94, respectively, or the transcription factors ATF4 and ATF6. Furthermore, no upregulation of the UPR stress sensor receptor PERK, no active splicing product of the transcription factor XBP1 and no upregulation of the transcription factor CHOP were detectable. In conclusion, the results of the present study indicate that effects of different-sized gold nanoparticles modified with various polymers were not related to the induction of ER stress in brain microvascular endothelial cells or led to apoptosis.
Collapse
Affiliation(s)
- Laura Anspach
- REPAIR-lab, Institute of Pathology, University Medical Center of the Johannes Gutenberg University Mainz and European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Mainz, Germany
| | - Ronald E. Unger
- REPAIR-lab, Institute of Pathology, University Medical Center of the Johannes Gutenberg University Mainz and European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Mainz, Germany
| | - Matthew I. Gibson
- University of Warwick, Department of Chemistry, Coventry, CV4 7AL, United Kingdom
| | - Harm-Anton Klok
- École Polytechnique Fédérale de Lausanne (EPFL), Institut des Matériaux and Institut des Sciences et Ingénierie Chimiques, Laboratoire des Polymères, Bâtiment MXD, Station 12, CH-1015 Lausanne, Switzerland
| | - C. James Kirkpatrick
- REPAIR-lab, Institute of Pathology, University Medical Center of the Johannes Gutenberg University Mainz and European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Mainz, Germany
| | - Christian Freese
- REPAIR-lab, Institute of Pathology, University Medical Center of the Johannes Gutenberg University Mainz and European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Mainz, Germany
| |
Collapse
|
62
|
Schuck F, Wolf D, Fellgiebel A, Endres K. Increase of α-Secretase ADAM10 in Platelets Along Cognitively Healthy Aging. J Alzheimers Dis 2016; 50:817-26. [PMID: 26757187 DOI: 10.3233/jad-150737] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
ADAM10 is one of the key players in ectodomain-shedding of the amyloid-β protein precursor (AβPP). Previous research with postmortem tissue has shown reduced expression and activity of ADAM10 within the central nervous system (CNS) of Alzheimer's disease (AD) patients. Determination of cerebral ADAM10 in living humans is hampered by its transmembrane property; only the physiological AβPP cleavage product generated by ADAM10, sAβPPα, can be assessed in cerebrospinal fluid. Establishment of surrogate markers in easily accessible material therefore is crucial. It has been demonstrated that ADAM10 is expressed in platelets and that platelet amount is decreased in AD patients. Just recently it has been shown that platelet ADAM10 and cognitive performance of AD patients positively correlate. In contrast to AD patients, to our knowledge almost no information has been published regarding ADAM10 expression during normal aging. We investigated ADAM10 amount and activity in platelets of cognitively healthy individuals from three different age groups ranging from 22-85 years. Interestingly, we observed an age-dependent increase in ADAM10 levels and activity in platelets.
Collapse
|
63
|
Bhoola NH, Kramvis A. Hepatitis B e Antigen Expression by Hepatitis B Virus Subgenotype A1 Relative to Subgenotypes A2 and D3 in Cultured Hepatocellular Carcinoma (Huh7) Cells. Intervirology 2016; 59:48-59. [PMID: 27553619 DOI: 10.1159/000446240] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 04/16/2016] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Hepatitis B virus (HBV) is hyperendemic in southern Africa, with subgenotype A1 prevailing. The precore/core (preC/C) region of A1, encoding for hepatitis B e antigen (HBeAg), has unique sequence characteristics, differentiating it from subgenotypes A2 and D3. Our aim was to follow the expression of HBeAg in vitro by the three subgenotypes. METHODS Huh7 cells were transfected with plasmids belonging to subgenotypes A1, A2, and D3. Using indirect immunofluorescence, the expression of HBeAg was followed, as was the activation of the unfolded protein response (UPR) and subsequent activation of apoptosis. RESULTS AND CONCLUSIONS Following transfection with D3, HBeAg passed through the secretory pathway earlier than cells transfected with genotype A. Cells transfected with A1 showed a lower expression of the preC/C precursor in the secretory pathway and a higher co-localization in the nucleus. Cells transfected with A1 showed greater endoplasmic reticulum (ER) stress and an earlier, prolonged activation of the UPR seen by the higher activity of three ER-localized transmembrane transducers (double-stranded RNA-dependent protein kinase-like ER kinase, activating transcription factor 6, and inositol-requiring enzyme 1), on day 3 compared to day 5. Moreover, our study also found that cells transfected with A1 had increased apoptosis.
Collapse
Affiliation(s)
- Nimisha Harshadrai Bhoola
- Hepatitis Virus Diversity Research Unit, Department of Internal Medicine, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | | |
Collapse
|
64
|
Vincent B. Regulation of the α-secretase ADAM10 at transcriptional, translational and post-translational levels. Brain Res Bull 2016; 126:154-169. [PMID: 27060611 DOI: 10.1016/j.brainresbull.2016.03.020] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 03/25/2016] [Accepted: 03/30/2016] [Indexed: 12/19/2022]
Abstract
A tremendous gain of interest in the biology of ADAM10 emerged during the past 15 years when it has first been shown that this protease was able to target the α-site of the β-amyloid precursor protein (βAPP) and later confirmed as the main physiological α-secretase activity. However, beside its well-established implication in the so-called non-amyloidogenic processing of βAPP and its probable protective role against Alzheimer's disease (AD), this metalloprotease also cleaves many other substrates, thereby being implicated in various physiological as well as pathological processes such as cancer and inflammation. Thus, in view of possible effective therapeutic interventions, a full comprehension of how ADAM10 is up and down regulated is required. This review discusses our current knowledge concerning the implication of this enzyme in AD as well as its more recently established roles in other brain disorders and provides a detailed up-date on its various transcriptional, translational and post-translational modulations.
Collapse
Affiliation(s)
- Bruno Vincent
- Mahidol University, Institute of Molecular Biosciences, Nakhon Pathom 73170, Thailand; Centre National de la Recherche Scientifique, 2 rue Michel Ange, 75016 Paris, France.
| |
Collapse
|
65
|
Ghani M, Reitz C, Cheng R, Vardarajan BN, Jun G, Sato C, Naj A, Rajbhandary R, Wang LS, Valladares O, Lin CF, Larson EB, Graff-Radford NR, Evans D, De Jager PL, Crane PK, Buxbaum JD, Murrell JR, Raj T, Ertekin-Taner N, Logue M, Baldwin CT, Green RC, Barnes LL, Cantwell LB, Fallin MD, Go RCP, Griffith PA, Obisesan TO, Manly JJ, Lunetta KL, Kamboh MI, Lopez OL, Bennett DA, Hendrie H, Hall KS, Goate AM, Byrd GS, Kukull WA, Foroud TM, Haines JL, Farrer LA, Pericak-Vance MA, Lee JH, Schellenberg GD, St George-Hyslop P, Mayeux R, Rogaeva E. Association of Long Runs of Homozygosity With Alzheimer Disease Among African American Individuals. JAMA Neurol 2016; 72:1313-23. [PMID: 26366463 DOI: 10.1001/jamaneurol.2015.1700] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
IMPORTANCE Mutations in known causal Alzheimer disease (AD) genes account for only 1% to 3% of patients and almost all are dominantly inherited. Recessive inheritance of complex phenotypes can be linked to long (>1-megabase [Mb]) runs of homozygosity (ROHs) detectable by single-nucleotide polymorphism (SNP) arrays. OBJECTIVE To evaluate the association between ROHs and AD in an African American population known to have a risk for AD up to 3 times higher than white individuals. DESIGN, SETTING, AND PARTICIPANTS Case-control study of a large African American data set previously genotyped on different genome-wide SNP arrays conducted from December 2013 to January 2015. Global and locus-based ROH measurements were analyzed using raw or imputed genotype data. We studied the raw genotypes from 2 case-control subsets grouped based on SNP array: Alzheimer's Disease Genetics Consortium data set (871 cases and 1620 control individuals) and Chicago Health and Aging Project-Indianapolis Ibadan Dementia Study data set (279 cases and 1367 control individuals). We then examined the entire data set using imputed genotypes from 1917 cases and 3858 control individuals. MAIN OUTCOMES AND MEASURES The ROHs larger than 1 Mb, 2 Mb, or 3 Mb were investigated separately for global burden evaluation, consensus regions, and gene-based analyses. RESULTS The African American cohort had a low degree of inbreeding (F ~ 0.006). In the Alzheimer's Disease Genetics Consortium data set, we detected a significantly higher proportion of cases with ROHs greater than 2 Mb (P = .004) or greater than 3 Mb (P = .02), as well as a significant 114-kilobase consensus region on chr4q31.3 (empirical P value 2 = .04; ROHs >2 Mb). In the Chicago Health and Aging Project-Indianapolis Ibadan Dementia Study data set, we identified a significant 202-kilobase consensus region on Chr15q24.1 (empirical P value 2 = .02; ROHs >1 Mb) and a cluster of 13 significant genes on Chr3p21.31 (empirical P value 2 = .03; ROHs >3 Mb). A total of 43 of 49 nominally significant genes common for both data sets also mapped to Chr3p21.31. Analyses of imputed SNP data from the entire data set confirmed the association of AD with global ROH measurements (12.38 ROHs >1 Mb in cases vs 12.11 in controls; 2.986 Mb average size of ROHs >2 Mb in cases vs 2.889 Mb in controls; and 22% of cases with ROHs >3 Mb vs 19% of controls) and a gene-cluster on Chr3p21.31 (empirical P value 2 = .006-.04; ROHs >3 Mb). Also, we detected a significant association between AD and CLDN17 (empirical P value 2 = .01; ROHs >1 Mb), encoding a protein from the Claudin family, members of which were previously suggested as AD biomarkers. CONCLUSIONS AND RELEVANCE To our knowledge, we discovered the first evidence of increased burden of ROHs among patients with AD from an outbred African American population, which could reflect either the cumulative effect of multiple ROHs to AD or the contribution of specific loci harboring recessive mutations and risk haplotypes in a subset of patients. Sequencing is required to uncover AD variants in these individuals.
Collapse
Affiliation(s)
- Mahdi Ghani
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
| | - Christiane Reitz
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, New York3Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, New York4
| | - Rong Cheng
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, New York
| | - Badri Narayan Vardarajan
- Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, New York
| | - Gyungah Jun
- Department of Medicine (Biomedical Genetics), Boston University, Boston, Massachusetts6Department of Biostatistics, Boston University, Boston, Massachusetts7Department of Ophthalmology, Boston University, Boston, Massachusetts
| | - Christine Sato
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
| | - Adam Naj
- The John P. Hussman Institute for Human Genomics, University of Miami, Miami, Florida
| | - Ruchita Rajbhandary
- The John P. Hussman Institute for Human Genomics, University of Miami, Miami, Florida
| | - Li-San Wang
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Otto Valladares
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Chiao-Feng Lin
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Eric B Larson
- Department of Medicine, University of Washington, Seattle11Group Health Research Institute, Group Health, Seattle, Washington
| | - Neill R Graff-Radford
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida13Department of Neurology, Mayo Clinic, Jacksonville, Florida
| | - Denis Evans
- Rush Institute for Healthy Aging, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois
| | - Philip L De Jager
- Program in Translational Neuropsychiatric Genomics, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts16Harvard Medical School, Boston, Massachusetts17Program in Medical and Population Genetics, The Broad Institute, Cambridge, Ma
| | - Paul K Crane
- Department of Medicine, University of Washington, Seattle
| | - Joseph D Buxbaum
- Department of Psychiatry, Mount Sinai School of Medicine, New York, New York19Department of Genetics and Genomics Sciences, Mount Sinai School of Medicine, New York, New York20Department of Neuroscience, Mount Sinai School of Medicine, New York, New York2
| | - Jill R Murrell
- Department of Medical and Molecular Genetics, Indiana University, Indianapolis
| | | | - Nilufer Ertekin-Taner
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida13Department of Neurology, Mayo Clinic, Jacksonville, Florida
| | - Mark Logue
- Department of Medicine (Biomedical Genetics), Boston University, Boston, Massachusetts
| | - Clinton T Baldwin
- Department of Medicine (Biomedical Genetics), Boston University, Boston, Massachusetts
| | - Robert C Green
- Harvard Medical School, Boston, Massachusetts23Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts24Partners Center for Personalized Genetic Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Lisa L Barnes
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois26Department of Behavioral Sciences, Rush University Medical Center, Chicago, Illinois
| | - Laura B Cantwell
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - M Daniele Fallin
- Department of Epidemiology, Johns Hopkins University School of Public Health, Baltimore, Maryland
| | - Rodney C P Go
- School of Public Health, University of Alabama at Birmingham
| | | | | | - Jennifer J Manly
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, New York4Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, New York
| | - Kathryn L Lunetta
- Department of Biostatistics, Boston University, Boston, Massachusetts
| | - M Ilyas Kamboh
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania32Alzheimer's Disease Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Oscar L Lopez
- Alzheimer's Disease Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - David A Bennett
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois33Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois
| | - Hugh Hendrie
- Indiana University Center for Aging Research, Indianapolis35Department of Psychiatry, Indiana University School of Medicine, Indianapolis36Regenstrief Institute Inc, Indianapolis, Indiana
| | - Kathleen S Hall
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis
| | - Alison M Goate
- Hope Center Program on Protein Aggregation and Neurodegeneration, Department of Psychiatry, Washington University School of Medicine, St Louis, Missouri
| | - Goldie S Byrd
- Department of Biology, North Carolina A & T University, Greensboro
| | - Walter A Kukull
- National Alzheimer's Coordinating Center, Department of Epidemiology, University of Washington, Seattle
| | - Tatiana M Foroud
- Department of Behavioral Sciences, Rush University Medical Center, Chicago, Illinois
| | - Jonathan L Haines
- Vanderbilt Center for Human Genetics Research, Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee
| | - Lindsay A Farrer
- Department of Medicine (Biomedical Genetics), Boston University, Boston, Massachusetts6Department of Biostatistics, Boston University, Boston, Massachusetts7Department of Ophthalmology, Boston University, Boston, Massachusetts41Department of Neurology, Bo
| | | | - Joseph H Lee
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, New York3Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, New York4
| | - Gerard D Schellenberg
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Peter St George-Hyslop
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
| | - Richard Mayeux
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, New York3Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, New York4
| | - Ekaterina Rogaeva
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
| | | |
Collapse
|
66
|
Increased susceptibility to metabolic dysregulation in a mouse model of Alzheimer's disease is associated with impaired hypothalamic insulin signaling and elevated BCAA levels. Alzheimers Dement 2016; 12:851-61. [PMID: 26928090 DOI: 10.1016/j.jalz.2016.01.008] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 01/20/2016] [Accepted: 01/26/2016] [Indexed: 01/21/2023]
Abstract
INTRODUCTION Epidemiologic studies have demonstrated an association between diabetes and dementia. Insulin signaling within the brain, in particular within the hypothalamus regulates carbohydrate, lipid, and branched chain amino acid (BCAA) metabolism in peripheral organs such as the liver and adipose tissue. We hypothesized that cerebral amyloidosis impairs central nervous system control of metabolism through disruption of insulin signaling in the hypothalamus, which dysregulates glucose and BCAA homeostasis resulting in increased susceptibility to diabetes. METHODS We examined whether APP/PS1 mice exhibit increased susceptibility to aging or high-fat diet (HFD)-induced metabolic impairment using metabolic phenotyping and insulin-signaling studies. RESULTS APP/PS1 mice were more susceptible to high-fat feeding and aging-induced metabolic dysregulation including disrupted BCAA homeostasis and exhibited impaired hypothalamic insulin signaling. DISCUSSION Our data suggest that AD pathology increases susceptibility to diabetes due to impaired hypothalamic insulin signaling, and that plasma BCAA levels could serve as a biomarker of hypothalamic insulin action in patients with AD.
Collapse
|
67
|
Turner RC, Lucke-Wold BP, Robson MJ, Lee JM, Bailes JE. Alzheimer's disease and chronic traumatic encephalopathy: Distinct but possibly overlapping disease entities. Brain Inj 2016; 30:1279-1292. [PMID: 27715315 PMCID: PMC5303562 DOI: 10.1080/02699052.2016.1193631] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) and chronic traumatic encephalopathy (CTE) have long been recognized as sharing some similar neuropathological features, mainly the presence of neurofibrilary tangles and hyperphosphorylated tau, but have generally been described as distinct entities. Evidence indicates that neurotrauma increases the risk of developing dementia and accelerates the progression of disease. Findings are emerging that CTE and AD may be present in the same patients. CLINICAL PRESENTATION This study presents a series of previously unpublished cases, with one case demonstrating possible neurotrauma-related AD, one pure CTE, and an example of a case exhibiting features of both AD and CTE. The future significance of this work lies not only in the confirmation of AD-CTE co-existence, but, more importantly, ways of generating a hypothesis about the possibility that CTE may accelerate AD development. Understanding the relationship between neurotrauma and neurodegenerative disease will help elucidate how distinct disease entities can co-exist in the same patient. It will ultimately require the use of pre-clinical animal models and repeat injury paradigms to investigate clinically relevant injury mechanisms. These models should produce a CTE-like phenotype that must be both neuropathologically and behaviourally similar to human disease. CONCLUSION This case series and review of the literature presents a discussion of AD and CTE in the context of neurotrauma. It highlights recent work from repetitive neurotrauma models with an emphasis on those exhibiting a CTE-like phenotype. Potential mechanisms of interest shared amongst AD and CTE are briefly addressed and future experiments are advocated for to enhance understanding of CTE pathophysiology and the relationship between CTE and AD.
Collapse
Affiliation(s)
- Ryan C. Turner
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV 26506
- Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506
| | - Brandon P. Lucke-Wold
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV 26506
- Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506
| | - Matthew J. Robson
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - John M. Lee
- NorthShore Neurological Institute, NorthShore University Health System, Evanston, IL 60201
| | - Julian E. Bailes
- NorthShore Neurological Institute, NorthShore University Health System, Evanston, IL 60201
| |
Collapse
|
68
|
The alpha secretase ADAM10: A metalloprotease with multiple functions in the brain. Prog Neurobiol 2015; 135:1-20. [PMID: 26522965 DOI: 10.1016/j.pneurobio.2015.10.003] [Citation(s) in RCA: 158] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 09/23/2015] [Accepted: 10/26/2015] [Indexed: 01/07/2023]
Abstract
Proteins belonging to the 'A Disintegrin And Metalloproteinase' (ADAM) family are membrane-anchored proteases that are able to cleave the extracellular domains of several membrane-bound proteins in a process known as 'ectodomain shedding'. In the central nervous system, ADAM10 has attracted the most attention, since it was described as the amyloid precursor protein α-secretase over ten years ago. Despite the excitement over the potential of ADAM10 as a novel drug target in Alzheimer disease, the physiological functions of ADAM10 in the brain are not yet well understood. This is largely because of the embryonic lethality of ADAM10-deficient mice, which results from the loss of cleavage and signaling of the Notch receptor, another ADAM10 substrate. However, the recent generation of conditional ADAM10-deficient mice and the identification of further ADAM10 substrates in the brain has revealed surprisingly numerous and fundamental functions of ADAM10 in the development of the embryonic brain and also in the homeostasis of adult neuronal networks. Mechanistically, ADAM10 controls these functions by utilizing unique postsynaptic substrates in the central nervous system, in particular synaptic cell adhesion molecules, such as neuroligin-1, N-cadherin, NCAM, Ephrin A2 and A5. Consequently, a dysregulation of ADAM10 activity is linked to psychiatric and neurological diseases, such as epilepsy, fragile X syndrome and Huntington disease. This review highlights the recent progress in understanding the substrates and function as well as the regulation and cell biology of ADAM10 in the central nervous system and discusses the value of ADAM10 as a drug target in brain diseases.
Collapse
|
69
|
Schuck F, Schmitt U, Reinhardt S, Freese C, Lee IS, Thines E, Efferth T, Endres K. Extract of Caragana sinica as a potential therapeutic option for increasing alpha-secretase gene expression. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2015; 22:1027-1036. [PMID: 26407945 DOI: 10.1016/j.phymed.2015.08.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 07/28/2015] [Accepted: 08/01/2015] [Indexed: 06/05/2023]
Abstract
BACKGROUND Alzheimer's disease represents one of the main neurological disorders in the aging population. Treatment options so far are only of symptomatic nature and efforts in developing disease modifying drugs by targeting amyloid beta peptide-generating enzymes remain fruitless in the majority of human studies. During the last years, an alternative approach emerged to target the physiological alpha-secretase ADAM10, which is not only able to prevent formation of toxic amyloid beta peptides but also provides a neuroprotective fragment of the amyloid precursor protein - sAPPalpha. PURPOSE To identify novel alpha-secretase enhancers from a library of 313 extracts of medicinal plants indigenous to Korea, a screening approach was used and hits were further evaluated for their therapeutic value. METHODS The extract library was screened for selective enhancers of ADAM10 gene expression using a luciferase-based promoter reporter gene assay in the human neuroblastoma cell line SH-SY5Y. Candidate extracts were then tested in wild type mice for acute behavioral effects using an open field paradigm. Brain and liver tissue from treated mice was biochemically analyzed for ADAM10 gene expression in vivo. An in vitro blood-brain barrier model and an in vitro ATPase assay were used to unravel transport properties of bioactive compounds from extract candidates. Finally, fractionation of the most promising extract was performed to identify biologically active components. RESULTS The extract of Caragana sinica (Buc'hoz) Rehder was identified as the best candidate from our screening approach. We were able to demonstrate that the extract is acutely applicable in mice without obvious side effects and induces ADAM10 gene expression in peripheral tissue. A hindered passage across the blood-brain barrier was detected explaining lack of cerebral induction of ADAM10 gene expression in treated mice. By fractionating C. sinica extract we identified alpha-viniferin as one of the biologically active components. CONCLUSION The extract of C. sinica and alpha-viniferin as one of its bioactive constituents might serve as novel therapeutic options for treating Alzheimer's disease by increasing ADAM10 gene expression. The identification of alpha-viniferin represents a promising starting point to achieve blood-brain barrier penetrance in the future.
Collapse
Affiliation(s)
- Florian Schuck
- Department of Psychiatry and Psychotherapy, University Medical Center of the Johannes Gutenberg University, Untere Zahlbacher Strasse 8, 55131 Mainz, Germany
| | - Ulrich Schmitt
- Department of Psychiatry and Psychotherapy, University Medical Center of the Johannes Gutenberg University, Untere Zahlbacher Strasse 8, 55131 Mainz, Germany
| | - Sven Reinhardt
- Department of Psychiatry and Psychotherapy, University Medical Center of the Johannes Gutenberg University, Untere Zahlbacher Strasse 8, 55131 Mainz, Germany
| | - Christian Freese
- REPAIR-lab, Institute of Pathology, University Medical Center of the Johannes Gutenberg University and European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Langenbeckstrasse 1, 55101 Mainz, Germany
| | - Ik-Soo Lee
- College of Pharmacy, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 500-757, South Korea
| | - Eckhard Thines
- Institute of Biotechnology and Drug Research, Johannes Gutenberg University, Duesbergweg 10-14, 55128 Mainz, Germany
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany
| | - Kristina Endres
- Department of Psychiatry and Psychotherapy, University Medical Center of the Johannes Gutenberg University, Untere Zahlbacher Strasse 8, 55131 Mainz, Germany.
| |
Collapse
|
70
|
Manzine PR, Marcello E, Borroni B, Kamphuis W, Hol E, Padovani A, Nascimento CC, de Godoy Bueno P, Assis Carvalho Vale F, Iost Pavarini SC, Di Luca M, Cominetti MR. ADAM10 gene expression in the blood cells of Alzheimer's disease patients and mild cognitive impairment subjects. Biomarkers 2015. [DOI: 10.3109/1354750x.2015.1062554] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
71
|
Lancellotti S, Dragani A, Ranalli P, Petrucci G, Basso M, Tartaglione R, Rocca B, De Cristofaro R. Qualitative and quantitative modifications of von Willebrand factor in patients with essential thrombocythemia and controlled platelet count. J Thromb Haemost 2015; 13:1226-37. [PMID: 25876231 DOI: 10.1111/jth.12967] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Indexed: 12/23/2022]
Abstract
BACKGROUND Essential thrombocythemia (ET) is characterized by increased platelets and prevalent thrombosis. An acquired von Willebrand factor (VWF) disease has been hypothesized and inconsistently associated with extreme thrombocytosis or rare bleeding in ET. Whether VWF is modified in ET patients with controlled platelet count remains unclear. OBJECTIVES We studied different VWF- and platelet-associated parameters in ET patients treated according to current recommendations. PATIENTS/METHODS Sixty-nine ET patients (M = 29; median age, 62 [48-70] years; platelets, 432 [337-620] × 10(3) μL(-1) ), 69 matched controls and 10 subjects with reactive thrombocytosis (RT) were studied. VWF:antigen (Ag), activity (act), electrophoretic patterns, VWF:propeptide, plasma glycocalycin (GC), glycoproteinV (GpV), ADAMTS-13, elastase, C-reactive protein and serum thromboxane (TX)B2 were measured. RESULTS In ET patients, VWF:Ag was increased by 31 ± 13% vs. controls (P < 0.01), without dependence of blood groups, while VWF:act was reduced by 21 ± 12% vs. controls and by 50 ± 24% vs. RT (P < 0.01). The VWF:act/VWF:Ag ratios in ET were reduced by 35 ± 17% vs. controls and RT patients (P < 0.001) and significantly associated with: immature or total platelet counts, GC, GpV and TXB2 . In multivariable analysis, only GC inversely predicted ET patients' VWF:act/VWF:Ag ratios (β = -0.42, P = 0.01). By electrophoresis analyses, high-molecular-weight VWF multimers were variably reduced with atypical cleavage bands in ET only. VWF:propeptide, ADAMTS-13 and elastase levels were normal in ET patients. Platelet-associated ADAM-10 and ADAM-17 hydrolyzed VWFm in vitro, showing patterns similar to those in ET samples. CONCLUSIONS In ET patients with controlled platelet counts, the VWF:act/VWF:Ag ratio is decreased and predicted by GC, a product of platelet activation. ADAM-10 and/or ADAM-17 might be involved. In vivo platelet activation, which characterizes ET, might contribute to disease-specific VWF alterations.
Collapse
Affiliation(s)
- S Lancellotti
- Center for Haemorrhagic and Thrombotic Diseases, Department of Medical Sciences, Catholic University School of Medicine, 'A. Gemelli' Hospital, Rome, Italy
| | - A Dragani
- Center for Haemorrhagic, Thrombotic and Rare Hematologic Diseases, Spirito Santo Hospital, Pescara, Italy
| | - P Ranalli
- Center for Haemorrhagic, Thrombotic and Rare Hematologic Diseases, Spirito Santo Hospital, Pescara, Italy
| | - G Petrucci
- Institute of Pharmacology, Catholic University School of Medicine, Rome, Italy
| | - M Basso
- Center for Haemorrhagic and Thrombotic Diseases, Department of Medical Sciences, Catholic University School of Medicine, 'A. Gemelli' Hospital, Rome, Italy
| | - R Tartaglione
- Institute of Haematology, Complesso Integrato Columbus, Catholic University School of Medicine, Rome, Italy
| | - B Rocca
- Institute of Pharmacology, Catholic University School of Medicine, Rome, Italy
| | - R De Cristofaro
- Center for Haemorrhagic and Thrombotic Diseases, Department of Medical Sciences, Catholic University School of Medicine, 'A. Gemelli' Hospital, Rome, Italy
| |
Collapse
|
72
|
Tanaka Y, Ishitsuka Y, Hayasaka M, Yamada Y, Miyata K, Endo M, Kondo Y, Moriuchi H, Irikura M, Tanaka KI, Mizushima T, Oike Y, Irie T. The exacerbating roles of CCAAT/enhancer-binding protein homologous protein (CHOP) in the development of bleomycin-induced pulmonary fibrosis and the preventive effects of tauroursodeoxycholic acid (TUDCA) against pulmonary fibrosis in mice. Pharmacol Res 2015; 99:52-62. [PMID: 26005208 DOI: 10.1016/j.phrs.2015.05.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 04/30/2015] [Accepted: 05/14/2015] [Indexed: 12/16/2022]
Abstract
The purpose of this study was to evaluate the role of CCAAT/enhancer-binding protein homologous protein (CHOP), an important transcription factor that regulates the inflammatory reaction during the endoplasmic reticulum (ER) stress response, in the development of pulmonary fibrosis induced by bleomycin (BLM) in mice. An intratracheal injection of BLM transiently increased the expression of CHOP mRNA and protein in an early phase (days 1 and 3) in mice lungs. BLM-induced pulmonary fibrosis was significantly attenuated in Chop gene deficient (Chop KO) mice, compared with wild-type (WT) mice. Furthermore, the inflammatory reactions evaluated by protein concentration, the total number of leucocytes and neutrophils in the bronchoalveolar lavage fluid (BALF), the mRNA expression of interleukin 1b and caspase 11, and the apoptotic cell death were suppressed in Chop KO mice compared with those in WT mice. In addition, administration of tauroursodeoxycholic acid (TUDCA), a pharmacological agent that can inhibit CHOP expression, inhibited the BLM-induced pulmonary fibrosis and inflammation, and the increase in Chop mRNA expression in WT mice in a dose-dependent manner. These results suggest that the ER stress-induced transcription factor, CHOP, at least in part, plays an important role in the development of BLM-induced pulmonary fibrosis in mice, and that the inhibition of CHOP expression by a pharmacological agent, such as TUDCA, may be a promising strategy for the prevention of pulmonary fibrosis.
Collapse
Affiliation(s)
- Yuta Tanaka
- Department of Clinical Chemistry and Informatics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Yoichi Ishitsuka
- Department of Clinical Chemistry and Informatics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan.
| | - Marina Hayasaka
- Department of Clinical Chemistry and Informatics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Yusei Yamada
- Department of Clinical Chemistry and Informatics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Keishi Miyata
- Department of Molecular Genetics, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556, Japan
| | - Motoyoshi Endo
- Department of Molecular Genetics, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556, Japan
| | - Yuki Kondo
- Department of Clinical Chemistry and Informatics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Hiroshi Moriuchi
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Science, Sojo University, 4-22-1 Ikeda, Kumamoto 860-0082, Japan
| | - Mitsuru Irikura
- Laboratory of Evidence-Based Pharmacotherapy, College of Pharmaceutical Sciences, Daiichi University, 22-1 Tamagawa-Cho, Minami-Ku, Fukuoka 815-8511, Japan
| | - Ken-ichiro Tanaka
- Department of Analytical Chemistry, Faculty of Pharmacy, Keio University, Tokyo 105-8512, Japan
| | - Tohru Mizushima
- Department of Analytical Chemistry, Faculty of Pharmacy, Keio University, Tokyo 105-8512, Japan
| | - Yuichi Oike
- Department of Molecular Genetics, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556, Japan
| | - Tetsumi Irie
- Department of Clinical Chemistry and Informatics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan; Center for Clinical Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| |
Collapse
|
73
|
Saftig P, Bovolenta P. Proteases at work: cues for understanding neural development and degeneration. Front Mol Neurosci 2015; 8:13. [PMID: 25999813 PMCID: PMC4419857 DOI: 10.3389/fnmol.2015.00013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 04/21/2015] [Indexed: 12/19/2022] Open
Abstract
Proteolytical processing of membrane bound molecules is a fundamental mechanism for the degradation of these proteins as well as for controlling cell-to-cell communication, which is at the basis of tissue development and homeostasis. Members of families of metalloproteinases and intra-membrane proteases are major effectors of these events. A recent workshop in Baeza, Spain, was devoted to discuss how this mechanism coordinates brain development and how its dysfunction leads to brain pathologies. Herein we summarize the findings presented during this workshop, which illuminate the role of metalloproteinases, including matrix metalloproteinase, A Disintegrin and Metalloproteinase-proteases and intra-membrane proteases, in the regulation of neurogenesis, axon guidance, and synaptogenesis as well as in neurodegeneration. Indeed, there is increasing evidence that proteolysis at the membrane is directly linked to neuropathologies such as Alzheimer Disease and autism spectrum or prion disorders. These proteolytic events are tightly regulated and we are just at the beginning of understanding how these processes could be exploited to design therapeutic treatments aimed at alleviating psychiatric and neurodegenerative pathologies.
Collapse
Affiliation(s)
- Paul Saftig
- Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, Kiel Germany
| | - Paola Bovolenta
- Centro de Biología Molecular "Severo Ochoa", Consejo Superior de Investigaciones Científicas - Universidad Autónoma de Madrid, Madrid Spain ; Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid Spain
| |
Collapse
|
74
|
Kim S, Joe Y, Kim HJ, Kim YS, Jeong SO, Pae HO, Ryter SW, Surh YJ, Chung HT. Endoplasmic reticulum stress-induced IRE1α activation mediates cross-talk of GSK-3β and XBP-1 to regulate inflammatory cytokine production. THE JOURNAL OF IMMUNOLOGY 2015; 194:4498-506. [PMID: 25821218 DOI: 10.4049/jimmunol.1401399] [Citation(s) in RCA: 110] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 02/24/2015] [Indexed: 12/22/2022]
Abstract
IL-1β and TNF-α are important proinflammatory cytokines that respond to mutated self-antigens of tissue damage and exogenous pathogens. The endoplasmic reticulum (ER) stress and unfolded protein responses are related to the induction of proinflammatory cytokines. However, the detailed molecular pathways by which ER stress mediates cytokine gene expression have not been investigated. In this study, we found that ER stress-induced inositol-requiring enzyme (IRE)1α activation differentially regulates proinflammatory cytokine gene expression via activation of glycogen synthase kinase (GSK)-3β and X-box binding protein (XBP)-1. Surprisingly, IL-1β gene expression was modulated by IRE1α-mediated GSK-3β activation, but not by XBP-1. However, IRE1α-mediated XBP-1 splicing regulated TNF-α gene expression. SB216763, a GSK-3 inhibitor, selectively inhibited IL-1β gene expression, whereas the IRE1α RNase inhibitor STF083010 suppressed only TNF-α production. Additionally, inhibition of GSK-3β greatly increased IRE1α-dependent XBP-1 splicing. Our results identify an unsuspected differential role of downstream mediators GSK-3β and XBP-1 in ER stress-induced IRE1α activation that regulates cytokine production through signaling cross-talk. These results have important implications in the regulation of inflammatory pathways during ER stress, and they suggest novel therapeutic targets for diseases in which meta-inflammation plays a key role.
Collapse
Affiliation(s)
- Sena Kim
- School of Biological Sciences, University of Ulsan, Ulsan 680-749, Korea
| | - Yeonsoo Joe
- School of Biological Sciences, University of Ulsan, Ulsan 680-749, Korea
| | - Hyo Jeong Kim
- School of Biological Sciences, University of Ulsan, Ulsan 680-749, Korea
| | - You-Sun Kim
- Department of Biochemistry, Ajou University School of Medicine, Graduate School, Ajou University, Suwon 443-721, Korea; Department of Biomedical Sciences, Graduate School, Ajou University, Suwon 443-721, Korea
| | - Sun Oh Jeong
- Department of Anesthesiology and Pain Medicine, Wonkwang University School of Medicine, Iksan 570-749, Korea
| | - Hyun-Ock Pae
- Department of Anesthesiology and Pain Medicine, Wonkwang University School of Medicine, Iksan 570-749, Korea
| | - Stefan W Ryter
- Department of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115; Joan and Sanford I. Weill Department of Medicine, New York-Presbyterian Hospital, Weill Cornell Medical College, New York, NY 10065; and
| | - Young-Joon Surh
- College of Pharmacy, Seoul National University, Seoul 151-742, Korea
| | - Hun Taeg Chung
- School of Biological Sciences, University of Ulsan, Ulsan 680-749, Korea;
| |
Collapse
|
75
|
Hosoi T, Nomura J, Ozawa K, Nishi A, Nomura Y. Possible involvement of endoplasmic reticulum stress in the pathogenesis of Alzheimer’s disease. ENDOPLASMIC RETICULUM STRESS IN DISEASES 2015. [DOI: 10.1515/ersc-2015-0008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
AbstractThe endoplasmic reticulum (ER) is an organelle that plays a crucial role in protein quality control such as protein folding. Evidence to indicate the involvement of ER in maintaining cellular homeostasis is increasing. However, when cells are exposed to stressful conditions, which perturb ER function, unfolded proteins accumulate leading to ER stress. Cells then activate the unfolded protein response (UPR) to cope with this stressful condition. In the present review, we will discuss and summarize recent advances in research on the basic mechanisms of the UPR. We also discuss the possible involvement of ER stress in the pathogenesis of Alzheimer’s disease (AD). Potential therapeutic opportunities for diseases targeting ER stress is also described.
Collapse
|
76
|
Huang HC, Tang D, Lu SY, Jiang ZF. Endoplasmic reticulum stress as a novel neuronal mediator in Alzheimer's disease. Neurol Res 2014; 37:366-74. [PMID: 25310352 DOI: 10.1179/1743132814y.0000000448] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Alzheimer's disease (AD) is one of the most common types of progressive dementias. The typical neuropathological changes in AD include extracellular senile plaques, intracellular neurofibrillary tangles, and loss of neurons. The pathogenetic mechanism of this disease is not comprehensively understood yet. Recently, endoplasmic reticulum stress (ER stress) has been considered as a potential event involved in AD development. Some AD-related factors, such as misfolded protein and Ca(2+) depletion, could disrupt the homeostasis of ER lumen. In AD, the aggregated amyloid-beta peptide (Abeta) could induce ER stress in an assembly dependent way. The presenilin has been identified as a Ca(2+) channel. Mutations of presenilin could change the balance of Ca(2+) in ER lumen and thus disrupts the ER homeostasis. Furthermore, the ER stress could lead to cellular disorders like inflammation. Through activating the expression of inflammatory factors, ER stress triggers inflammatory response in AD pathology. Herein, we reviewed the recent progress of ER stress-induced unfolded protein response (UPR) and the roles of ER stress in AD pathological process.
Collapse
|
77
|
Abstract
PURPOSE OF REVIEW Genome-wide association studies have been used as an unbiased tool to identify novel genes that contribute to variations in LDL cholesterol levels in the hopes of uncovering new biology and new therapeutic targets for the treatment of atherosclerotic cardiovascular disease. The locus identified by genome-wide association studies with the strongest association with LDL cholesterol and atherosclerotic cardiovascular disease is the 1p13 sortilin-1 (SORT1) locus. Here, we review the identification and characterization of this locus, the initial physiological studies describing the role of SORT1 in lipoprotein metabolism, and recent work that has begun to sort out the complexity of this role. RECENT FINDINGS Studies by several groups support an important role for sortilin in lipoprotein metabolism; however, the directionality of the effect of sortilin on plasma cholesterol and its role in the secretion of hepatic lipoproteins remains controversial. Studies by several groups support a role for sortilin in inhibiting lipoprotein export, whereas other studies suggest that sortilin facilitates lipoprotein export. SUMMARY Understanding the mechanism by which sortilin affects LDL cholesterol will increase our understanding of the regulation of lipoprotein metabolism and hepatic lipoprotein export and may also allow us to harness the power of the 1p13 SORT1 locus for the treatment of atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Alanna Strong
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | | |
Collapse
|
78
|
Dunys J, Duplan E, Checler F. The transcription factor X-box binding protein-1 in neurodegenerative diseases. Mol Neurodegener 2014; 9:35. [PMID: 25216759 PMCID: PMC4166022 DOI: 10.1186/1750-1326-9-35] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 08/14/2014] [Indexed: 12/12/2022] Open
Abstract
Endoplasmic reticulum (ER) is the cellular compartment where secreted and integral membrane proteins are folded and matured. The accumulation of unfolded or misfolded proteins triggers a stress that is physiologically controlled by an adaptative protective response called Unfolded Protein Response (UPR). UPR is primordial to induce a quality control response and to restore ER homeostasis. When this adaptative response is defective, protein aggregates overwhelm cells and affect, among other mechanisms, synaptic function, signaling transduction and cell survival. Such dysfunction likely contributes to several neurodegenerative diseases that are indeed characterized by exacerbated protein aggregation, protein folding impairment, increased ER stress and UPR activation. This review briefly documents various aspects of the biology of the transcription factor XBP-1 (X-box Binding Protein-1) and summarizes recent findings concerning its putative contribution to the altered UPR response observed in various neurodegenerative disorders including Parkinson’s and Alzheimer’s diseases.
Collapse
Affiliation(s)
| | | | - Frédéric Checler
- Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275 CNRS-UNS, Sophia Antipolis, Nice, Valbonne F-06560, France.
| |
Collapse
|
79
|
Hetz C, Mollereau B. Disturbance of endoplasmic reticulum proteostasis in neurodegenerative diseases. Nat Rev Neurosci 2014; 15:233-49. [PMID: 24619348 DOI: 10.1038/nrn3689] [Citation(s) in RCA: 513] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The unfolded protein response (UPR) is a homeostatic mechanism by which cells regulate levels of misfolded proteins in the endoplasmic reticulum (ER). Although it is well characterized in non-neuronal cells, a proliferation of papers over the past few years has revealed a key role for the UPR in normal neuronal function and as an important driver of neurodegenerative diseases. A complex scenario is emerging in which distinct UPR signalling modules have specific and even opposite effects on neurodegeneration depending on the disease context. Here, we provide an overview of the most recent findings addressing the biological relevance of ER stress in the nervous system.
Collapse
Affiliation(s)
- Claudio Hetz
- 1] Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile. [2] Institute of Biomedical Sciences, Center for Molecular Studies of the Cell, Program of Cellular and Molecular Biology, University of Chile, Santiago, Chile. [3] Neurounion Biomedical Foundation, Santiago, Chile. [4] Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, Massachusetts 02115, USA
| | - Bertrand Mollereau
- Laboratory of Molecular Biology of the Cell, CNRS UMR5239, Ecole Normale Supérieure de Lyon, UMS3444 Biosciences Lyon Gerland, University of Lyon, Lyon 69364, France
| |
Collapse
|
80
|
Duran-Aniotz C, Martínez G, Hetz C. Memory loss in Alzheimer's disease: are the alterations in the UPR network involved in the cognitive impairment? Front Aging Neurosci 2014; 6:8. [PMID: 24523695 PMCID: PMC3906588 DOI: 10.3389/fnagi.2014.00008] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Accepted: 01/12/2014] [Indexed: 11/13/2022] Open
Affiliation(s)
- Claudia Duran-Aniotz
- Faculty of Medicine, Biomedical Neuroscience Institute, University of Chile Santiago, Chile ; Program of Cellular and Molecular Biology, Center for Molecular Studies of the Cell, Institute of Biomedical Sciences, University of Chile Santiago, Chile
| | - Gabriela Martínez
- Faculty of Medicine, Biomedical Neuroscience Institute, University of Chile Santiago, Chile ; Program of Cellular and Molecular Biology, Center for Molecular Studies of the Cell, Institute of Biomedical Sciences, University of Chile Santiago, Chile
| | - Claudio Hetz
- Faculty of Medicine, Biomedical Neuroscience Institute, University of Chile Santiago, Chile ; Program of Cellular and Molecular Biology, Center for Molecular Studies of the Cell, Institute of Biomedical Sciences, University of Chile Santiago, Chile ; Neurounion Biomedical Foundation Santiago, Chile ; Department of Immunology and Infectious diseases, Harvard School of Public Health Boston, MA, USA
| |
Collapse
|