101
|
Louessard M, Bardou I, Lemarchand E, Thiebaut AM, Parcq J, Leprince J, Terrisse A, Carraro V, Fafournoux P, Bruhat A, Orset C, Vivien D, Ali C, Roussel BD. Activation of cell surface GRP78 decreases endoplasmic reticulum stress and neuronal death. Cell Death Differ 2017. [PMID: 28644439 DOI: 10.1038/cdd.2017.35] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The unfolded protein response (UPR) is an endoplasmic reticulum (ER) -related stress conserved pathway that aims to protect cells from being overwhelmed. However, when prolonged, UPR activation converts to a death signal, which relies on its PERK-eIF2α branch. Overactivation of the UPR has been implicated in many neurological diseases, including cerebral ischaemia. Here, by using an in vivo thromboembolic model of stroke on transgenic ER stress-reporter mice and neuronal in vitro models of ischaemia, we demonstrate that ischaemic stress leads to the deleterious activation of the PERK branch of the UPR. Moreover, we show that the serine protease tissue-type plasminogen activator (tPA) can bind to cell surface Grp78 (78 kD glucose-regulated protein), leading to a decrease of the PERK pathway activation, thus a decrease of the deleterious factor CHOP, and finally promotes neuroprotection. Altogether, this work highlights a new role and a therapeutic potential of the chaperone protein Grp78 as a membrane receptor of tPA capable to prevent from ER stress overactivation.
Collapse
Affiliation(s)
- Morgane Louessard
- Normandie Univ, UNICAEN, INSERM, Physiopathology and Imaging of Neurological Disorders, Caen, France
| | - Isabelle Bardou
- Normandie Univ, UNICAEN, INSERM, Physiopathology and Imaging of Neurological Disorders, Caen, France
| | - Eloïse Lemarchand
- Normandie Univ, UNICAEN, INSERM, Physiopathology and Imaging of Neurological Disorders, Caen, France
| | - Audrey M Thiebaut
- Normandie Univ, UNICAEN, INSERM, Physiopathology and Imaging of Neurological Disorders, Caen, France
| | - Jérôme Parcq
- Normandie Univ, UNICAEN, INSERM, Physiopathology and Imaging of Neurological Disorders, Caen, France
| | - Jérôme Leprince
- Normandie Univ, UNIROUEN, INSERM, Laboratoire Différenciation et Communication Neuronale et Neuroendocrine, Plate-forme de Recherche en Imagerie Cellulaire de Normandie (PRIMACEN), Rouen, France
| | - Anne Terrisse
- INRA, UMR 1019 Nutrition Humaine, Centre de Clermont-Ferrand-Theix, Saint Genès Champanelle, France
| | - Valérie Carraro
- INRA, UMR 1019 Nutrition Humaine, Centre de Clermont-Ferrand-Theix, Saint Genès Champanelle, France
| | - Pierre Fafournoux
- INRA, UMR 1019 Nutrition Humaine, Centre de Clermont-Ferrand-Theix, Saint Genès Champanelle, France
| | - Alain Bruhat
- INRA, UMR 1019 Nutrition Humaine, Centre de Clermont-Ferrand-Theix, Saint Genès Champanelle, France
| | - Cyrille Orset
- Normandie Univ, UNICAEN, INSERM, Physiopathology and Imaging of Neurological Disorders, Caen, France
| | - Denis Vivien
- Normandie Univ, UNICAEN, INSERM, Physiopathology and Imaging of Neurological Disorders, Caen, France.,Clinical Research Department, Medical Center, University Caen Normandy, Centre Hospitalo-Universitaire Caen Côte de Nacre, Caen, France
| | - Carine Ali
- Normandie Univ, UNICAEN, INSERM, Physiopathology and Imaging of Neurological Disorders, Caen, France
| | - Benoit D Roussel
- Normandie Univ, UNICAEN, INSERM, Physiopathology and Imaging of Neurological Disorders, Caen, France
| |
Collapse
|
102
|
Abstract
Macrophages are the main immune-competent cells that infiltrate in tumors. Tumor-associated macrophages (TAMs), termed M2 macrophages, facilitate tumor progress and promote metastasis. However, M2 macrophages always display an immunosuppressive phenotype, which is not in accordance with the tumor inflammatory microenvironment and inflammation-related metastasis. In this study, we established a macrophage polarization model with human monocytes and found that the conditioned medium from M2 macrophages increased GRP78 expression in tumor cells and facilitated tumor cell migration. Mechanistically, excessive GRP78 formed a protein complex with STAT3 and JAK2 to promote STAT3 phosphorylation. Furthermore, p-STAT3 facilitated the high expression of inflammatory factors IL-1β and TNF-α in tumor cells, which was important in M2 macrophage-induced metastasis. The present data demonstrate that M2 macrophages elevate tumor cell GRP78 expression to trigger an inflammatory response, which further facilitates tumor metastasis. Therefore, our study not only uncovered a new cause of GRP78 overexpression in tumor cell, but also, explained the antinomy of TAMs immunosuppressive properties and inflammation-related tumor metastasis.
Collapse
|
103
|
Yip CW, Lam CY, Poon TCW, Cheung TT, Cheung PFY, Fung SW, Wang XQ, Leung ICY, Ng LWC, Lo CM, Tsao GSW, Cheung ST. Granulin-epithelin precursor interacts with 78-kDa glucose-regulated protein in hepatocellular carcinoma. BMC Cancer 2017; 17:409. [PMID: 28601093 PMCID: PMC5466756 DOI: 10.1186/s12885-017-3399-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 06/01/2017] [Indexed: 02/18/2025] Open
Abstract
Background Granulin-epithelin precursor (GEP) is a secretory growth factor, which has been demonstrated to control cancer growth, invasion, drug resistance and immune escape. Our previous studies and others also demonstrated its potential in targeted therapy. Comprehensive characterization of GEP partner on cancer cells are warranted. We have previously shown that GEP interacted with heparan sulfate on the surface of liver cancer cells and the interaction is crucial for GEP-mediated signaling transduction. This study aims to characterize GEP protein partner at the cell membrane with the co-immunoprecipitation and mass spectrometry approach. Methods The membrane fraction from liver cancer model Hep3B was used for capturing binding partner with the specific monoclonal antibody against GEP. The precipitated proteins were analyzed by mass spectrometry. After identifying the GEP binding partner, this specific interaction was validated in additional liver cancer cell line HepG2 by co-immunoprecipitation using GRP78 and GEP antibodies, respectively, as the bait. GRP78 transcript levels in hepatocellular carcinoma (HCC) clinical samples (n = 77 pairs) were examined by real-time quantitative RT-PCR. GEP and GRP78 protein expressions were investigated by immunohistochemistry on paraffin sections. Results We identified the GEP-binding protein as 78-kDa glucose-regulated protein (GRP78, also named heat shock 70-kDa protein 5, HSPA5). This interaction was validated in independent HCC cell lines. Increased GRP78 mRNA levels were demonstrated in liver cancer tissues compared with the paralleled liver tissues (t-test, P = 0.002). GRP78 and GEP transcript levels were significantly correlated (Spearman’s correlation, P = 0.001), and the proteins were also detectable in the cytoplasm of liver cancer cells by immunohistochemical staining. Conclusions GRP78 and GEP are interacting protein partners in liver cancer cells and may play a role in GEP-mediated cancer progression in HCC.
Collapse
Affiliation(s)
- Chi Wai Yip
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong, China.,Department of Surgery, The University of Hong Kong, Hong Kong, China
| | - Ching Yan Lam
- Department of Surgery, The University of Hong Kong, Hong Kong, China.,Department of Health, The Government of the Hong Kong Special Administrative Region, Hong Kong, China
| | | | - Tan To Cheung
- Department of Surgery, The University of Hong Kong, Hong Kong, China
| | - Phyllis F Y Cheung
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong, China.,Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong, China
| | - Sze Wai Fung
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong, China.,Department of Surgery, The University of Hong Kong, Hong Kong, China.,School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Xiao Qi Wang
- Department of Surgery, The University of Hong Kong, Hong Kong, China
| | - Idy C Y Leung
- Department of Surgery, The University of Hong Kong, Hong Kong, China
| | - Linda W C Ng
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong, China
| | - Chung Mau Lo
- Department of Surgery, The University of Hong Kong, Hong Kong, China
| | - George S W Tsao
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Siu Tim Cheung
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong, China. .,Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China. .,Department of Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong.
| |
Collapse
|
104
|
Lindholm D, Korhonen L, Eriksson O, Kõks S. Recent Insights into the Role of Unfolded Protein Response in ER Stress in Health and Disease. Front Cell Dev Biol 2017; 5:48. [PMID: 28540288 PMCID: PMC5423914 DOI: 10.3389/fcell.2017.00048] [Citation(s) in RCA: 149] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 04/13/2017] [Indexed: 12/20/2022] Open
Abstract
Unfolded stress response (UPR) is a conserved cellular pathway involved in protein quality control to maintain homeostasis under different conditions and disease states characterized by cell stress. Although three general schemes of and genes induced by UPR are rather well-established, open questions remain including the precise role of UPR in human diseases and the interactions between different sensor systems during cell stress signaling. Particularly, the issue how the normally adaptive and pro-survival UPR pathway turns into a deleterious process causing sustained endoplasmic reticulum (ER) stress and cell death requires more studies. UPR is also named a friend with multiple personalities that we need to understand better to fully recognize its role in normal physiology and in disease pathology. UPR interacts with other organelles including mitochondria, and with cell stress signals and degradation pathways such as autophagy and the ubiquitin proteasome system. Here we review current concepts and mechanisms of UPR as studied in different cells and model systems and highlight the relevance of UPR and related stress signals in various human diseases.
Collapse
Affiliation(s)
- Dan Lindholm
- Medicum, Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of HelsinkiHelsinki, Finland.,Minerva Foundation Institute for Medical ResearchHelsinki, Finland
| | - Laura Korhonen
- Minerva Foundation Institute for Medical ResearchHelsinki, Finland.,Division of Child Psychiatry, Helsinki University Central HospitalHelsinki, Finland
| | - Ove Eriksson
- Medicum, Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of HelsinkiHelsinki, Finland
| | - Sulev Kõks
- Department of Pathophysiology, University of TartuTartu, Estonia.,Department of Reproductive Biology, Estonian University of Life SciencesTartu, Estonia
| |
Collapse
|
105
|
Casas C. GRP78 at the Centre of the Stage in Cancer and Neuroprotection. Front Neurosci 2017; 11:177. [PMID: 28424579 PMCID: PMC5380735 DOI: 10.3389/fnins.2017.00177] [Citation(s) in RCA: 172] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 03/17/2017] [Indexed: 12/21/2022] Open
Abstract
The 78-kDa glucose-regulated protein GRP78, also known as BiP and HSP5a, is a multifunctional protein with activities far beyond its well-known role in the unfolded protein response (UPR) which is activated after endoplasmic reticulum (ER) stress in the cells. Most of these newly discovered activities depend on its position within the cell. GRP78 is located mainly in the ER, but it has also been observed in the cytoplasm, the mitochondria, the nucleus, the plasma membrane, and secreted, although it is dedicated mostly to engage endogenous cytoprotective processes. Hence, GRP78 may control either UPR and macroautophagy or may activated phosphatidylinositol 3-kinase (PI3K)/AKT pro-survival pathways. GRP78 influences how tumor cells survive, proliferate, and develop chemoresistance. In neurodegeneration, endogenous mechanisms of neuroprotection are frequently insufficient or dysregulated. Lessons from tumor biology may give us clues about how boosting endogenous neuroprotective mechanisms in age-related neurodegeneration. Herein, the functions of GRP78 are revealed at the center of the stage of apparently opposite sites of the same coin regarding cytoprotection: neurodegeneration and cancer. The goal is to give a comprehensive and critical review that may serve to guide future experiments to identify interventions that will enhance neuroprotection.
Collapse
Affiliation(s)
- Caty Casas
- Department of Cell Biology, Physiology and Immunology, Institut de Neurociències, Universitat Autònoma de BarcelonaBarcelona, Spain
| |
Collapse
|
106
|
Linden R. The Biological Function of the Prion Protein: A Cell Surface Scaffold of Signaling Modules. Front Mol Neurosci 2017; 10:77. [PMID: 28373833 PMCID: PMC5357658 DOI: 10.3389/fnmol.2017.00077] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Accepted: 03/06/2017] [Indexed: 12/18/2022] Open
Abstract
The prion glycoprotein (PrPC) is mostly located at the cell surface, tethered to the plasma membrane through a glycosyl-phosphatydil inositol (GPI) anchor. Misfolding of PrPC is associated with the transmissible spongiform encephalopathies (TSEs), whereas its normal conformer serves as a receptor for oligomers of the β-amyloid peptide, which play a major role in the pathogenesis of Alzheimer’s Disease (AD). PrPC is highly expressed in both the nervous and immune systems, as well as in other organs, but its functions are controversial. Extensive experimental work disclosed multiple physiological roles of PrPC at the molecular, cellular and systemic levels, affecting the homeostasis of copper, neuroprotection, stem cell renewal and memory mechanisms, among others. Often each such process has been heralded as the bona fide function of PrPC, despite restricted attention paid to a selected phenotypic trait, associated with either modulation of gene expression or to the engagement of PrPC with a single ligand. In contrast, the GPI-anchored prion protein was shown to bind several extracellular and transmembrane ligands, which are required to endow that protein with the ability to play various roles in transmembrane signal transduction. In addition, differing sets of those ligands are available in cell type- and context-dependent scenarios. To account for such properties, we proposed that PrPC serves as a dynamic platform for the assembly of signaling modules at the cell surface, with widespread consequences for both physiology and behavior. The current review advances the hypothesis that the biological function of the prion protein is that of a cell surface scaffold protein, based on the striking similarities of its functional properties with those of scaffold proteins involved in the organization of intracellular signal transduction pathways. Those properties are: the ability to recruit spatially restricted sets of binding molecules involved in specific signaling; mediation of the crosstalk of signaling pathways; reciprocal allosteric regulation with binding partners; compartmentalized responses; dependence of signaling properties upon posttranslational modification; and stoichiometric requirements and/or oligomerization-dependent impact on signaling. The scaffold concept may contribute to novel approaches to the development of effective treatments to hitherto incurable neurodegenerative diseases, through informed modulation of prion protein-ligand interactions.
Collapse
Affiliation(s)
- Rafael Linden
- Laboratory of Neurogenesis, Institute of Biophysics, Federal University of Rio de Janeiro Rio de Janeiro, Brazil
| |
Collapse
|
107
|
HSPA5 Gene encoding Hsp70 chaperone BiP in the endoplasmic reticulum. Gene 2017; 618:14-23. [PMID: 28286085 DOI: 10.1016/j.gene.2017.03.005] [Citation(s) in RCA: 187] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 02/28/2017] [Accepted: 03/06/2017] [Indexed: 01/07/2023]
Abstract
The HSPA5 gene encodes the binding immunoglobulin protein (BiP), an Hsp70 family chaperone localized in the ER lumen. As a highly conserved molecular chaperone, BiP assists in a wide range of folding processes via its two structural domains, a nucleotide-binding domain (NBD) and substrate-binding domain (SBD). BiP is also an essential component of the translocation machinery for protein import into the ER, a regulator for Ca2+ homeostasis in the ER, as well as a facilitator of ER-associated protein degradation (ERAD) via retrograde transportation of aberrant proteins across the ER membrane. When unfolded/misfolded proteins in the ER overwhelm the capacity of protein folding machinery, BiP can initiate the unfolded protein response (UPR), decrease unfolded/misfolded protein load, induce autophagy, and crosstalk with apoptosis machinery to assist in the cell survival decision. Post-translational modifications (PTMs) of BiP have been shown to regulate BiP's activity, turnover, and availability upon different extrinsic or intrinsic stimuli. As a master regulator of ER function, BiP is associated with cancer, cardiovascular disease, neurodegenerative disease, and immunological diseases. BiP has been targeted in cancer therapies and shows promise for application in other relevant diseases.
Collapse
|
108
|
GRP78 Is an Important Host Factor for Japanese Encephalitis Virus Entry and Replication in Mammalian Cells. J Virol 2017; 91:JVI.02274-16. [PMID: 28053106 DOI: 10.1128/jvi.02274-16] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 12/27/2016] [Indexed: 12/25/2022] Open
Abstract
Japanese encephalitis virus (JEV), a mosquito-borne flavivirus, is the leading cause of viral encephalitis in Southeast Asia with potential to become a global pathogen. Here, we identify glucose-regulated protein 78 (GRP78) as an important host protein for virus entry and replication. Using the plasma membrane fractions from mouse neuronal (Neuro2a) cells, mass spectroscopy analysis identified GRP78 as a protein interacting with recombinant JEV envelope protein domain III. GRP78 was found to be expressed on the plasma membranes of Neuro2a cells, mouse primary neurons, and human epithelial Huh-7 cells. Antibodies against GRP78 significantly inhibited JEV entry in all three cell types, suggesting an important role of the protein in virus entry. Depletion of GRP78 by small interfering RNA (siRNA) significantly blocked JEV entry into Neuro2a cells, further supporting its role in virus uptake. Immunofluorescence studies showed extensive colocalization of GRP78 with JEV envelope protein in virus-infected cells. This interaction was also confirmed by immunoprecipitation studies. Additionally, GRP78 was shown to have an important role in JEV replication, as treatment of cells post-virus entry with subtilase cytotoxin that specifically cleaved GRP78 led to a substantial reduction in viral RNA replication and protein synthesis, resulting in significantly reduced extracellular virus titers. Our results indicate that GRP78, an endoplasmic reticulum chaperon of the HSP70 family, is a novel host factor involved at multiple steps of the JEV life cycle and could be a potential therapeutic target.IMPORTANCE Recent years have seen a rapid spread of mosquito-borne diseases caused by flaviviruses. The flavivirus family includes West Nile, dengue, Japanese encephalitis, and Zika viruses, which are major threats to public health with potential to become global pathogens. JEV is the major cause of viral encephalitis in several parts of Southeast Asia, affecting a predominantly pediatric population with a high mortality rate. This study is focused on identification of crucial host factors that could be targeted to cripple virus infection and ultimately lead to development of effective antivirals. We have identified a cellular protein, GRP78, that plays a dual role in virus entry and virus replication, two crucial steps of the virus life cycle, and thus is a novel host factor that could be a potential therapeutic target.
Collapse
|
109
|
Araujo TLS, Zeidler JD, Oliveira PVS, Dias MH, Armelin HA, Laurindo FRM. Protein disulfide isomerase externalization in endothelial cells follows classical and unconventional routes. Free Radic Biol Med 2017; 103:199-208. [PMID: 28034831 DOI: 10.1016/j.freeradbiomed.2016.12.021] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 12/09/2016] [Accepted: 12/17/2016] [Indexed: 12/22/2022]
Abstract
Extracellular protein disulfide isomerase (PDIA1) pool mediates thrombosis and vascular remodeling, however its externalization mechanisms remain unclear. We performed systematic pharmacological screening of secretory pathways affecting extracellular PDIA1 in endothelial cells (EC). We identified cell-surface (csPDIA1) and secreted non-particulated PDIA1 pools in EC. Such Golgi bypass also occurred for secreted PDIA1 in EC at baseline or after PMA, thrombin or ATP stimulation. Inhibitors of Type I, II and III unconventional routes, secretory lysosomes and recycling endosomes, including syntaxin-12 deletion, did not impair EC PDIA1 externalization. This suggests predominantly Golgi-independent unconventional secretory route(s), which were GRASP55-independent. Also, these data reinforce a vesicular-type traffic for PDIA1. We further showed that PDIA1 traffic is ATP-independent, while actin or tubulin cytoskeletal disruption markedly increased EC PDIA1 secretion. Clathrin inhibition enhanced extracellular soluble PDIA1, suggesting dynamic cycling. Externalized PDIA1 represents <2% of intracellular PDIA1. PDIA1 was robustly secreted by physiological levels of arterial laminar shear in EC and supported alpha 5 integrin thiol oxidation. Such results help clarify signaling and homeostatic mechanisms involved in multiple (patho)physiological extracellular PDIA1 functions.
Collapse
Affiliation(s)
- Thaís L S Araujo
- Vascular Biology Laboratory, Heart Institute (InCor), University of São Paulo School of Medicine, São Paulo 05403-000, Brazil
| | - Julianna D Zeidler
- Vascular Biology Laboratory, Heart Institute (InCor), University of São Paulo School of Medicine, São Paulo 05403-000, Brazil
| | - Percíllia V S Oliveira
- Vascular Biology Laboratory, Heart Institute (InCor), University of São Paulo School of Medicine, São Paulo 05403-000, Brazil
| | - Matheus H Dias
- Instituto de Química, Universidade de São Paulo, Brazil; Laboratório Especial de Ciclo Celular (LECC), Center of Toxins, Immune-Response and Cell Signaling - CeTICS-Cepid, Instituto Butantan, Brazil
| | - Hugo A Armelin
- Instituto de Química, Universidade de São Paulo, Brazil; Laboratório Especial de Ciclo Celular (LECC), Center of Toxins, Immune-Response and Cell Signaling - CeTICS-Cepid, Instituto Butantan, Brazil
| | - Francisco R M Laurindo
- Vascular Biology Laboratory, Heart Institute (InCor), University of São Paulo School of Medicine, São Paulo 05403-000, Brazil.
| |
Collapse
|
110
|
Soares Moretti AI, Martins Laurindo FR. Protein disulfide isomerases: Redox connections in and out of the endoplasmic reticulum. Arch Biochem Biophys 2016; 617:106-119. [PMID: 27889386 DOI: 10.1016/j.abb.2016.11.007] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 11/08/2016] [Accepted: 11/21/2016] [Indexed: 12/13/2022]
Abstract
Protein disulfide isomerases are thiol oxidoreductase chaperones from thioredoxin superfamily. As redox folding catalysts from the endoplasmic reticulum (ER), their roles in ER-related redox homeostasis and signaling are well-studied. PDIA1 exerts thiol oxidation/reduction and isomerization, plus chaperone effects. Also, substantial evidence indicates that PDIs regulate thiol-disulfide switches in other cell locations such as cell surface and possibly cytosol. Subcellular PDI translocation routes remain unclear and seem Golgi-independent. The list of signaling and structural proteins reportedly regulated by PDIs keeps growing, via thiol switches involving oxidation, reduction and isomerization, S-(de)nytrosylation, (de)glutathyonylation and protein oligomerization. PDIA1 is required for agonist-triggered Nox NADPH oxidase activation and cell migration in vascular cells and macrophages, while PDIA1-dependent cytoskeletal regulation appears a converging pathway. Extracellularly, PDIs crucially regulate thiol redox signaling of thrombosis/platelet activation, e.g., integrins, and PDIA1 supports expansive caliber remodeling during injury repair via matrix/cytoskeletal organization. Some proteins display regulatory PDI-like motifs. PDI effects are orchestrated by expression levels or post-translational modifications. PDI is redox-sensitive, although probably not a mass-effect redox sensor due to kinetic constraints. Rather, the "all-in-one" organization of its peculiar redox/chaperone properties likely provide PDIs with precision and versatility in redox signaling, making them promising therapeutic targets.
Collapse
Affiliation(s)
- Ana Iochabel Soares Moretti
- Vascular Biology Laboratory, Heart Institute (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil
| | | |
Collapse
|
111
|
Dadey DYA, Kapoor V, Hoye K, Khudanyan A, Collins A, Thotala D, Hallahan DE. Antibody Targeting GRP78 Enhances the Efficacy of Radiation Therapy in Human Glioblastoma and Non-Small Cell Lung Cancer Cell Lines and Tumor Models. Clin Cancer Res 2016; 23:2556-2564. [PMID: 27815359 DOI: 10.1158/1078-0432.ccr-16-1935] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 10/07/2016] [Accepted: 10/12/2016] [Indexed: 11/16/2022]
Abstract
Purpose: Non-small cell lung cancer (NSCLC) and glioblastoma multiforme (GBM) have poor median survival. NSCLC and GBM overexpress glucose regulated protein 78 (GRP78), which has a role in radioresistance and recurrence. In this study, we determined the effect of anti-GRP78 antibody and the combined effect of the anti-GRP78 antibody with ionizing radiation (XRT) on NSCLC and GBM cell lines both in vitro and in vivoExperimental Design: NSCLC and GBM cancer cell lines were treated with anti-GRP78 antibodies and evaluated for proliferation, colony formation, cell death, and PI3K/Akt/mTOR signaling. The efficacy of anti-GRP78 antibodies on tumor growth in combination with XRT was determined in vivo in mouse xenograft models.Results: GBM and NSCLC cells treated with anti-GRP78 antibodies showed attenuated cell proliferation, colony formation, and enhanced apoptosis. GBM and NSCLC cells treated with anti-GRP78 antibodies also showed global suppression of PI3K/Akt/mTOR signaling. Combining antibody with XRT resulted in significant tumor growth delay in both NSCLC and GBM heterotopic tumor models.Conclusions: Antibodies targeting GRP78 exhibited antitumor activity and enhanced the efficacy of radiation in NSCLC and GBM both in vitro and in vivo GRP78 is a promising novel target, and anti-GRP78 antibodies could be used as an effective cancer therapy alone or in combination with XRT. Clin Cancer Res; 23(10); 2556-64. ©2016 AACR.
Collapse
Affiliation(s)
- David Y A Dadey
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, Missouri.,Medical Scientist Training Program, Washington University in St. Louis, St. Louis, Missouri
| | - Vaishali Kapoor
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, Missouri
| | - Kelly Hoye
- Medical Guidance Systems, St. Louis, Missouri
| | - Arpine Khudanyan
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, Missouri
| | - Andrea Collins
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, Missouri
| | - Dinesh Thotala
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, Missouri. .,Siteman Cancer Center, School of Medicine, Washington University in St. Louis, St. Louis, Missouri
| | - Dennis E Hallahan
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, Missouri. .,Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri.,Siteman Cancer Center, School of Medicine, Washington University in St. Louis, St. Louis, Missouri
| |
Collapse
|
112
|
BOOTH LAURENCE, ROBERTS JANEL, ECROYD HEATH, TRITSCH SARAHR, BAVARI SINA, REID STPATRICK, PRONIUK STEFAN, ZUKIWSKI ALEXANDER, JACOB ABRAHAM, SEPÚLVEDA CLAUDIAS, GIOVANNONI FEDERICO, GARCÍA CYBELEC, DAMONTE ELSA, GONZÁLEZ-GALLEGO JAVIER, TUÑÓN MARÍAJ, DENT PAUL. AR-12 Inhibits Multiple Chaperones Concomitant With Stimulating Autophagosome Formation Collectively Preventing Virus Replication. J Cell Physiol 2016; 231:2286-302. [PMID: 27187154 PMCID: PMC6327852 DOI: 10.1002/jcp.25431] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 05/16/2016] [Indexed: 01/13/2023]
Abstract
We have recently demonstrated that AR-12 (OSU-03012) reduces the function and ATPase activities of multiple HSP90 and HSP70 family chaperones. Combined knock down of chaperones or AR-12 treatment acted to reduce the expression of virus receptors and essential glucosidase proteins. Combined knock down of chaperones or AR-12 treatment inactivated mTOR and elevated ATG13 S318 phosphorylation concomitant with inducing an endoplasmic reticulum stress response that in an eIF2α-dependent fashion increased Beclin1 and LC3 expression and autophagosome formation. Over-expression of chaperones prevented the reduction in receptor/glucosidase expression, mTOR inactivation, the ER stress response, and autophagosome formation. AR-12 reduced the reproduction of viruses including Mumps, Influenza, Measles, Junín, Rubella, HIV (wild type and protease resistant), and Ebola, an effect replicated by knock down of multiple chaperone proteins. AR-12-stimulated the co-localization of Influenza, EBV and HIV virus proteins with LC3 in autophagosomes and reduced viral protein association with the chaperones HSP90, HSP70, and GRP78. Knock down of Beclin1 suppressed drug-induced autophagosome formation and reduced the anti-viral protection afforded by AR-12. In an animal model of hemorrhagic fever virus, a transient exposure of animals to low doses of AR-12 doubled animal survival from ∼30% to ∼60% and suppressed liver damage as measured by ATL, GGT and LDH release. Thus through inhibition of chaperone protein functions; reducing the production, stability and processing of viral proteins; and stimulating autophagosome formation/viral protein degradation, AR-12 acts as a broad-specificity anti-viral drug in vitro and in vivo. We argue future patient studies with AR-12 are warranted. J. Cell. Physiol. 231: 2286-2302, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- LAURENCE BOOTH
- Department of Biochemistry and Molecular Biology, Virginia
Commonwealth University, Richmond, Virginia
| | - JANE L. ROBERTS
- Department of Biochemistry and Molecular Biology, Virginia
Commonwealth University, Richmond, Virginia
| | - HEATH ECROYD
- School of Biological Sciences and Illawarra Health and
Medical Research Institute, University of Wollongong, New South Wales,
Australia
| | - SARAH R. TRITSCH
- Molecular and Translational Science, United States Army
Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick,
Frederick, Maryland
| | - SINA BAVARI
- Molecular and Translational Science, United States Army
Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick,
Frederick, Maryland
| | - ST. PATRICK REID
- Molecular and Translational Science, United States Army
Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick,
Frederick, Maryland
| | | | | | - ABRAHAM JACOB
- Department of Otolaryngology, University of Arizona Ear
Institute, Tucson, Arizona
| | - CLAUDIA S. SEPÚLVEDA
- FCEN-UBA, Ciudad Universitaria, Pabellón 2 Piso 4,
lab QB-17, Buenos Aires, Argentina
| | - FEDERICO GIOVANNONI
- FCEN-UBA, Ciudad Universitaria, Pabellón 2 Piso 4,
lab QB-17, Buenos Aires, Argentina
| | - CYBELE C. GARCÍA
- FCEN-UBA, Ciudad Universitaria, Pabellón 2 Piso 4,
lab QB-17, Buenos Aires, Argentina
| | - ELSA DAMONTE
- FCEN-UBA, Ciudad Universitaria, Pabellón 2 Piso 4,
lab QB-17, Buenos Aires, Argentina
| | | | - MARÍA J. TUÑÓN
- Institute of Biomedicine and CIBEREhd, University of
León, León, Spain
| | - PAUL DENT
- Department of Biochemistry and Molecular Biology, Virginia
Commonwealth University, Richmond, Virginia
| |
Collapse
|
113
|
Kaliberov SA, Kaliberova LN, Yan H, Kapoor V, Hallahan DE. Retargeted adenoviruses for radiation-guided gene delivery. Cancer Gene Ther 2016; 23:303-14. [PMID: 27492853 PMCID: PMC5031535 DOI: 10.1038/cgt.2016.32] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 06/21/2016] [Accepted: 06/30/2016] [Indexed: 11/25/2022]
Abstract
The combination of radiation with radiosensitizing gene delivery or oncolytic viruses promises to provide an advantage that could improve the therapeutic results for glioblastoma. X-rays can induce significant molecular changes in cancer cells. We isolated the GIRLRG peptide that binds to radiation-inducible 78 kDa glucose-regulated protein (GRP78), which is overexpressed on the plasma membranes of irradiated cancer cells and tumor-associated microvascular endothelial cells. The goal of our study was to improve tumor-specific adenovirus-mediated gene delivery by selectively targeting the adenovirus binding to this radiation-inducible protein. We employed an adenoviral fiber replacement approach to conduct a study of the targeting utility of GRP78-binding peptide. We have developed fiber-modified adenoviruses encoding the GRP78-binding peptide inserted into the fiber-fibritin. We have evaluated the reporter gene expression of fiber-modified adenoviruses in vitro using a panel of glioma cells and a human D54MG tumor xenograft model. The obtained results demonstrated that employment of the GRP78-binding peptide resulted in increased gene expression in irradiated tumors following infection with fiber-modified adenoviruses, compared with untreated tumor cells. These studies demonstrate the feasibility of adenoviral retargeting using the GRP78-binding peptide that selectively recognizes tumor cells responding to radiation treatment.
Collapse
Affiliation(s)
- S A Kaliberov
- Department of Radiation Oncology, Washington University School of Medicine, St Louis, MO, USA.,Biologic Therapeutics Center, Washington University School of Medicine, St Louis, MO, USA
| | - L N Kaliberova
- Department of Radiation Oncology, Washington University School of Medicine, St Louis, MO, USA.,Biologic Therapeutics Center, Washington University School of Medicine, St Louis, MO, USA
| | - H Yan
- Department of Radiation Oncology, Washington University School of Medicine, St Louis, MO, USA.,Biologic Therapeutics Center, Washington University School of Medicine, St Louis, MO, USA
| | - V Kapoor
- Department of Radiation Oncology, Washington University School of Medicine, St Louis, MO, USA
| | - D E Hallahan
- Department of Radiation Oncology, Washington University School of Medicine, St Louis, MO, USA.,Biologic Therapeutics Center, Washington University School of Medicine, St Louis, MO, USA.,Siteman Cancer Center, St Louis, MO, USA
| |
Collapse
|
114
|
Zeng YQ, Cao RY, Yang JL, Li XZ, Li S, Zhong W. Design, synthesis and biological evaluation of novel HSP70 inhibitors: N, N′-disubstituted thiourea derivatives. Eur J Med Chem 2016; 119:83-95. [DOI: 10.1016/j.ejmech.2016.04.042] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 04/14/2016] [Accepted: 04/15/2016] [Indexed: 01/09/2023]
|
115
|
Rasche L, Menoret E, Dubljevic V, Menu E, Vanderkerken K, Lapa C, Steinbrunn T, Chatterjee M, Knop S, Düll J, Greenwood DL, Hensel F, Rosenwald A, Einsele H, Brändlein S. A GRP78-Directed Monoclonal Antibody Recaptures Response in Refractory Multiple Myeloma with Extramedullary Involvement. Clin Cancer Res 2016; 22:4341-9. [DOI: 10.1158/1078-0432.ccr-15-3111] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 03/16/2016] [Indexed: 11/16/2022]
|
116
|
Cirillo F, Ghiroldi A, Fania C, Piccoli M, Torretta E, Tettamanti G, Gelfi C, Anastasia L. NEU3 Sialidase Protein Interactors in the Plasma Membrane and in the Endosomes. J Biol Chem 2016; 291:10615-24. [PMID: 26987901 DOI: 10.1074/jbc.m116.719518] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Indexed: 11/06/2022] Open
Abstract
NEU3 sialidase has been shown to be a key player in many physio- and pathological processes, including cell differentiation, cellular response to hypoxic stress, and carcinogenesis. The enzyme, peculiarly localized on the outer leaflet of the plasma membrane, has been shown to be able to remove sialic acid residues from the gangliosides present on adjacent cells, thus creating cell to cell interactions. Nonetheless, herein we report that the enzyme localization is dynamically regulated between the plasma membrane and the endosomes, where a substantial amount of NEU3 is stored with low enzymatic activity. However, under opportune stimuli, NEU3 is shifted from the endosomes to the plasma membrane, where it greatly increases the sialidase activity. Finally, we found that NEU3 possesses also the ability to interact with specific proteins, many of which are different in each cell compartment. They were identified by mass spectrometry, and some selected ones were also confirmed by cross-immunoprecipitation with the enzyme, supporting NEU3 involvement in the cell stress response, protein folding, and intracellular trafficking.
Collapse
Affiliation(s)
- Federica Cirillo
- From the Istituto Di Ricovero e Cura a Carattere Scientifico Policlinico San Donato, 20097 San Donato Milanese, Milan and
| | - Andrea Ghiroldi
- From the Istituto Di Ricovero e Cura a Carattere Scientifico Policlinico San Donato, 20097 San Donato Milanese, Milan and
| | - Chiara Fania
- From the Istituto Di Ricovero e Cura a Carattere Scientifico Policlinico San Donato, 20097 San Donato Milanese, Milan and
| | - Marco Piccoli
- From the Istituto Di Ricovero e Cura a Carattere Scientifico Policlinico San Donato, 20097 San Donato Milanese, Milan and
| | - Enrica Torretta
- the Department of Biomedical Sciences for Health, University of Milan, 20122 Milan, Italy
| | - Guido Tettamanti
- From the Istituto Di Ricovero e Cura a Carattere Scientifico Policlinico San Donato, 20097 San Donato Milanese, Milan and
| | - Cecilia Gelfi
- From the Istituto Di Ricovero e Cura a Carattere Scientifico Policlinico San Donato, 20097 San Donato Milanese, Milan and the Department of Biomedical Sciences for Health, University of Milan, 20122 Milan, Italy
| | - Luigi Anastasia
- From the Istituto Di Ricovero e Cura a Carattere Scientifico Policlinico San Donato, 20097 San Donato Milanese, Milan and the Department of Biomedical Sciences for Health, University of Milan, 20122 Milan, Italy
| |
Collapse
|
117
|
Rangel MC, Bertolette D, Castro NP, Klauzinska M, Cuttitta F, Salomon DS. Developmental signaling pathways regulating mammary stem cells and contributing to the etiology of triple-negative breast cancer. Breast Cancer Res Treat 2016; 156:211-26. [PMID: 26968398 PMCID: PMC4819564 DOI: 10.1007/s10549-016-3746-7] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 03/04/2016] [Indexed: 12/17/2022]
Abstract
Cancer has been considered as temporal and spatial aberrations of normal development in tissues. Similarities between mammary embryonic development and cell transformation suggest that the underlying processes required for mammary gland development are also those perturbed during various stages of mammary tumorigenesis and breast cancer (BC) development. The master regulators of embryonic development Cripto-1, Notch/CSL, and Wnt/β-catenin play key roles in modulating mammary gland morphogenesis and cell fate specification in the embryo through fetal mammary stem cells (fMaSC) and in the adult organism particularly within the adult mammary stem cells (aMaSC), which determine mammary progenitor cell lineages that generate the basal/myoepithelial and luminal compartments of the adult mammary gland. Together with recognized transcription factors and embryonic stem cell markers, these embryonic regulatory molecules can be inappropriately augmented during tumorigenesis to support the tumor-initiating cell (TIC)/cancer stem cell (CSC) compartment, and the effects of their deregulation may contribute for the etiology of BC, in particular the most aggressive subtype of BC, triple-negative breast cancer (TNBC). This in depth review will present evidence of the involvement of Cripto-1, Notch/CSL, and Wnt/β-catenin in the normal mammary gland morphogenesis and tumorigenesis, from fMaSC/aMaSC regulation to TIC generation and maintenance in TNBC. Specific therapies for treating TNBC by targeting these embryonic pathways in TICs will be further discussed, providing new opportunities to destroy not only the bulk tumor, but also TICs that initiate and promote the metastatic spread and recurrence of this aggressive subtype of BC.
Collapse
Affiliation(s)
- Maria Cristina Rangel
- Tumor Growth Factor Section, Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Building 560, Room 32-40B, 1050 Boyles Street, Ft. Detrick, Frederick, MD, 21702, USA
| | - Daniel Bertolette
- Tumor Growth Factor Section, Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Building 560, Room 32-40B, 1050 Boyles Street, Ft. Detrick, Frederick, MD, 21702, USA
| | - Nadia P Castro
- Tumor Growth Factor Section, Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Building 560, Room 32-40B, 1050 Boyles Street, Ft. Detrick, Frederick, MD, 21702, USA
| | - Malgorzata Klauzinska
- Tumor Growth Factor Section, Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Building 560, Room 32-40B, 1050 Boyles Street, Ft. Detrick, Frederick, MD, 21702, USA
| | - Frank Cuttitta
- Tumor Growth Factor Section, Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Building 560, Room 32-40B, 1050 Boyles Street, Ft. Detrick, Frederick, MD, 21702, USA
| | - David S Salomon
- Tumor Growth Factor Section, Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Building 560, Room 32-40B, 1050 Boyles Street, Ft. Detrick, Frederick, MD, 21702, USA.
| |
Collapse
|
118
|
Yamada K, Miyamoto Y, Tsujii A, Moriyama T, Ikuno Y, Shiromizu T, Serada S, Fujimoto M, Tomonaga T, Naka T, Yoneda Y, Oka M. Cell surface localization of importin α1/KPNA2 affects cancer cell proliferation by regulating FGF1 signalling. Sci Rep 2016; 6:21410. [PMID: 26887791 PMCID: PMC4757827 DOI: 10.1038/srep21410] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 01/22/2016] [Indexed: 02/07/2023] Open
Abstract
Importin α1 is involved in nuclear import as a receptor for proteins with a classical nuclear localization signal (cNLS). Here, we report that importin α1 is localized to the cell surface in several cancer cell lines and detected in their cultured medium. We also found that exogenously added importin α1 is associated with the cell membrane via interaction with heparan sulfate. Furthermore, we revealed that the cell surface importin α1 recognizes cNLS-containing substrates. More particularly, importin α1 bound directly to FGF1 and FGF2, secreted cNLS-containing growth factors, and addition of exogenous importin α1 enhanced the activation of ERK1/2, downstream targets of FGF1 signalling, in FGF1-stimulated cancer cells. Additionally, anti-importin α1 antibody treatment suppressed the importin α1-FGF1 complex formation and ERK1/2 activation, resulting in decreased cell growth. This study provides novel evidence that functional importin α1 is located at the cell surface, where it accelerates the proliferation of cancer cells.
Collapse
Affiliation(s)
- Kohji Yamada
- Laboratory of Nuclear Transport Dynamics, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito-Asagi, Ibaraki, Osaka 567-0085, Japan
| | - Yoichi Miyamoto
- Laboratory of Nuclear Transport Dynamics, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito-Asagi, Ibaraki, Osaka 567-0085, Japan
| | - Akira Tsujii
- Laboratory of Nuclear Transport Dynamics, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito-Asagi, Ibaraki, Osaka 567-0085, Japan.,Department of Genetics, Graduate School of Medicine, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Tetsuji Moriyama
- Laboratory of Nuclear Transport Dynamics, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito-Asagi, Ibaraki, Osaka 567-0085, Japan
| | - Yudai Ikuno
- Laboratory of Nuclear Transport Dynamics, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito-Asagi, Ibaraki, Osaka 567-0085, Japan
| | - Takashi Shiromizu
- Laboratory of Proteome Research, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito-Asagi, Ibaraki, Osaka 567-0085, Japan
| | - Satoshi Serada
- Laboratory of Immune Signal, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito-Asagi, Ibaraki, Osaka 567-0085, Japan
| | - Minoru Fujimoto
- Laboratory of Immune Signal, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito-Asagi, Ibaraki, Osaka 567-0085, Japan
| | - Takeshi Tomonaga
- Laboratory of Proteome Research, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito-Asagi, Ibaraki, Osaka 567-0085, Japan
| | - Tetsuji Naka
- Laboratory of Immune Signal, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito-Asagi, Ibaraki, Osaka 567-0085, Japan
| | - Yoshihiro Yoneda
- National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito-Asagi, Ibaraki, Osaka 567-0085, Japan.,Laboratory of Biomedical Innovation, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka 565-0871, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka 567-0085, Japan
| | - Masahiro Oka
- Laboratory of Nuclear Transport Dynamics, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito-Asagi, Ibaraki, Osaka 567-0085, Japan.,Laboratory of Biomedical Innovation, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka 565-0871, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka 567-0085, Japan
| |
Collapse
|
119
|
Kamada Y, Nawata Y, Sugimoto Y. Lysozyme Mutants Accumulate in Cells while Associated at their N-terminal Alpha-domain with the Endoplasmic Reticulum Chaperone GRP78/BiP. Int J Biol Sci 2016; 12:184-97. [PMID: 26884716 PMCID: PMC4737675 DOI: 10.7150/ijbs.13710] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 11/09/2015] [Indexed: 01/07/2023] Open
Abstract
Amyloidogenic human lysozyme variants deposit in cells and cause systemic amyloidosis. We recently observed that such lysozymes accumulate in the endoplasmic reticulum (ER) with the ER chaperone GRP78/BiP, accompanying the ER stress response. Here we investigated the region of lysozyme that is critical to its association with GRP78/BiP. In addition to the above-mentioned variants of lysozyme, we constructed lysozyme truncation or substitution mutants. These were co-expressed with GRP78/BiP (tagged with FLAG) in cultured human embryonic kidney cells, which were analyzed by western blotting and immunocytochemistry using anti-lysozyme and anti-FLAG antibodies. The amyloidogenic variants were confirmed to be strongly associated with GRP78/BiP as revealed by the co-immunoprecipitation assay, whereas N-terminal mutants pruned of 1-41 or 1-51 residues were found not to be associated with the chaperone. Single amino acid substitutions for the leucine array along the α-helices in the N-terminal region resulted in wild-type lysozyme remaining attached to GRP78/BiP. These mutations also tended to show lowered secretion ability. We conclude that the N-terminal α-helices region of the lysozyme is pivotal for its strong adhesion to GRP78/BiP. We suspect that wild-type lysozyme interacts with the GRP at this region as a step in the proper folding monitored by the ER chaperone.
Collapse
Affiliation(s)
- Yoshiki Kamada
- Laboratory of Biochemistry and Bioscience, The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima 890-0065, Japan
| | - Yusuke Nawata
- Laboratory of Biochemistry and Bioscience, The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima 890-0065, Japan
| | - Yasushi Sugimoto
- Laboratory of Biochemistry and Bioscience, The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima 890-0065, Japan
| |
Collapse
|
120
|
Wang J, Nie Y, Li Y, Hou Y, Zhao W, Deng J, Wang PG, Bai G. Identification of target proteins of mangiferin in mice with acute lung injury using functionalized magnetic microspheres based on click chemistry. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2015; 63:10013-10021. [PMID: 26488336 DOI: 10.1021/acs.jafc.5b04439] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Prevention of the occurrence and development of inflammation is a vital therapeutic strategy for treating acute lung injury (ALI). Increasing evidence has shown that a wealth of ingredients from natural foods and plants have potential anti-inflammatory activity. In the present study, mangiferin, a natural C-glucosyl xanthone that is primarily obtained from the peels and kernels of mango fruits and the bark of the Mangifera indica L. tree, alleviated the inflammatory responses in lipopolysaccharide (LPS)-induced ALI mice. Mangiferin-modified magnetic microspheres (MMs) were developed on the basis of click chemistry to capture the target proteins of mangiferin. Mass spectrometry and molecular docking identified 70 kDa heat-shock protein 5 (Hspa5) and tyrosine 3-monooxygenase (Ywhae) as mangiferin-binding proteins. Furthermore, an enzyme-linked immunosorbent assay (ELISA) indicated that mangiferin exerted its anti-inflammatory effect by binding Hspa5 and Ywhae to suppress downstream mitogen-activated protein kinase (MAPK) signaling pathways. Thoroughly revealing the mechanism and function of mangiferin will contribute to the development and utilization of agricultural resources from M. indica L.
Collapse
Affiliation(s)
- Jiajia Wang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, Nankai University , Tianjin 300071, People's Republic of China
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University , Beijing 100871, People's Republic of China
| | - Yan Nie
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, Nankai University , Tianjin 300071, People's Republic of China
| | - Yunjuan Li
- Guangxi University of Chinese Medicine , Nanning, Guangxi 530001, People's Republic of China
| | - Yuanyuan Hou
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, Nankai University , Tianjin 300071, People's Republic of China
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University , Beijing 100871, People's Republic of China
| | - Wei Zhao
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, Nankai University , Tianjin 300071, People's Republic of China
| | - Jiagang Deng
- Guangxi University of Chinese Medicine , Nanning, Guangxi 530001, People's Republic of China
| | - Peng George Wang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, Nankai University , Tianjin 300071, People's Republic of China
| | - Gang Bai
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, Nankai University , Tianjin 300071, People's Republic of China
| |
Collapse
|
121
|
Wang Y, Wang X, Ferrone CR, Schwab JH, Ferrone S. Intracellular antigens as targets for antibody based immunotherapy of malignant diseases. Mol Oncol 2015; 9:1982-93. [PMID: 26597109 DOI: 10.1016/j.molonc.2015.10.019] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 10/19/2015] [Accepted: 10/20/2015] [Indexed: 12/13/2022] Open
Abstract
This review discusses the potential use of intracellular tumor antigens as targets of antibody-based immunotherapy for the treatment of solid tumors. In addition, it describes the characteristics of the intracellular tumor antigens targeted with antibodies which have been described in the literature and have been identified in the authors' laboratory. Finally, the mechanism underlying the trafficking of the intracellular tumor antigens to the plasma membrane of tumor cells are reviewed.
Collapse
Affiliation(s)
- Yangyang Wang
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, United States
| | - Xinhui Wang
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, United States
| | - Cristina R Ferrone
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, United States
| | - Joseph H Schwab
- Department of Orthopaedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, United States
| | - Soldano Ferrone
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, United States; Department of Orthopaedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, United States.
| |
Collapse
|
122
|
Roberts JL, Tavallai M, Nourbakhsh A, Fidanza A, Cruz-Luna T, Smith E, Siembida P, Plamondon P, Cycon KA, Doern CD, Booth L, Dent P. GRP78/Dna K Is a Target for Nexavar/Stivarga/Votrient in the Treatment of Human Malignancies, Viral Infections and Bacterial Diseases. J Cell Physiol 2015; 230:2552-78. [PMID: 25858032 PMCID: PMC4843173 DOI: 10.1002/jcp.25014] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 04/06/2015] [Indexed: 01/10/2023]
Abstract
Prior tumor cell studies have shown that the drugs sorafenib (Nexavar) and regorafenib (Stivarga) reduce expression of the chaperone GRP78. Sorafenib/regorafenib and the multi‐kinase inhibitor pazopanib (Votrient) interacted with sildenafil (Viagra) to further rapidly reduce GRP78 levels in eukaryotes and as single agents to reduce Dna K levels in prokaryotes. Similar data were obtained in tumor cells in vitro and in drug‐treated mice for: HSP70, mitochondrial HSP70, HSP60, HSP56, HSP40, HSP10, and cyclophilin A. Prolonged ‘rafenib/sildenafil treatment killed tumor cells and also rapidly decreased the expression of: the drug efflux pumps ABCB1 and ABCG2; and NPC1 and NTCP, receptors for Ebola/Hepatitis A and B viruses, respectively. Pre‐treatment with the ‘Rafenib/sildenafil combination reduced expression of the Coxsackie and Adenovirus receptor in parallel with it also reducing the ability of a serotype 5 Adenovirus or Coxsackie virus B4 to infect and to reproduce. Sorafenib/pazopanib and sildenafil was much more potent than sorafenib/pazopanib as single agents at preventing Adenovirus, Mumps, Chikungunya, Dengue, Rabies, West Nile, Yellow Fever, and Enterovirus 71 infection and reproduction. ‘Rafenib drugs/pazopanib as single agents killed laboratory generated antibiotic resistant E. coli which was associated with reduced Dna K and Rec A expression. Marginally toxic doses of ‘Rafenib drugs/pazopanib restored antibiotic sensitivity in pan‐antibiotic resistant bacteria including multiple strains of blakpcKlebsiella pneumoniae. Thus, Dna K is an antibiotic target for sorafenib, and inhibition of GRP78/Dna K has therapeutic utility for cancer and for bacterial and viral infections. J. Cell. Physiol. 230: 2552–2578, 2015. © 2015 The Authors. Journal of Cellular Physiology published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Jane L Roberts
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia
| | - Mehrad Tavallai
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia
| | - Aida Nourbakhsh
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia
| | | | | | | | | | | | | | - Christopher D Doern
- Department of Pathology, Virginia Commonwealth University, Richmond, Virginia
| | - Laurence Booth
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia
| | - Paul Dent
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
123
|
Abstract
Over 230,000 new cases of invasive breast cancer are diagnosed annually within the USA. Recurrent breast cancer remains a mostly incurable disease with drug resistance, tumor latency and distant metastases driving breast tumor recurrence and morbidity. Understanding drug resistance is a critical component of combating breast cancer. Recently, the protein chaperone GRP78 and the unfolded protein response were implicated as drivers of drug resistance. Preclinical studies show inhibiting GRP78 can reverse drug resistance. Furthermore, drugs developed to target GRP78 show clinical promise in several ongoing clinical trials.
Collapse
|
124
|
Booth L, Roberts JL, Tavallai M, Nourbakhsh A, Chuckalovcak J, Carter J, Poklepovic A, Dent P. OSU-03012 and Viagra Treatment Inhibits the Activity of Multiple Chaperone Proteins and Disrupts the Blood-Brain Barrier: Implications for Anti-Cancer Therapies. J Cell Physiol 2015; 230:1982-98. [PMID: 25736380 PMCID: PMC4835175 DOI: 10.1002/jcp.24977] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 02/26/2015] [Indexed: 12/14/2022]
Abstract
We examined the interaction between OSU‐03012 (also called AR‐12) with phosphodiesterase 5 (PDE5) inhibitors to determine the role of the chaperone glucose‐regulated protein (GRP78)/BiP/HSPA5 in the cellular response. Sildenafil (Viagra) interacted in a greater than additive fashion with OSU‐03012 to kill stem‐like GBM cells. Treatment of cells with OSU‐03012/sildenafil: abolished the expression of multiple oncogenic growth factor receptors and plasma membrane drug efflux pumps and caused a rapid degradation of GRP78 and other HSP70 and HSP90 family chaperone proteins. Decreased expression of plasma membrane receptors and drug efflux pumps was dependent upon enhanced PERK‐eIF2α‐ATF4‐CHOP signaling and was blocked by GRP78 over‐expression. In vivo OSU‐03012/sildenafil was more efficacious than treatment with celecoxib and sildenafil at killing tumor cells without damaging normal tissues and in parallel reduced expression of ABCB1 and ABCG2 in the normal brain. The combination of OSU‐03012/sildenafil synergized with low concentrations of sorafenib to kill tumor cells, and with lapatinib to kill ERBB1 over‐expressing tumor cells. In multiplex assays on plasma and human tumor tissue from an OSU‐03012/sildenafil treated mouse, we noted a profound reduction in uPA signaling and identified FGF and JAK1/2 as response biomarkers for potentially suppressing the killing response. Inhibition of FGFR signaling and to a lesser extent JAK1/2 signaling profoundly enhanced OSU‐03012/sildenafil lethality. J. Cell. Physiol. 230: 1982–1998, 2015. © 2015 The Authors. Journal of Cellular Physiology Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Laurence Booth
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia
| | | | | | | | | | | | | | | |
Collapse
|