51
|
Kiss B, Ecsédi P, Simon M, Nyitray L. Isolation and Characterization of S100 Protein-Protein Complexes. Methods Mol Biol 2019; 1929:325-338. [PMID: 30710283 DOI: 10.1007/978-1-4939-9030-6_21] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
S100 proteins are small, mostly dimeric, EF-hand Ca2+-binding proteins. Upon Ca2+ binding, a conformational change occurs resulting in the exposure of a shallow hydrophobic binding groove in each subunit. Interestingly, S100 proteins can interact with their partners in two ways: symmetrically, when the two partners identically bind into each groove, or asymmetrically, when only one partner binds to the S100 dimer occupying both binding pockets. Here we present a heterologous expression and purification protocol for all known human S100 proteins as well as for their partner peptides. Moreover, we provide a detailed description of three in vitro methods to determine the affinity, stoichiometry, and kinetics of S100 protein-protein interactions.
Collapse
Affiliation(s)
- Bence Kiss
- Department of Biochemistry, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Péter Ecsédi
- Department of Biochemistry, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Márton Simon
- Department of Biochemistry, ELTE Eötvös Loránd University, Budapest, Hungary
| | - László Nyitray
- Department of Biochemistry, ELTE Eötvös Loránd University, Budapest, Hungary.
| |
Collapse
|
52
|
Wang Y, Tadayon R, Shaw GS. Monitoring Interactions Between S100B and the Dopamine D2 Receptor Using NMR Spectroscopy. Methods Mol Biol 2019; 1929:311-324. [PMID: 30710282 DOI: 10.1007/978-1-4939-9030-6_20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
S100B is a dimeric EF-hand protein that undergoes a calcium-induced conformational change and interacts with a wide range of proteins to modulate their functions. The dopamine D2 receptor is one potential S100B binding partner that may play a key role in neurological processing. In this chapter, we describe the use of NMR spectroscopy to examine the interaction between calcium-bound S100B and the third intracellular loop (IC3) from the dopamine D2 receptor. We provide details that allow the strength of the interaction (K d) between the two proteins to be determined and the IC3 site of interaction on the structure of S100B to be identified. Both these characteristics can be identified from a single series of nondestructive experiments.
Collapse
Affiliation(s)
- Yuning Wang
- Department of Biochemistry, The University of Western Ontario, London, ON, Canada
| | - Roya Tadayon
- Department of Biochemistry, The University of Western Ontario, London, ON, Canada
| | - Gary S Shaw
- Department of Biochemistry, The University of Western Ontario, London, ON, Canada.
| |
Collapse
|
53
|
Bresnick AR. S100 proteins as therapeutic targets. Biophys Rev 2018; 10:1617-1629. [PMID: 30382555 PMCID: PMC6297089 DOI: 10.1007/s12551-018-0471-y] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 10/21/2018] [Indexed: 12/13/2022] Open
Abstract
The human genome codes for 21 S100 protein family members, which exhibit cell- and tissue-specific expression patterns. Despite sharing a high degree of sequence and structural similarity, the S100 proteins bind a diverse range of protein targets and contribute to a broad array of intracellular and extracellular functions. Consequently, the S100 proteins regulate multiple cellular processes such as proliferation, migration and/or invasion, and differentiation, and play important roles in a variety of cancers, autoimmune diseases, and chronic inflammatory disorders. This review focuses on the development of S100 neutralizing antibodies and small molecule inhibitors and their potential therapeutic use in controlling disease progression and severity.
Collapse
Affiliation(s)
- Anne R Bresnick
- Department of Biochemistry, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10461, USA.
| |
Collapse
|
54
|
Zhu Q, Smith EA. Diaphanous-1 affects the nanoscale clustering and lateral diffusion of receptor for advanced glycation endproducts (RAGE). BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2018; 1861:43-49. [PMID: 30401627 DOI: 10.1016/j.bbamem.2018.10.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/24/2018] [Accepted: 10/24/2018] [Indexed: 12/22/2022]
Abstract
The interactions between the cytoplasmic protein diaphanous-1 (Diaph1) and the receptor for advanced glycation endproducts (RAGE) drive the negative consequences of RAGE signaling in several disease processes. Reported in this work is how Diaph1 affects the nanoscale clustering and diffusion of RAGE measured using super-resolution stochastic optical reconstruction microscopy (STORM) and single particle tracking (SPT). Altering the Diaph1 binding site has a different impact on RAGE diffusion compared to when Diaph1 expression is reduced in HEK293 cells. In cells with reduced Diaph1 expression (RAGE-Diaph1-/-), the average RAGE diffusion coefficient is increased by 35%. RAGE diffusion is known to be influenced by the dynamics of the actin cytoskeleton. Actin labeling shows that a reduced Diaph1 expression leads to cells with reduced filopodia density and length. In contrast, when two RAGE amino acids that interact with Diaph1 are mutated (RAGERQ/AA), the average RAGE diffusion coefficient is decreased by 16%. Since RAGE diffusion is slowed when the interaction between Diaph1 and RAGE is disrupted, the interaction of the two proteins results in faster RAGE diffusion. In both RAGERQ/AA and RAGE-Diaph1-/- cells the number and size of RAGE clusters are decreased compared to cells expressing RAGE and native concentrations of Diaph1. This work shows that Diaph1 has a role in affecting RAGE clusters and diffusion.
Collapse
Affiliation(s)
- Qiaochu Zhu
- Department of Chemistry, Iowa State University, Ames, IA 50011, United States
| | - Emily A Smith
- Department of Chemistry, Iowa State University, Ames, IA 50011, United States.
| |
Collapse
|
55
|
Abstract
The S100B protein is an intra- and extracellular signaling protein that
plays a role in a multitude of cellular processes and abnormal S100B is
associated with various neurological diseases and cancers. S100B recognizes and
binds effector proteins in a calcium-dependent manner. S100B has been shown to
interact with the actin capping protein CapZ, protein kinase C, Hdm2 and 4, RAGE
receptor, and p53, among others. These protein partners interact with
a common area on the S100B protein surface, validating the method of using the
consensus sequence for S100B target search. In addition, each S100B target
protein distinguishes itself by additional contacts with S100B. This perspective
suggests that the combination of sequence homology search and structural
analysis promises to identify newer S100B-binding partners beyond the use of the
consensus sequence alone as the given example in the XPB subunit of the TFIIH
general transcription factor. XPB is a helicase required for both transcription
and DNA repair. Inherited xpb mutations are associated with human disease
Xeroderma Pigmentasum, Cockayne syndrome, and trichothiodystrophy. S100B protein
is likely associated with much more biological pathways and processes. We
believe that S100B will attract more and more attentions in the scientific
community and S100B related studies will have important implications in human
health and medicine.
Collapse
Affiliation(s)
- K D Prez
- Department of Biochemistry, University of California Riverside, 900 University Ave, Riverside, California, USA
| | - L Fan
- Department of Biochemistry, University of California Riverside, 900 University Ave, Riverside, California, USA
| |
Collapse
|
56
|
Dowarha D, Chou RH, Yu C. S100B as an Antagonist To Interfere with the Interface Area Flanked by S100A11 and RAGE V Domain. ACS OMEGA 2018; 3:9689-9698. [PMID: 31459098 PMCID: PMC6644751 DOI: 10.1021/acsomega.8b00922] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 08/08/2018] [Indexed: 05/03/2023]
Abstract
The Ca2+-sensing protein S100A11 of the S100 family is an important mediator of numerous biological functions and pathological conditions including cancer. The receptor for advanced glycation end products (RAGE) has been well accepted as the major receptor for several S100 family members. Here, we take the S100B protein as an antagonist to interfere with the interaction flanked by S100A11 and the RAGE V domain. We employed NMR spectroscopy to describe the interactions between the S100A11 and S100B proteins. 1H-15N heteronuclear single-quantum correlation-NMR titrations showed the potential binding dynamics of S100A11 and S100B interactions. In the HADDOCK program, we constructed the S100A11-S100B heterodimer complex that was then superimposed with the S100A11-S100B complex structure in the same orientation as the S100A11-RAGE V domain complex. This overlay analysis showed that S100B could interfere in the binding section of S100A11 and the RAGE V domain. Additionally, water-soluble tetrazolium-1 assay provided a functional read-out of the effects of these proteins in an in vitro cancer model. Our study establishes that the development of an S100B antagonist could perform a vital part in the treatment of S100- and RAGE-dependent human diseases.
Collapse
Affiliation(s)
- Deepu Dowarha
- Department
of Chemistry, National Tsing Hua University, 101, Section 2, Kuang-Fu Road, Hsinchu 30013, Taiwan
| | - Ruey-Hwang Chou
- Graduate
Institute of Cancer Biology and Center for Molecular Medicine, China Medical University, No. 91, Hsueh-Shih Road, Taichung 40402, Taiwan
| | - Chin Yu
- Department
of Chemistry, National Tsing Hua University, 101, Section 2, Kuang-Fu Road, Hsinchu 30013, Taiwan
- E-mail: . Fax: 886-35-711082
| |
Collapse
|
57
|
Jangde N, Ray R, Sinha S, Rana K, Singh SK, Khandagale P, Acharya N, Rai V. Cysteine mediated disulfide bond formation in RAGE V domain facilitates its functionally relevant dimerization. Biochimie 2018; 154:55-61. [PMID: 30076903 DOI: 10.1016/j.biochi.2018.07.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 07/30/2018] [Indexed: 02/06/2023]
Abstract
Receptor for Advanced Glycation End product (RAGE) is a multiligand receptor implicated in diverse pathological conditions such as diabetes, atherosclerosis, cancer and neural diseases. Extracellular, RAGE consists of V, C1 and C2 domains. Here, we show RAGE exists as a monomer in equilibrium with a fraction of a covalently linked dimer of monomers via its V domain through cysteine. In order to understand the functional implication of this dimer, we examined the binding capacity and functional potential of RAGE dimer via advanced glycation end products (AGEs) which shows enhanced binding capacity towards V domain, ERK phosphorylation, cytokine release and actin polymerization ability of the dimeric form for AGEs compared with the reduced monomeric form. Our data, suggests that the dimeric state of RAGE controls its function and ligand mediated signaling which may play important role in RAGE mediated various diseases.
Collapse
Affiliation(s)
- Nitish Jangde
- Laboratory of Vascular Immunology, Institute of Life Sciences, Bhubaneswar, 751023, India; Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Rashmi Ray
- Laboratory of Vascular Immunology, Institute of Life Sciences, Bhubaneswar, 751023, India; Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Sunita Sinha
- Laboratory of Vascular Immunology, Institute of Life Sciences, Bhubaneswar, 751023, India
| | - Khokan Rana
- Laboratory of Vascular Immunology, Institute of Life Sciences, Bhubaneswar, 751023, India
| | - Satyendra Kumar Singh
- Laboratory of Vascular Immunology, Institute of Life Sciences, Bhubaneswar, 751023, India
| | - Prashant Khandagale
- Laboratory of Genomic Instability and Diseases, Institute of Life Sciences, Bhubaneswar, 751023, India
| | - Narottam Acharya
- Laboratory of Genomic Instability and Diseases, Institute of Life Sciences, Bhubaneswar, 751023, India
| | - Vivek Rai
- Laboratory of Vascular Immunology, Institute of Life Sciences, Bhubaneswar, 751023, India.
| |
Collapse
|
58
|
Impaired oligodendrogenesis and myelination by elevated S100B levels during neurodevelopment. Neuropharmacology 2018; 129:69-83. [DOI: 10.1016/j.neuropharm.2017.11.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 10/22/2017] [Accepted: 11/03/2017] [Indexed: 11/23/2022]
|
59
|
Hagmeyer S, Cristóvão JS, Mulvihill JJE, Boeckers TM, Gomes CM, Grabrucker AM. Zinc Binding to S100B Affords Regulation of Trace Metal Homeostasis and Excitotoxicity in the Brain. Front Mol Neurosci 2018; 10:456. [PMID: 29386995 PMCID: PMC5776125 DOI: 10.3389/fnmol.2017.00456] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 12/26/2017] [Indexed: 12/16/2022] Open
Abstract
Neuronal metal ions such as zinc are essential for brain function. In particular synaptic processes are tightly related to metal and protein homeostasis, for example through extracellular metal-binding proteins. One such protein is neuronal S100B, a calcium and zinc binding damage-associated molecular pattern (DAMP), whose chronic upregulation is associated with aging, Alzheimer’s disease (AD), motor neuron disease and traumatic brain injury (TBI). Despite gained insights on the structure of S100B, it remains unclear how its calcium and zinc binding properties regulate its function on cellular level. Here we report a novel role of S100B in trace metal homeostasis, in particular the regulation of zinc levels in the brain. Our results show that S100B at increased extracellular levels is not toxic, persists at high levels, and is taken up into neurons, as shown by cell culture and biochemical analysis. Combining protein bioimaging and zinc quantitation, along with a zinc-binding impaired S100B variant, we conclude that S100B effectively scavenges zinc ions through specific binding, resulting in a redistribution of the intracellular zinc pool. Our results indicate that scavenging of zinc by increased levels of S100B affects calcium levels in vitro. Thereby S100B is able to mediate the cross talk between calcium and zinc homeostasis. Further, we investigated a possible new neuro-protective role of S100B in excitotoxicity via its effects on calcium and zinc homeostasis. Exposure of cells to zinc-S100B but not the zinc-binding impaired S100B results in an inhibition of excitotoxicity. We conclude that in addition to its known functions, S100B acts as sensor and regulator of elevated zinc levels in the brain and this metal-buffering activity is tied to a neuroprotective role.
Collapse
Affiliation(s)
- Simone Hagmeyer
- WG Molecular Analysis of Synaptopathies, Department of Neurology, Neurocenter of Ulm University, Ulm, Germany.,Cellular Neurobiology and Neuro-Nanotechnology Lab, Department of Biological Sciences, University of Limerick, Limerick, Ireland.,Bernal Institute, University of Limerick, Limerick, Ireland
| | - Joana S Cristóvão
- Biosystems and Integrative Sciences Institute, Faculdade de Ciências, Universidade de Lisboa, and Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - John J E Mulvihill
- Bernal Institute, University of Limerick, Limerick, Ireland.,Health Research Institute (HRI), University of Limerick, Limerick, Ireland
| | - Tobias M Boeckers
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | - Cláudio M Gomes
- Biosystems and Integrative Sciences Institute, Faculdade de Ciências, Universidade de Lisboa, and Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - Andreas M Grabrucker
- Cellular Neurobiology and Neuro-Nanotechnology Lab, Department of Biological Sciences, University of Limerick, Limerick, Ireland.,Bernal Institute, University of Limerick, Limerick, Ireland.,Health Research Institute (HRI), University of Limerick, Limerick, Ireland
| |
Collapse
|
60
|
Abstract
The receptor for advanced glycation end-products (RAGE) is a multiligand pattern recognition receptor implicated in diverse chronic inflammatory states. RAGE binds and mediates the cellular response to a range of damage-associated molecular pattern molecules (DAMPs) including AGEs, HMGB1, S100s, and DNA. RAGE can also act as an innate immune sensor of microbial pathogen-associated molecular pattern molecules (PAMPs) including bacterial endotoxin, respiratory viruses, and microbial DNA. RAGE is expressed at low levels under normal physiology, but it is highly upregulated under chronic inflammation because of the accumulation of various RAGE ligands. Blocking RAGE signaling in cell and animal models has revealed that targeting RAGE impairs inflammation and progression of diabetic vascular complications, cardiovascular disease (CVD), and cancer progression and metastasis. The clinical relevance of RAGE in inflammatory disease is being demonstrated in emerging clinical trials of novel small-molecule RAGE inhibitors.
Collapse
Affiliation(s)
- Barry I Hudson
- Department of Cell Biology, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida 33136, USA; .,University of Miami Sylvester Comprehensive Cancer Center, Miami, Florida 33136, USA
| | - Marc E Lippman
- University of Miami Sylvester Comprehensive Cancer Center, Miami, Florida 33136, USA.,Department of Medicine, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida 33136, USA;
| |
Collapse
|
61
|
Azizan N, Suter MA, Liu Y, Logsdon CD. RAGE maintains high levels of NFκB and oncogenic Kras activity in pancreatic cancer. Biochem Biophys Res Commun 2017; 493:592-597. [PMID: 28867179 DOI: 10.1016/j.bbrc.2017.08.147] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 08/30/2017] [Indexed: 01/31/2023]
Abstract
Oncogenic KRas activity is central to several cancer types including pancreatic ductal adenocarcinoma (PDAC) but has been determined to be "undruggable". Recent studies have indicated that oncogenic KRas is not constitutively active but relies on a feed-forward stimulatory mechanism involving NFκB mediated inflammation. In the current study, we investigated the role of the receptor for advanced glycation end-products (RAGE) in maintaining oncogenic signaling in PDAC. We observed that there was a shift in the levels of specific RAGE isoforms and altered cellular localization in PDAC. Furthermore, inhibition of RAGE using a pharmacological antagonist, FPS-ZM1, or a blocking antibody, decreased phosphorylation of IKBα and inhibited Erk activity down-stream of Kras in PDAC cell lines. In vivo, inhibition of RAGE using FPS-ZM1 reduced the growth of PDAC syngeneic orthotopic xenografts and prolonged survival. These data indicate that RAGE plays a central role in maintaining inflammatory signaling in PDAC that benefits tumor growth. These observations support the development of approaches to inhibit the carcinogenic actions of Kras indirectly by blocking the mechanisms which maintain its activity.
Collapse
Affiliation(s)
- Nancy Azizan
- Department of Cancer Biology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Melissa A Suter
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, 77030, USA
| | - Yan Liu
- Department of Cancer Biology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Craig D Logsdon
- Department of Cancer Biology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
62
|
Bongarzone S, Savickas V, Luzi F, Gee AD. Targeting the Receptor for Advanced Glycation Endproducts (RAGE): A Medicinal Chemistry Perspective. J Med Chem 2017; 60:7213-7232. [PMID: 28482155 PMCID: PMC5601361 DOI: 10.1021/acs.jmedchem.7b00058] [Citation(s) in RCA: 224] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
![]()
The
receptor for advanced glycation endproducts (RAGE) is an ubiquitous,
transmembrane, immunoglobulin-like receptor that exists in multiple
isoforms and binds to a diverse range of endogenous extracellular
ligands and intracellular effectors. Ligand binding at the extracellular
domain of RAGE initiates a complex intracellular signaling cascade,
resulting in the production of reactive oxygen species (ROS), immunoinflammatory
effects, cellular proliferation, or apoptosis with concomitant upregulation
of RAGE itself. To date, research has mainly focused on the correlation
between RAGE activity and pathological conditions, such as cancer,
diabetes, cardiovascular diseases, and neurodegeneration. Because
RAGE plays a role in many pathological disorders, it has become an
attractive target for the development of inhibitors at the extracellular
and intracellular domains. This review describes the role of endogenous
RAGE ligands/effectors in normo- and pathophysiological processes,
summarizes the current status of exogenous small-molecule inhibitors
of RAGE and concludes by identifying key strategies for future therapeutic
intervention.
Collapse
Affiliation(s)
- Salvatore Bongarzone
- Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners , St. Thomas' Hospital, London, SE1 7EH, United Kingdom
| | - Vilius Savickas
- Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners , St. Thomas' Hospital, London, SE1 7EH, United Kingdom
| | - Federico Luzi
- Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners , St. Thomas' Hospital, London, SE1 7EH, United Kingdom
| | - Antony D Gee
- Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners , St. Thomas' Hospital, London, SE1 7EH, United Kingdom
| |
Collapse
|
63
|
Braun M, Vaibhav K, Saad NM, Fatima S, Vender JR, Baban B, Hoda MN, Dhandapani KM. White matter damage after traumatic brain injury: A role for damage associated molecular patterns. Biochim Biophys Acta Mol Basis Dis 2017; 1863:2614-2626. [PMID: 28533056 DOI: 10.1016/j.bbadis.2017.05.020] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 05/09/2017] [Accepted: 05/16/2017] [Indexed: 12/12/2022]
Abstract
Traumatic brain injury (TBI) is a leading cause of mortality and long-term morbidity worldwide. Despite decades of pre-clinical investigation, therapeutic strategies focused on acute neuroprotection failed to improve TBI outcomes. This lack of translational success has necessitated a reassessment of the optimal targets for intervention, including a heightened focus on secondary injury mechanisms. Chronic immune activation correlates with progressive neurodegeneration for decades after TBI; however, significant challenges remain in functionally and mechanistically defining immune activation after TBI. In this review, we explore the burgeoning evidence implicating the acute release of damage associated molecular patterns (DAMPs), such as adenosine 5'-triphosphate (ATP), high mobility group box protein 1 (HMGB1), S100 proteins, and hyaluronic acid in the initiation of progressive neurological injury, including white matter loss after TBI. The role that pattern recognition receptors, including toll-like receptor and purinergic receptors, play in progressive neurological injury after TBI is detailed. Finally, we provide support for the notion that resident and infiltrating macrophages are critical cellular targets linking acute DAMP release with adaptive immune responses and chronic injury after TBI. The therapeutic potential of targeting DAMPs and barriers to clinical translational, in the context of TBI patient management, are discussed.
Collapse
Affiliation(s)
- Molly Braun
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Kumar Vaibhav
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA, United States; Department of Medical Laboratory, Imaging & Radiologic Sciences, College of Allied Health Science, Augusta University, Augusta, GA, United States
| | - Nancy M Saad
- Department of Oral Biology, Dental College of Georgia, Augusta University, Augusta, GA, United States
| | - Sumbul Fatima
- Department of Medical Laboratory, Imaging & Radiologic Sciences, College of Allied Health Science, Augusta University, Augusta, GA, United States
| | - John R Vender
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Babak Baban
- Department of Oral Biology, Dental College of Georgia, Augusta University, Augusta, GA, United States; Department of Neurology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Md Nasrul Hoda
- Department of Medical Laboratory, Imaging & Radiologic Sciences, College of Allied Health Science, Augusta University, Augusta, GA, United States; Department of Neurology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Krishnan M Dhandapani
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA, United States.
| |
Collapse
|
64
|
Zhou H, Ding L, Wu Z, Cao X, Zhang Q, Lin L, Bian JS. Hydrogen sulfide reduces RAGE toxicity through inhibition of its dimer formation. Free Radic Biol Med 2017; 104:262-271. [PMID: 28108276 DOI: 10.1016/j.freeradbiomed.2017.01.026] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2016] [Revised: 12/30/2016] [Accepted: 01/16/2017] [Indexed: 12/11/2022]
Abstract
RAGE is important in the development of neurodegenerative diseases. The present study was designed to investigate the effect of hydrogen sulfide (H2S, an endogenous gaseous mediator) on the cytotoxicity caused by RAGE activation during the chronic oxidative stress. Aβ1-42 decreased cell viability and induced cell senescence in SH-SY5Y cells. Treatment with advanced glycation end products (AGEs) induced cell injury in HEK293 cells stably expressing RAGE (HEK293-RAGE) and stimulated inflammatory responses in SH-SY5Y cells. Pretreatment of SH-SY5Y cells with an H2S donor, NaHS, significantly attenuated the above harmful effects caused by Aβ1-42 or AGEs. Western blotting analysis shows that oxidative stress enhanced RAGE protein expression which was attenuated by either NaHS or over-expression of cystathionine β-synthase (CBS), a critical enzyme for producing H2S in brain cells. Both Western blots and split GFP complementation analysis demonstrate that NaHS reduced H2O2-enhanced RAGE dimerization. Immunofluorescence analysis shows that H2O2 up-regulated the membrane expression of wild-type RAGE. However, H2O2-enhanced expression of the RAGE harboring C259S/C310S double mutation (DM-RAGE) was observed in the endoplasmic reticulum. Treatment with NaHS attenuated the effects of H2O2 on the protein expression of WT-RAGE, but not that of DM-RAGE. Cycloheximide chase and ubiquitination assays show that NaHS reduced the half-life of WT-RAGE to a similar level of DM-RAGE. S-sulfhydration assay with the tag-switch technique demonstrate that H2S may directly S-sulfhydrate the C259/C301 residues. Our data suggest that H2S reduces RAGE dimer formation and impairs its membrane stability. The lowered plasma membrane abundance of RAGE therefore helps to protect cells against various RAGE mediated pathological effects.
Collapse
Affiliation(s)
- Hong Zhou
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600 Singapore, Singapore
| | - Lei Ding
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600 Singapore, Singapore
| | - Zhiyuan Wu
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600 Singapore, Singapore; Life Science Institute, National University of Singapore, Singapore
| | - Xu Cao
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600 Singapore, Singapore
| | - Qichun Zhang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600 Singapore, Singapore
| | - Li Lin
- Laboratory of Cardiovascular Sciences, National Institute on Aging, Baltimore, MD 21224, USA
| | - Jin-Song Bian
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600 Singapore, Singapore; Life Science Institute, National University of Singapore, Singapore.
| |
Collapse
|
65
|
Anggayasti WL, Mancera RL, Bottomley S, Helmerhorst E. The self-association of HMGB1 and its possible role in the binding to DNA and cell membrane receptors. FEBS Lett 2017; 591:282-294. [PMID: 28027393 DOI: 10.1002/1873-3468.12545] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 12/04/2016] [Accepted: 12/22/2016] [Indexed: 01/09/2023]
Abstract
High mobility group box 1 (HMGB1), a chromatin protein, interacts with DNA and controls gene expression. However, when HMGB1 is released from apoptotic or damaged cells, it triggers proinflammatory reactions by interacting with various receptors, mainly receptor for advanced glycation end-products (RAGE) and toll-like receptors (TLRs). The self-association of HMGB1 has been found to be crucial for its DNA-related biological functions. It is influenced by several factors, such as ionic strength, pH, specific divalent metal cations, redox environment and acetylation. This self-association may also play a role in the interaction with RAGE and TLRs and the concomitant inflammatory responses. Future studies should address the potential role of HMGB1 self-association on its interactions with DNA, RAGE and TLRs, as well as the influence of physicochemical factors in different cellular environments on these interactions.
Collapse
Affiliation(s)
- Wresti L Anggayasti
- School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, Australia
| | - Ricardo L Mancera
- School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, Australia
| | - Steve Bottomley
- School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, Australia
| | - Erik Helmerhorst
- School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, Australia
| |
Collapse
|
66
|
Kim I, Lee KO, Yun YJ, Jeong JY, Kim EH, Cheong H, Ryu KS, Kim NK, Suh JY. Biophysical characterization of Ca 2+-binding of S100A5 and Ca 2+-induced interaction with RAGE. Biochem Biophys Res Commun 2016; 483:332-338. [PMID: 28017722 DOI: 10.1016/j.bbrc.2016.12.143] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 12/21/2016] [Indexed: 12/12/2022]
Abstract
S100A5 is a calcium-binding protein of S100 family, which represents a major ligand to the receptor for advanced glycation end product (RAGE), a pattern recognition receptor engaged in diverse pathological processes. Here we have characterized calcium binding of S100A5 and the complex formation between S100A5 and RAGE using calorimetry and NMR spectroscopy. S100A5 binds to calcium ions in a sequential manner with the equilibrium dissociation constants (KD) of 1.3 μM and 3.5 μM, which corresponds to the calcium-binding at the C-terminal and N-terminal EF-hands. Upon calcium binding, S100A5 interacts with the V domain of RAGE (RAGE-v) to form a heterotrimer (KD ∼5.9 μM) that is distinct among the S100 family proteins. Chemical shift perturbation data from NMR titration experiments indicates that S100A5 employs the periphery of the dimer interface to interact with RAGE-v. Distinct binding mode and stoichiometry of RAGE against different S100 family proteins could be important to modulate diverse RAGE signaling.
Collapse
Affiliation(s)
- Iktae Kim
- Department of Agricultural Biotechnology, Seoul National University, 599 Gwanak-ro, Gwanak-gu, Seoul 151-921, South Korea.
| | - Ko On Lee
- Department of Agricultural Biotechnology, Seoul National University, 599 Gwanak-ro, Gwanak-gu, Seoul 151-921, South Korea.
| | - Young-Joo Yun
- Department of Agricultural Biotechnology, Seoul National University, 599 Gwanak-ro, Gwanak-gu, Seoul 151-921, South Korea
| | - Jea Yeon Jeong
- Department of Agricultural Biotechnology, Seoul National University, 599 Gwanak-ro, Gwanak-gu, Seoul 151-921, South Korea
| | - Eun-Hee Kim
- Division of Magnetic Resonance, Korea Basic Science Institute, 16 Yeongudanji-Ro, Ochang, Chungbuk 363-883, South Korea
| | - Haekap Cheong
- Division of Magnetic Resonance, Korea Basic Science Institute, 16 Yeongudanji-Ro, Ochang, Chungbuk 363-883, South Korea
| | - Kyoung-Seok Ryu
- Division of Magnetic Resonance, Korea Basic Science Institute, 16 Yeongudanji-Ro, Ochang, Chungbuk 363-883, South Korea
| | - Nak-Kyoon Kim
- Advanced Analysis Center, Korea Institute of Science and Technology, Seoul 02792, South Korea
| | - Jeong-Yong Suh
- Department of Agricultural Biotechnology, Seoul National University, 599 Gwanak-ro, Gwanak-gu, Seoul 151-921, South Korea.
| |
Collapse
|
67
|
Syed A, Zhu Q, Smith EA. Ligand binding affinity and changes in the lateral diffusion of receptor for advanced glycation endproducts (RAGE). BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2016; 1858:3141-3149. [DOI: 10.1016/j.bbamem.2016.10.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 09/22/2016] [Accepted: 10/03/2016] [Indexed: 12/16/2022]
|
68
|
Blocking the interaction between S100A9 and RAGE V domain using CHAPS molecule: A novel route to drug development against cell proliferation. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2016; 1864:1558-69. [DOI: 10.1016/j.bbapap.2016.08.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 08/03/2016] [Accepted: 08/09/2016] [Indexed: 11/18/2022]
|
69
|
Abstract
The receptor for advanced glycation end products (RAGE) is a novel protein increasingly studied in the pathogenesis of type 1 diabetes (T1D). RAGE is expressed by several immune cell types, including T cells, antigen-presenting cells, endothelial cells, and the endocrine cells of the pancreatic islets. RAGE binds various ligands including advanced glycation end products (AGEs), high-mobility group box protein 1 (HMGB1), S100 proteins, β-amyloid, β-sheet fibrils, and lipopolysaccharide. AGEs are a particularly interesting ligand because their exogenous introduction into the body can be accelerated by the consumption of AGE-rich processed foods. This review will detail RAGE isoforms and its ligands and discuss how RAGE binding on the aforementioned cells could be linked to T1D pathogenesis.
Collapse
Affiliation(s)
- Sherman S Leung
- Glycation and Diabetes, Mater Research Institute, Translational Research Institute, The University of Queensland, 37 Kent St, Woolloongabba, Brisbane, Queensland, Australia
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Josephine M Forbes
- Glycation and Diabetes, Mater Research Institute, Translational Research Institute, The University of Queensland, 37 Kent St, Woolloongabba, Brisbane, Queensland, Australia.
- Mater Clinical School, School of Medicine, The University of Queensland, Brisbane, Queensland, Australia.
| | - Danielle J Borg
- Glycation and Diabetes, Mater Research Institute, Translational Research Institute, The University of Queensland, 37 Kent St, Woolloongabba, Brisbane, Queensland, Australia
| |
Collapse
|
70
|
HMGB1, IL-1α, IL-33 and S100 proteins: dual-function alarmins. Cell Mol Immunol 2016; 14:43-64. [PMID: 27569562 PMCID: PMC5214941 DOI: 10.1038/cmi.2016.34] [Citation(s) in RCA: 344] [Impact Index Per Article: 38.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 05/16/2016] [Accepted: 05/17/2016] [Indexed: 02/08/2023] Open
Abstract
Our immune system is based on the close collaboration of the innate and adaptive immune systems for the rapid detection of any threats to the host. Recognition of pathogen-derived molecules is entrusted to specific germline-encoded signaling receptors. The same receptors have now also emerged as efficient detectors of misplaced or altered self-molecules that signal tissue damage and cell death following, for example, disruption of the blood supply and subsequent hypoxia. Many types of endogenous molecules have been shown to provoke such sterile inflammatory states when released from dying cells. However, a group of proteins referred to as alarmins have both intracellular and extracellular functions which have been the subject of intense research. Indeed, alarmins can either exert beneficial cell housekeeping functions, leading to tissue repair, or provoke deleterious uncontrolled inflammation. This group of proteins includes the high-mobility group box 1 protein (HMGB1), interleukin (IL)-1α, IL-33 and the Ca2+-binding S100 proteins. These dual-function proteins share conserved regulatory mechanisms, such as secretory routes, post-translational modifications and enzymatic processing, that govern their extracellular functions in time and space. Release of alarmins from mesenchymal cells is a highly relevant mechanism by which immune cells can be alerted of tissue damage, and alarmins play a key role in the development of acute or chronic inflammatory diseases and in cancer development.
Collapse
|
71
|
Xue J, Manigrasso M, Scalabrin M, Rai V, Reverdatto S, Burz DS, Fabris D, Schmidt AM, Shekhtman A. Change in the Molecular Dimension of a RAGE-Ligand Complex Triggers RAGE Signaling. Structure 2016; 24:1509-22. [PMID: 27524199 DOI: 10.1016/j.str.2016.06.021] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 05/20/2016] [Accepted: 06/16/2016] [Indexed: 01/13/2023]
Abstract
The weak oligomerization exhibited by many transmembrane receptors has a profound effect on signal transduction. The phenomenon is difficult to characterize structurally due to the large sizes of and transient interactions between monomers. The receptor for advanced glycation end products (RAGE), a signaling molecule central to the induction and perpetuation of inflammatory responses, is a weak constitutive oligomer. The RAGE domain interaction surfaces that mediate homo-dimerization were identified by combining segmental isotopic labeling of extracellular soluble RAGE (sRAGE) and nuclear magnetic resonance spectroscopy with chemical cross-linking and mass spectrometry. Molecular modeling suggests that two sRAGE monomers orient head to head forming an asymmetric dimer with the C termini directed toward the cell membrane. Ligand-induced association of RAGE homo-dimers on the cell surface increases the molecular dimension of the receptor, recruiting Diaphanous 1 (DIAPH1) and activating signaling pathways.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/chemistry
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/metabolism
- Amino Acid Sequence
- Animals
- Antigens, Neoplasm/chemistry
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/metabolism
- Bacterial Proteins/genetics
- Bacterial Proteins/metabolism
- Binding Sites
- Cross-Linking Reagents/chemistry
- Formins
- Gene Expression
- Genes, Reporter
- Green Fluorescent Proteins/genetics
- Green Fluorescent Proteins/metabolism
- HEK293 Cells
- Humans
- Ligands
- Luminescent Proteins/genetics
- Luminescent Proteins/metabolism
- Maleimides/chemistry
- Mitogen-Activated Protein Kinases/chemistry
- Mitogen-Activated Protein Kinases/genetics
- Mitogen-Activated Protein Kinases/metabolism
- Molecular Docking Simulation
- Nuclear Magnetic Resonance, Biomolecular
- Protein Binding
- Protein Interaction Domains and Motifs
- Protein Structure, Secondary
- Recombinant Proteins/chemistry
- Recombinant Proteins/genetics
- Recombinant Proteins/metabolism
- Sequence Alignment
- Sequence Homology, Amino Acid
- Signal Transduction
- Thermodynamics
Collapse
Affiliation(s)
- Jing Xue
- Department of Chemistry, State University of New York at Albany, Albany, NY 12222, USA
| | | | - Matteo Scalabrin
- Department of Chemistry, State University of New York at Albany, Albany, NY 12222, USA
| | - Vivek Rai
- Institute of Life Sciences, Bhubaneswar, Odisha 751023, India
| | - Sergey Reverdatto
- Department of Chemistry, State University of New York at Albany, Albany, NY 12222, USA
| | - David S Burz
- Department of Chemistry, State University of New York at Albany, Albany, NY 12222, USA
| | - Daniele Fabris
- Department of Chemistry, State University of New York at Albany, Albany, NY 12222, USA
| | - Ann Marie Schmidt
- New York University, Langone Medical Center, New York, NY 10016, USA
| | - Alexander Shekhtman
- Department of Chemistry, State University of New York at Albany, Albany, NY 12222, USA.
| |
Collapse
|
72
|
Gilston BA, Skaar EP, Chazin WJ. Binding of transition metals to S100 proteins. SCIENCE CHINA. LIFE SCIENCES 2016; 59:792-801. [PMID: 27430886 PMCID: PMC5123432 DOI: 10.1007/s11427-016-5088-4] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Accepted: 05/02/2016] [Indexed: 12/13/2022]
Abstract
The S100 proteins are a unique class of EF-hand Ca(2+) binding proteins distributed in a cell-specific, tissue-specific, and cell cycle-specific manner in humans and other vertebrates. These proteins are distinguished by their distinctive homodimeric structure, both intracellular and extracellular functions, and the ability to bind transition metals at the dimer interface. Here we summarize current knowledge of S100 protein binding of Zn(2+), Cu(2+) and Mn(2+) ions, focusing on binding affinities, conformational changes that arise from metal binding, and the roles of transition metal binding in S100 protein function.
Collapse
Affiliation(s)
- Benjamin A Gilston
- Departments of Biochemistry and Chemistry, and Center for Structural Biology, Vanderbilt University, Nashville, TN, 37232-9717, USA
| | - Eric P Skaar
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232-2561, USA
| | - Walter J Chazin
- Departments of Biochemistry and Chemistry, and Center for Structural Biology, Vanderbilt University, Nashville, TN, 37232-9717, USA.
| |
Collapse
|
73
|
Anggayasti WL, Mancera RL, Bottomley S, Helmerhorst E. The effect of physicochemical factors on the self-association of HMGB1: A surface plasmon resonance study. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2016; 1864:1620-9. [PMID: 27476953 DOI: 10.1016/j.bbapap.2016.07.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 07/22/2016] [Accepted: 07/26/2016] [Indexed: 11/17/2022]
Abstract
HMGB1 triggers proinflammatory reactions by interacting extracellularly with various receptors. HMGB1 also acts in the nucleus by interacting with DNA and controlling DNA transcription, a process which involves its self-association. The self-association of HMGB1 was characterized using surface plasmon resonance (SPR). A dimer/tetramer binding model was developed that provided a good fit to the SPR sensorgrams and enabled the kinetics of self-association of different HMGB1 oligomers to be evaluated under a variety of physicochemical conditions. The formation of HMGB1 tetramers, and not dimers, was strongly influenced by ionic strength. HMGB1 self-association increased as the pH was decreased from 7.4 to 4.8 but was abolished at pH4.0, suggesting the involvement of acidic amino acids of HMGB1 in its self-association. HMGB1 dimers were found to predominate in the absence of zinc, but addition of zinc promoted the formation of HMGB1 tetramers. More reducing conditions favored dimerization but diminished tetramer formation. In contrast, oxidizing conditions favored tetramer formation. Physicochemical factors modulate the extent of self-association of HMGB1. We speculate that HMGB1 dimers may preferentially bind DNA, whereas HMGB1 tetramers may promote inflammatory responses by binding to RAGE and TLRs. The self-association of HMGB1, regulated by variations of physicochemical factors, may influence its roles in DNA rearrangement and regulation of pathophysiological diseases.
Collapse
Affiliation(s)
- Wresti L Anggayasti
- School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, GPO Box U1987, Perth, WA 6845, Australia.
| | - Ricardo L Mancera
- School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, GPO Box U1987, Perth, WA 6845, Australia
| | - Steven Bottomley
- School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, GPO Box U1987, Perth, WA 6845, Australia
| | - Erik Helmerhorst
- School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, GPO Box U1987, Perth, WA 6845, Australia.
| |
Collapse
|
74
|
AGE-RAGE interaction in the TGFβ2-mediated epithelial to mesenchymal transition of human lens epithelial cells. Glycoconj J 2016; 33:631-43. [PMID: 27263094 DOI: 10.1007/s10719-016-9686-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 05/18/2016] [Accepted: 05/24/2016] [Indexed: 01/12/2023]
Abstract
Basement membrane (BM) proteins accumulate chemical modifications with age. One such modification is glycation, which results in the formation of advanced glycation endproducts (AGEs). In a previous study, we reported that AGEs in the human lens capsule (BM) promote the TGFβ2-mediated epithelial-to-mesenchymal transition (EMT) of lens epithelial cells, which we proposed as a mechanism for posterior capsule opacification (PCO) or secondary cataract formation. In this study, we investigated the role of a receptor for AGEs (RAGE) in the TGFβ2-mediated EMT in a human lens epithelial cell line (FHL124). RAGE was present in FHL124 cells, and its levels were unaltered in cells cultured on either native or AGE-modified BM or upon treatment with TGFβ2. RAGE overexpression significantly enhanced the TGFβ2-mediated EMT responses in cells cultured on AGE-modified BM compared with the unmodified matrix. In contrast, treatment of cells with a RAGE antibody or EN-RAGE (an endogenous ligand for RAGE) resulted in a significant reduction in the TGFβ2-mediated EMT response. This was accompanied by a reduction in TGFβ2-mediated Smad signaling and ROS generation. These results imply that the interaction of matrix AGEs with RAGE plays a role in the TGFβ2-mediated EMT of lens epithelial cells and suggest that the blockade of RAGE could be a strategy to prevent PCO and other age-associated fibrosis.
Collapse
|
75
|
Huang YK, Chou RH, Yu C. Tranilast Blocks the Interaction between the Protein S100A11 and Receptor for Advanced Glycation End Products (RAGE) V Domain and Inhibits Cell Proliferation. J Biol Chem 2016; 291:14300-14310. [PMID: 27226584 DOI: 10.1074/jbc.m116.722215] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Indexed: 01/13/2023] Open
Abstract
The human S100 calcium-binding protein A11 (S100A11) is a member of the S100 protein family. Once S100A11 proteins bind to calcium ions at EF-hand motifs, S100A11 changes its conformation, promoting interaction with target proteins. The receptor for advanced glycation end products (RAGE) consists of three extracellular domains, including the V domain, C1 domain, and C2 domain. In this case, the V domain is the target for mutant S100A11 (mS100A11) binding. RAGE binds to the ligands, resulting in cell proliferation, cell growth, and several signal transduction cascades. We used NMR and fluorescence spectroscopy to demonstrate the interactions between mS100A11and RAGE V domain. The tranilast molecule is a drug used for treating allergic disorders. We discovered that the RAGE V domain and tranilast would interact with mS100A11 by using (1)H-(15)N HSQC NMR titrations. According to the results, we obtained two binary complex models from the HADDOCK program, S100A11-RAGE V domain and S100A11-tranilast, respectively. We overlapped two binary complex models with the same orientation of S100A11 homodimer and demonstrated that tranilast would block the binding site between S100A11 and the RAGE V domain. We further utilized a water-soluble tetrazolium-1 assay to confirm this result. We think that the results will be potentially useful in the development of new anti-cancer drugs.
Collapse
Affiliation(s)
- Yen-Kai Huang
- Department of Chemistry, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Ruey-Hwang Chou
- Graduate Institute of Cancer Biology and Center for Molecular Medicine, China Medical University, Taichung 40454, Taiwan
| | - Chin Yu
- Department of Chemistry, National Tsing Hua University, Hsinchu 30013, Taiwan.
| |
Collapse
|
76
|
Kurzątkowska K, Jankowska A, Wysłouch-Cieszyńska A, Zhukova L, Puchalska M, Dehaen W, Radecka H, Radecki J. Voltammetric detection of the S100B protein using His-tagged RAGE domain immobilized onto a gold electrode modified with a dipyrromethene–Cu(II) complex and different diluents. J Electroanal Chem (Lausanne) 2016. [DOI: 10.1016/j.jelechem.2016.02.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
77
|
Polymorphisms in the receptor for advanced glycation end products gene are associated with susceptibility to drug-resistant epilepsy. Neurosci Lett 2016; 619:137-41. [PMID: 26828298 DOI: 10.1016/j.neulet.2016.01.043] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 12/21/2015] [Accepted: 01/25/2016] [Indexed: 01/14/2023]
|
78
|
Stroth N. A surface plasmon resonance-based method for monitoring interactions between G protein-coupled receptors and interacting proteins. J Biol Methods 2016; 3:e36. [PMID: 31453205 PMCID: PMC6706161 DOI: 10.14440/jbm.2016.97] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 01/13/2016] [Accepted: 01/14/2016] [Indexed: 12/18/2022] Open
Abstract
The present protocol describes a method by which interactions between G protein-coupled receptors (GPCR) and intracellular proteins can be monitored in real-time and without the use of exogenous labels. The method is based on surface plasmon resonance (SPR) and uses synthetic peptides as mimics of intracellular GPCR domains. These peptides are covalently immobilized onto sensor chips and brought into contact with putative interacting proteins in the flow cells of the SPR instrument. The method allows flexible experimental designs, rapid testing of hypotheses and quantitative analysis of interactions. Relative to other established methods, it provides both an alternative and a complementary approach with several key advantages. The present protocol describes the method step-by-step, using the interaction between the serotonin 5-HT7 receptor and the calcium-binding protein S100B as an example.
Collapse
Affiliation(s)
- Nikolas Stroth
- Center for Molecular Medicine, Karolinska Institute and University Hospital, Stockholm, Sweden
| |
Collapse
|
79
|
Changyaleket B, Xu H, Vetri F, Valyi-Nagy T, Paisansathan C, Chong ZZ, Pelligrino DA, Testai FD. Intracerebroventricular application of S100B selectively impairs pial arteriolar dilating function in rats. Brain Res 2016; 1634:171-178. [PMID: 26773687 DOI: 10.1016/j.brainres.2015.12.061] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 12/22/2015] [Accepted: 12/29/2015] [Indexed: 10/22/2022]
Abstract
S100B is an astrocyte-derived protein that can act through the receptor for advanced glycation endproducts (RAGE) to mediate either "trophic" or "toxic" responses. Its levels increase in many neurological conditions with associated microvascular dysregulation, such as subarachnoid hemorrhage (SAH) and traumatic brain injury. The role of S100B in the pathogenesis of microvasculopathy has not been addressed. This study was designed to examine whether S100B alters pial arteriolar vasodilating function. Rats were randomized to receive (1) artificial cerebrospinal fluid (aCSF), (2) exogenous S100B, and (3) exogenous S100B+the decoy soluble RAGE (sRAGE). S100B was infused intracerebroventricularly (icv) using an osmotic pump and its levels in the CSF were adjusted to achieve a concentration similar to what we observed in SAH. After 48 h of continuous icv infusion, a cranial window/intravital microscopy was applied to animals for evaluation of pial arteriolar dilating responses to sciatic nerve stimulation (SNS), hypercapnia, and topical suffusion of vasodilators including acetylcholine (ACh), s-nitroso-N-acetyl penicillamine (SNAP), or adenosine (ADO). Pial arteriolar dilating responses were calculated as the percentage change of arteriolar diameter in relation to baseline. The continuous S100B infusion for 48 h was associated with reduced responses to the neuronal-dependent vasodilator SNS (p<0.05) and the endothelial-dependent vasodilator ACh (p<0.05), compared to controls. The inhibitory effects of S100B were prevented by sRAGE. On the other hand, S100B did not alter the responses elicited by vascular smooth muscle cell-dependent vasodilators, namely hypercapnia, SNAP, or ADO. These findings indicate that S100B regulates neuronal and endothelial dependent cerebral arteriolar dilation and suggest that this phenomenon is mediated through RAGE-associated pathways.
Collapse
Affiliation(s)
- Benjarat Changyaleket
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, IL, United States
| | - Haoliang Xu
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, IL, United States; Department of Pathology, University of Illinois at Chicago, Chicago, IL, United States.
| | - Francesco Vetri
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, IL, United States
| | - Tibor Valyi-Nagy
- Department of Pathology, University of Illinois at Chicago, Chicago, IL, United States
| | | | - Zhao Zhong Chong
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, IL, United States
| | - Dale A Pelligrino
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, IL, United States
| | - Fernando D Testai
- Department of Neurology and Rehabilitation, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
80
|
Yamaguchi F, Tsuchiya M, Shimamoto S, Fujimoto T, Tokumitsu H, Tokuda M, Kobayashi R. Oxidative Stress Impairs the Stimulatory Effect of S100 Proteins on Protein Phosphatase 5 Activity. TOHOKU J EXP MED 2016; 240:67-78. [DOI: 10.1620/tjem.240.67] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
| | - Mitsumasa Tsuchiya
- Department of Signal Transduction Sciences, Faculty of Medicine, Kagawa University
| | - Seiko Shimamoto
- Laboratory of Oncology, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation
| | - Tomohito Fujimoto
- Department of Signal Transduction Sciences, Faculty of Medicine, Kagawa University
| | - Hiroshi Tokumitsu
- Department of Signal Transduction Sciences, Faculty of Medicine, Kagawa University
| | - Masaaki Tokuda
- Department of Cell Physiology, Faculty of Medicine, Kagawa University
| | - Ryoji Kobayashi
- Department of Signal Transduction Sciences, Faculty of Medicine, Kagawa University
| |
Collapse
|
81
|
Maltais JS, Simard E, Froehlich U, Denault JB, Gendron L, Grandbois M. iRAGE as a novel carboxymethylated peptide that prevents advanced glycation end product-induced apoptosis and endoplasmic reticulum stress in vascular smooth muscle cells. Pharmacol Res 2015; 104:176-85. [PMID: 26707030 DOI: 10.1016/j.phrs.2015.12.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 11/06/2015] [Accepted: 12/15/2015] [Indexed: 01/11/2023]
Abstract
Advanced glycation end-products (AGE) and the receptor for AGE (RAGE) have been linked to numerous diabetic vascular complications. RAGE activation promotes a self-sustaining state of chronic inflammation and has been shown to induce apoptosis in various cell types. Although previous studies in vascular smooth muscle cells (VSMC) showed that RAGE activation increases vascular calcification and interferes with their contractile phenotype, little is known on the potential of RAGE to induce apoptosis in VSMC. Using a combination of apoptotic assays, we showed that RAGE stimulation with its ligand CML-HSA promotes apoptosis of VSMC. The formation of stress granules and the increase in the level of the associated protein HuR point toward RAGE-dependent endoplasmic reticulum (ER) stress, which is proposed as a key contributor of RAGE-induced apoptosis in VSMC as it has been shown to promote cell death via numerous mechanisms, including up-regulation of caspase-9. Chronic NF-κB activation and modulation of Bcl-2 homologs are also suspected to contribute to RAGE-dependent apoptosis in VSMC. With the goal of reducing RAGE signaling and its detrimental impact on VSMC, we designed a RAGE antagonist (iRAGE) derived from the primary amino acid sequence of HSA. The resulting CML peptide was selected for the high glycation frequency of the primary sequence in the native protein in vivo. Pretreatment with iRAGE blocked 69.6% of the increase in NF-κB signaling caused by RAGE activation with CML-HSA after 48h. Preincubation with iRAGE was successful in reducing RAGE-induced apoptosis, as seen through enhanced cell survival by SPR and reduced PARP cleavage. Activation of executioner caspases was 63.5% lower in cells treated with iRAGE before stimulation with CML-HSA. To our knowledge, iRAGE is the first antagonist shown to block AGE-RAGE interaction and we propose the molecule as an initial candidate for drug discovery.
Collapse
Affiliation(s)
- Jean-Sébastien Maltais
- Département de pharmacologie et physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, 3001 12e Avenue Nord, Sherbrooke, Québec J1H 5N4, Canada
| | - Elie Simard
- Département de pharmacologie et physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, 3001 12e Avenue Nord, Sherbrooke, Québec J1H 5N4, Canada
| | - Ulrike Froehlich
- Département de pharmacologie et physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, 3001 12e Avenue Nord, Sherbrooke, Québec J1H 5N4, Canada
| | - Jean-Bernard Denault
- Département de pharmacologie et physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, 3001 12e Avenue Nord, Sherbrooke, Québec J1H 5N4, Canada
| | - Louis Gendron
- Département de pharmacologie et physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, 3001 12e Avenue Nord, Sherbrooke, Québec J1H 5N4, Canada
| | - Michel Grandbois
- Département de pharmacologie et physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, 3001 12e Avenue Nord, Sherbrooke, Québec J1H 5N4, Canada.
| |
Collapse
|
82
|
Wang J, Wang H, Shi J, Ding Y. Effects of bone marrow MSCs transfected with sRAGE on the intervention of HMGB1 induced immuno-inflammatory reaction. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:12028-12040. [PMID: 26722388 PMCID: PMC4680333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 09/24/2015] [Indexed: 06/05/2023]
Abstract
BACKGROUND High mobility group box chromosomal protein 1 (HMGB1) is an important proinflammatory molecule in many inflammatory disorders, but little is known about its role in acute liver failure (ALF). In this study, we determined the plasma and hepatic tissue levels of HMGB1 in a d-galactosamine-induced rat ALF model and investigated the effect of soluble receptor for advanced glycation end products (sRAGE) on ALF successfully. METHODS Male Sprague-Dawley rats were divided into five groups randomly. Group A (Control group, n=20) received administrated saline via peritoneal cavity. Group B (ALF group, n=20) induced by d-galactosamine (0.6 g/kg) via peritoneal cavity. Group C (HMGB1 group, n=20) were treated with HMGB1 recombination protein (200 μg/kg) via penile vein after ALF model induced. Group D (sRAGE group, n=20) received administrated sRAGE recombination protein (400 μg/kg) via penile vein after ALF model induced. Group E (sRAGE-MSC group, n=20) received 3 × 10(6) MSC transplantation which could maintain a stable expression of sRAGE via penile vein after ALF model induced. Liver function, level of cytokines and liver pathological changes were measured. RESULTS We determined that the plasma levels and hepatic tissue levels of HMGB1 were significant increased in ALF model (P<0.05). SRAGE group and sRAGE-MSC group could significantly prolong ALF rat survival time, as well as improve its liver functions, inflammatory cytokines level and hepatocytes necrosis. CONCLUSION SRAGE as a ligand decoy has illustrated largely beneficial effects on reducing immuno-inflammatory response, which holds promise for the identification of potential therapeutic targets and/or biomarkers of RAGE activity in ALF.
Collapse
Affiliation(s)
- Jun Wang
- Department of Hepatobiliary Surgery, Drum Tower Clinical Medical College of Nanjing Medical UniversityNanjing, China
| | - Hao Wang
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjing, China
- Jiangsu Province’s Key Medical Center for Digestive DiseaseNanjing, China
- Institute of Hepatobiliary Surgery, Nanjing UniversityNanjing, China
| | - Jiong Shi
- Department of Pathology, Drum Tower Clinical Medical College of Nanjing Medical UniversityNanjing, China
| | - Yitao Ding
- Department of Hepatobiliary Surgery, Drum Tower Clinical Medical College of Nanjing Medical UniversityNanjing, China
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjing, China
- Jiangsu Province’s Key Medical Center for Digestive DiseaseNanjing, China
- Institute of Hepatobiliary Surgery, Nanjing UniversityNanjing, China
| |
Collapse
|
83
|
Wu X, Wu J, Thompson CW, Li Y. Adaptive evolution of the MHC class III-encoded receptor RAGE in primates and murine rodents. Int J Immunogenet 2015; 42:461-8. [DOI: 10.1111/iji.12230] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 06/25/2015] [Accepted: 07/19/2015] [Indexed: 12/27/2022]
Affiliation(s)
- X. Wu
- Department of Nephrology; The Second Affiliated Hospital of Nanjing Medical University; Nanjing China
| | - J. Wu
- College of Animal Science and Technology; Sichuan Agricultural University; Yaan China
| | - C. W. Thompson
- Department of Ecology and Evolutionary Biology and Museum of Zoology; University of Michigan; Ann Arbor MI USA
| | - Y. Li
- College of Animal Science and Technology; Sichuan Agricultural University; Yaan China
| |
Collapse
|
84
|
Barateiro A, Afonso V, Santos G, Cerqueira JJ, Brites D, van Horssen J, Fernandes A. S100B as a Potential Biomarker and Therapeutic Target in Multiple Sclerosis. Mol Neurobiol 2015; 53:3976-3991. [DOI: 10.1007/s12035-015-9336-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 07/01/2015] [Indexed: 12/30/2022]
|
85
|
Medapati MR, Dahlmann M, Ghavami S, Pathak KA, Lucman L, Klonisch T, Hoang-Vu C, Stein U, Hombach-Klonisch S. RAGE Mediates the Pro-Migratory Response of Extracellular S100A4 in Human Thyroid Cancer Cells. Thyroid 2015; 25:514-27. [PMID: 25744544 DOI: 10.1089/thy.2014.0257] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Expression of the small calcium-binding protein S100A4 is associated with poor prognosis in patients with thyroid cancer (TC). The authors have previously shown that S100A4 is a target for relaxin and insulin-like peptide 3 signaling in TC cells and that S100A4 is secreted from human TC cells. Although the pro-migratory role of intracellular S100A4 in binding to non-muscle myosin is well known, this study investigated here whether extracellular S100A4 contributes to TC migration. METHODS Human cell lines of follicular, papillary, and undifferentiated thyroid cancer, primary patient TC cells, and TC tissues were utilized to discover the presence of the receptor of advanced glycation end products (RAGE) in TC cells and TC tissues. Fluorescence imaging, protein pull-down assays, Western blot, siRNA protein silencing, small GTPase inhibitors, cell proliferation, and cell migration assays were used to investigate the interaction of extracellular S100A4 with RAGE in promoting a TC migratory response. RESULTS It was demonstrated that RAGE served as receptor for extracellular S100A4 mediating cell migration in TC cells. The RAGE-mediated increase in cell migration was dependent on the intracellular RAGE signaling partner diaphanous-1 (Dia-1) and involved the activation of the small GTPases Cdc42 and RhoA. Although extracellular S100A4 consistently activated ERK signaling in TC cells, it was shown that ERK signaling was not mediated by RAGE and not essential for the migratory response in TC cells. CONCLUSION The data have identified the RAGE/Dia-1 signaling system as a mediator for the pro-migratory response of extracellular S100A4 in human TC. Thus, therapeutic targeting of the RAGE/Dia-1/small GTPases signaling may successfully reduce local invasion and metastasis in TC.
Collapse
Affiliation(s)
- Manoj Reddy Medapati
- 1 Department of Human Anatomy and Cell Science, University of Manitoba , Winnipeg, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
86
|
Jensen JL, Indurthi VSK, Neau DB, Vetter SW, Colbert CL. Structural insights into the binding of the human receptor for advanced glycation end products (RAGE) by S100B, as revealed by an S100B-RAGE-derived peptide complex. ACTA CRYSTALLOGRAPHICA. SECTION D, BIOLOGICAL CRYSTALLOGRAPHY 2015; 71:1176-83. [PMID: 25945582 PMCID: PMC4427201 DOI: 10.1107/s1399004715004216] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Accepted: 03/01/2015] [Indexed: 12/30/2022]
Abstract
S100B is a damage-associated molecular pattern protein that, when released into the extracellular milieu, triggers initiation of the inflammatory response through the receptor for advanced glycation end products (RAGE). Recognition of S100B is accomplished via the amino-terminal variable immunoglobulin domain (V-domain) of RAGE. To gain insights into this interaction, a complex between S100B and a 15-amino-acid peptide derived from residues 54-68 of the V-domain was crystallized. The X-ray crystal structure was solved to 2.55 Å resolution. There are two dimers of S100B and one peptide in the asymmetric unit. The binding interface of this peptide is compared with that found in the complex between S100B and the 12-amino-acid CapZ-derived peptide TRTK-12. This comparison reveals that although the peptides adopt completely different backbone structures, the residues buried at the interface interact with S100B in similar regions to form stable complexes. The binding affinities of S100B for the intact wild-type V-domain and a W61A V-domain mutant were determined to be 2.7 ± 0.5 and 1.3 ± 0.7 µM, respectively, using fluorescence titration experiments. These observations lead to a model whereby conformational flexibility in the RAGE receptor allows the adoption of a binding conformation for interaction with the stable hydrophobic groove on the surface of S100B.
Collapse
Affiliation(s)
- Jaime L. Jensen
- Department of Chemistry and Biochemistry, North Dakota State University, PO Box 6050, Fargo, ND 58108-6050, USA
| | - Venkata S. K. Indurthi
- Department of Pharmaceutical Sciences, North Dakota State University, PO Box 6050, Fargo, ND 58108-6050, USA
| | - David B. Neau
- NE-CAT, Bldg. 436E, Department of Chemistry and Chemical Biology, Cornell University, 9700 South Cass Avenue,, Argonne, IL 60439, USA
| | - Stefan W. Vetter
- Department of Pharmaceutical Sciences, North Dakota State University, PO Box 6050, Fargo, ND 58108-6050, USA
| | - Christopher L. Colbert
- Department of Chemistry and Biochemistry, North Dakota State University, PO Box 6050, Fargo, ND 58108-6050, USA
| |
Collapse
|
87
|
Downs CA, Kreiner LH, Johnson NM, Brown LA, Helms MN. Receptor for advanced glycation end-products regulates lung fluid balance via protein kinase C-gp91(phox) signaling to epithelial sodium channels. Am J Respir Cell Mol Biol 2015; 52:75-87. [PMID: 24978055 DOI: 10.1165/rcmb.2014-0002oc] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The receptor for advanced glycation end-products (RAGE), a multiligand member of the Ig family, may play a crucial role in the regulation of lung fluid balance. We quantified soluble RAGE (sRAGE), a decoy isoform, and advanced glycation end-products (AGEs) from the bronchoalveolar lavage fluid of smokers and nonsmokers, and tested the hypothesis that AGEs regulate lung fluid balance through protein kinase C (PKC)-gp91(phox) signaling to the epithelial sodium channel (ENaC). Human bronchoalveolar lavage samples from smokers showed increased AGEs (9.02 ± 3.03 μg versus 2.48 ± 0.53 μg), lower sRAGE (1,205 ± 292 pg/ml versus 1,910 ± 263 pg/ml), and lower volume(s) of epithelial lining fluid (97 ± 14 ml versus 133 ± 17 ml). sRAGE levels did not predict ELF volumes in nonsmokers; however, in smokers, higher volumes of ELF were predicted with higher levels of sRAGE. Single-channel patch clamp analysis of rat alveolar epithelial type 1 cells showed that AGEs increased ENaC activity measured as the product of the number of channels (N) and the open probability (Po) (NPo) from 0.19 ± 0.08 to 0.83 ± 0.22 (P = 0.017) and the subsequent addition of 4-hydroxy-2, 2, 6, 6-tetramethylpiperidine-N-oxyl decreased ENaC NPo to 0.15 ± 0.07 (P = 0.01). In type 2 cells, human AGEs increased ENaC NPo from 0.12 ± 0.05 to 0.53 ± 0.16 (P = 0.025) and the addition of 4-hydroxy-2, 2, 6, 6-tetramethylpiperidine-N-oxyl decreased ENaC NPo to 0.10 ± 0.03 (P = 0.013). Using molecular and biochemical techniques, we observed that inhibition of RAGE and PKC activity attenuated AGE-induced activation of ENaC. AGEs induced phosphorylation of p47(phox) and increased gp91(phox)-dependent reactive oxygen species production, a response that was abrogated with RAGE or PKC inhibition. Finally, tracheal instillation of AGEs promoted clearance of lung fluid, whereas concomitant inhibition of RAGE, PKC, and gp91(phox) abrogated the response.
Collapse
|
88
|
Modeling the interaction between quinolinate and the receptor for advanced glycation end products (RAGE): relevance for early neuropathological processes. PLoS One 2015; 10:e0120221. [PMID: 25757085 PMCID: PMC4354912 DOI: 10.1371/journal.pone.0120221] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 01/20/2015] [Indexed: 01/13/2023] Open
Abstract
The receptor for advanced glycation end products (RAGE) is a pattern-recognition receptor involved in neurodegenerative and inflammatory disorders. RAGE induces cellular signaling upon binding to a variety of ligands. Evidence suggests that RAGE up-regulation is involved in quinolinate (QUIN)-induced toxicity. We investigated the QUIN-induced toxic events associated with early noxious responses, which might be linked to signaling cascades leading to cell death. The extent of early cellular damage caused by this receptor in the rat striatum was characterized by image processing methods. To document the direct interaction between QUIN and RAGE, we determined the binding constant (Kb) of RAGE (VC1 domain) with QUIN through a fluorescence assay. We modeled possible binding sites of QUIN to the VC1 domain for both rat and human RAGE. QUIN was found to bind at multiple sites to the VC1 dimer, each leading to particular mechanistic scenarios for the signaling evoked by QUIN binding, some of which directly alter RAGE oligomerization. This work contributes to the understanding of the phenomenon of RAGE-QUIN recognition, leading to the modulation of RAGE function.
Collapse
|
89
|
Cerofolini L, Amato J, Borsi V, Pagano B, Randazzo A, Fragai M. Probing the interaction of distamycin A with S100β: the "unexpected" ability of S100β to bind to DNA-binding ligands. J Mol Recognit 2015; 28:376-84. [PMID: 25694263 DOI: 10.1002/jmr.2452] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Revised: 11/20/2014] [Accepted: 11/24/2014] [Indexed: 11/11/2022]
Abstract
DNA-minor-groove-binding ligands are potent antineoplastic molecules. The antibiotic distamycin A is the prototype of one class of these DNA-interfering molecules that have been largely used in vitro. The affinity of distamycin A for DNA is well known, and the structural details of the complexes with some B-DNA and G-quadruplex-forming DNA sequences have been already elucidated. Here, we show that distamycin A binds S100β, a protein involved in the regulation of several cellular processes. The reported affinity of distamycin A for the calcium(II)-loaded S100β reinforces the idea that some biological activities of the DNA-minor-groove-binding ligands arise from the binding to cellular proteins.
Collapse
Affiliation(s)
- Linda Cerofolini
- Giotto Biotech, Via Madonna del Piano 6, Sesto Fiorentino, Florence, 50019, Italy
| | | | | | | | | | | |
Collapse
|
90
|
Batkulwar KB, Bansode SB, Patil GV, Godbole RK, Kazi RS, Chinnathambi S, Shanmugam D, Kulkarni MJ. Investigation of phosphoproteome in RAGE signaling. Proteomics 2015; 15:245-259. [PMID: 25315903 DOI: 10.1002/pmic.201400169] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 08/14/2014] [Accepted: 10/06/2014] [Indexed: 01/03/2025]
Abstract
The receptor for advanced glycation end products (RAGE) is one of the most important proteins implicated in diabetes, cardiovascular diseases, neurodegenerative diseases, and cancer. It is a pattern recognition receptor by virtue of its ability to interact with multiple ligands, RAGE activates several signal transduction pathways through involvement of various kinases that phosphorylate their respective substrates. Only few substrates have been known to be phosphorylated in response to activation by RAGE (e.g., nuclear factor kappa B); however, it is possible that these kinases can phosphorylate multiple substrates depending upon their expression and localization, leading to altered cellular responses in different cell types and conditions. One such example is, glycogen synthase kinase 3 beta which is known to phosphorylate glycogen synthase, acts downstream to RAGE, and hyperphosphorylates microtubule-associated protein tau causing neuronal damage. Thus, it is important to understand the role of various RAGE-activated kinases and their substrates. Therefore, we have reviewed here the details of RAGE-activated kinases in response to different ligands and their respective phosphoproteome. Furthermore, we discuss the analysis of the data mined for known substrates of these kinases from the PhosphoSitePlus (http://www.phosphosite.org) database, and the role of some of the important substrates involved in cancer, diabetes, cardiovascular diseases, and neurodegenerative diseases. In summary, this review provides information on RAGE-activated kinases and their phosphoproteome, which will be helpful in understanding the possible role of RAGE and its ligands in progression of diseases.
Collapse
Affiliation(s)
- Kedar B Batkulwar
- Proteomics Facility, Division of Biochemical Sciences, CSIR-National Chemical Laboratory, Pune, India
| | | | | | | | | | | | | | | |
Collapse
|
91
|
Voziyan P, Brown KL, Chetyrkin S, Hudson B. Site-specific AGE modifications in the extracellular matrix: a role for glyoxal in protein damage in diabetes. Clin Chem Lab Med 2014; 52:39-45. [PMID: 23492568 DOI: 10.1515/cclm-2012-0818] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Accepted: 02/07/2013] [Indexed: 11/15/2022]
Abstract
Non-enzymatic modification of proteins in hyperglycemia is a major proposed mechanism of diabetic complications. Specifically, advanced glycation end products (AGEs) derived from hyperglycemia-induced reactive carbonyl species (RCS) can have pathogenic consequences when they target functionally critical protein residues. Modification of a small number of these critical residues, often undetectable by the methodologies relying on measurements of total AGE levels, can cause significant functional damage. Therefore, detection of specific sites of protein damage in diabetes is central to understanding the molecular basis of diabetic complications and for identification of biomarkers which are mechanistically linked to the disease. The current paradigm of RCS-derived protein damage places a major focus on methylglyoxal (MGO), an intermediate of cellular glycolysis. We propose that glyoxal (GO) is a major contributor to extracellular matrix (ECM) damage in diabetes. Here, we review the current knowledge and provide new data about GO-derived site-specific ECM modification in experimental diabetes.
Collapse
|
92
|
Munesue S, Yamamoto Y, Urushihara R, Inomata K, Saito H, Motoyoshi S, Watanabe T, Yonekura H, Yamamoto H. Low-molecular weight fractions of Japanese soy sauce act as a RAGE antagonist via inhibition of RAGE trafficking to lipid rafts. Food Funct 2014; 4:1835-42. [PMID: 24191276 DOI: 10.1039/c2fo30359k] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Advanced glycation end-products (AGE) have been implicated in aging and the pathogenesis of diabetic complications, inflammation, Alzheimer's disease, and cancer. AGE engage the cell surface receptor for AGE (RAGE), which in turn elicits intracellular signaling, leading to activation of NF-κB to cause deterioration of tissue homeostasis. AGE are not only formed within our bodies but are also derived from foods, endowing them with flavor. In the present study, we assessed the agonistic/antagonistic effects of food-derived AGE on RAGE signaling in a reporter assay system and found that low-molecular weight AGE can antagonize the action of AGE-BSA. Foods tested were Japanese soy sauce, coffee, cola, and red wine, all of which showed fluorescence characteristics of AGE. Soy sauce and coffee contained N(ε)-carboxymethyl-lysine (CML). Soy sauce, coffee, and red wine inhibited the RAGE ligand-induced activation of NF-κB, whereas cola had no effect on the ligand induction of NF-κB. The liquids were then fractionated into high-molecular weight (HMW) fractions and low-molecular weight (LMW) fractions. Soy sauce-, coffee-, and red wine-derived LMW fractions consistently inhibited the RAGE ligand induction of NF-κB, whereas the HMW fractions of these foods activated RAGE signaling. Using the LMW fraction of soy sauce as a model food-derived RAGE antagonist, we performed a plate-binding assay and found that the soy sauce LMW fractions competitively inhibited AGE-RAGE association. Further, this fraction significantly reduced AGE-dependent monocyte chemoattractant protein-1 (MCP-1) secretion from murine peritoneal macrophages. The LMF from soy sauce suppressed the AGE-induced RAGE trafficking to lipid rafts. These results indicate that small components in some, if not all, foods antagonize RAGE signaling and could exhibit beneficial effects on RAGE-related diseases.
Collapse
Affiliation(s)
- Seiichi Munesue
- Department of Biochemistry and Molecular Vascular Biology, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8640, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
93
|
RAGE overexpression confers a metastatic phenotype to the WM115 human primary melanoma cell line. Biochim Biophys Acta Mol Basis Dis 2014; 1842:1017-27. [DOI: 10.1016/j.bbadis.2014.02.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Revised: 02/16/2014] [Accepted: 02/26/2014] [Indexed: 12/19/2022]
|
94
|
Hombach-Klonisch S, Natarajan S, Thanasupawat T, Medapati M, Pathak A, Ghavami S, Klonisch T. Mechanisms of therapeutic resistance in cancer (stem) cells with emphasis on thyroid cancer cells. Front Endocrinol (Lausanne) 2014; 5:37. [PMID: 24723911 PMCID: PMC3971176 DOI: 10.3389/fendo.2014.00037] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 03/11/2014] [Indexed: 12/11/2022] Open
Abstract
The two main reasons for death of cancer patients, tumor recurrence and metastasis, are multi-stage cellular processes that involve increased cell plasticity and coincide with elevated resistance to anti-cancer treatments. Epithelial-to-mesenchymal transition (EMT) is a key contributor to metastasis in many cancer types, including thyroid cancer and is known to confer stem cell-like properties onto cancer cells. This review provides an overview of molecular mechanisms and factors known to contribute to cancer cell plasticity and capable of enhancing cancer cell resistance to radio- and chemotherapy. We elucidate the role of DNA repair mechanisms in contributing to therapeutic resistance, with a special emphasis on thyroid cancer. Next, we explore the emerging roles of autophagy and damage-associated molecular pattern responses in EMT and chemoresistance in tumor cells. Finally, we demonstrate how cancer cells, including thyroid cancer cells, can highjack the oncofetal nucleoprotein high-mobility group A2 to gain increased transformative cell plasticity, prevent apoptosis, and enhance metastasis of chemoresistant tumor cells.
Collapse
Affiliation(s)
- Sabine Hombach-Klonisch
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, MB, Canada
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Suchitra Natarajan
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, MB, Canada
| | | | - Manoj Medapati
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, MB, Canada
| | - Alok Pathak
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, MB, Canada
- Department of Surgery, University of Manitoba, Winnipeg, MB, Canada
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, MB, Canada
- Manitoba Institute of Child Health, University of Manitoba, Winnipeg, MB, Canada
| | - Thomas Klonisch
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, MB, Canada
- Department of Surgery, University of Manitoba, Winnipeg, MB, Canada
- Manitoba Institute of Child Health, University of Manitoba, Winnipeg, MB, Canada
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
95
|
Chen H, Xu C, Jin Q, Liu Z. S100 protein family in human cancer. Am J Cancer Res 2014; 4:89-115. [PMID: 24660101 PMCID: PMC3960449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Accepted: 02/10/2014] [Indexed: 06/03/2023] Open
Abstract
S100 protein family has been implicated in multiple stages of tumorigenesis and progression. Among the S100 genes, 22 are clustered at chromosome locus 1q21, a region frequently rearranged in cancers. S100 protein possesses a wide range of intracellular and extracellular functions such as regulation of calcium homeostasis, cell proliferation, apoptosis, cell invasion and motility, cytoskeleton interactions, protein phosphorylation, regulation of transcriptional factors, autoimmunity, chemotaxis, inflammation and pluripotency. Many lines of evidence suggest that altered expression of S100 proteins was associated with tumor progression and prognosis. Therefore, S100 proteins might also represent potential tumor biomarkers and therapeutic targets. In this review, we summarize the evidence connecting S100 protein family and cancer and discuss the mechanisms by which S100 exerts its diverse functions.
Collapse
Affiliation(s)
- Hongyan Chen
- The State Key Laboratory of Molecular Oncology, Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing 100021, China
| | - Chengshan Xu
- The State Key Laboratory of Molecular Oncology, Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing 100021, China
| | - Qing'e Jin
- The State Key Laboratory of Molecular Oncology, Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing 100021, China
| | - Zhihua Liu
- The State Key Laboratory of Molecular Oncology, Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing 100021, China
| |
Collapse
|
96
|
Musumeci D, Roviello GN, Montesarchio D. An overview on HMGB1 inhibitors as potential therapeutic agents in HMGB1-related pathologies. Pharmacol Ther 2013; 141:347-57. [PMID: 24220159 DOI: 10.1016/j.pharmthera.2013.11.001] [Citation(s) in RCA: 262] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Accepted: 10/25/2013] [Indexed: 12/13/2022]
Abstract
HMGB1 (High-Mobility Group Box-1) is a nuclear protein that acts as an architectural chromatin-binding factor involved in the maintenance of nucleosome structure and regulation of gene transcription. It can be released into the extracellular milieu from immune and non-immune cells in response to various stimuli. Extracellular HMGB1 contributes to the pathogenesis of numerous chronic inflammatory and autoimmune diseases, including sepsis, rheumatoid arthritis, atherosclerosis, chronic kidney disease, systemic lupus erythematosus (SLE), as well as cancer pathogenesis. Interaction of released HMGB1 with the cell-surface receptor for advanced glycation end products (RAGE) is one of the main signaling pathways triggering these diseases. It has been also demonstrated that the inhibition of the HMGB1-RAGE interaction represents a promising approach for the modulation of the inflammatory and tumor-facilitating activity of HMGB1. In this review we describe various approaches recently proposed in the literature to inhibit HMGB1 and the related inflammatory processes, especially focusing on the block of RAGE-HMGB1 signaling. Several strategies are based on molecules which mainly interact with RAGE as competitive antagonists of HMGB1. As an alternative, encouraging results have been obtained with HMGB1-targeting, leading to the identification of compounds that directly bind to HMGB1, ranging from small natural or synthetic molecules, such as glycyrrhizin and gabexate mesilate, to HMGB1-specific antibodies, peptides, proteins as well as bent DNA-based duplexes. Future perspectives are discussed in the light of the overall body of knowledge acquired by a large number of research groups operating in different but related fields.
Collapse
Affiliation(s)
- Domenica Musumeci
- Dipartimento di Scienze Chimiche, Università di Napoli "Federico II", via Cintia 21, Complesso Universitario di Monte Sant'Angelo, I-80126 Napoli, Italy.
| | - Giovanni N Roviello
- Istituto di Biostrutture e Bioimmagini - CNR, via Mezzocannone 16, I-80134 Napoli, Italy
| | - Daniela Montesarchio
- Dipartimento di Scienze Chimiche, Università di Napoli "Federico II", via Cintia 21, Complesso Universitario di Monte Sant'Angelo, I-80126 Napoli, Italy
| |
Collapse
|
97
|
Su PC, Berger BW. A Novel Assay for Assessing Juxtamembrane and Transmembrane Domain Interactions Important for Receptor Heterodimerization. J Mol Biol 2013; 425:4652-8. [DOI: 10.1016/j.jmb.2013.07.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Revised: 07/12/2013] [Accepted: 07/15/2013] [Indexed: 10/26/2022]
|
98
|
Cohen MM. Perspectives on RAGE signaling and its role in cardiovascular disease. Am J Med Genet A 2013; 161A:2750-5. [PMID: 24123885 DOI: 10.1002/ajmg.a.36181] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Accepted: 07/16/2013] [Indexed: 01/14/2023]
Abstract
RAGE stands for Receptor of Advanced Glycation Endproducts. The two main topics discussed are (1) the nature of RAGE signaling and (2) its role in cardiovascular disease. RAGE may occur in membrane-bound form or in secretory form. RAGE signaling involves multiple ligands: (1) several AGEs (2) amyloid β pecursor protein (APP), (3) high mobility group box 1 (HMGB1), (4) S100A4, (5) S100A8/A9, and (6) S100A12, which are calcium-binding proteins, and (7) S100B, a glial-derived protein. RAGE ligands and various diseases involving RAGE signaling are summarized in tabular form.
Collapse
Affiliation(s)
- M Michael Cohen
- Department of Pediatrics, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
99
|
Carvalho SB, Botelho HM, Leal SS, Cardoso I, Fritz G, Gomes CM. Intrinsically disordered and aggregation prone regions underlie β-aggregation in S100 proteins. PLoS One 2013; 8:e76629. [PMID: 24098542 PMCID: PMC3788126 DOI: 10.1371/journal.pone.0076629] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Accepted: 08/24/2013] [Indexed: 12/19/2022] Open
Abstract
S100 proteins are small dimeric calcium-binding proteins which control cell cycle, growth and differentiation via interactions with different target proteins. Intrinsic disorder is a hallmark among many signaling proteins and S100 proteins have been proposed to contain disorder-prone regions. Interestingly, some S100 proteins also form amyloids: S100A8/A9 forms fibrils in prostatic inclusions and S100A6 fibrillates in vitro and seeds SOD1 aggregation. Here we report a study designed to investigate whether β-aggregation is a feature extensive to more members of S100 family. In silico analysis of seven human S100 proteins revealed a direct correlation between aggregation and intrinsic disorder propensity scores, suggesting a relationship between these two independent properties. Averaged position-specific analysis and structural mapping showed that disorder-prone segments are contiguous to aggregation-prone regions and that whereas disorder is prominent on the hinge and target protein-interaction regions, segments with high aggregation propensity are found in ordered regions within the dimer interface. Acidic conditions likely destabilize the seven S100 studied by decreasing the shielding of aggregation-prone regions afforded by the quaternary structure. In agreement with the in silico analysis, hydrophobic moieties become accessible as indicated by strong ANS fluorescence. ATR-FTIR spectra support a structural inter-conversion from α-helices to intermolecular β-sheets, and prompt ThT-binding takes place with no noticeable lag phase. Dot blot analysis using amyloid conformational antibodies denotes a high diversity of conformers; subsequent analysis by TEM shows fibrils as dominant species. Altogether, our data suggests that β-aggregation and disorder-propensity are related properties in S100 proteins, and that the onset of aggregation is likely triggered by loss of protective tertiary and quaternary interactions.
Collapse
Affiliation(s)
- Sofia B. Carvalho
- Instituto Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Hugo M. Botelho
- Instituto Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Sónia S. Leal
- Instituto Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Isabel Cardoso
- Molecular Neurobiology Unit, Instituto de Biologia Molecular e Celular, Porto, Portugal
- Escola Superior Tecnologia Saúde Porto, Instituto Politécnico, Porto, Vila Nova de Gaia, Portugal
| | - Günter Fritz
- Department of Neuropathology, University of Freiburg, Freiburg, Germany
| | - Cláudio M. Gomes
- Instituto Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
- * E-mail:
| |
Collapse
|
100
|
Sitkiewicz E, Tarnowski K, Poznański J, Kulma M, Dadlez M. Oligomerization interface of RAGE receptor revealed by MS-monitored hydrogen deuterium exchange. PLoS One 2013; 8:e76353. [PMID: 24098480 PMCID: PMC3788119 DOI: 10.1371/journal.pone.0076353] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Accepted: 08/26/2013] [Indexed: 01/10/2023] Open
Abstract
Activation of the receptor for advanced glycation end products (RAGE) leads to a chronic proinflammatory signal, affecting patients with a variety of diseases. Potentially beneficial modification of RAGE activity requires understanding the signal transduction mechanism at the molecular level. The ligand binding domain is structurally uncoupled from the cytoplasmic domain, suggesting receptor oligomerization is a requirement for receptor activation. In this study, we used hydrogen-deuterium exchange and mass spectrometry to map structural differences between the monomeric and oligomeric forms of RAGE. Our results indicated the presence of a region shielded from exchange in the oligomeric form of RAGE and led to the identification of a new oligomerization interface localized at the linker region between domains C1 and C2. Based on this finding, a model of a RAGE dimer and higher oligomeric state was constructed.
Collapse
Affiliation(s)
- Ewa Sitkiewicz
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warszawa, Poland
| | - Krzysztof Tarnowski
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warszawa, Poland
| | - Jarosław Poznański
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warszawa, Poland
| | - Magdalena Kulma
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warszawa, Poland
| | - Michal Dadlez
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warszawa, Poland
- Institute of Genetics and Biotechnology, Biology Department, Warsaw University, Warszawa, Poland
- * E-mail:
| |
Collapse
|