701
|
Xie J, Burz DS, He W, Bronstein IB, Lednev I, Shekhtman A. Hexameric Calgranulin C (S100A12) Binds to the Receptor for Advanced Glycated End Products (RAGE) Using Symmetric Hydrophobic Target-binding Patches. J Biol Chem 2007; 282:4218-31. [PMID: 17158877 DOI: 10.1074/jbc.m608888200] [Citation(s) in RCA: 135] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Calgranulin C (S100A12) is a member of the S100 family of proteins that undergoes a conformational change upon calcium binding allowing them to interact with target molecules and initiate biological responses; one such target is the receptor for advanced glycation products (RAGE). The RAGE-calgranulin C interaction mediates a pro-inflammatory response to cellular stress and can contribute to the pathogenesis of inflammatory lesions. The soluble extracellular part of RAGE (sRAGE) was shown to decrease the inflammation response possibly by scavenging RAGE-activating ligands. Here, by using high resolution NMR spectroscopy, we identified the sRAGE-calgranulin C interaction surface. Ca2+ binding creates two symmetric hydrophobic surfaces on Ca2+-calgranulin C that allow calgranulin C to bind to the C-type immunoglobulin domain of RAGE. Apo-calgranulin C also binds to sRAGE using a completely different surface and with substantially lower affinity, thus underscoring the role of Ca2+ binding to S100 proteins as a molecular switch. By using native gel electrophoresis, chromatography, and fluorescence spectroscopy, we established that sRAGE forms tetramers that bind to hexamers of Ca2+-calgranulin C. This arrangement creates a large platform for effectively transmitting RAGE-dependent signals from extracellular S100 proteins to the cytoplasmic signaling complexes.
Collapse
Affiliation(s)
- Jingjing Xie
- Department of Chemistry, State University of New York, Albany, New York 12222, USA
| | | | | | | | | | | |
Collapse
|
702
|
Yoon SJ, Park S, Shim CY, Park CM, Ko YG, Choi D, Park HY, Oh B, Kim H, Jang Y, Chung N. Association of RAGE gene polymorphisms with coronary artery disease in the Korean population. Coron Artery Dis 2007; 18:1-8. [PMID: 17172923 DOI: 10.1097/mca.0b013e3280105b4d] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND AND OBJECTIVES AGEs (advanced glycation end products)-RAGE (receptor for advanced glycation end products) interaction in vessel wall may lead to inflammation, smooth muscle cell proliferation and extracellular matrix production, culminating in exaggerated intimal hyperplasia and restenosis. We focused on the putative association of five candidate RAGE gene polymorphisms on the risk of coronary artery disease in the Korean population. METHODS A total of 1555 male patients who underwent coronary angiography were enrolled in the study; 805 patients (mean age: 53.18+/-9.74 years) had normal coronary artery and 750 patients (mean age: 55.73+/-8.31 years) had significant coronary artery disease. Among the coronary artery disease patients, 269 had single-vessel disease (35.87%), 242 had two-vessel disease (32.27%) and 239 had three-vessel disease (31.87%). The genotypes of RAGE were determined by the methods of single base extension with amplifying primers and probes for TaqMan. Genotype analysis was performed on five single nucleotide polymorphisms of the RAGE gene, namely -443T>C, -388T>A, -257G>A, +557G>A and +1704G>T. Analysis for the association with coronary artery disease was performed. RESULTS Analysis of four single nucleotide polymorphisms, except +557G>A (G82S), with regard to the association with coronary artery disease was not significant. Only the +557 gene allele (G/A) showed significant association with coronary artery disease (coronary artery disease vs. normal; G allele: 0.87 vs. 0.84, A allele: 0.13 vs. 0.16, P=0.0326). The +557G>A (G82S) showed strong tendency of association with coronary artery disease (coronary artery disease vs. normal; GG: 75.2 vs. 69.8%, GA: 23.2 vs. 28.6%, AA: 1.6 vs. 1.6%, P=0.0524). The presence of AA or GA genotype, assuming codominant effect of the A allele, was independently associated with decreased risk of coronary artery disease when controlled for age, body mass index, smoking and diabetes mellitus [odds ratio=0.749 (95% confidence interval, 0.579-0.969), P=0.0280]. Subgroup analysis demonstrated the significant protective effect of AA or GA genotype in nondiabetic patients as well [odds ratio=0.741 (0.570-0.962), P=0.0244]. CONCLUSIONS The results of this large population study demonstrate that the AA/GA genotypes of the RAGE +557G>A polymorphism are associated with a significantly decreased risk of significant coronary artery disease. Other polymorphisms of RAGE were not significantly associated with the risk of coronary artery disease in this study population.
Collapse
Affiliation(s)
- Se-Jung Yoon
- Division of Cardiology, Yonsei Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
703
|
Moser B, Szabolcs MJ, Ankersmit HJ, Lu Y, Qu W, Weinberg A, Herold KC, Schmidt AM. Blockade of RAGE suppresses alloimmune reactions in vitro and delays allograft rejection in murine heart transplantation. Am J Transplant 2007; 7:293-302. [PMID: 17241110 DOI: 10.1111/j.1600-6143.2006.01617.x] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The receptor for advanced glycation endproducts (RAGE), a multiligand member of the immunoglobulin superfamily, interacts with proinflammatory AGEs, the products of nonenzymatic glycation and oxidation of proteins; high-mobility group box 1 (HMGB1), also known as amphoterin and S100/calgranulins to amplify inflammation and tissue injury. Previous studies showed that blockade of RAGE suppressed recruitment of proinflammatory mechanisms in murine models. We tested the hypothesis that RAGE contributes to alloimmune responses and report that in vivo, acute rejection of fully allogeneic cardiac allografts in a murine model of heterotopic cardiac transplantation is significantly delayed by pharmacological antagonism of RAGE. In parallel, allogeneic T-cell proliferation in the mixed lymphocyte reaction is, at least in part, RAGE-dependent. These data provide the first insights into key roles for RAGE in allorecognition responses and suggest that antagonism of this receptor may exert beneficial effects in allogeneic organ transplantation.
Collapse
Affiliation(s)
- B Moser
- Department of Surgery, Columbia University Medical Center, New York, New York, USA. [corrected]
| | | | | | | | | | | | | | | |
Collapse
|
704
|
Thierry S, Gozlan J, Jaulmes A, Boniface R, Nasreddine N, Strauss F, Maréchal V. High-mobility group box 1 protein induces HIV-1 expression from persistently infected cells. AIDS 2007; 21:283-92. [PMID: 17255735 DOI: 10.1097/qad.0b013e3280115b50] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Necrosis is a frequent condition during AIDS, notably in organs targetted by opportunistic infections. Soluble factors released by necrotic cells are important for signalling cell damage, but little is known concerning their effect on HIV-1 replication. We focused on HMGB1, an abundant component of the chromatin that is released from necrotic cells and can act as a pro-inflammatory mediator. MATERIALS AND METHODS A native form of HMGB1 was obtained from necrotic Hela cells, whereas a purified recombinant HMGB1 was generated in Escherichia coli. ACH-2 and U1 cells were used as models of persistent HIV-1 infection in lymphocytes and monocytes. Reactivation from latency was also investigated ex vivo using peripheral blood mononuclear cells (PBMC) collected from HIV-1-infected patients controlled by HAART. HIV-1 expression was quantified by enzyme-linked immunosorbent assay, real-time reverse transcription-polymerase chain reaction and branched DNA techniques. Flow cytometry and blocking experiments were used to identify the receptor used by HMGB1. Chromatin immunoprecipitation was used to investigate long-terminal repeat activation upon stimulation by HMGB1. RESULTS HMGB1 increased HIV-1 transcription in chronically infected cells, a process that did not require de-novo protein synthesis. HIV-1 induction relied on HMGB1 interaction with the receptor for advanced glycation end-products. The activation pathway involved p38 and extracellular signal-related kinase as well as nuclear factor kappa B binding to the HIV-1 promoter. Finally, HMGB1 reactivated HIV-1 from latently infected PBMC collected in aviraemic HIV-infected patients. CONCLUSION This work establishes for the first time a link between necrosis and HIV-1 replication, which involves HMGB1, a soluble mediator released by damaged cells.
Collapse
|
705
|
Kobayashi S, Kubo H, Suzuki T, Ishizawa K, Yamada M, He M, Yamamoto Y, Yamamoto H, Sasano H, Sasaki H, Suzuki S. Endogenous Secretory Receptor for Advanced Glycation End Products in Non–Small Cell Lung Carcinoma. Am J Respir Crit Care Med 2007; 175:184-9. [PMID: 17023736 DOI: 10.1164/rccm.200602-212oc] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE The receptor for advanced glycation end products is a multiligand receptor that plays an important role in regulating the invasiveness and metastatic potential of cancer cells. A recently discovered novel splice variant, the endogenous secretory receptor for advanced glycation end products, mediates the receptor for advanced glycation end-product-associated cell responses by functioning as a decoy receptor. OBJECTIVES To evaluate the expression pattern of endogenous secretory receptor for advanced glycation end products in non-small cell lung carcinoma, and analyze its impact on prognosis. METHODS We performed immunohistochemical evaluation in 182 non-small cell lung carcinoma surgical specimens. The effect of an overexpressed receptor in cancer cell proliferation was also evaluated. MEASUREMENTS AND MAIN RESULTS The endogenous secretory receptor for advanced glycation end-product expression in cytoplasm was reduced or absent in 137 of the 182 (75%) carcinomas in contrast to normal lung tissues. mRNA expression was also suppressed in cancer cells. Overexpression of the secretory receptor in lung cancer cell lines had an inhibitory effect on cell proliferation, suggesting the reduced receptor expression accelerated tumor growth. Among patients with low expression of the cytoplasmic secretory receptor, the overall survival rate was significantly lower than that of patients with normal expression (p = 0.0003). This association was most prominent in TNM stage I patients (p = 0.0001). In a multivariate analysis, endogenous secretory receptor immunoreactivity was an independent prognostic factor with a relative risk of 3.1. CONCLUSIONS The cytoplasmic endogenous secretory receptor for advanced glycation end-product expression has the potential to be a prognostic factor for predicting the outcome of curative surgery in patients with non-small cell lung carcinoma.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Alternative Splicing
- Carcinoma, Non-Small-Cell Lung/chemistry
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/mortality
- Cell Proliferation
- Cytoplasm/chemistry
- Cytoplasm/metabolism
- Down-Regulation
- Female
- Glycation End Products, Advanced/metabolism
- Humans
- Immunohistochemistry
- Lung Neoplasms/chemistry
- Lung Neoplasms/metabolism
- Lung Neoplasms/mortality
- Male
- Middle Aged
- Prognosis
- RNA, Messenger/analysis
- RNA, Messenger/metabolism
- Receptor for Advanced Glycation End Products
- Receptors, Immunologic/analysis
- Receptors, Immunologic/genetics
- Receptors, Immunologic/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Treatment Outcome
Collapse
Affiliation(s)
- Seiichi Kobayashi
- Department of Geriatric and Respiratory Medicine, Tohoku University School of Medicine, 1-1 Seiryoumachi, Aobaku, Sendai, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
706
|
Chavakis E, Hain A, Vinci M, Carmona G, Bianchi ME, Vajkoczy P, Zeiher AM, Chavakis T, Dimmeler S. High-mobility group box 1 activates integrin-dependent homing of endothelial progenitor cells. Circ Res 2007; 100:204-12. [PMID: 17218606 DOI: 10.1161/01.res.0000257774.55970.f4] [Citation(s) in RCA: 235] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Endothelial progenitor cells (EPCs) are recruited to ischemic regions and improve neovascularization. Integrins contribute to EPC homing. High-mobility group box 1 (HMGB1) is a nuclear protein that is released extracellularly on cell necrosis and tissue damage, eliciting a proinflammatory response and stimulating tissue repair. In the present study, we investigated the effects of HMGB1 on EPC homing. EPCs express the HMGB1 receptors RAGE (receptor for advanced glycation end products) and TLR2 (Toll-like receptor 2). EPC migration was stimulated by HMGB1 in a RAGE-dependent manner. In addition, the HMGB1-induced migration of EPCs on fibronectin and fibrinogen was significantly inhibited by antibodies against beta1 and beta2 integrins, respectively. Short-term prestimulation of EPCs with HMGB1 also increased EPC adhesion to endothelial cell monolayers, and this effect was blocked by antibodies to beta2 integrins or RAGE. HMGB1 increased EPC adhesion to the immobilized integrin ligands intercellular adhesion molecule-1 and fibronectin in a RAGE-dependent manner. Strikingly, HMGB1 rapidly increased integrin affinity and induced integrin polarization. Using intravital microscopy in a tumor model of neovascularization, prestimulation of EPCs with HMGB1 enhanced the initial in vivo adhesion of EPCs to microvessels and the recruitment of EPCs in the tumor tissue. In addition, prestimulation of EPCs with HMGB1 increased the homing of EPCs to ischemic muscles. In conclusion, these data represent a link between HMGB1 and integrin functions of EPCs and demonstrate that HMGB1 stimulates EPC homing to ischemic tissues. These results may provide a platform for the development of novel therapeutic approaches to improve EPC homing.
Collapse
Affiliation(s)
- Emmanouil Chavakis
- Molecular Cardiology, Department of Internal Medicine III, J. W. Goethe University of Frankfurt, Frankfurt, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
707
|
Yan SF, Naka Y, Hudson BI, Herold K, Yan SD, Ramasamy R, Schmidt AM. The ligand/RAGE axis: lighting the fuse and igniting vascular stress. Curr Atheroscler Rep 2007; 8:232-9. [PMID: 16640960 DOI: 10.1007/s11883-006-0078-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Vascular inflammation contributes critically to the initiation and progression of atherosclerosis. These processes are accelerated in hyperglycemia and play key roles in the increased incidence and severity of myocardial infarction and stroke observed in diabetes. Evidence suggests that the ligands of the receptor for advanced glycation endproducts (RAGE), a multiligand member of the immunoglobulin superfamily, interact with this receptor to play important roles in both early development and progression of atherosclerosis and vascular inflammation. Studies in animal models of vascular injury underscored the potent impact of RAGE blockade; administration of ligand-binding decoys of RAGE or antibodies to the receptor reduced the consequences of diabetes, hyperlipidemia, and physical injury to the vessel wall. This review focuses on the ligand repertoire of RAGE, the impact of ligand-RAGE interaction, and the potent effect of RAGE blockade in rodent models of vascular injury.
Collapse
Affiliation(s)
- Shi Fang Yan
- Division of Surgical Science, Department of Surgery, Columbia University Medical Center, 630 West 168th Street, P&S 17-501, New York, NY 10032, USA
| | | | | | | | | | | | | |
Collapse
|
708
|
Diamanti-Kandarakis E, Piperi C, Patsouris E, Korkolopoulou P, Panidis D, Pawelczyk L, Papavassiliou AG, Duleba AJ. Immunohistochemical localization of advanced glycation end-products (AGEs) and their receptor (RAGE) in polycystic and normal ovaries. Histochem Cell Biol 2007; 127:581-9. [PMID: 17205306 DOI: 10.1007/s00418-006-0265-3] [Citation(s) in RCA: 126] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/07/2006] [Indexed: 10/23/2022]
Abstract
The aim of the present study was to investigate the localization/immunohistochemical distribution of AGEs and RAGE, as well as their putative signalling mediator NF-kappaB in ovaries of women with polycystic ovary syndrome (PCOS) compared to normal. Archival ovarian-tissue samples from biopsies of six women with PCOS and from six healthy of similar age women, were examined immunohistochemically with monoclonal anti-AGEs, anti-RAGE and anti-NF-kappaB(p50/p65) specific antibodies. In healthy women, AGE immunoreactivity was observed in follicular cell layers (granulosa and theca) and luteinized cells, but not in endothelial cells. PCOS specimens displayed AGE immunoexpression in theca interna and granulosa cells as well as in endothelial cells, but staining of granulosa cells was stronger than in that of normal ovaries. RAGE was highly expressed in normal and PCOS tissues. Normal tissue exhibited no staining differences between granulosa cell layer and theca interna. However, in PCOS ovaries, granulosa cells displayed stronger RAGE expression compared to theca interna cells in comparison to controls. NF-kappaB(p50/p65) was expressed in the cytoplasm of theca interna and granulosa cells of both normal and PCOS ovaries; whereas the NF-kappaB p65 subunit was only observed in granulosa cells nuclei in PCOS tissue. In conclusion, these findings demonstrate for the first time that RAGE and AGE-modified proteins with activated NF-kappaB are expressed in human ovarian tissue. Furthermore, a differential qualitative distribution of AGE, RAGE and NF-kappaB p65 subunit was observed in women with PCOS compared to healthy controls, where a stronger localization of both AGE and RAGE was observed in the granulosa cell layer of PCOS ovaries.
Collapse
Affiliation(s)
- Evanthia Diamanti-Kandarakis
- First Department of Internal Medicine, Endocrine section, University of Athens Medical School, 75, M. Asias Str., 11527, Athens, Greece.
| | | | | | | | | | | | | | | |
Collapse
|
709
|
Inoue K, Kawahara KI, Biswas KK, Ando K, Mitsudo K, Nobuyoshi M, Maruyama I. HMGB1 expression by activated vascular smooth muscle cells in advanced human atherosclerosis plaques. Cardiovasc Pathol 2007; 16:136-43. [PMID: 17502242 DOI: 10.1016/j.carpath.2006.11.006] [Citation(s) in RCA: 110] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2006] [Revised: 09/26/2006] [Accepted: 11/16/2006] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Chronic inflammation plays a key role in atherogenesis, which is followed by atheromatous plaque instability. High-mobility group box 1 is released by activated macrophages as a late-phase mediator during prolonged inflammation. However, the expression of high-mobility group box 1 and its effect on the production of C-reactive protein and matrix metalloproteinases, particularly on human vascular smooth muscle cells, still remain unknown. METHODS AND RESULTS Immunohistochemical studies revealed that high-mobility group box 1 was abundantly expressed in vascular smooth muscle cells of carotid and coronary atheromatous plaques, but not in atrophic vascular smooth muscle cells of fibrous plaques and normal medial vascular smooth muscle cells. Receptor for advanced glycation end products was also detected in vascular smooth muscle cells positive for high-mobility group box 1. Moreover, vascular smooth muscle cells positive for high-mobility group box 1 were found to express both C-reactive protein and matrix metalloproteinases (2, 3, and 9). Administration of exogenous high-mobility group box 1 to cultured vascular smooth muscle cells caused a marked elevation of C-reactive protein mRNA by reverse transcriptase-polymerase chain reaction and of C-reactive protein levels by enzyme-linked immunosorbent assay. Conversely, C-reactive protein also triggered a significant release of high-mobility group box 1 in vascular smooth muscle cell culture medium as determined by immunoblot. CONCLUSIONS Activated vascular smooth muscle cells are the source of high-mobility group box 1 in human advanced atherosclerotic lesions. High-mobility group box 1 directly stimulates the production of both C-reactive protein and matrix metalloproteinase through receptor for advanced glycation end product. These findings provide new evidence that high-mobility group box 1 produced by activated vascular smooth muscle cells may contribute to the progression and vulnerability of human atherosclerotic lesions toward rupture.
Collapse
Affiliation(s)
- Katsumi Inoue
- Department of Cardiology, Kurashiki Central Hospital, Kurashiki, Japan
| | | | | | | | | | | | | |
Collapse
|
710
|
Abstract
The S100 proteins are exclusively expressed in vertebrates and are the largest subgroup within the superfamily of EF-hand Ca2(+)-binding proteins Generally, S100 proteins are organized as tight homodimers (some as heterodimers). Each subunit is composed of a C-terminal, 'canonical' EF-hand, common to all EF-hand proteins, and a N-terminal, 'pseudo' EF-hand, characteristic of S100 proteins. Upon Ca2(+)-binding, the C-terminal EF-hand undergoes a large conformational change resulting in the exposure of a hydrophobic surface responsible for target binding A unique feature of this protein family is that some members are secreted from cells upon stimulation, exerting cytokine- and chemokine-like extracellular activities via the Receptor for Advanced Glycation Endproducts, RAGE. Recently, larger assemblies of some S100 proteins (hexamers, tetramers, octamers) have been also observed and are suggested to be the active extracellular species required for receptor binding and activation through receptor multimerization Most S100 genes are located in a gene cluster on human chromosome 1q21, a region frequently rearranged in human cancer The functional diversification of S100 proteins is achieved by their specific cell- and tissue-expression patterns, structural variations, different metal ion binding properties (Ca2+, Zn2+ and Cu2+) as well as their ability to form homo-, hetero- and oligomeric assemblies Here, we review the most recent developments focussing on the biological functions of the S100 proteins and we discuss the presently available S100-specific mouse models and their possible use as human disease models In addition, the S100-RAGE interaction and the activation of various cellular pathways will be discussed. Finally, the close association of S100 proteins with cardiomyopathy, cancer, inflammation and brain diseases is summarized as well as their use in diagnosis and their potential as drug targets to improve therapies in the future.
Collapse
Affiliation(s)
- C W Heizmann
- Division of Clinical Chemistry and Biochemistry, Department of Pediatrics, University of Zurich, Switzerland.
| | | | | |
Collapse
|
711
|
Lutterloh EC, Opal SM, Pittman DD, Keith JC, Tan XY, Clancy BM, Palmer H, Milarski K, Sun Y, Palardy JE, Parejo NA, Kessimian N. Inhibition of the RAGE products increases survival in experimental models of severe sepsis and systemic infection. Crit Care 2007; 11:R122. [PMID: 18042296 PMCID: PMC2246216 DOI: 10.1186/cc6184] [Citation(s) in RCA: 131] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2007] [Revised: 09/25/2007] [Accepted: 11/06/2007] [Indexed: 01/26/2023] Open
Abstract
INTRODUCTION The receptor for advanced glycation end products (RAGE), a multi-ligand member of the immunoglobulin superfamily, contributes to acute and chronic disease processes, including sepsis. METHODS We studied the possible therapeutic role of RAGE inhibition in the cecal ligation and puncture (CLP) model of polymicrobial sepsis and a model of systemic listeriosis using mice genetically deficient in RAGE expression or mice injected with a rat anti-murine RAGE monoclonal antibody. RESULTS The 7-day survival rates after CLP were 80% for RAGE-/- mice (n = 15) (P < 0.01 versus wild-type), 69% for RAGE+/- mice (n = 23), and 37% for wild-type mice (n = 27). Survival benefits were evident in BALB/c mice given anti-RAGE antibody (n = 15 per group) over serum-treated control animals (P < 0.05). Moreover, delayed treatment with anti-RAGE antibody up to 24 hours after CLP resulted in a significant survival benefit compared with control mice. There was no significant increase in tissue colony counts from enteric Gram-negative or Gram-positive bacteria in animals treated with anti-RAGE antibody. RAGE-/-, RAGE+/-, and anti-RAGE antibody-treated animals were resistant to lethality from Listeria monocytogenes by almost two orders of magnitude compared with wild-type mice. CONCLUSION Further studies are warranted to determine the clinical utility of anti-RAGE antibody as a novel treatment for sepsis.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/therapeutic use
- Disease Models, Animal
- Glycation End Products, Advanced/antagonists & inhibitors
- Glycation End Products, Advanced/biosynthesis
- Glycation End Products, Advanced/genetics
- Listeriosis/metabolism
- Listeriosis/mortality
- Listeriosis/therapy
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Knockout
- Receptor for Advanced Glycation End Products
- Receptors, Immunologic/antagonists & inhibitors
- Receptors, Immunologic/biosynthesis
- Receptors, Immunologic/genetics
- Receptors, Immunologic/immunology
- Sepsis/genetics
- Sepsis/mortality
- Sepsis/therapy
- Survival Rate
- Systemic Inflammatory Response Syndrome/metabolism
- Systemic Inflammatory Response Syndrome/mortality
- Systemic Inflammatory Response Syndrome/therapy
Collapse
Affiliation(s)
- Emily C Lutterloh
- Division of Infectious Diseases, Memorial Hospital of Rhode Island, 111 Brewster St., Pawtucket, RI 02860
| | - Steven M Opal
- Division of Infectious Diseases, Memorial Hospital of Rhode Island, 111 Brewster St., Pawtucket, RI 02860
| | | | - James C Keith
- Wyeth Research, 35 Cambridge Park Dr., Cambridge, MA 02140
| | - Xiang-Yang Tan
- Wyeth Research, 35 Cambridge Park Dr., Cambridge, MA 02140
| | - Brian M Clancy
- Wyeth Research, 35 Cambridge Park Dr., Cambridge, MA 02140
| | - Helen Palmer
- Wyeth Research, 35 Cambridge Park Dr., Cambridge, MA 02140
| | - Kim Milarski
- Wyeth Research, 35 Cambridge Park Dr., Cambridge, MA 02140
| | - Ying Sun
- Wyeth Research, 35 Cambridge Park Dr., Cambridge, MA 02140
| | - John E Palardy
- Division of Infectious Diseases, Memorial Hospital of Rhode Island, 111 Brewster St., Pawtucket, RI 02860
| | - Nicholas A Parejo
- Division of Infectious Diseases, Memorial Hospital of Rhode Island, 111 Brewster St., Pawtucket, RI 02860
| | - Noubar Kessimian
- Department of Pathology, Memorial Hospital of Rhode Island, 111 Brewster St., Pawtucket, RI 02860
| |
Collapse
|
712
|
Sessa L, Bianchi ME. The evolution of High Mobility Group Box (HMGB) chromatin proteins in multicellular animals. Gene 2007; 387:133-40. [PMID: 17156942 DOI: 10.1016/j.gene.2006.08.034] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2006] [Revised: 07/14/2006] [Accepted: 08/31/2006] [Indexed: 11/29/2022]
Abstract
Mammalian HMGB proteins are abundant chromatin components, and are characterized by the presence of 2 HMG-box domains and an acidic tail. HMG boxes are present in a large number of DNA-binding proteins, and HMGB chromatin proteins represent a small and specific subset of HMG-box proteins. The comparison of DNA sequences that code for HMG-box proteins suggests that the ancestral HMG box was coded by an intronless gene, which picked up one or more introns during its radiation. Canonical HMGB proteins are only present in multicellular animals, from sponges onwards, and appear to have arisen through the fusion of two different genes, each coding for one of the boxes. The organization of HMGB genes was very conserved during Metazoan evolution, with the only deviations appearing in Caenorhabditis and Dipteran (Drosophila and Anopheles) species.
Collapse
Affiliation(s)
- Luca Sessa
- Vita-Salute San Raffaele University, Chromatin Dynamics Unit, via Olgettina 58, 20132 Milano, Italy
| | | |
Collapse
|
713
|
Ramji N, Toth C, Kennedy J, Zochodne DW. Does diabetes mellitus target motor neurons? Neurobiol Dis 2006; 26:301-11. [PMID: 17337195 DOI: 10.1016/j.nbd.2006.11.016] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2006] [Revised: 11/02/2006] [Accepted: 11/04/2006] [Indexed: 11/18/2022] Open
Abstract
A pattern of peripheral neurodegeneration occurs in chronic diabetes mellitus in which an early, but selective retraction of distal axons may occur prior to any irretrievable neuronal loss. Clinical observations suggest that sensory systems undergo damage before those of motor neurons. In this work, we examined the fate of the spinal motor neuron in a long-term chronic model of experimental (streptozotocin-induced) diabetes already known to be associated with substantial loss of sensory neurons. The integrity, physiological function, and critical forms of protein expression of the full motor neuron tree was examined in mice exposed to 8 months of diabetes. Motor neurons developed progressive features of distal loss of axonal terminals but without perikaryal dropout, indicating distal axon retraction. While numbers and caliber of motor neuron perikarya and their nerve trunk axons were preserved, axons developed conduction velocity slowing, loss of motor units and neuromuscular junctions, and compensatory single motor unit action potential enlargement. Four critical proteins directly linked to diabetic complications were altered in motor neurons of diabetic mice: an elevated perikaryal expression of RAGE and PARP, molecules associated with cellular stress, along with concurrent rises in HSP-27 and pAKT, molecules alternatively identified with neuroprotective survival. Moreover, Akt mRNA was increased in diabetic lumbar spinal cords. Overall these findings indicate that although motor neurons are resistant to irretrievable dropout, they are targeted nonetheless by diabetes and gradually withdraw their terminals from distal innervation.
Collapse
Affiliation(s)
- Noor Ramji
- University of Calgary, Department of Clinical Neurosciences, Room 168, 3330 Hospital Drive, N.W., Calgary, Alberta, Canada T2N 4N1
| | | | | | | |
Collapse
|
714
|
Sawa H, Ueda T, Takeyama Y, Yasuda T, Shinzeki M, Nakajima T, Kuroda Y. Blockade of high mobility group box-1 protein attenuates experimental severe acute pancreatitis. World J Gastroenterol 2006; 12:7666-70. [PMID: 17171797 PMCID: PMC4088050 DOI: 10.3748/wjg.v12.i47.7666] [Citation(s) in RCA: 115] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To examine the effects of anti-high mobility group box 1 (HMGB1) neutralizing antibody in experimental severe acute pancreatitis (SAP).
METHODS: SAP was induced by creating closed duodenal loop in C3H/HeN mice. SAP was induced immediately after intraperitoneal injection of anti-HMGB1 neutralizing antibody (200 μg). Severity of pancreatitis, organ injury (liver, kidney and lung), and bacterial translocation to pancreas was examined 12 h after induction of SAP.
RESULTS: Anti-HMGB1 neutralizing antibody significantly improved the elevation of the serum amylase level and the histological alterations of pancreas and lung in SAP. Anti-HMGB1 antibody also significantly ameliorated the elevations of serum alanine aminotransferase and creatinine in SAP. However, anti-HMGB1 antibody worsened the bacterial translocation to pancreas.
CONCLUSION: Blockade of HMGB1 attenuated the development of SAP and associated organ dysfunction, suggesting that HMGB1 may act as a key mediator for inflammatory response and organ injury in SAP.
Collapse
Affiliation(s)
- Hidehiro Sawa
- Department of Gastroenterological Surgery, Kobe University Graduate School of Medical Sciences, Kobe 650-0017, Japan.
| | | | | | | | | | | | | |
Collapse
|
715
|
Yang R, Harada T, Mollen KP, Prince JM, Levy RM, Englert JA, Gallowitsch-Puerta M, Yang L, Yang H, Tracey KJ, Harbrecht BG, Billiar TR, Fink MP. Anti-HMGB1 neutralizing antibody ameliorates gut barrier dysfunction and improves survival after hemorrhagic shock. Mol Med 2006; 12:105-14. [PMID: 16953558 PMCID: PMC1578769 DOI: 10.2119/2006-00010.yang] [Citation(s) in RCA: 184] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2006] [Accepted: 04/15/2006] [Indexed: 01/29/2023] Open
Abstract
Intestinal barrier dysfunction occurs following hemorrhagic shock and resuscitation (HS/R). High-mobility group B1 (HMGB1) has been shown to increase the permeability of Caco-2 human enterocyte-like epithelial monolayers in vitro. In this study, we found that serum concentrations of HMGB1 were higher in blood samples obtained from 25 trauma victims with hemorrhagic shock than in 9 normal volunteers. We also studied whether treatment with anti-HMGB1 antibody can ameliorate HS/R-induced gut barrier dysfunction in mice. Animals were shocked by withdrawal of blood to maintain mean arterial pressure at 25 to 30 mmHg for 2 h. After resuscitation with shed blood plus Ringer's lactate solution, the mice were treated with either anti-HMGB1 antibody or nonimmune rabbit IgG. Serum HMGB1 concentrations were significantly higher in trauma victims than control mice. Treatment with anti-HMGB1 antibody improved survival at 24 h and ameliorated the development of ileal mucosal hyperpermeability to FITC-labeled dextran. At 24 h after HS/R, treatment with anti-HMGB1 antibody decreased bacterial translocation to mesenteric lymph nodes and was associated with lower circulating concentrations of IL-6 and IL-10. These data support the notion that HMGB1 is a mediator of HS/R-induced gut barrier dysfunction and suggest that anti-HMGB1 antibodies warrant further evaluation as a therapeutic to ameliorate the morbidity of HS/R in trauma patients.
Collapse
Affiliation(s)
| | | | - Kevin P Mollen
- Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jose M Prince
- Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Ryan M Levy
- Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | | | - LiHong Yang
- Laboratory of Biomedical Science, The Feinstein Institute for Medical Research, Manhasset, NY, USA
| | - Huan Yang
- Laboratory of Biomedical Science, The Feinstein Institute for Medical Research, Manhasset, NY, USA
| | - Kevin J Tracey
- Laboratory of Biomedical Science, The Feinstein Institute for Medical Research, Manhasset, NY, USA
| | - Brian G Harbrecht
- Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Timothy R Billiar
- Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Mitchell P Fink
- Departments of Critical Care Medicine and
- Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Address correspondence and reprint requests to Mitchell P. Fink, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, 616 Scaife Hall, 3550 Terrace St, Pittsburgh PA 15261. Phone: 412-647-6965; fax: 412-647-5258;
| |
Collapse
|
716
|
Pullerits R, Brisslert M, Jonsson IM, Tarkowski A. Soluble receptor for advanced glycation end products triggers a proinflammatory cytokine cascade via beta2 integrin Mac-1. ARTHRITIS AND RHEUMATISM 2006; 54:3898-3907. [PMID: 17133598 DOI: 10.1002/art.22217] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
OBJECTIVE Receptor for advanced glycation end products (RAGE) is a cell surface molecule that binds a variety of ligands, including high mobility group box chromosomal protein 1 (HMGB-1), a potent proinflammatory cytokine. RAGE-ligand interaction leads to an inflammatory response. A truncated form of the receptor, soluble RAGE (sRAGE), has been suggested to function as a decoy abrogating cellular activation, but its endogenous activity is not fully understood. We undertook this study to assess the properties of sRAGE in vivo and in vitro and to analyze the role of sRAGE in HMGB-1-induced arthritis. METHODS Mice were injected intraarticularly with HMGB-1 and treated systemically with sRAGE prior to histologic joint evaluation. All animals were subjected to peritoneal lavage to assess the local effect of sRAGE treatment. For in vitro studies, mouse splenocytes were incubated with sRAGE followed by assessment of NF-kappaB activation and cytokine production. The chemotactic properties of sRAGE were investigated using in vitro migration assay. RESULTS Soluble RAGE was determined to have proinflammatory properties since it gave rise to production of interleukin-6, tumor necrosis factor alpha, and macrophage inflammatory protein 2. This effect was triggered by interaction with leukocyte beta2 integrin Mac-1 and was mediated via NF-kappaB. Systemic treatment with sRAGE significantly down-regulated HMGB-1-triggered arthritis, but the observed effect was due to a deviation of the inflammatory response from the joint to the peritoneal cavity rather than a genuine antiinflammatory effect. Apart from its proinflammatory properties, sRAGE was proven to act as a chemotactic stimulus for neutrophils. CONCLUSION We conclude that sRAGE interacts with Mac-1, thereby acting as an important proinflammatory and chemotactic molecule.
Collapse
Affiliation(s)
- Rille Pullerits
- Department of Rheumatology and Inflammation Research, The Sahlgrenska Academy at Göteborg University, Göteborg, Sweden.
| | | | | | | |
Collapse
|
717
|
Marubuchi S, Okuda T, Tagawa K, Enokido Y, Horiuchi D, Shimokawa R, Tamura T, Qi ML, Eishi Y, Watabe K, Shibata M, Nakagawa M, Okazawa H. Hepatoma-derived growth factor, a new trophic factor for motor neurons, is up-regulated in the spinal cord of PQBP-1 transgenic mice before onset of degeneration. J Neurochem 2006; 99:70-83. [PMID: 16987236 DOI: 10.1111/j.1471-4159.2006.04021.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Hepatoma-derived growth factor (HDGF) is a nuclear protein homologous to the high-mobility group B1 family of proteins. It is known to be released from cells and to act as a trophic factor for dividing cells. In this study HDGF was increased in spinal motor neurons of a mouse model of motor neuron degeneration, polyglutamine-tract-binding protein-1 (PQBP-1) transgenic mice, before onset of degeneration. HDGF promoted neurite extension and survival of spinal motor neurons in primary culture. HDGF repressed cell death of motor neurons after facial nerve section in newborn rats in vivo. We also found a significant increase in p53 in spinal motor neurons of the transgenic mice. p53 bound to a sequence in the upstream of the HDGF gene in a gel mobility shift assay, and promoted gene expression through the cis-element in chloramphenicol acetyl transfer (CAT) assay. Finally, we found that HDGF was increased in CSF of PQBP-1 transgenic mice. Collectively, our results show that HDGF is a novel trophic factor for motor neurons and suggest that it might play a protective role against motor neuron degeneration in PQBP-1 transgenic mice.
Collapse
Affiliation(s)
- Shigeki Marubuchi
- Department of Neuropathology, Medical Research Institute and 21st Century Center of Excellence Program for Brain Integration and Its Disorders, Tokyo Medical and Dental University, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
718
|
Yasuda T, Ueda T, Takeyama Y, Shinzeki M, Sawa H, Nakajima T, Ajiki T, Fujino Y, Suzuki Y, Kuroda Y. Significant increase of serum high-mobility group box chromosomal protein 1 levels in patients with severe acute pancreatitis. Pancreas 2006; 33:359-63. [PMID: 17079940 DOI: 10.1097/01.mpa.0000236741.15477.8b] [Citation(s) in RCA: 124] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Multiple organ failure because of systemic inflammatory response in the early phase and sepsis in the late phase is the main contributor to high mortality in severe acute pancreatitis (SAP). High-mobility group box chromosomal protein 1 (HMGB1) was recently identified as a potent proinflammatory mediator and increases in various pathological conditions such as sepsis. The aim of this study was to investigate contributions of HMGB1 in SAP. METHODS We measured serum HMGB1 concentrations by an enzyme-linked immunosorbent assay in 45 patients with SAP at the time of admission. Furthermore, relationship between their serum HMGB1 levels and clinical factors was analyzed. RESULTS The mean value of serum HMGB1 levels was significantly higher in patients with SAP (5.4 +/- 1.3 ng/mL) than that in healthy volunteers (1.7 +/- 0.3 ng/mL). Serum HMGB1 levels were significantly positively correlated with the Japanese severity score and Glasgow score. Serum HMGB1 levels were significantly positively correlated with lactate dehydrogenase, C-reactive protein, and total bilirubin. The HMGB1 levels were higher in patients with organ dysfunction and infection during the clinical course. The HMGB1 levels in nonsurvivors were higher than those in survivors. Serum HMGB1 levels gradually declined after the admission. CONCLUSIONS Serum HMGB1 levels were significantly increased in patients with SAP and were correlated with disease severity. These results suggest that HMGB1 may act as a key mediator for inflammation and organ failure in SAP.
Collapse
Affiliation(s)
- Takeo Yasuda
- Department of Gastroenterological Surgery, Kobe University Graduate School of Medical Sciences, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
719
|
Yu M, Wang H, Ding A, Golenbock DT, Latz E, Czura CJ, Fenton MJ, Tracey KJ, Yang H. HMGB1 signals through toll-like receptor (TLR) 4 and TLR2. Shock 2006; 26:174-9. [PMID: 16878026 DOI: 10.1097/01.shk.0000225404.51320.82] [Citation(s) in RCA: 669] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
In response to bacterial endotoxin (e.g., LPS) or endogenous proinflammatory cytokines (e.g., TNF and IL-1beta), innate immune cells release HMGB1, a late cytokine mediator of lethal endotoxemia and sepsis. The delayed kinetics of HMGB1 release makes it an attractive therapeutic target with a wider window of opportunity for the treatment of lethal systemic inflammation. However, the receptor(s) responsible for HMGB1-mediated production of proinflammatory cytokines has not been well characterized. Here we demonstrate that in human whole blood, neutralizing antibodies against Toll-like receptor 4 (TLR4, but not TLR2 or receptor for advanced glycation end product) dose-dependently attenuate HMGB1-induced IL-8 release. Similarly, in primary human macrophages, HMGB1-induced TNF release is dose-dependently inhibited by anti-TLR4 antibodies. In primary macrophages from knockout mice, HMGB1 activates significantly less TNF release in cells obtained from MyD88 and TLR4 knockout mice as compared with cells from TLR2 knockout and wild-type controls. However, in human embryonic kidney 293 cells transfected with TLR2 or TLR4, HMGB1 effectively induces IL-8 release only from TLR2 overexpressing cells. Consistently, anti-TLR2 antibodies dose-dependently attenuate HMGB1-induced IL-8 release in human embryonic kidney/TLR2-expressing cells and markedly reduce HMGB1 cell surface binding on murine macrophage-like RAW 264.7 cells. Taken together, our data suggest that there is a differential usage of TLR2 and TLR4 in HMGB1 signaling in primary cells and in established cell lines, adding complexity to studies of HMGB1 signaling which was not previously expected.
Collapse
Affiliation(s)
- Man Yu
- Laboratories of Biomedical Science, The Feinstein Institute for Medical Research, Manhasset, NY 11030, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
720
|
Sugars R, Karlström E, Christersson C, Olsson ML, Wendel M, Fried K. Expression of HMGB1 during tooth development. Cell Tissue Res 2006; 327:511-9. [PMID: 17043795 DOI: 10.1007/s00441-006-0293-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2006] [Accepted: 06/20/2006] [Indexed: 10/24/2022]
Abstract
High mobility group box 1 (HMGB1) is a nuclear and cytosolic protein that can act as a transcription factor, a growth factor, or a cytokine. To elucidate a possible role for HMGB1 in tooth development, we have studied the expression of HMGB1 and its receptor RAGE (receptor for advanced glycation end-products) during the late fetal and early postnatal period of rat by using light- and electron-microscopic immunohistochemistry. Low HMGB1 protein expression was observed during fetal and newborn stages of tooth development. However, from postnatal day 5 (P5) onward, a marked increase occurred in the levels of the protein in most dental cell types. Expression was particularly high in ameloblasts and odontoblasts at regions of ongoing mineralization. Although most HMGB1 immunoreactivity was confined to cell nuclei, it was also present in odontoblast cytoplasm. At P5, ameloblasts and odontoblasts also showed RAGE immunoreactivity, and reverse transcription-polymerase chain reaction demonstrated both HMGB1 and RAGE mRNA in human dental pulp cells in vitro. Immunoblots performed on extracts from bovine dentin demonstrated a principal band at approximately 27 kDa, indicating that HMGB1 participates in tooth mineralization. The expression of both ligand and receptor suggests an autocrine/paracrine HMGB1 signalling axis in odontoblasts.
Collapse
Affiliation(s)
- R Sugars
- Center for Oral Biology, Novum, Karolinska Institutet, Huddinge, Sweden
| | | | | | | | | | | |
Collapse
|
721
|
Rauvala H. Interview with Dr. Heikki Rauvala regarding pivotal advance: analysis of proinflammatory activity of highly purified eukaryotic recombinant HMGB1 (amphoterin). Interview by Marco E Bianchi. J Leukoc Biol 2006; 81:46-48. [PMID: 17038581 DOI: 10.1189/jlb.1306200] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
722
|
Abstract
Multicellular animals detect pathogens via a set of receptors that recognize pathogen-associated molecular patterns (PAMPs). However, pathogens are not the only causative agents of tissue and cell damage: trauma is another one. Evidence is accumulating that trauma and its associated tissue damage are recognized at the cell level via receptor-mediated detection of intracellular proteins released by the dead cells. The term "alarmin" is proposed to categorize such endogenous molecules that signal tissue and cell damage. Intriguingly, effector cells of innate and adaptive immunity can secrete alarmins via nonclassical pathways and often do so when they are activated by PAMPs or other alarmins. Endogenous alarmins and exogenous PAMPs therefore convey a similar message and elicit similar responses; they can be considered subgroups of a larger set, the damage-associated molecular patterns (DAMPs).
Collapse
Affiliation(s)
- Marco E Bianchi
- San Raffaele University, Chromatin Dynamics Unit, via Olgettina 58, 20132 Milan, Italy.
| |
Collapse
|
723
|
Yamada S, Yakabe K, Ishii J, Imaizumi H, Maruyama I. New high mobility group box 1 assay system. Clin Chim Acta 2006; 372:173-8. [PMID: 16797518 DOI: 10.1016/j.cca.2006.04.016] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2006] [Revised: 04/08/2006] [Accepted: 04/10/2006] [Indexed: 10/24/2022]
Abstract
BACKGROUND High-sensitivity sandwich ELISA methods have been developed using chemiluminescent substrates. HMGB1 (high mobility group box 1) protein has been shown to play a critical role in several inflammatory diseases and it may be involved in the development of atherosclerosis. METHODS Anti-human HMGB1 monoclonal antibodies and anti-peptide polyclonal antibodies against the peptide sequence (KPDAAKKGVVKAEK) with high antigenicity and different from the sequence of HMGB2 were developed, and the antibodies were used to construct sandwich ELISA methods with a chromogenic substrate (TMBZ) and a chemiluminescent substrate (PS-atto). Highly purified human HMGB1 was used as a standard material and high-sensitivity CRP was measured to compare with HMGB1. RESULTS The analytical characteristics of the ELISA method we developed were validated inter-assay and intra-assay CVs were <10%, and the detection limit was 0.3 microg/l by the chemiluminescent method and 1 microg/l with the chromogenic substrates. HMGB1 was detected in the serum of patients with acute coronary syndrome (ACS). When a cut-off of 0.6 microg/l HMGB1 upon admission to the intensive care unit (ICU) was used, the risk of developing an acute cardiac event within 1 month after discharge of ACS patients with an abnormal HMGB1 was significantly higher than for the patients with normal values (P<0.0001). The usefulness of HMGB1 as an acute prognostic marker was suggested. CONCLUSIONS The assay is easy to perform and suitable for use in the hospital laboratory and for screening large populations. HMGB1 is detectable in the serum of ACS patients and that the serum concentration of HMGB1 may be a prognostic indicator in ACS patients.
Collapse
|
724
|
Raman KG, Sappington PL, Yang R, Levy RM, Prince JM, Liu S, Watkins SK, Schmidt AM, Billiar TR, Fink MP. The role of RAGE in the pathogenesis of intestinal barrier dysfunction after hemorrhagic shock. Am J Physiol Gastrointest Liver Physiol 2006; 291:G556-65. [PMID: 16751175 DOI: 10.1152/ajpgi.00055.2006] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The receptor for advanced glycation end products (RAGE) has been implicated in the pathogenesis of numerous conditions associated with excessive inflammation. To determine whether RAGE-dependent signaling is important in the development of intestinal barrier dysfunction after hemorrhagic shock and resuscitation (HS/R), C57Bl/6, rage(-/-), or congenic rage(+/+) mice were subjected to HS/R (mean arterial pressure of 25 mmHg for 3 h) or a sham procedure. Twenty-four hours later, bacterial translocation to mesenteric lymph nodes and ileal mucosal permeability to FITC-labeled dextran were assessed. Additionally, samples of ileum were obtained for immunofluorescence microscopy, and plasma was collected for measuring IL-6 and IL-10 levels. HS/R in C57Bl/6 mice was associated with increased bacterial translocation, ileal mucosal hyperpermeability, and high circulating levels of IL-6. All of these effects were prevented when C57Bl/6 mice were treated with recombinant human soluble RAGE (sRAGE; the extracellular ligand-binding domain of RAGE). HS/R induced bacterial translocation, ileal mucosal hyperpermeability, and high plasma IL-6 levels in rage(+/+) but not rage(-/-) mice. Circulating IL-10 levels were higher in rage(-/-) compared with rage(+/+) mice. These results suggest that activation of RAGE-dependent signaling is a key factor leading to gut mucosal barrier dysfunction after HS/R.
Collapse
Affiliation(s)
- Kathleen G Raman
- Univ. of Pittsburgh School of Medicine, 616 Scaife Hall, 3550 Terrace St., Pittsburgh, PA 15213, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
725
|
Yang D, Chen Q, Yang H, Tracey KJ, Bustin M, Oppenheim JJ. High mobility group box-1 protein induces the migration and activation of human dendritic cells and acts as an alarmin. J Leukoc Biol 2006; 81:59-66. [PMID: 16966386 DOI: 10.1189/jlb.0306180] [Citation(s) in RCA: 302] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
High mobility group box-1 (HMGB1) protein is a nonhistone, DNA-binding protein that plays a critical role in regulating gene transcription. Recently, HMGB1 has also been shown to act as a late mediator of endotoxic shock and to exert a variety of proinflammatory, extracellular activities. Here, we report that HMGB1 simultaneously acts as a chemoattractant and activator of dendritic cells (DCs). HMGB1 induced the migration of monocyte-derived, immature DCs (Mo-iDCs) but not mature DCs. The chemotactic effect of HMGB1 on iDCs was pertussis toxin-inhibitable and also inhibited by antibody against the receptor of advanced glycation end products (RAGE), suggesting that HMGB1 chemoattraction of iDCs is mediated by RAGE in a Gi protein-dependent manner. In addition, HMGB1 treatment of Mo-iDCs up-regulated DC surface markers (CD80, CD83, CD86, and HLA-A,B,C), enhanced DC production of cytokines (IL-6, CXCL8, IL-12p70, and TNF-alpha), switched DC chemokine responsiveness from CCL5-sensitive to CCL21-sensitive, and acquired the capacity to stimulate allogeneic T cell proliferation. Based on its dual DC-attracting and -activating activities as well as its reported capacity to promote an antigen-specific immune response, we consider HMGB1 to have the properties of an immune alarmin.
Collapse
Affiliation(s)
- De Yang
- Laboratory of Molecular Immunoregulation, Center for Cancer Research, and Basic Research Program, Rm. 31-19/Bldg. 560, 1050 Boyles Street, Frederick, MD 21702, USA.
| | | | | | | | | | | |
Collapse
|
726
|
Shim CY, Park S, Yoon SJ, Park HY, Kim HT, Oh B, Park C, Ko YG, Choi D, Jang Y, Chung N. Association of RAGE gene polymorphisms with in-stent restenosis in non-diabetic Korean population. Cardiology 2006; 107:261-8. [PMID: 16954682 DOI: 10.1159/000095516] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2005] [Accepted: 05/23/2006] [Indexed: 11/19/2022]
Abstract
BACKGROUND Interaction between advanced glycosylation end products (AGEs) and receptor for AGEs (RAGE) in vessel wall may lead to inflammation, smooth muscle cell proliferation, and extracellular matrix production, culminating in exaggerated intimal hyperplasia and restenosis. We investigated the possibility that single nucleotide polymorphisms of the genes encoding RAGE are associated with in-stent restenosis after coronary stenting. METHODS Our study included 334 consecutive non-diabetic male patients with symptomatic coronary artery disease who underwent bare metal stent implantation. Follow-up angiography was performed in 297 patients (88.9%), 6 months after intervention. We screened -1106T-->C, -443T-->C, -388T-->A, -257G-->A, +557G-->A and +1704G-->T RAGE polymorphisms in these patients. Genotyping was performed by single base extension with amplifying primers and probes for TaqMan. RESULTS A total of 355 target lesions were evaluated, in 297 patients. There was no significant association of the -1106T-->C, -443T-->C, -388T-->A, -257G-->A, +557G-->A or +1704G-->T polymorphisms with in-stent restenosis after coronary artery stenting. We did not observe a significant difference between the genotype distributions and the rates of angiographic restenosis between the polymorphisms. In a multivariate analysis of angiographic restenosis, we examined the possible influence of the baseline, lesion-related, and procedural variables. After adjustment for these potentially confounding factors, the multivariate analysis did not reveal an association of polymorphism with angiographic restenosis. CONCLUSION Our observation suggests that the RAGE gene polymorphism is not associated with in-stent restenosis after coronary artery stenting in non-diabetic patients in the Korean population.
Collapse
Affiliation(s)
- Chi Young Shim
- Division of Cardiology, Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, South Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
727
|
Yan SF, Yan SD, Herold K, Ramsamy R, Schmidt AM. Receptor for advanced glycation end products and the cardiovascular complications of diabetes and beyond: lessons from AGEing. Endocrinol Metab Clin North Am 2006; 35:511-24, viii. [PMID: 16959583 DOI: 10.1016/j.ecl.2006.06.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The presence of elevated blood glucose levels characterizes the diabetic state. Hyperglycemia may be caused by a number of underlying factors; however, the consequences of chronically elevated glucose are similar. Both the macrovasculature and microvasculature are exquisitely sensitive to the long-term effects of elevated blood glucose. Cardiovascular disease remains the leading cause of morbidity and mortality in diabetes, regardless of the underlying cause of hyperglycemia. Although other substrates, such as DNA, are susceptible to glycation, this article addresses the impact of nonenzymatic glycation on the proteome. The impact of Advanced Glycation End products (AGEs) on alteration of protein function and signal transduction mechanisms contributes to the pathogenesis of diabetes complications. This suggests that blocking the generation or molecular impact of AGEs may modulate the complications of diabetes.
Collapse
Affiliation(s)
- Shi Fang Yan
- Columbia University Medical Center, 630 West 168th Street, P&S 17-501, New York, NY 10032, USA
| | | | | | | | | |
Collapse
|
728
|
Germani A, Limana F, Capogrossi MC. Pivotal advances: high-mobility group box 1 protein--a cytokine with a role in cardiac repair. J Leukoc Biol 2006; 81:41-5. [PMID: 16940333 DOI: 10.1189/jlb.0306165] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The nuclear protein high-mobility group box 1 (HMGB1) has been largely characterized for its role in inflammation. However, HMGB1 released by inflammatory cells, as well as by necrotic cells, may also act as a signal of tissue damage and participate in tissue repair by recruiting stem cells to the injury site. The emergence of this function has focused the interest on HMGB1 as a molecule with an active role in tissue regeneration. We recently demonstrated that HMGB1 administration in a mouse model of myocardial infarction activates cardiac stem cells and promotes their differentiation into cardiomyocytes. The regenerative effect results in the improvement of cardiac function. In this review, we highlight the beneficial role of HMGB1 and discuss growth factor-based therapeutic approaches for the treatment of myocardial infarction.
Collapse
Affiliation(s)
- Antonia Germani
- Laboratorio di Biologia Vascolare e Terapia Genica, Centro Cardiologico Fondazione Monzino, Istituto di Ricovero e Cura a Carattere Scientifico, Milan, Italy
| | | | | |
Collapse
|
729
|
Sitia G, Iannacone M, Müller S, Bianchi ME, Guidotti LG. Treatment with HMGB1 inhibitors diminishes CTL-induced liver disease in HBV transgenic mice. J Leukoc Biol 2006; 81:100-7. [PMID: 16935945 DOI: 10.1189/jlb.0306173] [Citation(s) in RCA: 108] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Using hepatitis B virus (HBV) transgenic mice as recipients of virus-specific cytotoxic T lymphocytes (CTLs), we recently showed that polymorphonuclear neutrophils (PMNs) and the matrix-degrading metalloproteinases (MMPs) they produce are necessary for the intrahepatic recruitment of antigen nonspecific mononuclear cells that amplify the liver damage initiated by the CTLs. We now report that the high-mobility group box 1 protein (HMGB1) is also involved in this process. Transfer of CTLs in HBV transgenic mice induces the translocation of HMGB1 from the nucleus to the cytoplasm of hepatocytes surrounding CTL-containing necroinflammatory liver foci, without significant net synthesis of HMGB1. Treatment of CTL-injected HBV transgenic mice with either recombinant Box-A or glycyrrhizin, two functional inhibitors of extracellular HMGB1, significantly decreases the intrahepatic recruitment of PMNs and all other inflammatory cells, in the face of intact homing of virus-specific CTLs into the liver. The inhibition of PMN chemoattraction explains the mode of action of glycyrrhizin, which has long been used in Japan for the treatment of hepatitis, and suggests that new and more potent inhibitors of HMGB1 may be useful for the treatment of patients chronically infected with HBV.
Collapse
Affiliation(s)
- Giovanni Sitia
- San Raffaele Scientific Institute, Chromatin Dynamics Unit, Via Olgettina 58, Milan 20132, Italy.
| | | | | | | | | |
Collapse
|
730
|
Pedrazzi M, Raiteri L, Bonanno G, Patrone M, Ledda S, Passalacqua M, Milanese M, Melloni E, Raiteri M, Pontremoli S, Sparatore B. Stimulation of excitatory amino acid release from adult mouse brain glia subcellular particles by high mobility group box 1 protein. J Neurochem 2006; 99:827-38. [PMID: 16911580 DOI: 10.1111/j.1471-4159.2006.04120.x] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The multifunctional protein high mobility group box 1 (HMGB1) is expressed in hippocampus and cerebellum of adult mouse brain. Our aim was to determine whether HMGB1 affects glutamatergic transmission by monitoring neurotransmitter release from glial (gliosomes) and neuronal (synaptosomes) re-sealed subcellular particles isolated from cerebellum and hippocampus. HMGB1 induced release of the glutamate analogue [(3)H]d-aspartate form gliosomes in a concentration-dependent manner, whereas nerve terminals were insensitive to the protein. The HMGB1-evoked release of [(3)H]d-aspartate was independent of modifications of cytosolic Ca(2+) , but it was blocked by dl-threo-beta-benzyloxyaspartate (dl-TBOA), an inhibitor of glutamate transporters. HMGB1 also stimulated the release of endogenous glutamate in a Ca(2+)-independent and dl-TBOA-sensitive manner. These findings suggest the involvement of carrier-mediated release. Moreover, dihydrokainic acid, a selective inhibitor of glutamate transporter 1 (GLT1), does not block the effect of HMGB1, indicating a role for the glial glutamate-aspartate transporter (GLAST) subtype in this response. We also demonstrate that HMGB1/glial particles association is promoted by Ca(2+). Furthermore, although HMGB1 can physically interact with GLAST and the receptor for advanced glycation end products (RAGE), only its binding with RAGE is promoted by Ca(2+). These results suggest that the HMGB1 cytokine could act as a modulator of glutamate homeostasis in adult mammal brain.
Collapse
Affiliation(s)
- Marco Pedrazzi
- Department of Experimental Medicine and Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
731
|
O'Callaghan A, Wang J, Redmond HP. HMGB1 as a key mediator of tissue response to injury: roles in inflammation and tissue repair. Eur Surg 2006. [DOI: 10.1007/s10353-006-0255-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
732
|
Li J, Wang H, Mason JM, Levine J, Yu M, Ulloa L, Czura CJ, Tracey KJ, Yang H. Recombinant HMGB1 with cytokine-stimulating activity. J Immunol Methods 2006; 289:211-23. [PMID: 15251426 DOI: 10.1016/j.jim.2004.04.019] [Citation(s) in RCA: 114] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2004] [Revised: 04/16/2004] [Accepted: 04/24/2004] [Indexed: 11/29/2022]
Abstract
We describe methods for the isolation, purification, and characterization of full-length high-mobility group box 1 (HMGB1) and truncated mutants expressed in bacteria and in mammalian Chinese Hamster Ovary (CHO) cells. HMGB1 is an abundant nuclear and cytoplasmic protein, highly conserved across species and widely distributed in eukaryotic cells from yeast to man. As a ubiquitous nuclear DNA binding protein, HMGB1 binds DNA, facilitates gene transcription, and stabilizes nucleosome structure. In addition to these intracellular roles, HMGB1 can be released into the extracellular milieu by activated innate immune cells (i.e., macrophages, monocytes) and functions as a mediator of lethal endotoxemia and sepsis. The proinflammatory cytokine activity of HMGB1 has become an intense area of research and recombinant protein can be a useful tool to probe HMGB1 functions. Due to its dipolar charged properties, HMGB1 isolated by some methods can be contaminated with bacterial products (such as CpG DNA or lipopolysaccharide [LPS]) that may interfere with immunological analyses. Here we report our newly developed methods for the isolation and purification of biologically active HMGB1 from bacteria or mammalian CHO cells that is essentially free of contaminants. This strategy provides an important advance in methodology to facilitate future HMGB1 studies.
Collapse
Affiliation(s)
- Jianhua Li
- Laboratories of Biomedical Science, North Shore-Long Island Jewish Research Institute, Manhasset, NY 11030, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
733
|
Jaulmes A, Thierry S, Janvier B, Raymondjean M, Maréchal V. Activation of sPLA2-IIA and PGE2 production by high mobility group protein B1 in vascular smooth muscle cells sensitized by IL-1beta. FASEB J 2006; 20:1727-9. [PMID: 16807371 DOI: 10.1096/fj.05-5514fje] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Lipid mediators such as prostaglandin E2 (PGE2) play a central role during atherogenesis as a consequence of inflammation. PGE2 is produced from phospholipids by a cascade of enzymatic reactions involving phospholipase A2 (PLA2), cyclooxygenase (COX), and prostaglandin E synthase (PGES). It is released by several cell types, including vascular smooth muscle cells (VSMCs). Recent work has shown that the secretory PLA2-IIA (sPLA2-IIA), the most abundant isoform of secreted PLA2 in VSMCs, acts as a potent cytokine and activates VSMCs through a positive feedback loop. High mobility group protein 1 (HMGB1), also known as amphoterin, is a ubiquitous protein that plays various roles in the nucleus. HMGB1 is released by necrotic cells and by immune cells in response to various inflammatory mediators and acts as a potent proinflammatory cytokine. The present study investigates the role of HMGB1 in the activation of sPLA2-IIA expression and PGE2 production in VSMCs. Recombinant HMGB1 slightly activated the sPLA2-IIA, COX-2, and mPGES-1 genes but dramatically stimulated these genes in VSMCs that had been incubated with the proinflammatory cytokine IL-1beta for 24 h. This effect was accompanied by significantly increased PGE2 release. Induction of the three known receptors of HMGB1, namely RAGE, TLR-2, and TLR-4, by IL-1beta suggests that proinflammatory cytokines sensitize VSMCs to HMGB1. This provides new insights into the role of HMGB1 in VSMCs, suggesting it may be essential for the progression of atherosclerosis.
Collapse
Affiliation(s)
- Amandine Jaulmes
- UMR Physiologie et Physiopathologie, Université Pierre et Marie Curie, CNRS, Paris, France
| | | | | | | | | |
Collapse
|
734
|
Goldstein RS, Gallowitsch-Puerta M, Yang L, Rosas-Ballina M, Huston JM, Czura CJ, Lee DC, Ward MF, Bruchfeld AN, Wang H, Lesser ML, Church AL, Litroff AH, Sama AE, Tracey KJ. ELEVATED HIGH-MOBILITY GROUP BOX 1 LEVELS IN PATIENTS WITH CEREBRAL AND MYOCARDIAL ISCHEMIA. Shock 2006; 25:571-4. [PMID: 16721263 DOI: 10.1097/01.shk.0000209540.99176.72] [Citation(s) in RCA: 202] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Cerebral and myocardial ischemia, two of the leading causes of morbidity and mortality worldwide, are associated with inflammation that can lead to multiple organ failure and death. High-mobility group box 1(HMGB1), a recently described mediator of lethal systemic inflammation, has been detected in individuals with severe sepsis and hemorrhagic shock, but its role during ischemic injury in humans is unknown. To determine whether systemic HMGB1 levels are elevated after ischemic injury, a prospective observational study was performed in subjects with a diagnosis of either Acute Coronary Syndrome (ACS) or cerebral vascular ischemia (transient ischemic attack or cerebral vascular accident). Subjects (n, 16; age [mean], 67+/-16.3 years) were enrolled in the North Shore-LIJ emergency department within 24 h of symptom onset. Blood samples were collected, and HMGB1 levels analyzed by Western blot analysis using previously described methods (Wang et al. Science. 1999). Control samples were obtained from healthy age- and sex-matched volunteers (n, 16; age [mean], 68+/-15.8 years). Here, we report that serum HMGB1 levels were significantly elevated in both myocardial ischemia subjects (myocardial control serum HMGB1, 1.94+/-2.05 ng/mL, vs. myocardial ischemia serum HMGB1, 159+/-54.3 ng/mL; P<0.001); and in cerebral ischemia subjects (cerebral control serum HMGB1, 16.8+/-10.9 ng/mL, vs. cerebral ischemia serum HMGB1, 218+/-18.8 ng/mL; P<0.001). These results suggest that systemic HMGB1 levels are elevated in human ischemic disease.
Collapse
Affiliation(s)
- Richard S Goldstein
- Department of Emergency Medicine, North Shore University Hospital, North Shore-LIJ, Manhasset, NY 11030, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
735
|
Huebschmann AG, Regensteiner JG, Vlassara H, Reusch JEB. Diabetes and advanced glycoxidation end products. Diabetes Care 2006; 29:1420-32. [PMID: 16732039 DOI: 10.2337/dc05-2096] [Citation(s) in RCA: 196] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Amy G Huebschmann
- Division of General Internal Medicine, Department of Medicine, University of Colorado Denver and Health Sciences Center, Mailstop F-729, Aurora, CO 80045, USA.
| | | | | | | |
Collapse
|
736
|
Harashima A, Yamamoto Y, Cheng C, Tsuneyama K, Myint K, Takeuchi A, Yoshimura K, Li H, Watanabe T, Takasawa S, Okamoto H, Yonekura H, Yamamoto H. Identification of mouse orthologue of endogenous secretory receptor for advanced glycation end-products: structure, function and expression. Biochem J 2006; 396:109-15. [PMID: 16503878 PMCID: PMC1450004 DOI: 10.1042/bj20051573] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The cell-surface RAGE [receptor for AGE (advanced glycation end-products)] is associated with the development of diabetic vascular complications, neurodegenerative disorders and inflammation. Recently, we isolated a human RAGE splice variant, which can work as a decoy receptor for RAGE ligands, and named it esRAGE (endogenous secretory RAGE). In the present study, we have isolated the murine equivalent of esRAGE from brain polysomal poly(A)+ (polyadenylated) RNA by RT (reverse transcription)-PCR cloning. The mRNA was generated by alternative splicing, and it encoded a 334-amino-acid protein with a signal sequence, but lacking the transmembrane domain. A transfection experiment revealed that the mRNA was actually translated as deduced to yield the secretory protein working as a decoy in AGE-induced NF-kappaB (nuclear factor kappaB) activation. RT-PCR and immunoblotting detected esRAGE mRNA and protein in the brain, lung, kidney and small intestine of wild-type mice, but not of RAGE-null mice. The esRAGE expression was increased in the kidney of diabetic wild-type mice. The present study has thus provided an animal orthologue of esRAGE for clarification of its roles in health and disease.
Collapse
MESH Headings
- Alternative Splicing
- Amino Acid Sequence
- Animals
- Blotting, Western
- COS Cells
- Cell Line, Tumor
- Chlorocebus aethiops
- Diabetes Mellitus, Experimental/metabolism
- Glioma/pathology
- Glycation End Products, Advanced/metabolism
- Humans
- Intestine, Small/metabolism
- Kidney/metabolism
- Ligands
- Lung/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Molecular Sequence Data
- NF-kappa B/metabolism
- Nerve Tissue Proteins/analysis
- Organ Specificity
- RNA, Messenger/genetics
- Rats
- Receptor for Advanced Glycation End Products
- Receptors, Immunologic/analysis
- Receptors, Immunologic/deficiency
- Receptors, Immunologic/genetics
- Receptors, Immunologic/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Sequence Alignment
- Sequence Homology, Amino Acid
- Solubility
- Species Specificity
- Structure-Activity Relationship
- Surface Plasmon Resonance
- Transfection
Collapse
Affiliation(s)
- Ai Harashima
- *Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Science, Kanazawa 920-8640, Japan
| | - Yasuhiko Yamamoto
- *Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Science, Kanazawa 920-8640, Japan
- To whom correspondence should be addressed (email )
| | - Chunmei Cheng
- †Department of Pathology, School of Medicine, University of Toyama, Toyama 930-0194, Japan
| | - Koichi Tsuneyama
- †Department of Pathology, School of Medicine, University of Toyama, Toyama 930-0194, Japan
| | - Khin Mar Myint
- *Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Science, Kanazawa 920-8640, Japan
| | - Akihiko Takeuchi
- *Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Science, Kanazawa 920-8640, Japan
| | - Kazunobu Yoshimura
- *Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Science, Kanazawa 920-8640, Japan
| | - Hui Li
- *Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Science, Kanazawa 920-8640, Japan
| | - Takuo Watanabe
- *Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Science, Kanazawa 920-8640, Japan
| | - Shin Takasawa
- ‡Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Hiroshi Okamoto
- §Department of Advanced Biological Sciences for Regeneration (Kotobiken Medical Laboratories), Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Hideto Yonekura
- *Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Science, Kanazawa 920-8640, Japan
| | - Hiroshi Yamamoto
- *Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Science, Kanazawa 920-8640, Japan
| |
Collapse
|
737
|
Monteiro FA, Cardoso I, Sousa MM, Saraiva MJ. In vitro inhibition of transthyretin aggregate-induced cytotoxicity by full and peptide derived forms of the soluble receptor for advanced glycation end products (RAGE). FEBS Lett 2006; 580:3451-6. [PMID: 16716307 DOI: 10.1016/j.febslet.2006.05.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2006] [Revised: 04/28/2006] [Accepted: 05/08/2006] [Indexed: 01/01/2023]
Abstract
Familial amyloidotic polyneuropathy is a neurodegenerative disorder characterized by systemic extracellular deposition of transthyretin (TTR) amyloid fibrils. The latter have been proposed to trigger neurodegeneration through engagement of the receptor for advanced glycation end products (RAGE). Here we show that TTR interaction with RAGE is conserved across mouse and human species and is not dependent on RAGE glycosylation. Moreover, strand D of TTR structure seems important for the TTR-RAGE interaction as well as a motif in RAGE (residues 102-118) located within the V-domain; this motif suppressed TTR aggregate-induced cytotoxicity in cell culture.
Collapse
|
738
|
Wilton R, Yousef MA, Saxena P, Szpunar M, Stevens FJ. Expression and purification of recombinant human receptor for advanced glycation endproducts in Escherichia coli. Protein Expr Purif 2006; 47:25-35. [PMID: 16510295 DOI: 10.1016/j.pep.2006.01.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2005] [Revised: 01/10/2006] [Accepted: 01/11/2006] [Indexed: 01/21/2023]
Abstract
The receptor for advanced glycation endproducts (RAGE) is a multiligand receptor that binds a variety of structurally and functionally unrelated ligands, including advanced glycation endproducts (AGEs), amyloid fibrils, amphoterin, and members of the S100 family of proteins. The receptor has been implicated in the pathology of diabetes as well as in inflammatory processes and tumor cell metastasis. For the present study, the extracellular region of RAGE (exRAGE) was expressed as a soluble, C-terminal hexahistidine-tagged fusion protein in the periplasmic space of Escherichia coli. Proper processing and folding of the purified protein, predicted to contain three immunoglobulin-type domains, was supported by the results of electrospray mass spectroscopy and circular dichroism experiments. Sedimentation velocity experiments showed that exRAGE was primarily monomeric in solution. Binding to several RAGE ligands, including AGE-BSA, immunoglobulin light chain amyloid fibrils, and glycosaminoglycans, was demonstrated using pull-down, dot-blot, or enzyme-linked microplate assays. Using surface plasmon resonance, the interaction of exRAGE with AGE-BSA was shown to fit a two-site model, with KD values of 88 nM and 1.4 microM. The E. coli-derived exRAGE did not bind the advanced glycation endproduct Nepsilon-(carboxymethyl)lysine, as reported for the cellular receptor, and the possible role of RAGE glycosylation in recognition of this ligand is discussed. This new RAGE construct will facilitate detailed studies of RAGE-ligand interactions and provides a platform for preparation of site-directed mutants for future structure/function studies.
Collapse
MESH Headings
- Amyloid/genetics
- Amyloid/metabolism
- Amyloidosis/metabolism
- Cloning, Molecular
- Escherichia coli/genetics
- Escherichia coli/metabolism
- Extracellular Space/chemistry
- Extracellular Space/genetics
- Extracellular Space/metabolism
- Glycation End Products, Advanced/biosynthesis
- Glycation End Products, Advanced/genetics
- Glycation End Products, Advanced/metabolism
- Humans
- Immunoglobulin Variable Region/genetics
- Ligands
- Models, Chemical
- Mutagenesis, Site-Directed
- Peptide Fragments/biosynthesis
- Peptide Fragments/chemistry
- Peptide Fragments/genetics
- Peptide Fragments/metabolism
- Protein Binding/genetics
- Protein Structure, Tertiary/genetics
- Receptor for Advanced Glycation End Products
- Receptors, Immunologic/biosynthesis
- Receptors, Immunologic/genetics
- Receptors, Immunologic/isolation & purification
Collapse
Affiliation(s)
- Rosemarie Wilton
- Biosciences Division, Argonne National Laboratory, Argonne, IL 60439, and Department of Biochemistry and Molecular Biology and Biophysics Core Facility, University of Chicago 60637, USA.
| | | | | | | | | |
Collapse
|
739
|
Nakano N, Fukuhara-Takaki K, Jono T, Nakajou K, Eto N, Horiuchi S, Takeya M, Nagai R. Association of Advanced Glycation End Products with A549 Cells, a Human Pulmonary Epithelial Cell Line, Is Mediated by a Receptor Distinct from the Scavenger Receptor Family and RAGE. ACTA ACUST UNITED AC 2006; 139:821-9. [PMID: 16751589 DOI: 10.1093/jb/mvj092] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Cellular interactions with advanced glycation end products (AGE)-modified proteins are known to induce several biological responses, not only endocytic uptake and degradation, but also the induction of cytokines and growth factors, combined responses that may be linked to the development of diabetic vascular complications. In this study we demonstrate that A549 cells, a human pulmonary epithelial cell line, possess a specific binding site for AGE-modified bovine serum albumin (AGE-BSA) (K(d) = 27.8 nM), and additionally for EN-RAGE (extracellular newly identified RAGE binding protein) (K(d) = 118 nM). Western blot and RT-PCR analysis showed that RAGE (receptor for AGE) is highly expressed on A549 cells, while the expression of other known AGE-receptors such as galectin-3 and SR-A (class A scavenger receptor), are below the level of detection. The binding of (125)I-AGE-BSA to these cells is inhibited by unlabeled AGE-BSA, but not by EN-RAGE. In contrast, the binding of (125)I-EN-RAGE is significantly inhibited by unlabeled EN-RAGE and soluble RAGE, but not by AGE-BSA. Our results indicate that A549 cells possess at least two binding sites, one specific for EN-RAGE and the other specific for AGE-BSA. The latter receptor on A549 cells is distinct from the scavenger receptor family and RAGE.
Collapse
MESH Headings
- Animals
- Cattle
- Cells, Cultured
- Epithelial Cells/metabolism
- Glycation End Products, Advanced/metabolism
- Glycation End Products, Advanced/pharmacokinetics
- Humans
- Lung/metabolism
- Protein Binding
- Receptor for Advanced Glycation End Products
- Receptors, Immunologic/agonists
- Receptors, Immunologic/classification
- Receptors, Scavenger/agonists
- Receptors, Scavenger/classification
- Receptors, Scavenger/metabolism
- Recombinant Proteins/metabolism
- Serum Albumin, Bovine/metabolism
- Serum Albumin, Bovine/pharmacokinetics
- Signal Transduction
- Substrate Specificity
Collapse
Affiliation(s)
- Nahoko Nakano
- Departments of Medical Biochemistry, Psychiatry, and Cell Pathology, Graduate School of Medical and Pharmaceutical Sciences, Kumamoto University, Honjo 1-1-1, Kumamoto 860-8556
| | | | | | | | | | | | | | | |
Collapse
|
740
|
Kiryushko D, Novitskaya V, Soroka V, Klingelhofer J, Lukanidin E, Berezin V, Bock E. Molecular mechanisms of Ca(2+) signaling in neurons induced by the S100A4 protein. Mol Cell Biol 2006; 26:3625-38. [PMID: 16612001 PMCID: PMC1447425 DOI: 10.1128/mcb.26.9.3625-3638.2006] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2005] [Revised: 10/13/2005] [Accepted: 02/08/2006] [Indexed: 12/14/2022] Open
Abstract
The S100A4 protein belongs to the S100 family of vertebrate-specific proteins possessing both intra- and extracellular functions. In the nervous system, high levels of S100A4 expression are observed at sites of neurogenesis and lesions, suggesting a role of the protein in neuronal plasticity. Extracellular oligomeric S100A4 is a potent promoter of neurite outgrowth and survival from cultured primary neurons; however, the molecular mechanism of this effect has not been established. Here we demonstrate that oligomeric S100A4 increases the intracellular calcium concentration in primary neurons. We present evidence that both S100A4-induced Ca(2+) signaling and neurite extension require activation of a cascade including a heterotrimeric G protein(s), phosphoinositide-specific phospholipase C, and diacylglycerol-lipase, resulting in Ca(2+) entry via nonselective cation channels and via T- and L-type voltage-gated Ca(2+) channels. We demonstrate that S100A4-induced neurite outgrowth is not mediated by the receptor for advanced glycation end products, a known target for other extracellular S100 proteins. However, S100A4-induced signaling depends on interactions with heparan sulfate proteoglycans at the cell surface. Thus, glycosaminoglycans may act as coreceptors of S100 proteins in neurons. This may provide a mechanism by which S100 proteins could locally regulate neuronal plasticity in connection with brain lesions and neurological disorders.
Collapse
Affiliation(s)
- Darya Kiryushko
- Protein Laboratory, Panum Institute, Blegdamsvej 3C, Bld. 6.2, Copenhagen 2200N, Denmark.
| | | | | | | | | | | | | |
Collapse
|
741
|
Leach ST, Day AS. S100 proteins in the pathogenesis and diagnosis of inflammatory bowel disease. Expert Rev Clin Immunol 2006; 2:471-480. [DOI: 10.1586/1744666x.2.3.471] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
|
742
|
Ramasamy R, Yan SF, Schmidt AM. The RAGE axis and endothelial dysfunction: maladaptive roles in the diabetic vasculature and beyond. Trends Cardiovasc Med 2006; 15:237-43. [PMID: 16226677 DOI: 10.1016/j.tcm.2005.08.003] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2005] [Revised: 08/01/2005] [Accepted: 08/04/2005] [Indexed: 12/20/2022]
Abstract
Receptor for advanced glycation end product (RAGE) is a member of the immunoglobulin superfamily of cell surface molecules. The ligand-RAGE axis is emerging as a central mechanism linked to vascular injury and atherosclerosis in diabetes and in euglycemia. The repertoire of RAGE ligands, including advanced glycation end products, S100/calgranulins, high-mobility group box 1, amyloid-beta peptide, and Mac-1, transcends RAGE biology from specifically the science of diabetic complications to central aspects of the inflammatory response and oxidative stress. Experiments in cell culture and in vivo support the notion that interaction of RAGE ligands with RAGE activates key signal transduction pathways that modulate fundamental cellular properties, thereby leading to vascular and inflammatory cell perturbation. These considerations support the premise that the ligand-RAGE axis may be an important target for therapeutic intervention in cardiovascular disease and, fundamentally, in initiation and amplification of inflammatory responses.
Collapse
Affiliation(s)
- Ravichandran Ramasamy
- Division of Surgical Science, Department of Surgery, Columbia University Medical Center, 630 West 168th Street, New York, NY 10032, USA
| | | | | |
Collapse
|
743
|
Schmitt HP. ε-Glycation, APP and Aβ in ageing and Alzheimer disease: A hypothesis. Med Hypotheses 2006; 66:898-906. [PMID: 16442744 DOI: 10.1016/j.mehy.2005.11.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2005] [Accepted: 11/17/2005] [Indexed: 01/30/2023]
Abstract
The post-translational modifications of protein molecules include glycation, which may not only occur enzymatically controlled in N and O position, but also wherever proteins meet reducing sugars non-enzymatically in epsilon position at lysines (non-enzymatic (epsilon) glycation (NEG)). The formation of keto-amines from the amine-sugar compounds (Amadori re-arrangement) and further processing of the largely undigestible Amadori compounds eventually results in insoluble advanced glycation end products (AGEs). The latter can induce or favour disease including mental disorders. Preferential targets of NEG include large cell surface proteins. Ample evidence has been provided that NEG also occurs in the brain where cross-linking of epsilon-glycated proteins, induction of oxidative stress and signalling of AGEs through their specific receptor (RAGE) likely play a role in (brain) ageing and Alzheimer disease (AD). This is underscored by the demonstration of particular interactions between AGE/RAGE and amyloid-beta (Abeta) that favour the aggregation and deposition of Abeta and, perhaps, the formation of Abeta itself. The close relationship between NEG and Abeta, as well as other facts foster the hypothesis that NEG of the large trans-membrane amyloid precursor protein (APP) might be a significant factor in the induction of aberrant APP cleavage with production of Abeta, not only in normal ageing, but also in AD. Blockade of lysine cleavage sites on APP by sugar chains or marker effects induced by NEG akin to ubiquitination of proteins for degradation at lysines could be expected to contribute to altered processing of APP. The hypothesis of epsilon-glycation in APP proposed here and the review of evidences for the significance of NEG in brain ageing and AD are aimed at the stimulation of investigations into the still open question which role NEG plays with respect to APP and its abnormal processing in AD. It can be rendered likely that such research might open new avenues towards decreasing the risk of AD and/or slowing its progression through the prevention of NEG in APP with aberrant APP processing, increased generation of Abeta and the formation of AGEs from epsilon-glycated APP.
Collapse
Affiliation(s)
- H Peter Schmitt
- Institute of Pathology, Department for Neuropathology, University of Heidelberg, Germany.
| |
Collapse
|
744
|
Devi Ramnath R, Weing S, He M, Sun J, Zhang H, Singh Bawa M, Bhatia M. Inflammatory mediators in sepsis: Cytokines, chemokines, adhesion molecules and gases. JOURNAL OF ORGAN DYSFUNCTION 2006; 2:80-92. [DOI: 10.1080/17471060500435662] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
745
|
Demling N, Ehrhardt C, Kasper M, Laue M, Knels L, Rieber EP. Promotion of cell adherence and spreading: a novel function of RAGE, the highly selective differentiation marker of human alveolar epithelial type I cells. Cell Tissue Res 2005; 323:475-88. [PMID: 16315007 DOI: 10.1007/s00441-005-0069-0] [Citation(s) in RCA: 185] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2005] [Accepted: 07/27/2005] [Indexed: 12/19/2022]
Abstract
The receptor for advanced glycation endproducts (RAGE) is expressed under pathological conditions in many tissues and has been assigned many functions. We demonstrate, in normal human lung tissue, the preferential and highly abundant expression of RAGE by quantitative polymerase chain reaction. In addition, RAGE expression, as a specific differentiation marker of alveolar epithelial type I cells (AT I cells), and its localization to the basolateral plasma membrane have been confirmed by means of newly raised monoclonal antibodies. The physiological function of RAGE on AT I cells has previously remained elusive. By using HEK293 cells transfected with cDNA encoding for full-length RAGE, we show that RAGE enhances the adherence of epithelial cells to collagen-coated surfaces and has a striking capacity for inducing cell spreading. The preferential binding of RAGE to collagen has been confirmed by assaying the binding of soluble RAGE to various substrates. RAGE might thus assist AT I cells to acquire a spreading morphology, thereby ensuring effective gas exchange and alveolar stability.
Collapse
Affiliation(s)
- Nina Demling
- Institute of Immunology, Medical Faculty, Technical University of Dresden, Fetscherstrasse 42, 01307, Dresden, Germany
| | | | | | | | | | | |
Collapse
|
746
|
Hermani A, Hess J, De Servi B, Medunjanin S, Grobholz R, Trojan L, Angel P, Mayer D. Calcium-binding proteins S100A8 and S100A9 as novel diagnostic markers in human prostate cancer. Clin Cancer Res 2005; 11:5146-52. [PMID: 16033829 DOI: 10.1158/1078-0432.ccr-05-0352] [Citation(s) in RCA: 200] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE S100 proteins comprise a family of calcium-modulated proteins that have recently been associated with epithelial tumors. We examined the expression of two members of this family, S100A8 and S100A9, together with the S100 receptor RAGE (receptor for advanced glycation end products) in human prostate adenocarcinomas and in prostatic intraepithelial neoplasia. EXPERIMENTAL DESIGN Tissue specimens of 75 patients with organ-confined prostate cancer of different grades were analyzed by immunohistochemistry for expression of S100A8, S100A9, and RAGE. In addition, in situ hybridization of S100A8 and S100A9 was done for 20 cases. An ELISA was applied to determine serum concentrations of S100A9 in cancer patients compared with healthy controls or to patients with benign prostatic hyperplasia (BPH). RESULTS S100A8, S100A9, and RAGE were up-regulated in prostatic intraepithelial neoplasia and preferentially in high-grade adenocarcinomas, whereas benign tissue was negative or showed weak expression of the proteins. There was a high degree of overlap of S100A8 and S100A9 expression patterns and of S100A8 or S100A9 and RAGE, respectively. Frequently, a gradient within the tumor tissue with an increased expression toward the invaded stroma of the prostate was observed. S100A9 serum levels were significantly elevated in cancer patients compared with BPH patients or healthy individuals. CONCLUSION Our data suggest that enhanced expression of S100A8, S100A9, and RAGE is an early event in prostate tumorigenesis and may contribute to development and progression or extension of prostate carcinomas. Furthermore, S100A9 in serum may serve as useful marker to discriminate between prostate cancer and BPH.
Collapse
Affiliation(s)
- Alexander Hermani
- Research Group Hormones and Signal Transduction, German Cancer Research Center, Heidelberg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
747
|
Park JS, Gamboni-Robertson F, He Q, Svetkauskaite D, Kim JY, Strassheim D, Sohn JW, Yamada S, Maruyama I, Banerjee A, Ishizaka A, Abraham E. High mobility group box 1 protein interacts with multiple Toll-like receptors. Am J Physiol Cell Physiol 2005; 290:C917-24. [PMID: 16267105 DOI: 10.1152/ajpcell.00401.2005] [Citation(s) in RCA: 739] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
High mobility group box 1 (HMGB1), originally described as a DNA-binding protein, can also be released extracellularly and functions as a late mediator of inflammatory responses. Although recent reports have indicated that the receptor for advanced glycation end products (RAGE) as well as Toll-like receptor (TLR)2 and TLR4 are involved in cellular activation by HMGB1, there has been little evidence of direct association between HMGB1 and these receptors. To examine this issue, we used fluorescence resonance energy transfer (FRET) and immunoprecipitation to directly investigate cell surface interactions of HMGB1 with TLR2, TLR4, and RAGE. FRET images in RAW264.7 macrophages demonstrated association of HMGB1 with TLR2 and TLR4 but not RAGE. Transient transfections into human embryonic kidney-293 cells showed that HMGB1 induced cellular activation and NF-kappaB-dependent transcription through TLR2 or TLR4 but not RAGE. Coimmunoprecipitation also found interaction between HMGB1 and TLR2 as well as TLR4, but not with RAGE. These studies provide the first direct evidence that HMGB1 can interact with both TLR2 and TLR4 and also supply an explanation for the ability of HMGB1 to induce cellular activation and generate inflammatory responses that are similar to those initiated by LPS.
Collapse
Affiliation(s)
- Jong Sung Park
- Division of Pulmonary Sciences and Critical Care Medicine, Box C272, University of Colorado Health Sciences Center, 4200 E. Ninth Ave., Denver, CO 80262, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
748
|
Hudson BI, Wendt T, Bucciarelli LG, Rong LL, Naka Y, Yan SF, Schmidt AM. Diabetic vascular disease: it's all the RAGE. Antioxid Redox Signal 2005; 7:1588-600. [PMID: 16356122 DOI: 10.1089/ars.2005.7.1588] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The major consequence of long-term diabetes is the increased incidence of disease of the vasculature. Of the underlying mechanisms leading to disease, the accumulation of advanced glycation end products (AGEs), resulting from the associated hyperglycemia, is the most convincing. Interaction of AGEs with their receptor, RAGE, activates numerous signaling pathways leading to activation of proinflammatory and procoagulatory genes. Studies in rodent models of macro- and microvascular disease have demonstrated that blockade of RAGE can prevent development of disease. These observations highlight RAGE as a therapeutic target for treatment of diabetic vascular disease.
Collapse
Affiliation(s)
- Barry I Hudson
- Department of Surgery, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA.
| | | | | | | | | | | | | |
Collapse
|
749
|
Kim W, Hudson BI, Moser B, Guo J, Rong LL, Lu Y, Qu W, Lalla E, Lerner S, Chen Y, Yan SSD, D'Agati V, Naka Y, Ramasamy R, Herold K, Yan SF, Schmidt AM. Receptor for advanced glycation end products and its ligands: a journey from the complications of diabetes to its pathogenesis. Ann N Y Acad Sci 2005; 1043:553-61. [PMID: 16037278 DOI: 10.1196/annals.1338.063] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Many studies have suggested that the expression of RAGE (receptor for advanced glycation end products) is upregulated in human tissues susceptible to the long-term complications of diabetes. From the kidneys to the macrovessels of the aorta, RAGE expression is upregulated in a diverse array of cell types, from glomerular epithelial cells (podocytes) to endothelial cells, vascular smooth muscle cells, and inflammatory mononuclear phagocytes and lymphocytes. Although RAGE was first described as a receptor for advanced glycation end products (AGEs), the key finding that RAGE was also a signaling receptor for proinflammatory S100/calgranulins and amphoterin, led to the premise that even in euglycemia, ligand-RAGE interaction propagated inflammatory mechanisms linked to chronic cellular perturbation and tissue injury. Indeed, such considerations suggested that RAGE might even participate in the pathogenesis of type 1 diabetes. Our studies have shown that pharmacological and/or genetic deletion/mutation of the receptor attenuates the development of hyperglycemia in NOD mice; in mice with myriad complications of diabetes, interruption of ligand-RAGE interaction prevents or delays the chronic complications of the disease in both macro- and microvessel structures. Taken together, these findings suggest that RAGE is "at the right place and time" to contribute to the pathogenesis of diabetes and it complications. Studies are in progress to test the premise that antagonism of this interaction is a logical strategy for the prevention and treatment of diabetes.
Collapse
Affiliation(s)
- William Kim
- Division of Surgical Science, Department of Surgery, Columbia University Medical Center, 630 West 168th St., P&S 17-501, New York, New York 10032, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
750
|
Zamora R, Grishin A, Wong C, Boyle P, Wang J, Hackam D, Upperman JS, Tracey KJ, Ford HR. High-mobility group box 1 protein is an inflammatory mediator in necrotizing enterocolitis: protective effect of the macrophage deactivator semapimod. Am J Physiol Gastrointest Liver Physiol 2005; 289:G643-52. [PMID: 15947118 DOI: 10.1152/ajpgi.00067.2005] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
High-mobility group box 1 (HMGB1) is a late mediator of endotoxemia known to stimulate the production of proinflammatory cytokines that are putative mediators of intestinal inflammation associated with necrotizing enterocolitis (NEC). We hypothesized that HMGB1 is also involved in the pathogenesis of NEC. We examined the expression of HMGB1 and the effect of the novel drug semapimod on intestinal inflammation in an experimental model of NEC in neonatal rats. Newborn rats were subjected to hypoxia and fed a conventional formula by gavage (FFH) or were breast fed (BF). Rats were killed on day 4, and the distal ileum was harvested for morphological studies and Western blot analysis. FFH newborn rats but not BF controls developed intestinal inflammation similar to the histological changes observed in human NEC. We found that the expression of HMGB1 and its receptor for advanced glycation end products (RAGE) as well as that of other apoptosis/inflammation-related proteins (Bad, Bax, inducible nitric oxide synthase, and cyclooxygenase 2) was upregulated in the ileal mucosa of FFH newborn rats compared with BF animals. Administration of the drug semapimod inhibited the upregulation of those proteins and partially protected the animals against the FFH-induced intestinal injury. Elevated levels of HMGB1 were also found in ileal samples from infants undergoing intestinal resection for acute NEC. Our results implicate HMGB1 and RAGE as important mediators of enterocyte cell death and hypoxia-induced injury in NEC and support the hypothesis that inhibitors such as semapimod might play a therapeutic role in chronic intestinal inflammation characterized by this animal model.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Anti-Inflammatory Agents, Non-Steroidal/pharmacology
- Apoptosis/drug effects
- Blotting, Western
- Cell Death/drug effects
- Cells, Cultured
- Cytokines/antagonists & inhibitors
- Enterocolitis, Necrotizing/mortality
- Enterocolitis, Necrotizing/pathology
- Enterocolitis, Necrotizing/prevention & control
- Female
- Genes, bcl-2
- Glycation End Products, Advanced/metabolism
- HMGB1 Protein/biosynthesis
- HMGB1 Protein/physiology
- Hydrazones/pharmacology
- Inflammation Mediators/physiology
- Intestines/pathology
- Lipopolysaccharides/pharmacology
- Macrophages/drug effects
- Nitric Oxide Synthase/biosynthesis
- Nitric Oxide Synthase/genetics
- Nitric Oxide Synthase Type II
- Pregnancy
- Rats
- Rats, Sprague-Dawley
- p38 Mitogen-Activated Protein Kinases/metabolism
Collapse
Affiliation(s)
- Ruben Zamora
- Dept. of Pediatric Surgery, Children's Hospital of Pittsburgh, PA 15213, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|