1
|
Li W, Chen Q, Peng C, Yang D, Liu S, Lv Y, Jiang L, Xu S, Huang L. Roles of the Receptor for Advanced Glycation End Products and Its Ligands in the Pathogenesis of Alzheimer's Disease. Int J Mol Sci 2025; 26:403. [PMID: 39796257 PMCID: PMC11721675 DOI: 10.3390/ijms26010403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/02/2025] [Accepted: 01/04/2025] [Indexed: 01/13/2025] Open
Abstract
The Receptor for Advanced Glycation End Products (RAGE), part of the immunoglobulin superfamily, plays a significant role in various essential functions under both normal and pathological conditions, especially in the progression of Alzheimer's disease (AD). RAGE engages with several damage-associated molecular patterns (DAMPs), including advanced glycation end products (AGEs), beta-amyloid peptide (Aβ), high mobility group box 1 (HMGB1), and S100 calcium-binding proteins. This interaction impairs the brain's ability to clear Aβ, resulting in increased Aβ accumulation, neuronal injury, and mitochondrial dysfunction. This further promotes inflammatory responses and oxidative stress, ultimately leading to a range of age-related diseases. Given RAGE's significant role in AD, inhibitors that target RAGE and its ligands hold promise as new strategies for treating AD, offering new possibilities for alleviating and treating this serious neurodegenerative disease. This article reviews the various pathogenic mechanisms of AD and summarizes the literature on the interaction between RAGE and its ligands in various AD-related pathological processes, with a particular focus on the evidence and mechanisms by which RAGE interactions with AGEs, HMGB1, Aβ, and S100 proteins induce cognitive impairment in AD. Furthermore, the article discusses the principles of action of RAGE inhibitors and inhibitors targeting RAGE-ligand interactions, along with relevant clinical trials.
Collapse
Affiliation(s)
- Wen Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (W.L.); (Q.C.); (C.P.); (D.Y.); (S.L.); (Y.L.); (L.J.)
| | - Qiuping Chen
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (W.L.); (Q.C.); (C.P.); (D.Y.); (S.L.); (Y.L.); (L.J.)
| | - Chengjie Peng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (W.L.); (Q.C.); (C.P.); (D.Y.); (S.L.); (Y.L.); (L.J.)
| | - Dan Yang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (W.L.); (Q.C.); (C.P.); (D.Y.); (S.L.); (Y.L.); (L.J.)
| | - Si Liu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (W.L.); (Q.C.); (C.P.); (D.Y.); (S.L.); (Y.L.); (L.J.)
| | - Yanwen Lv
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (W.L.); (Q.C.); (C.P.); (D.Y.); (S.L.); (Y.L.); (L.J.)
| | - Langqi Jiang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (W.L.); (Q.C.); (C.P.); (D.Y.); (S.L.); (Y.L.); (L.J.)
| | - Shijun Xu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (W.L.); (Q.C.); (C.P.); (D.Y.); (S.L.); (Y.L.); (L.J.)
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu 611137, China
- Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Lihua Huang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (W.L.); (Q.C.); (C.P.); (D.Y.); (S.L.); (Y.L.); (L.J.)
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu 611137, China
| |
Collapse
|
2
|
RAGE Inhibitors for Targeted Therapy of Cancer: A Comprehensive Review. Int J Mol Sci 2022; 24:ijms24010266. [PMID: 36613714 PMCID: PMC9820344 DOI: 10.3390/ijms24010266] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 10/28/2022] [Accepted: 12/16/2022] [Indexed: 12/28/2022] Open
Abstract
The receptor for advanced glycation end products (RAGE) is a member of the immunoglobulin family that is overexpressed in several cancers. RAGE is highly expressed in the lung, and its expression increases proportionally at the site of inflammation. This receptor can bind a variety of ligands, including advanced glycation end products, high mobility group box 1, S100 proteins, adhesion molecules, complement components, advanced lipoxidation end products, lipopolysaccharides, and other molecules that mediate cellular responses related to acute and chronic inflammation. RAGE serves as an important node for the initiation and stimulation of cell stress and growth signaling mechanisms that promote carcinogenesis, tumor propagation, and metastatic potential. In this review, we discuss different aspects of RAGE and its prominent ligands implicated in cancer pathogenesis and describe current findings that provide insights into the significant role played by RAGE in cancer. Cancer development can be hindered by inhibiting the interaction of RAGE with its ligands, and this could provide an effective strategy for cancer treatment.
Collapse
|
3
|
Increased cerebrospinal fluid S100B protein levels in patients with trigeminal neuralgia and hemifacial spasm. Acta Neurochir (Wien) 2022; 165:959-965. [PMID: 36459237 DOI: 10.1007/s00701-022-05434-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 11/19/2022] [Indexed: 12/04/2022]
Abstract
BACKGROUND The pathophysiology of neurovascular compression syndrome has not been fully elucidated, and cerebrospinal fluid levels of nerve tissue-related markers involved in this disorder have not yet been reported. METHODS We measured cerebrospinal fluid levels of S100B protein, neuron-specific enolase, and myelin basic protein in 21 patients with trigeminal neuralgia, 9 patients with hemifacial spasms, and 10 patients with non-ruptured intracranial aneurysms (control). Cerebrospinal fluid levels of these markers were determined using commercially available assay kits. RESULTS Both trigeminal neuralgia and hemifacial spasm groups showed significantly increased cerebrospinal fluid levels of S100B compared with the control group (1120 [IQR 391-1420], 766 [IQR 583-1500], and 255 [IQR 190-285] pg/mL, respectively; p = 0.001). There were no statistically significant differences in cerebrospinal fluid levels of neuron-specific enolase or myelin basic protein among the groups. CONCLUSION Cerebrospinal fluid S100B levels were significantly higher in patients with trigeminal neuralgia and hemifacial spasm than in controls, which suggests the involvement of S100B in the underlying pathophysiology of neurovascular compression syndrome.
Collapse
|
4
|
Daneshvar N, Anderson JE. Preliminary Study of S100B and Sema3A Expression Patterns in Regenerating Muscle Implicates P75-Expressing Terminal Schwann Cells and Muscle Satellite Cells in Neuromuscular Junction Restoration. Front Cell Dev Biol 2022; 10:874756. [PMID: 35923848 PMCID: PMC9340223 DOI: 10.3389/fcell.2022.874756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 05/31/2022] [Indexed: 12/02/2022] Open
Abstract
Terminal Schwann cells (TSCs) help regulate the formation, maintenance, function, and repair of neuromuscular junctions (NMJs) and axon guidance after muscle injury. Premature activation of muscle satellite cells (SCs), induced by isosorbide dinitrate (ISDN) before injury, accelerates myogenic regeneration, disrupts NMJ remodeling and maturation, decreases Sema3A protein-induced neuro-repulsion, and is accompanied by time-dependent changes in S100B protein levels. Here, to study the effects of premature SC activation on TSCs and SCs, both expressing P75 nerve growth-factor receptor, in situ hybridization was used to identify transcripts of S100B and Sema3A, and the number, intensity, and diameter of expression sites were analyzed. The number of sites/fields expressing S100B and Sema3A increased with regeneration time (both p < 0.001). Expression-site intensity (S100B) and diameter (S100B and Sema3A) decreased during regeneration (p = 0.005; p < 0.05, p = 0.006, respectively). P75 protein colocalized with a subset of S100B and Sema3A expression sites. Principal component analyses of gene expression, protein levels, and histological variables (fiber diameter, vascular density) in control and ISDN-pretreated groups explained 83% and 64% of the dataset variance, respectively. A very strong loading coefficient for colocalization of P75 protein with S100B and Sema3A mRNAs (0.91) in control regenerating muscle dropped markedly during regeneration disrupted by premature SC activation (-0.10 in Factor 1 to 0.55 in Factor 3). These findings strongly implicate the triple-expression profile by TSCs and/or SCs as a strong correlate of the important synchrony of muscle and nerve regeneration after muscle tissue injury. The results have the potential to focus future research on the complex interplay of TSCs and SCs in neuromuscular tissue repair and help promote effective function after traumatic muscle injury.
Collapse
Affiliation(s)
| | - Judy E. Anderson
- Department of Biological Sciences, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
5
|
Zaręba-Kozioł M, Burdukiewicz M, Wysłouch-Cieszyńska A. Intracellular Protein S-Nitrosylation—A Cells Response to Extracellular S100B and RAGE Receptor. Biomolecules 2022; 12:biom12050613. [PMID: 35625541 PMCID: PMC9138530 DOI: 10.3390/biom12050613] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 02/18/2022] [Accepted: 03/09/2022] [Indexed: 02/04/2023] Open
Abstract
Human S100B is a small, multifunctional protein. Its activity, inside and outside cells, contributes to the biology of the brain, muscle, skin, and adipocyte tissues. Overexpression of S100B occurs in Down Syndrome, Alzheimer’s disease, Creutzfeldt–Jakob disease, schizophrenia, multiple sclerosis, brain tumors, epilepsy, melanoma, myocardial infarction, muscle disorders, and sarcopenia. Modulating the activities of S100B, related to human diseases, without disturbing its physiological functions, is vital for drug and therapy design. This work focuses on the extracellular activity of S100B and one of its receptors, the Receptor for Advanced Glycation End products (RAGE). The functional outcome of extracellular S100B, partially, depends on the activation of intracellular signaling pathways. Here, we used Biotin Switch Technique enrichment and mass-spectrometry-based proteomics to show that the appearance of the S100B protein in the extracellular milieu of the mammalian Chinese Hamster Ovary (CHO) cells, and expression of the membrane-bound RAGE receptor, lead to changes in the intracellular S-nitrosylation of, at least, more than a hundred proteins. Treatment of the wild-type CHO cells with nanomolar or micromolar concentrations of extracellular S100B modulates the sets of S-nitrosylation targets inside cells. The cellular S-nitrosome is tuned differently, depending on the presence or absence of stable RAGE receptor expression. The presented results are a proof-of-concept study, suggesting that S-nitrosylation, like other post-translational modifications, should be considered in future research, and in developing tailored therapies for S100B and RAGE receptor-related diseases.
Collapse
Affiliation(s)
- Monika Zaręba-Kozioł
- Mass Spectrometry Laboratory, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawińskiego 5a, 02-106 Warsaw, Poland;
- Laboratory of Cell Biophysics, Nencki Institute of Experimental Biology, Polish Academy of Science, Pasteura 3, 02-093 Warsaw, Poland
| | - Michał Burdukiewicz
- Clinical Research Centre, Medical University of Białystok, Kilińskiego 1, 15-369 Białystok, Poland;
| | - Aleksandra Wysłouch-Cieszyńska
- Mass Spectrometry Laboratory, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawińskiego 5a, 02-106 Warsaw, Poland;
- Correspondence:
| |
Collapse
|
6
|
Larouche JA, Mohiuddin M, Choi JJ, Ulintz PJ, Fraczek P, Sabin K, Pitchiaya S, Kurpiers SJ, Castor-Macias J, Liu W, Hastings RL, Brown LA, Markworth JF, De Silva K, Levi B, Merajver SD, Valdez G, Chakkalakal JV, Jang YC, Brooks SV, Aguilar CA. Murine muscle stem cell response to perturbations of the neuromuscular junction are attenuated with aging. eLife 2021; 10:e66749. [PMID: 34323217 PMCID: PMC8360658 DOI: 10.7554/elife.66749] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 07/28/2021] [Indexed: 01/29/2023] Open
Abstract
During aging and neuromuscular diseases, there is a progressive loss of skeletal muscle volume and function impacting mobility and quality of life. Muscle loss is often associated with denervation and a loss of resident muscle stem cells (satellite cells or MuSCs); however, the relationship between MuSCs and innervation has not been established. Herein, we administered severe neuromuscular trauma to a transgenic murine model that permits MuSC lineage tracing. We show that a subset of MuSCs specifically engraft in a position proximal to the neuromuscular junction (NMJ), the synapse between myofibers and motor neurons, in healthy young adult muscles. In aging and in a mouse model of neuromuscular degeneration (Cu/Zn superoxide dismutase knockout - Sod1-/-), this localized engraftment behavior was reduced. Genetic rescue of motor neurons in Sod1-/- mice reestablished integrity of the NMJ in a manner akin to young muscle and partially restored MuSC ability to engraft into positions proximal to the NMJ. Using single cell RNA-sequencing of MuSCs isolated from aged muscle, we demonstrate that a subset of MuSCs are molecularly distinguishable from MuSCs responding to myofiber injury and share similarity to synaptic myonuclei. Collectively, these data reveal unique features of MuSCs that respond to synaptic perturbations caused by aging and other stressors.
Collapse
Affiliation(s)
- Jacqueline A Larouche
- Department of Biomedical Engineering, University of MichiganAnn ArborUnited States
- Biointerfaces Institute, University of MichiganAnn ArborUnited States
| | - Mahir Mohiuddin
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of TechnologyAtlantaUnited States
- School of Biological Sciences, Georgia Institute of TechnologyAtlantaUnited States
- Wallace Coulter Departmentof Biomedical Engineering, Georgia Institute of TechnologyAtlantaUnited States
| | - Jeongmoon J Choi
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of TechnologyAtlantaUnited States
- School of Biological Sciences, Georgia Institute of TechnologyAtlantaUnited States
- Wallace Coulter Departmentof Biomedical Engineering, Georgia Institute of TechnologyAtlantaUnited States
| | - Peter J Ulintz
- Department of Biomedical Engineering, University of MichiganAnn ArborUnited States
- Biointerfaces Institute, University of MichiganAnn ArborUnited States
- Internal Medicine-Hematology/Oncology, University of MichiganAnn ArborUnited States
| | - Paula Fraczek
- Department of Biomedical Engineering, University of MichiganAnn ArborUnited States
- Biointerfaces Institute, University of MichiganAnn ArborUnited States
| | - Kaitlyn Sabin
- Department of Biomedical Engineering, University of MichiganAnn ArborUnited States
- Biointerfaces Institute, University of MichiganAnn ArborUnited States
| | | | - Sarah J Kurpiers
- Department of Biomedical Engineering, University of MichiganAnn ArborUnited States
- Biointerfaces Institute, University of MichiganAnn ArborUnited States
| | - Jesus Castor-Macias
- Department of Biomedical Engineering, University of MichiganAnn ArborUnited States
- Biointerfaces Institute, University of MichiganAnn ArborUnited States
| | - Wenxuan Liu
- Department of Pharmacology and Physiology, University of Rochester Medical CenterRochesterUnited States
- Department of Biomedical Engineering, University of Rochester Medical CenterRochesterUnited States
- Wilmot Cancer Institute, Stem Cell and Regenerative Medicine Institute, and The Rochester Aging Research Center, University of Rochester Medical CenterRochesterUnited States
| | - Robert Louis Hastings
- Departmentof Molecular Biology, Cell Biology and Biochemistry, Brown UniversityProvidenceUnited States
- Center for Translational Neuroscience, Robert J. and Nancy D. Carney Institute for Brain Science and Brown Institute for Translational Science, Brown UniversityProvidenceUnited States
| | - Lemuel A Brown
- Department of Molecular & Integrative Physiology, University of MichiganAnn ArborUnited States
| | - James F Markworth
- Department of Molecular & Integrative Physiology, University of MichiganAnn ArborUnited States
| | - Kanishka De Silva
- Department of Biomedical Engineering, University of MichiganAnn ArborUnited States
- Biointerfaces Institute, University of MichiganAnn ArborUnited States
| | - Benjamin Levi
- Department of Surgery, University of Texas SouthwesternDallasUnited States
- Childrens Research Institute and Center for Mineral MetabolismDallasUnited States
- Program in Cellular and Molecular Biology, University of MichiganAnn ArborUnited States
| | - Sofia D Merajver
- Department of Biomedical Engineering, University of MichiganAnn ArborUnited States
- Internal Medicine-Hematology/Oncology, University of MichiganAnn ArborUnited States
| | - Gregorio Valdez
- Departmentof Molecular Biology, Cell Biology and Biochemistry, Brown UniversityProvidenceUnited States
- Center for Translational Neuroscience, Robert J. and Nancy D. Carney Institute for Brain Science and Brown Institute for Translational Science, Brown UniversityProvidenceUnited States
| | - Joe V Chakkalakal
- Department of Pharmacology and Physiology, University of Rochester Medical CenterRochesterUnited States
- Department of Biomedical Engineering, University of Rochester Medical CenterRochesterUnited States
- Wilmot Cancer Institute, Stem Cell and Regenerative Medicine Institute, and The Rochester Aging Research Center, University of Rochester Medical CenterRochesterUnited States
| | - Young C Jang
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of TechnologyAtlantaUnited States
- School of Biological Sciences, Georgia Institute of TechnologyAtlantaUnited States
- Wallace Coulter Departmentof Biomedical Engineering, Georgia Institute of TechnologyAtlantaUnited States
| | - Susan V Brooks
- Department of Biomedical Engineering, University of MichiganAnn ArborUnited States
- Department of Molecular & Integrative Physiology, University of MichiganAnn ArborUnited States
| | - Carlos A Aguilar
- Department of Biomedical Engineering, University of MichiganAnn ArborUnited States
- Biointerfaces Institute, University of MichiganAnn ArborUnited States
- Childrens Research Institute and Center for Mineral MetabolismDallasUnited States
- Program in Cellular and Molecular Biology, University of MichiganAnn ArborUnited States
| |
Collapse
|
7
|
Angelopoulou E, Paudel YN, Piperi C. Emerging role of S100B protein implication in Parkinson's disease pathogenesis. Cell Mol Life Sci 2021; 78:1445-1453. [PMID: 33052436 PMCID: PMC11073186 DOI: 10.1007/s00018-020-03673-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 09/10/2020] [Accepted: 10/05/2020] [Indexed: 01/08/2023]
Abstract
The exact etiology of Parkinson's disease (PD) remains obscure, lacking effective diagnostic and prognostic biomarkers. In search of novel molecular factors that may contribute to PD pathogenesis, emerging evidence highlights the multifunctional role of the calcium-binding protein S100B that is widely expressed in the brain and predominantly in astrocytes. Preclinical evidence points towards the possible time-specific contributing role of S100B in the pathogenesis of neurodegenerative disorders including PD, mainly by regulating neuroinflammation and dopamine metabolism. Although existing clinical evidence presents some contradictions, estimation of S100B in the serum and cerebrospinal fluid seems to hold a great promise as a potential PD biomarker, particularly regarding the severity of motor and non-motor PD symptoms. Furthermore, given the recent development of S100B inhibitors that are able to cross the blood brain barrier, novel opportunities are arising in the research field of PD therapeutics. In this review, we provide an update on recent advances in the implication of S100B protein in the pathogenesis of PD and discuss relevant studies investigating the biomarker potential of S100B in PD, aiming to shed more light on clinical targeting approaches related to this incurable disorder.
Collapse
Affiliation(s)
- Efthalia Angelopoulou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 M. Asias Street, 11527, Athens, Greece
| | - Yam Nath Paudel
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia
| | - Christina Piperi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 M. Asias Street, 11527, Athens, Greece.
| |
Collapse
|
8
|
Riuzzi F, Sorci G, Arcuri C, Giambanco I, Bellezza I, Minelli A, Donato R. Cellular and molecular mechanisms of sarcopenia: the S100B perspective. J Cachexia Sarcopenia Muscle 2018; 9:1255-1268. [PMID: 30499235 PMCID: PMC6351675 DOI: 10.1002/jcsm.12363] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 09/27/2018] [Indexed: 12/11/2022] Open
Abstract
Primary sarcopenia is a condition of reduced skeletal muscle mass and strength, reduced agility, and increased fatigability and risk of bone fractures characteristic of aged, otherwise healthy people. The pathogenesis of primary sarcopenia is not completely understood. Herein, we review the essentials of the cellular and molecular mechanisms of skeletal mass maintenance; the alterations of myofiber metabolism and deranged properties of muscle satellite cells (the adult stem cells of skeletal muscles) that underpin the pathophysiology of primary sarcopenia; the role of the Ca2+ -sensor protein, S100B, as an intracellular factor and an extracellular signal regulating cell functions; and the functional role of S100B in muscle tissue. Lastly, building on recent results pointing to S100B as to a molecular determinant of myoblast-brown adipocyte transition, we propose S100B as a transducer of the deleterious effects of accumulation of reactive oxygen species in myoblasts and, potentially, myofibers concurring to the pathophysiology of sarcopenia.
Collapse
Affiliation(s)
- Francesca Riuzzi
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy.,Interuniversity Institute of Myology
| | - Guglielmo Sorci
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy.,Interuniversity Institute of Myology
| | - Cataldo Arcuri
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy.,Interuniversity Institute of Myology
| | - Ileana Giambanco
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy.,Interuniversity Institute of Myology
| | - Ilaria Bellezza
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy
| | - Alba Minelli
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy
| | - Rosario Donato
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy.,Interuniversity Institute of Myology.,Centro Universitario di Ricerca sulla Genomica Funzionale, University of Perugia, Perugia, 06132, Italy
| |
Collapse
|
9
|
Padilla L, Dakhel S, Adan J, Masa M, Martinez JM, Roque L, Coll T, Hervas R, Calvis C, Llinas L, Buenestado S, Castellsague J, Messeguer R, Mitjans F, Hernandez JL. S100A7: from mechanism to cancer therapy. Oncogene 2017; 36:6749-6761. [PMID: 28825725 DOI: 10.1038/onc.2017.283] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 06/02/2017] [Accepted: 07/08/2017] [Indexed: 12/21/2022]
Abstract
Within the tumor, malignant and stromal cells support each other by secreting a wide variety of growth factors and cytokines, allowing tumor growth and disease progression. The identification and regulation of those key factors in this crosstalk has opened the opportunity to develop new therapeutic strategies that not only act on the tumor cells but also on the stroma. Among these factors, S100A7 protein has gained interest in the last years. With key roles in cell motility its expression correlates with increased tumor growth, angiogenesis and metastatic potential. This work aims to deepen in the role played by extracellular S100A7 in the tumor microenvironment, offering a new integrative insight of its mechanism of action on each cellular compartment (tumor, endothelial, immune and fibroblast). As a result, we demonstrate its implication in cell migration and invasion, and its important contribution to the formation of a proinflammatory and proangiogenic environment that favors tumor progression and metastasis. Furthermore, we define its possible role in the pre-metastatic niche formation. Considering the relevance of S100A7 in cancer progression, we have developed neutralizing monoclonal antibodies, reporting for the first time the proof of principle of this promising therapeutic strategy for cancer treatment.
Collapse
Affiliation(s)
- L Padilla
- Biomed Division, LEITAT Technological Center, Barcelona, Spain
| | - S Dakhel
- Biomed Division, LEITAT Technological Center, Barcelona, Spain
| | - J Adan
- Biomed Division, LEITAT Technological Center, Barcelona, Spain
| | - M Masa
- Biomed Division, LEITAT Technological Center, Barcelona, Spain
| | - J M Martinez
- Biomed Division, LEITAT Technological Center, Barcelona, Spain
| | - L Roque
- Biomed Division, LEITAT Technological Center, Barcelona, Spain
| | - T Coll
- Biomed Division, LEITAT Technological Center, Barcelona, Spain
| | - R Hervas
- Biomed Division, LEITAT Technological Center, Barcelona, Spain
| | - C Calvis
- Biomed Division, LEITAT Technological Center, Barcelona, Spain
| | - L Llinas
- Biomed Division, LEITAT Technological Center, Barcelona, Spain
| | - S Buenestado
- Biomed Division, LEITAT Technological Center, Barcelona, Spain
| | - J Castellsague
- Biomed Division, LEITAT Technological Center, Barcelona, Spain
| | - R Messeguer
- Biomed Division, LEITAT Technological Center, Barcelona, Spain
| | - F Mitjans
- Biomed Division, LEITAT Technological Center, Barcelona, Spain
| | - J L Hernandez
- Biomed Division, LEITAT Technological Center, Barcelona, Spain
| |
Collapse
|
10
|
Hosokawa K, Hamada Y, Fujiya A, Murase M, Maekawa R, Niwa Y, Izumoto T, Seino Y, Tsunekawa S, Arima H. S100B impairs glycolysis via enhanced poly(ADP-ribosyl)ation of glyceraldehyde-3-phosphate dehydrogenase in rodent muscle cells. Am J Physiol Endocrinol Metab 2017; 312:E471-E481. [PMID: 28174179 DOI: 10.1152/ajpendo.00328.2016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 01/31/2017] [Accepted: 01/31/2017] [Indexed: 02/07/2023]
Abstract
S100 calcium-binding protein B (S100B), a multifunctional macromolecule mainly expressed in nerve tissues and adipocytes, has been suggested to contribute to the pathogenesis of obesity. To clarify the role of S100B in insulin action and glucose metabolism in peripheral tissues, we investigated the effect of S100B on glycolysis in myoblast and myotube cells. Rat myoblast L6 cells were treated with recombinant mouse S100B to examine glucose consumption, lactate production, glycogen accumulation, glycolytic metabolites and enzyme activity, insulin signaling, and poly(ADP-ribosyl)ation of glyceraldehyde-3-phosphate dehydrogenase (GAPDH). Glycolytic metabolites were investigated by enzyme assays or metabolome analysis, and insulin signaling was assessed by Western blot analysis. Enzyme activity and poly(ADP-ribosyl)ation of GAPDH was evaluated by an enzyme assay and immunoprecipitation followed by dot blot with an anti-poly(ADP-ribose) antibody, respectively. S100B significantly decreased glucose consumption, glucose analog uptake, and lactate production in L6 cells, in either the presence or absence of insulin. In contrast, S100B had no effect on glycogen accumulation and insulin signaling. Metabolome analysis revealed that S100B increased the concentration of glycolytic intermediates upstream of GAPDH. S100B impaired GAPDH activity and increased poly(ADP-ribosyl)ated GAPDH proteins. The effects of S100B on glucose metabolism were mostly canceled by a poly(ADP-ribose) polymerase inhibitor. Similar results were obtained in C2C12 myotube cells. We conclude that S100B as a humoral factor may impair glycolysis in muscle cells independent of insulin action, and the effect may be attributed to the inhibition of GAPDH activity from enhanced poly(ADP-ribosyl)ation of the enzyme.
Collapse
MESH Headings
- Animals
- Cell Line
- Cells, Cultured
- Enzyme Induction/drug effects
- Glyceraldehyde-3-Phosphate Dehydrogenases/antagonists & inhibitors
- Glyceraldehyde-3-Phosphate Dehydrogenases/genetics
- Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism
- Glycolysis/drug effects
- Hexokinase/chemistry
- Hexokinase/genetics
- Hexokinase/metabolism
- Insulin/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle Fibers, Skeletal/drug effects
- Muscle Fibers, Skeletal/enzymology
- Muscle Fibers, Skeletal/metabolism
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/enzymology
- Muscle, Skeletal/metabolism
- Myoblasts/drug effects
- Myoblasts/enzymology
- Myoblasts/metabolism
- Poly(ADP-ribose) Polymerase Inhibitors/pharmacology
- Poly(ADP-ribose) Polymerases/chemistry
- Poly(ADP-ribose) Polymerases/metabolism
- Protein Processing, Post-Translational/drug effects
- Rats
- Recombinant Proteins/metabolism
- S100 Calcium Binding Protein beta Subunit/genetics
- S100 Calcium Binding Protein beta Subunit/metabolism
Collapse
Affiliation(s)
- Kaori Hosokawa
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoji Hamada
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan;
| | - Atsushi Fujiya
- Department of Diabetology and Nephrology, Ogaki Municipal Hospital, Ogaki City, Gifu Prefecture, Japan
| | - Masatoshi Murase
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Ryuya Maekawa
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yasuhiro Niwa
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takako Izumoto
- Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan; and
| | - Yusuke Seino
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shin Tsunekawa
- Division of Diabetes, Department of Internal Medicine, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Hiroshi Arima
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
11
|
S100B raises the alert in subarachnoid hemorrhage. Rev Neurosci 2016; 27:745-759. [DOI: 10.1515/revneuro-2016-0021] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 05/26/2016] [Indexed: 12/19/2022]
Abstract
AbstractSubarachnoid hemorrhage (SAH) is a devastating disease with high mortality and mobility, the novel therapeutic strategies of which are essentially required. The calcium binding protein S100B has emerged as a brain injury biomarker that is implicated in pathogenic process of SAH. S100B is mainly expressed in astrocytes of the central nervous system and functions through initiating intracellular signaling or via interacting with cell surface receptor, such as the receptor of advanced glycation end products. The biological roles of S100B in neurons have been closely associated with its concentrations, resulting in either neuroprotection or neurotoxicity. The levels of S100B in the blood have been suggested as a biomarker to predict the progress or the prognosis of SAH. The role of S100B in the development of cerebral vasospasm and brain damage may result from the induction of oxidative stress and neuroinflammation after SAH. To get further insight into mechanisms underlying the role of S100B in SAH based on this review might help us to find novel therapeutic targets for SAH.
Collapse
|
12
|
Park YW, Kim SG, Jo YY. S100 and p65 expression are increased in the masseter muscle after botulinum toxin-A injection. Maxillofac Plast Reconstr Surg 2016; 38:33. [PMID: 27635391 PMCID: PMC5002051 DOI: 10.1186/s40902-016-0079-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 08/22/2016] [Indexed: 11/10/2022] Open
Abstract
Background The purpose of this study was to compare the expression levels of p65 and S100 in the rat masseter muscle after the injection of different concentrations of botulinum toxin-A (BTX-A). Methods We injected either 5 or 10 U of BTX-A into both masseter muscle of rats. As a control group, the same volume of saline was injected. After 14 days, the animals were sacrificed. Subsequently, a biopsy and immunohistochemical staining of the samples were performed using a p65 or S100 antibody. Results The cross-sectional area of each myofibril was significantly reduced by BTX-A injection (P < 0.001). The expression of p65 and S100 increased significantly with increasing concentrations of BTX-A (P < 0.001). Conclusions The injection of BTX-A into the masseter muscle induced muscle atrophy. Subsequently, p65 and S100 expression in myoblasts were increased for the protection of muscle cells.
Collapse
Affiliation(s)
- Young-Wook Park
- Department of Oral and Maxillofacial Surgery, College of Dentistry, Gangneung-Wonju National University, 7 Jukhyun-gil, Gangneung, 210-702 South Korea
| | - Seong-Gon Kim
- Department of Oral and Maxillofacial Surgery, College of Dentistry, Gangneung-Wonju National University, 7 Jukhyun-gil, Gangneung, 210-702 South Korea
| | - You-Young Jo
- Sericultural and Apicultural Materials Division, National Academy of Agricultural Science, Suwon, South Korea
| |
Collapse
|
13
|
Nam DH, Han JH, Lee TJ, Shishido T, Lim JH, Kim GY, Woo CH. CHOP deficiency prevents methylglyoxal-induced myocyte apoptosis and cardiac dysfunction. J Mol Cell Cardiol 2015; 85:168-77. [PMID: 26027784 DOI: 10.1016/j.yjmcc.2015.05.016] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Revised: 05/05/2015] [Accepted: 05/22/2015] [Indexed: 12/23/2022]
Abstract
Epidemiological studies indicate that methylglyoxal (MGO) plasma levels are closely linked to diabetes and the exacerbation of diabetic cardiovascular complications. Recently, it was established that endoplasmic reticulum (ER) stress importantly contributes to the pathogenesis of diabetes and its cardiovascular complications. The objective of this study was to explore the mechanism by which diabetes instigates cardiomyocyte apoptosis and cardiac dysfunction via MGO-mediated myocyte apoptosis. Intriguingly, the MGO activated unfolded protein response pathway accompanying apoptotic events, such as cleavages of PARP-1 and caspase-3. In addition, Western blot analysis revealed that MGO-induced myocyte apoptosis was inhibited by depletion of CHOP with siRNA against Ddit3, the gene name for rat CHOP. To investigate the physiologic roles of CHOP in vivo, glucose tolerance and cardiac dysfunction were assessed in CHOP-deficient mice. No significant difference was observed between CHOP KO and littermate naïve controls in terms of the MGO-induced impairment of glucose tolerance. In contrast, myocyte apoptosis, inflammation, and cardiac dysfunction were significantly diminished in CHOP KO compared with littermate naïve controls. These results showed that CHOP is the key signal for myocyte apoptosis and cardiac dysfunction induced by MGO. These findings suggest a therapeutic potential of CHOP inhibition in the management of diabetic cardiovascular complications including diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Dae-Hwan Nam
- Department of Pharmacology, Yeungnam University College of Medicine, 317-1 Daemyung-dong, Daegu, Republic of Korea
| | - Jung-Hwa Han
- Department of Pharmacology, Yeungnam University College of Medicine, 317-1 Daemyung-dong, Daegu, Republic of Korea
| | - Tae-Jin Lee
- Department of Anatomy, Yeungnam University College of Medicine, 317-1 Daemyung-dong, Daegu, Republic of Korea
| | - Tetsuro Shishido
- Department of Cardiology, Pulmonology and Nephrology, Yamagata University School of Medicine, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Jae Hyang Lim
- Department of Microbiology, Ewha Womans University School of Medicine, 911-1 Mok-dong, Seoul, Republic of Korea
| | - Geun-Young Kim
- Division of Cardiovascular and Rare Diseases, Center for Biomedical Sciences, Korea National Institute of Health, Cheongju-si, Chungcheongbuk-do, Republic of Korea
| | - Chang-Hoon Woo
- Department of Pharmacology, Yeungnam University College of Medicine, 317-1 Daemyung-dong, Daegu, Republic of Korea.
| |
Collapse
|
14
|
Cheong KA, Noh M, Kim CH, Lee AY. S100B as a potential biomarker for the detection of cytotoxicity of melanocytes. Exp Dermatol 2014; 23:165-71. [PMID: 24451020 DOI: 10.1111/exd.12332] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/17/2014] [Indexed: 12/30/2022]
Abstract
Skin irritation is one of the most common adverse reactions in hydroquinone (HQ) and retinoic acid (RA). Although melanocytes have rarely been considered to be involved in skin irritation, RA and particularly HQ could induce melanocyte toxicity, resulting in depigmentation. We chose S100B as a candidate gene for melanocytotoxicity from a genome-wide transcriptional profiling analysis after applying irritant doses of HQ, RA and sodium lauryl sulphate (SLS) to cultures of keratinocytes and/or melanocytes. In this study, the role of S100B on melanocyte viability and cytotoxicity was examined. S100B was detected in melanocytes, but not in keratinocytes or fibroblasts. Melanocytes after treatment with increasing concentrations of HQ, RA, SLS and urushiol showed significant increases in intracellular and extracellular S100B expression with reduced viable cell number and increased release of lactate dehydrogenase. No RAGE expression and no significant function of CD166/ALCAM in melanocyte survival and cytotoxicity favoured the role of intracellular S100B in chemically irritated melanocytes. S100B knock-down increased apoptosis through inhibition of PI3K/AKT, NF-κB and ERK activation, suggesting the increased intracellular S100B expression by chemical irritation as a compensatory reaction to reduce cytotoxicity. The numerical decrease in S100B/c-kit-double-positive melanocytes was also examined in human skin epidermis irritated by HQ or RA with stronger staining intensities of S100B. Collectively, the decrease in viable cell number by reduced intracellular S100B levels in vitro and by chemical irritation in vivo suggests that S100B could be a potential biomarker for melanocytes cytotoxicity.
Collapse
Affiliation(s)
- Kyung Ah Cheong
- Department of Dermatology, Graduate School of Medicine, Dongguk University Seoul, Gyeonggi-do, South Korea
| | | | | | | |
Collapse
|
15
|
Yao B, Zhang LN, Ai YH, Liu ZY, Huang L. Serum S100β is a better biomarker than neuron-specific enolase for sepsis-associated encephalopathy and determining its prognosis: a prospective and observational study. Neurochem Res 2014; 39:1263-9. [PMID: 24760429 DOI: 10.1007/s11064-014-1308-0] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2013] [Revised: 03/27/2014] [Accepted: 04/10/2014] [Indexed: 11/29/2022]
Abstract
S100β and neuron-specific enolase (NSE) are brain injury biomarkers, mainly used in brain trauma, cerebral stroke and hypoxic ischemia encephalopathy. The aim of this study was to study the clinical significance of serum S100β and NSE in diagnosing sepsis-associated encephalopathy (SAE) and predicting its prognosis. This was a prospective and observational study. Clinical data of septic patients were collected within 24 h after ICU admission from May 2012 to April 2013. We evaluated the level of consciousness twice per day. SAE was defined as cerebral dysfunction in the presence of sepsis that fulfilled the exclusion criteria. The infection biochemical indicators, Glasgow coma scale (GCS) score, acute physiology and chronic health evaluation score II, serum NSE and S100β were newly measured or evaluated for SAE patients. Finally, hospital mortality, bacteriological categories, length of ICU stay and length of hospital stay were also recorded for all enrolled patients. The data was analyzed with the Chi square test, two-sample t test or Mann-Whitney U test between two groups. The correlation between two factors was analyzed using the Pearson or Spearman analysis. Receiver operating characteristic (ROC) curves were used to determine the ability of S100β and NSE in diagnosing SAE and predicting the hospital mortality. In addition, cut-off points were obtained from the curves to determine the highest sum of sensitivity and specificity. Of 112 enrolled patients, 48 patients were diagnosed with SAE. The serum S100β and NSE concentrations in SAE patients were both significantly higher than in non-SAE patients 0.306 (IQR 0.157-0.880) μg/L vs. 0.095 (IQR 0.066-0.177) μg/L, 24.87 (IQR 31.73-12.73) ng/mL vs. 15.49 (IQR 9.88-21.46) ng/mL, P < 0.01]. GCS scores were related more closely to S100β than NSE (-0.595 vs. -0.337). S100β levels of 0.131 μg/L diagnosed SAE with 67.2% specificity and 85.4% sensitivity in the ROC curve, the area under the curve was 0.824 (95% confidence interval 0.750-0.898). NSE levels of 24.15 ng/mL diagnosed SAE with 82.8% specificity and 54.2% sensitivity, and the area under the curve was 0.664 (95 % confidence interval 0.561-0.767). In addition, the area under the curve for S100β for predicting hospital mortality was larger than for NSE (0.730 vs. 0.590). Serum S100β concentrations in SAE patients were significantly higher than in non-SAE patients. These may be related to the severity of SAE and may predict the outcome of sepsis. The efficacy and sensitivity of serum S100β in diagnosing SAE were high, but it had a low specificity. Moreover, compared to NSE, serum S100β was better for both diagnosing SAE and predicting the outcome of sepsis.
Collapse
Affiliation(s)
- Bo Yao
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
| | | | | | | | | |
Collapse
|
16
|
Cheng P, Alberts I, Li X. The role of ERK1/2 in the regulation of proliferation and differentiation of astrocytes in developing brain. Int J Dev Neurosci 2013; 31:783-9. [DOI: 10.1016/j.ijdevneu.2013.09.008] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Revised: 09/19/2013] [Accepted: 09/20/2013] [Indexed: 10/26/2022] Open
Affiliation(s)
- Peipei Cheng
- Shanghai Mental Health CenterShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Ian Alberts
- Department of Natural Sciences, LaGuardia CCCity University of New YorkNY11101USA
| | - Xiaohong Li
- Department of NeurochemistryNY State Institute for Basic Research in Developmental DisabilitiesNew YorkNY10314USA
| |
Collapse
|
17
|
Gupta AA, Chou RH, Li H, Yang LW, Yu C. Structural insights into the interaction of human S100B and basic fibroblast growth factor (FGF2): Effects on FGFR1 receptor signaling. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2013; 1834:2606-19. [PMID: 24063890 DOI: 10.1016/j.bbapap.2013.09.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Revised: 09/06/2013] [Accepted: 09/17/2013] [Indexed: 01/11/2023]
Abstract
S100B is a calcium sensing protein belonging to the S100 protein family with intracellular and extracellular roles. It is one of the EF hand homodimeric proteins, which is known to interact with various protein targets to regulate varied biological functions. Extracellular S100B has been recently reported to interact with FGF2 in a RAGE-independent manner. However, the recognition mechanism of S100B-FGF2 interaction at the molecular level remains unclear. In this study, the critical residues on S100B-FGF2 interface were mapped by combined information derived from NMR spectroscopy and site directed mutagenesis experiments. Utilizing NMR titration data, we generated the structural models of S100B-FGF2 complex from the computational docking program, HADDOCK which were further proved stable during 15ns unrestrained molecular dynamics (MD) simulations. Isothermal titration calorimetry studies indicated S100B interaction with FGF2 is an entropically favored process implying dominant role of hydrophobic contacts at the protein-protein interface. Residue level information of S100B interaction with FGF2 was useful to understand the varied target recognition ability of S100B and further explained its role in effecting extracellular signaling diversity. Mechanistic insights into the S100B-FGF2 complex interface and cell-based assay studies involving mutants led us to conclude the novel role of S100B in FGF2 mediated FGFR1 receptor inactivation.
Collapse
Affiliation(s)
- Arun A Gupta
- Department of Chemistry, National Tsing Hua University, Hsinchu, Taiwan
| | | | | | | | | |
Collapse
|
18
|
Ondruschka B, Pohlers D, Sommer G, Schober K, Teupser D, Franke H, Dressler J. S100B and NSE as useful postmortem biochemical markers of traumatic brain injury in autopsy cases. J Neurotrauma 2013; 30:1862-71. [PMID: 23796187 DOI: 10.1089/neu.2013.2895] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Postmortem analysis of relevant biomarkers might aid in characterizing causes of death and survival times in legal medicine. However, there are still no sufficiently established results of practical postmortem biochemical investigations in cases of traumatic brain injury (TBI). The two biomarkers--S100 protein subunit B (S100B) and neuronal specific enolase (NSE)--could be of special interest. Therefore, the aim of the present study was to investigate changes in their postmortem levels for further determination of brain damage in TBI. In 17 cases of TBI (average age, 58 years) and in 23 controls with different causes of death (average age, 59 years), serum and cerebrospinal fluid (CSF) samples were analyzed with a chemiluminescence immunoassay for marker expression. An increase in serum S100B, as well as a subsequent decrease after survival times>4 days, were detected in TBI cases (p<0.01). CSF NSE values >6,000 ng/mL and CSF S100B levels >10,000 ng/mL seem to indicate a TBI survival time of at least 15 min (p<0.01). It is of particular interest that CSF S100B levels (p<0.01) and serum S100B levels (p<0.05) as well as CSF NSE values (p<0.01) were significantly higher in TBI cases in comparison to the controls, especially when compared with fatal non-head injuries. In conclusion, the present findings emphasize that S100B and NSE are useful markers in postmortem biochemistry in cases of suspected TBI. Further, S100B may be helpful to estimate the survival time of fatal injuries in legal medicine.
Collapse
Affiliation(s)
- Benjamin Ondruschka
- 1 Institute of Legal Medicine, Medical Faculty University of Leipzig, Leipzig , Germany
| | | | | | | | | | | | | |
Collapse
|
19
|
Neuroinflammation in the aging down syndrome brain; lessons from Alzheimer's disease. Curr Gerontol Geriatr Res 2012; 2012:170276. [PMID: 22454637 PMCID: PMC3290800 DOI: 10.1155/2012/170276] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Accepted: 11/15/2011] [Indexed: 12/25/2022] Open
Abstract
Down syndrome (DS) is the most genetic cause of mental retardation and is caused by the triplication of chromosome 21. In addition to the disabilities caused early in life, DS is also noted as causing Alzheimer's-disease-like pathological changes in the brain, leading to 50-70% of DS patients showing dementia by 60-70 years of age. Inflammation is a complex process that has a key role to play in the pathogenesis of Alzheimer's disease. There is relatively little understood about inflammation in the DS brain and how the genetics of DS may alter this inflammatory response and change the course of disease in the DS brain. The goal of this review is to highlight our current understanding of inflammation in Alzheimer's disease and predict how inflammation may affect the pathology of the DS brain based on this information and the known genetic changes that occur due to triplication of chromosome 21.
Collapse
|
20
|
Li JP, Lu L, Wang LJ, Zhang FR, Shen WF. Increased serum levels of S100B are related to the severity of cardiac dysfunction, renal insufficiency and major cardiac events in patients with chronic heart failure. Clin Biochem 2011; 44:984-8. [PMID: 21640093 DOI: 10.1016/j.clinbiochem.2011.05.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2011] [Revised: 04/08/2011] [Accepted: 05/13/2011] [Indexed: 10/18/2022]
Abstract
OBJECTIVE To investigate the correlations between S100B and the severity of cardiac dysfunction, renal insufficiency (RI) and prognosis in chronic heart failure (CHF). METHOD Serum levels of S100B, TNF-α, high sensitivity CRP and NT-proBNP were determined in CHF patients with (n=96) and without RI (n=146). Patients with RI only (n=62) and control subjects (n=64) served for comparison. Patients were followed up for one year. RESULTS S100B levels were higher in CHF patients with a further elevation in those with RI (P<0.01). Serum S100B levels correlated with left ventricular ejection fraction, left ventricular end-diastolic volume and NT-proBNP in CHF patients, and eGFR in patients with RI (all P<0.05). Increased S100B levels were associated with major cardiac events (MCE), and were independently associated with the presence of CHF (all P<0.05). CONCLUSION Increased serum S100B levels were associated with the severity of cardiac dysfunction, RI and an adverse prognosis in CHF patients. It represents an independent risk factor for CHF.
Collapse
Affiliation(s)
- Jin Ping Li
- Department of Cardiology, Rui Jin Hospital, Shanghai Jiaotong University School of Medicine, PR China
| | | | | | | | | |
Collapse
|
21
|
Jin Q, Chen H, Luo A, Ding F, Liu Z. S100A14 stimulates cell proliferation and induces cell apoptosis at different concentrations via receptor for advanced glycation end products (RAGE). PLoS One 2011; 6:e19375. [PMID: 21559403 PMCID: PMC3084824 DOI: 10.1371/journal.pone.0019375] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2010] [Accepted: 03/31/2011] [Indexed: 01/09/2023] Open
Abstract
S100A14 is an EF-hand containing calcium-binding protein of the S100 protein family that exerts its biological effects on different types of cells. However, exact extracellular roles of S100A14 have not been clarified yet. Here we investigated the effects of S100A14 on esophageal squamous cell carcinoma (ESCC) cell lines. Results demonstrated that low doses of extracellular S100A14 stimulate cell proliferation and promote survival in KYSE180 cells through activating ERK1/2 MAPK and NF-κB signaling pathways. Immunoprecipitation assay showed that S100A14 binds to receptor for advanced glycation end products (RAGE) in KYSE180 cells. Inhibition of RAGE signaling by different approaches including siRNA for RAGE, overexpression of a dominant-negative RAGE construct or a RAGE antagonist peptide (AmphP) significantly blocked S100A14-induced effects, suggesting that S100A14 acts via RAGE ligation. Furthermore, mutation of the N-EF hand of S100A14 (E39A, E45A) virtually reduced 10 µg/ml S100A14-induced cell proliferation and ERK1/2 activation. However, high dose (80 µg/ml) of S100A14 causes apoptosis via the mitochondrial pathway with activation of caspase-3, caspase-9, and poly(ADP-ribose) polymerase. High dose S100A14 induces cell apoptosis is partially in a RAGE-dependent manner. This is the first study to demonstrate that S100A14 binds to RAGE and stimulates RAGE-dependent signaling cascades, promoting cell proliferation or triggering cell apoptosis at different doses.
Collapse
Affiliation(s)
- Qing'e Jin
- State Key Laboratory of Molecular Oncology, Cancer Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hongyan Chen
- State Key Laboratory of Molecular Oncology, Cancer Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Aiping Luo
- State Key Laboratory of Molecular Oncology, Cancer Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fang Ding
- State Key Laboratory of Molecular Oncology, Cancer Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhihua Liu
- State Key Laboratory of Molecular Oncology, Cancer Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- * E-mail:
| |
Collapse
|
22
|
Sorci G, Bianchi R, Riuzzi F, Tubaro C, Arcuri C, Giambanco I, Donato R. S100B Protein, A Damage-Associated Molecular Pattern Protein in the Brain and Heart, and Beyond. Cardiovasc Psychiatry Neurol 2010; 2010:656481. [PMID: 20827421 PMCID: PMC2933911 DOI: 10.1155/2010/656481] [Citation(s) in RCA: 115] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2010] [Accepted: 06/08/2010] [Indexed: 12/15/2022] Open
Abstract
S100B belongs to a multigenic family of Ca(2+)-binding proteins of the EF-hand type and is expressed in high abundance in the brain. S100B interacts with target proteins within cells thereby altering their functions once secreted/released with the multiligand receptor RAGE. As an intracellular regulator, S100B affects protein phosphorylation, energy metabolism, the dynamics of cytoskeleton constituents (and hence, of cell shape and migration), Ca(2+) homeostasis, and cell proliferation and differentiation. As an extracellular signal, at low, physiological concentrations, S100B protects neurons against apoptosis, stimulates neurite outgrowth and astrocyte proliferation, and negatively regulates astrocytic and microglial responses to neurotoxic agents, while at high doses S100B causes neuronal death and exhibits properties of a damage-associated molecular pattern protein. S100B also exerts effects outside the brain; as an intracellular regulator, S100B inhibits the postinfarction hypertrophic response in cardiomyocytes, while as an extracellular signal, (high) S100B causes cardiomyocyte death, activates endothelial cells, and stimulates vascular smooth muscle cell proliferation.
Collapse
Affiliation(s)
- Guglielmo Sorci
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Via del Giochetto, 06122 Perugia, Italy
| | - Roberta Bianchi
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Via del Giochetto, 06122 Perugia, Italy
| | - Francesca Riuzzi
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Via del Giochetto, 06122 Perugia, Italy
| | - Claudia Tubaro
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Via del Giochetto, 06122 Perugia, Italy
| | - Cataldo Arcuri
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Via del Giochetto, 06122 Perugia, Italy
| | - Ileana Giambanco
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Via del Giochetto, 06122 Perugia, Italy
| | - Rosario Donato
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Via del Giochetto, 06122 Perugia, Italy
| |
Collapse
|
23
|
Goyette J, Geczy CL. Inflammation-associated S100 proteins: new mechanisms that regulate function. Amino Acids 2010; 41:821-42. [PMID: 20213444 DOI: 10.1007/s00726-010-0528-0] [Citation(s) in RCA: 258] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2009] [Accepted: 02/12/2010] [Indexed: 01/11/2023]
Abstract
This review focuses on new aspects of extracellular roles of the calgranulins. S100A8, S100A9 and S100A12 are constitutively expressed in neutrophils and induced in several cell types. The S100A8 and S100A9 genes are regulated by pro- and anti-inflammatory mediators and their functions may depend on cell type, mediators within a particular inflammatory milieu, receptors involved in their recognition and their post-translational modification. The S100A8 gene induction in macrophages is dependent on IL-10 and potentiated by immunosuppressive agents. S100A8 and S100A9 are oxidized by peroxide, hypochlorite and nitric oxide (NO). HOCl generates intra-chain sulfinamide bonds; stronger oxidation promotes cross-linked forms that are seen in human atheroma. S100A8 is >200-fold more sensitive to oxidative cross-linking than low-density lipoprotein and may reduce oxidative damage. S100A8 and S100A9 can be S-nitrosylated. S100A8-SNO suppresses mast cell activation and inflammation in the microcirculation and may act as an NO transporter to regulate vessel tone in inflammatory lesions. S100A12 activates mast cells and is a monocyte and mast cell chemoattractant; a G-protein-coupled mechanism may be involved. Structure-function studies are discussed in relation to conservation and divergence of functions in S100A8. S100A12 induces cytokines in mast cells, but not monocytes/macrophages. It forms complexes with Zn(2+) and, by chelating Zn(2+), S100A12 significantly inhibits MMPs. Zn(2+) in S100A12 complexes co-localize with MMP-9 in foam cells in atheroma. In summary, S100A12 has pro-inflammatory properties that are likely to be stable in an oxidative environment, because it lacks Cys and Met residues. Conversely, S100A8 and S100A9 oxidation and S-nitrosylation may have important protective mechanisms in inflammation.
Collapse
Affiliation(s)
- Jesse Goyette
- Centre for Infection and Inflammation Research, School of Medical Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | | |
Collapse
|
24
|
Bernardini C, Lattanzi W, Businaro R, Leone S, Corvino V, Sorci G, Lauro G, Fumagalli L, Donato FR, Michetti F. Transcritpional effects of S100B on neuroblastoma cells: perturbation of cholesterol homeostasis and interference on the cell cycle. Gene Expr 2010; 14:345-59. [PMID: 20635576 PMCID: PMC6042022 DOI: 10.3727/105221610x12718619643013] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
S100B is a Ca2+ binding protein mainly secreted by astrocytes in the vertebrate brain that is considered a multifunctional cytokine and/or a damage-associated molecular pattern (DAMP) protein and a marker of brain injury and neurodegeneration when measured in different body fluids. It has been widely shown that this protein can exert diverse effects in neural cultures depending on its concentration, having detrimental effects at micromolar concentrations. The molecular mechanisms underlying this effect are still largely unknown. This study attempts to delineate the genome-wide gene expression analysis of the events associated with exposure to micromolar concentration of S100B in a human neuroblastoma cell line. In this experimental condition cells undergo a severe perturbation of lipid homeostasis along with cell cycle arrest. These mechanisms might reasonably mediate some aspects of the S100B-related detrimental effects of S100B, although obvious differences between mature neurons and neuroblastoma cells have to be considered.
Collapse
Affiliation(s)
- C Bernardini
- Institute of Anatomy and Cell Biology, Catholic University, Rome, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
S100A8/A9 induces autophagy and apoptosis via ROS-mediated cross-talk between mitochondria and lysosomes that involves BNIP3. Cell Res 2009; 20:314-31. [PMID: 19935772 DOI: 10.1038/cr.2009.129] [Citation(s) in RCA: 196] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The complex formed by two members of the S100 calcium-binding protein family, S100A8/A9, exerts apoptosis-inducing activity in various cells of different origins. Here, we present evidence that the underlying molecular mechanisms involve both programmed cell death I (PCD I, apoptosis) and PCD II (autophagy)-like death. Treatment of cells with S100A8/A9 caused the increase of Beclin-1 expression as well as Atg12-Atg5 formation. S100A8/A9-induced cell death was partially inhibited by the specific PI3-kinase class III inhibitor, 3-methyladenine (3-MA), and by the vacuole H(+)-ATPase inhibitor, bafilomycin-A1 (Baf-A1). S100A8/A9 provoked the translocation of BNIP3, a BH3 only pro-apoptotic Bcl2 family member, to mitochondria. Consistent with this finding, DeltaTM-BNIP3 overexpression partially inhibited S100A8/A9-induced cell death, decreased reactive oxygen species (ROS) generation, and partially protected against the decrease in mitochondrial transmembrane potential in S100A8/A9-treated cells. In addition, either DeltaTM-BNIP3 overexpression or N-acetyl-L-cysteine co-treatment decreased lysosomal activation in cells treated with S100A8/A9. Our data indicate that S100A8/A9-promoted cell death occurs through the cross-talk of mitochondria and lysosomes via ROS and the process involves BNIP3.
Collapse
|
26
|
Tsoporis JN, Izhar S, Leong-Poi H, Desjardins JF, Huttunen HJ, Parker TG. S100B interaction with the receptor for advanced glycation end products (RAGE): a novel receptor-mediated mechanism for myocyte apoptosis postinfarction. Circ Res 2009; 106:93-101. [PMID: 19910580 DOI: 10.1161/circresaha.109.195834] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
RATIONALE Post-myocardial infarction ventricular remodeling is associated with the expression of a variety of factors including S100B that can potentially modulate myocyte apoptosis. OBJECTIVE This study was undertaken to investigate the expression and function of S100B and its receptor, the receptor for advanced glycation end products (RAGE) in both postinfarction myocardium and in a rat neonatal myocyte culture model. METHODS AND RESULTS In a rat model of myocardial infarction following coronary artery ligation, we demonstrate in periinfarct myocytes, upregulation of RAGE, induction of S100B, and release into plasma with consequent myocyte apoptosis. Using a coimmunoprecipitation strategy, we demonstrate a direct interaction between S100B and RAGE. In rat neonatal cardiac myocyte cultures, S100B at concentrations > or = 50 nmol/L induced myocyte apoptosis, as evidenced by increased terminal DNA fragmentation, TUNEL, cytochrome c release from mitochondria to cytoplasm, phosphorylation of extracellular signal-regulated kinase (ERK)1/2 and p53, increased expression and activity of proapoptotic caspase-3, and decreased expression of antiapoptotic Bcl-2. Transfection of a full-length cDNA of RAGE or a dominant-negative mutant of RAGE resulted in increased or attenuated S100B-induced myocyte apoptosis, respectively. Inhibition of ERK1/2 by U0126/PD-98059 or overexpression of a dominant negative p53 comparably inhibited S100B-induced myocyte apoptosis. CONCLUSIONS These results suggest that interaction of RAGE and its ligand S100B after myocardial infarction may play a role in myocyte apoptosis by activating ERK1/2 and p53 signaling. This receptor-mediated mechanism is uniquely amenable to therapeutic intervention.
Collapse
Affiliation(s)
- J N Tsoporis
- Division of Cardiology, Department of Medicine, Keenan Research Centre, Li Ka Shing Knowledge Institute, St Michael's Hospital, University of Toronto, Canada
| | | | | | | | | | | |
Collapse
|
27
|
Comparison of distinct protein isoforms of the receptor for advanced glycation end-products expressed in murine tissues and cell lines. Cell Tissue Res 2009; 337:79-89. [PMID: 19415334 DOI: 10.1007/s00441-009-0791-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2008] [Accepted: 03/09/2009] [Indexed: 02/02/2023]
Abstract
The receptor for advanced glycation end-products (RAGE) is thought to be expressed ubiquitously as various protein isoforms. Our objective was to use Northern blotting, immunoblotting, and sensitivity to N-glycanase digestion to survey RAGE isoforms expressed in cell lines and mouse tissues in order to obtain a more comprehensive view of the RAGE expressome. Pulmonary RAGE mRNA (1.4 kb) was smaller than cell-line and tissue RAGE mRNA (6 kb-10 kb). Three anti-RAGE antibodies that recognized three distinct RAGE epitopes were used for protein studies (N-16, H-300, and alphaES). Lung expressed three predominant protein isoforms with apparent molecular masses of 45.1, 52.6, and 57.4 kDa (N-16/H-300) and four isoforms at 25.0, 46.9, 52.5, and 54.2 kDa (alphaES). These isoforms were expressed exclusively in lung. Heart, ileum, and kidney expressed a 44.0-kDa isoform (N-16), whereas aorta and pancreas expressed a 53.3-kDa isoform (alphaES). Each of these isoforms were absent in tissue extracts prepared from RAGE(-/-) mice. Cell lines expressed a 70.0-kDa isoform, and a subset expressed a 30.0-kDa isoform (alphaES). Lung RAGE appeared to contain two N-linked glycans. Tissue and cell-line RAGE isoforms were completely insensitive to PNGase F digestion. Thus, numerous RAGE protein isoforms are detectable in tissues and cell lines. Canonical transmembrane and soluble RAGE appear to be expressed solely in lung (N-16/H-300). Non-pulmonary tissues and cell lines, regardless of the source tissue, both express distinct RAGE protein isoforms containing the N-terminal N-16 epitope or the alphaES RAGE epitope encoded by alternate exon 9, but lacking the H-300 epitope.
Collapse
|
28
|
|
29
|
Grau E, Balaguer J, Canete A, Martinez F, Orellana C, Oltra S, Hernandez M, Castel V. Subtelomeric analysis of pediatric astrocytoma: subchromosomal instability is a distinctive feature of pleomorphic xanthoastrocytoma. J Neurooncol 2008; 93:175-82. [PMID: 19099200 DOI: 10.1007/s11060-008-9763-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2008] [Accepted: 12/08/2008] [Indexed: 10/21/2022]
Abstract
Astrocytic neoplasms are genetically heterogeneous; however a low frequency of genomic changes has been found in juvenile pilocytic astrocytoma (PA) in molecular studies. Concerning pleomorphic xanthoastrocytomas (PXA), recent studies have given heterogeneous results for chromosomal alterations. We studied the subtelomeric regions of 19 primary astrocytoma tumors. Results were near normality for the PA group with relative scarcity of chromosomal imbalances, except for the duplication of 3pter in 4/15 and deletion of 21qter in 5/15 of them. In contrast, a specific profile was observed in the 4 PXA tumoral samples. This involved 3pter, 14qter and 19pter duplication and 4qter, 6qter, 9qter, 13cen, 17pter, 18qter and 21qter deletion. Our results indicate that the chromosomal and genetic aberrations in PXAs differed from those typically associated with the diffusely infiltrating astrocytic and oligodendroglial gliomas. These genetic differences would likely contribute to the more favorable behavior of PXAs and may be helpful for molecular differential diagnosis of pediatric cerebral tumors.
Collapse
Affiliation(s)
- Elena Grau
- Hospital Universitario La Fe, Valencia, Spain.
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Leclerc E, Fritz G, Vetter SW, Heizmann CW. Binding of S100 proteins to RAGE: an update. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2008; 1793:993-1007. [PMID: 19121341 DOI: 10.1016/j.bbamcr.2008.11.016] [Citation(s) in RCA: 382] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2008] [Revised: 11/24/2008] [Accepted: 11/28/2008] [Indexed: 12/21/2022]
Abstract
The Receptor for Advanced Glycation Endproducts (RAGE) is a multi-ligand receptor of the immunoglobulin family. RAGE interacts with structurally different ligands probably through the oligomerization of the receptor on the cell surface. However, the exact mechanism is unknown. Among RAGE ligands are members of the S100 protein family. S100 proteins are small calcium binding proteins with high structural homology. Several members of the family have been shown to interact with RAGE in vitro or in cell-based assays. Interestingly, many RAGE ligands appear to interact with distinct domains of the extracellular portion of RAGE and to trigger various cellular effects. In this review, we summarize the modes of S100 protein-RAGE interaction with regard to their cellular functions.
Collapse
Affiliation(s)
- Estelle Leclerc
- Department of Chemistry and Biochemistry, Florida Atlantic University, 777 Glades Road, Boca Raton, Fl 33431, USA
| | | | | | | |
Collapse
|
31
|
Donato R, Sorci G, Riuzzi F, Arcuri C, Bianchi R, Brozzi F, Tubaro C, Giambanco I. S100B's double life: intracellular regulator and extracellular signal. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2008; 1793:1008-22. [PMID: 19110011 DOI: 10.1016/j.bbamcr.2008.11.009] [Citation(s) in RCA: 537] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2008] [Revised: 11/12/2008] [Accepted: 11/21/2008] [Indexed: 12/22/2022]
Abstract
The Ca2+-binding protein of the EF-hand type, S100B, exerts both intracellular and extracellular functions. Recent studies have provided more detailed information concerning the mechanism(s) of action of S100B as an intracellular regulator and an extracellular signal. Indeed, intracellular S100B acts as a stimulator of cell proliferation and migration and an inhibitor of apoptosis and differentiation, which might have important implications during brain, cartilage and skeletal muscle development and repair, activation of astrocytes in the course of brain damage and neurodegenerative processes, and of cardiomyocyte remodeling after infarction, as well as in melanomagenesis and gliomagenesis. As an extracellular factor, S100B engages RAGE (receptor for advanced glycation end products) in a variety of cell types with different outcomes (i.e. beneficial or detrimental, pro-proliferative or pro-differentiative) depending on the concentration attained by the protein, the cell type and the microenvironment. Yet, RAGE might not be the sole S100B receptor, and S100B's ability to engage RAGE might be regulated by its interaction with other extracellular factors. Future studies using S100B transgenic and S100B null mice might shed more light on the functional role(s) of the protein.
Collapse
Affiliation(s)
- Rosario Donato
- Department of Experimental Medicine and Biochemical Sciences, Section Anatomy, University of Perugia, Via del Giochetto C.P. 81 Succ. 3, 06122 Perugia, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Neural-activity-dependent release of S100B from astrocytes enhances kainate-induced gamma oscillations in vivo. J Neurosci 2008; 28:10928-36. [PMID: 18945900 DOI: 10.1523/jneurosci.3693-08.2008] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
S100B is the principal calcium-binding protein of astrocytes and known to be secreted to extracellular space. Although secreted S100B has been reported to promote neurite extension and cell survival via its receptor [receptor for advanced glycation end products (RAGE)], effects of extracellular S100B on neural activity have been mostly unexplored. Here, we demonstrate that secreted S100B enhances kainate-induced gamma oscillations. Local infusion of S100B in S100B(-/-) mice enhanced hippocampal kainate-induced gamma oscillations in vivo. In a complementary set of experiments, local application of anti-S100B antibody in wild-type mice attenuated the gamma oscillations. Both results indicate that the presence of extracellular S100B enhances the kainate-induced gamma oscillations. In acutely isolated hippocampal slices, kainate application increased S100B secretion in a neural-activity-dependent manner. Further pharmacological experiments revealed that S100B secretion was critically dependent on presynaptic release of neurotransmitter and activation of metabotropic glutamate receptor 3. Moreover, the kainate-induced gamma oscillations were attenuated by the genetic deletion or antibody blockade of RAGE in vivo. These results suggest RAGE activation by S100B enhances the gamma oscillations. Together, we propose a novel pathway of neuron-glia communications--astrocytic release of S100B modulates neural network activity through RAGE activation.
Collapse
|
33
|
RAGE expression is up-regulated in human cerebral ischemia and pMCAO rats. Neurosci Lett 2008; 445:117-21. [PMID: 18782604 DOI: 10.1016/j.neulet.2008.08.077] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2008] [Revised: 08/26/2008] [Accepted: 08/27/2008] [Indexed: 11/22/2022]
Abstract
To determine whether the receptor for advanced glycation endproducts (RAGE) contributes to cerebral ischemia, we evaluated RAGE expression in human cerebral ischemia and a model of permanent middle cerebral artery occlusion (pMCAO) in rats. Biopsy specimens were obtained from 12 patients with unilateral cerebral infarction. For the pMCAO model, the middle cerebral artery (MCA) of Sprague-Dawley (SD) rats was permanently occluded. Immunohistochemistry and Western blotting were used to measure RAGE expression in the ischemic hemisphere relative to the normal hemisphere. PC12 cells subjected to oxygen and glucose deprivation (OGD) were used to evaluate the role of RAGE in cell injury. As expected, cerebral ischemia patients expressed elevated levels of RAGE in the ischemic hemisphere. In 1 and 2 days pMCAO rats, levels of RAGE were higher in the ischemic hemisphere relative to the non-ischemic hemisphere, and expression was primarily located in the penumbra of the ischemic hemisphere. In PC12 cells, levels of RAGE increased after 7h of OGD culture. Notably, blockade of RAGE with a selective RAGE antibody in vitro reduced the cytotoxicity caused by OGD. The present data suggest that RAGE is up-regulated in human cerebral ischemia and pMCAO rats, suggesting a role for RAGE in brain ischemia.
Collapse
|
34
|
Herczenik E, Gebbink MFBG. Molecular and cellular aspects of protein misfolding and disease. FASEB J 2008; 22:2115-33. [PMID: 18303094 DOI: 10.1096/fj.07-099671] [Citation(s) in RCA: 134] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Proteins are essential elements for life. They are building blocks of all organisms and the operators of cellular functions. Humans produce a repertoire of at least 30,000 different proteins, each with a different role. Each protein has its own unique sequence and shape (native conformation) to fulfill its specific function. The appearance of incorrectly shaped (misfolded) proteins occurs on exposure to environmental changes. Protein misfolding and the subsequent aggregation is associated with various, often highly debilitating, diseases for which no sufficient cure is available yet. In the first part of this review we summarize the structural composition of proteins and the current knowledge of underlying forces that lead proteins to lose their native structure. In the second and third parts we describe the molecular and cellular mechanisms that are associated with protein misfolding in disease. Finally, in the last part we portray recent efforts to develop treatments for protein misfolding diseases.
Collapse
Affiliation(s)
- Eszter Herczenik
- Laboratory of Thrombosis and Haemostasis, Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | | |
Collapse
|
35
|
Ghavami S, Kerkhoff C, Chazin WJ, Kadkhoda K, Xiao W, Zuse A, Hashemi M, Eshraghi M, Schulze-Osthoff K, Klonisch T, Los M. S100A8/9 induces cell death via a novel, RAGE-independent pathway that involves selective release of Smac/DIABLO and Omi/HtrA2. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2007; 1783:297-311. [PMID: 18060880 DOI: 10.1016/j.bbamcr.2007.10.015] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2007] [Revised: 10/19/2007] [Accepted: 10/23/2007] [Indexed: 11/18/2022]
Abstract
A complex of two S100 EF-hand calcium-binding proteins S100A8/A9 induces apoptosis in various cells, especially tumor cells. Using several cell lines, we have shown that S100A8/A9-induced cell death is not mediated by the receptor for advanced glycation endproducts (RAGE), a receptor previously demonstrated to engage S100 proteins. Investigation of cell lines either deficient in, or over-expressing components of the death signaling machinery provided insight into the S100A8/A9-mediated cell death pathway. Treatment of cells with S100A8/A9 caused a rapid decrease in the mitochondrial membrane potential (DeltaPsi(m)) and activated Bak, but did not cause release of apoptosis-inducing factor (AIF), endonuclease G (Endo G) or cytochrome c. However, both Smac/DIABLO and Omi/HtrA2 were selectively released into the cytoplasm concomitantly with a decrease in Drp1 expression, which inhibits mitochondrial fission machinery. S100A8/A9 treatment also resulted in decreased expression of the anti-apoptotic proteins Bcl2 and Bcl-X(L), whereas expression of the pro-apoptotic proteins Bax, Bad and BNIP3 was not altered. Over-expression of Bcl2 partially reversed the cytotoxicity of S100A8/A9. Together, these data indicate that S100A8/A9-induced cell death involves Bak, selective release of Smac/DIABLO and Omi/HtrA2 from mitochondria, and modulation of the balance between pro- and anti-apoptotic proteins.
Collapse
|
36
|
Zreiqat H, Howlett CR, Gronthos S, Hume D, Geczy CL. S100A8/S100A9 and their association with cartilage and bone. J Mol Histol 2007; 38:381-91. [PMID: 17636430 DOI: 10.1007/s10735-007-9117-2] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2007] [Accepted: 06/20/2007] [Indexed: 10/23/2022]
Abstract
S100A8 and S100A9 are calcium-binding proteins expressed in myeloid cells and are markers of numerous inflammatory diseases in humans. S100A9 has been associated with dystrophic calcification in human atherosclerosis. Here we demonstrate S100A8 and S100A9 expression in murine and human bone and cartilage cells. Only S100A8 was seen in preosteogenic cells whereas osteoblasts had variable, but generally weak expression of both proteins. In keeping with their reported high-mRNA expression, S100A8 and S100A9 were prominent in osteoclasts. S100A8 was expressed in alkaline phosphatase-positive hypertrophic chondrocytes, but not in proliferating chondrocytes within the growth plate where the cartilaginous matrix was calcifying. S100A9 was only evident in the invading vascular osteogenic tissue penetrating the degenerating chondrocytic zone adjacent to the primary spongiosa, where S100A8 was also expressed. Whilst, S100A8 has been shown to be associated with osteoblast differentiation, both S100A8 and S100A9 may contribute to calcification of the cartilage matrix and its replacement with trabecular bone, and to regulation of redox in bone resorption.
Collapse
Affiliation(s)
- H Zreiqat
- Biomaterials and Tissue Engineering Research Unit, Biomedical Engineering, School of AMME, The University of Sydney, Sydney, NSW, Australia.
| | | | | | | | | |
Collapse
|
37
|
Abstract
In recent years there has been a proliferation of interest in the brain-specific protein S100B, its many physiologic roles, and its behaviour in various neuropathologic conditions. Since the mid-1960s, its wide variety of intracellular and extracellular activities has been elucidated, and it has also been implicated in an increasing number of central nervous system (CNS) disorders. S100B is part of a superfamily of proteins, some of which (including S100B) have been implicated as calcium-dependent regulatory proteins that modulate the activity of effector proteins or cells. S100B is primarily an astrocytic protein. Within cells, it may have a role in signal transduction, and it is involved in calcium homeostasis. Information about the functional implication of S100B secretion by astrocytes into the extracellular space is scant but there is substantial evidence that secreted glial S100B exerts trophic or toxic effects depending on its concentration. This review summarises the historic development and current knowledge of S100B, including recent interesting findings relating S100B to a diversity of CNS pathologies such as traumatic brain injury, Alzheimer's disease, Down's syndrome, schizophrenia, and Tourette's syndrome. These broad implications have led some workers to describe S100B as 'the CRP (C-reactive protein) of the brain.' This review also examines S100B's potential role as a neurologic screening tool, or biomarker of CNS injury, analogous to the role of CRP as a marker of systemic inflammation.
Collapse
Affiliation(s)
- Jon Sen
- Institute of Neurology, University College London, Queen Square, London, United Kingdom.
| | | |
Collapse
|
38
|
Breyer MD, Tchekneva E, Qi Z, Takahashi T, Fogo AB, Zhao HJ, Harris RC. Genetics of diabetic nephropathy: lessons from mice. Semin Nephrol 2007; 27:237-47. [PMID: 17418691 DOI: 10.1016/j.semnephrol.2007.01.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Although diabetic nephropathy occurs only in a minority of diabetic patients (approximately 30%), it is the major single cause of end-stage renal disease in the United States. Hyperglycemia and hypertension are important factors predisposing patients to nephropathy, however, accumulating evidence points to critical genetic factors that predispose only a subset of diabetic patients to nephropathy. Defining the genes responsible for nephropathy risk in human populations has proven challenging. Comparative genomics using the robust genetic reagents available in the laboratory mouse should provide a complementary approach to defining genes that may predispose to diabetic nephropathy in mice and human beings. In this article we review studies that have started to identify genetic risk factors for diabetic nephropathy in mice and the multiple approaches that may be used to elucidate the genetic pathogenesis of this disorder.
Collapse
Affiliation(s)
- Matthew D Breyer
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University, Nashville, TN 37232, USA.
| | | | | | | | | | | | | |
Collapse
|
39
|
Zen K, Chen CXJ, Chen YT, Wilton R, Liu Y. Receptor for advanced glycation endproducts mediates neutrophil migration across intestinal epithelium. THE JOURNAL OF IMMUNOLOGY 2007; 178:2483-90. [PMID: 17277156 DOI: 10.4049/jimmunol.178.4.2483] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Receptor for advanced glycation endproducts (RAGE) is an Ig superfamily cell surface receptor that interacts with a diverse array of ligands associated with inflammatory responses. In this study, we provide evidence demonstrating that RAGE is involved in inflammatory responses in the intestines. We showed that RAGE is expressed in intestinal epithelial cells, primarily concentrated at the lateral membranes close to the apical cell junction complexes. Although RAGE expression was low in epithelium under normal conditions, this protein was up-regulated after treatment with the inflammatory cytokines IFN-gamma and/or TNF-alpha. RAGE expression was also elevated in colon tissue samples from patients with inflammatory bowel diseases. Using in vitro transmigration assays, we found that RAGE mediates neutrophil (polymorphonuclear leukocytes (PMN)) adhesion to, and subsequent migration across, intestinal epithelial monolayers. This activity appears to be mediated by the binding of RAGE to the PMN-specific beta(2) integrin CD11b/CD18. Thus, these results provide a novel mechanism for the regulation of PMN transepithelial migration and may suggest a new therapeutic target for intestinal inflammation.
Collapse
Affiliation(s)
- Ke Zen
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA
| | | | | | | | | |
Collapse
|
40
|
Druse MJ, Gillespie RA, Tajuddin NF, Rich M. S100B-mediated protection against the pro-apoptotic effects of ethanol on fetal rhombencephalic neurons. Brain Res 2007; 1150:46-54. [PMID: 17400198 PMCID: PMC2077982 DOI: 10.1016/j.brainres.2007.02.092] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2007] [Revised: 02/27/2007] [Accepted: 02/28/2007] [Indexed: 01/07/2023]
Abstract
Previously, this laboratory demonstrated that ethanol treatment significantly reduces the number of developing serotonin (5-HT) and other fetal rhombencephalic neurons in rats by augmenting apoptosis. Using a 5-HT(1A) agonist we were able to attenuate the ethanol-associated reduction and apoptosis of 5-HT and rhombencephalic neurons. The downstream pro-survival effects of 5-HT(1A) stimulation were associated with the activation of phosphatidylinositol 3'kinase (PI-3K) and its subsequent up-regulation of specific NF-kappaB-dependent pro-survival genes. Using an in vitro model, we investigated the hypothesis that S100B, a protein which is released from astrocytes following 5-HT(1A) agonist stimulation, can reduce apoptosis in ethanol-treated rat fetal rhombencephalic neurons. We also evaluated whether the anti-apoptotic effects of S100B on fetal rhombencephalic neurons were linked to the activation of the PI-3K-->pAkt pro-survival pathway and the expression of two NF-kappaB-dependent pro-survival genes: XIAP and Bcl-2. Moreover, we determined whether S100B's pro-survival effects were associated with mitogen activated protein kinase kinase (MAPKK)-->p42/p44 MAPK. The results of these investigations demonstrated that S100B treatment prevented ethanol-associated apoptosis of fetal rhombencephalic neurons. In addition, it appears that these neuroprotective effects are linked to activation of the PI-3K pathways, because the PI-3K inhibitor LY294002 blocks the neuroprotective effects of S100B. Moreover, S100B increases the formation of pAkt and the up-regulation of two downstream NF-kappaB-dependent pro-survival genes: XIAP and Bcl-2. Although the MAPKK inhibitor PD98059 reduced the number of surviving neurons in S100B-treated cultures, S100B did not activate MAPKK.
Collapse
Affiliation(s)
- Mary J. Druse
- Department of Cell Biology, Neurobiology, and Anatomy, Division of Molecular and Cellular Biochemistry, Loyola University, Stritch School of Medicine, 2160 S. First Avenue, Maywood, IL 60153
- Department of Cell Biology, Neurobiology, and Anatomy, The Alcohol Research Program, Loyola University, Stritch School of Medicine, 2160 S. First Avenue, Maywood, IL 60153
- Please address all communications to Mary Druse Manteuffel, Ph.D., Professor of Cell Biology, Neurobiology, and Anatomy, LUSSOM, Maywood, IL 60153. Phone: 708-216-3370, Fax: 708-216-8523,
| | - Roberta A. Gillespie
- Department of Cell Biology, Neurobiology, and Anatomy, Division of Molecular and Cellular Biochemistry, Loyola University, Stritch School of Medicine, 2160 S. First Avenue, Maywood, IL 60153
- Department of Cell Biology, Neurobiology, and Anatomy, The Alcohol Research Program, Loyola University, Stritch School of Medicine, 2160 S. First Avenue, Maywood, IL 60153
- Please address all communications to Mary Druse Manteuffel, Ph.D., Professor of Cell Biology, Neurobiology, and Anatomy, LUSSOM, Maywood, IL 60153. Phone: 708-216-3370, Fax: 708-216-8523,
| | - Nuzhath F. Tajuddin
- Department of Cell Biology, Neurobiology, and Anatomy, Division of Molecular and Cellular Biochemistry, Loyola University, Stritch School of Medicine, 2160 S. First Avenue, Maywood, IL 60153
- Department of Cell Biology, Neurobiology, and Anatomy, The Alcohol Research Program, Loyola University, Stritch School of Medicine, 2160 S. First Avenue, Maywood, IL 60153
- Please address all communications to Mary Druse Manteuffel, Ph.D., Professor of Cell Biology, Neurobiology, and Anatomy, LUSSOM, Maywood, IL 60153. Phone: 708-216-3370, Fax: 708-216-8523,
| | - Megan Rich
- Department of Cell Biology, Neurobiology, and Anatomy, Stritch School of Medicine, Loyola University, Stritch School of Medicine, 2160 S. First Avenue, Maywood, IL 60153
| |
Collapse
|
41
|
Basso D, Greco E, Fogar P, Pucci P, Flagiello A, Baldo G, Giunco S, Valerio A, Navaglia F, Zambon CF, Falda A, Pedrazzoli S, Plebani M. Pancreatic cancer-derived S-100A8 N-terminal peptide: A diabetes cause? Clin Chim Acta 2006; 372:120-8. [PMID: 16678810 DOI: 10.1016/j.cca.2006.03.027] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2006] [Revised: 03/24/2006] [Accepted: 03/24/2006] [Indexed: 10/24/2022]
Abstract
BACKGROUND Our aim was to identify the pancreatic cancer diabetogenic peptide. METHODS Pancreatic tumor samples from patients with (n=15) or without (n=7) diabetes were compared with 6 non-neoplastic pancreas samples using SDS-PAGE. RESULTS A band measuring approximately 1500 Da was detected in tumors from diabetics, but not in neoplastic samples from non-diabetics or samples from non-neoplastic subjects. Sequence analysis revealed a 14 amino acid peptide (1589.88 Da), corresponding to the N-terminal of the S100A8. At 50 nmol/L and 2 mmol/L, this peptide significantly reduced glucose consumption and lactate production by cultured C(2)C(12) myoblasts. The 14 amino acid peptide caused a lack of myotubular differentiation, the presence of polynucleated cells and caspase-3 activation. CONCLUSIONS The 14 amino acid peptide from S100A8 impairs the catabolism of glucose by myoblasts in vitro and may cause hyperglycemia in vivo. Its identification in biological fluids might be helpful in diagnosing pancreatic cancer in patients with recent onset diabetes mellitus.
Collapse
Affiliation(s)
- Daniela Basso
- Department of Laboratory Medicine, University of Padua, Via Giustiniani 2, 35128 Padova, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Riuzzi F, Sorci G, Donato R. S100B stimulates myoblast proliferation and inhibits myoblast differentiation by independently stimulating ERK1/2 and inhibiting p38 MAPK. J Cell Physiol 2006; 207:461-70. [PMID: 16419039 DOI: 10.1002/jcp.20580] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The Ca2+-modulated protein of the EF-hand type, S100B, was shown to inhibit rat L6 myoblast differentiation and myotube formation by interacting with a high affinity with an unidentified receptor (Sorci et al., 2003). We show here that S100B independently inhibits the MKK6-p38 MAPK pathway and stimulates the Ras-MEK-ERK1/2 pathway. The inhibitory effect of S100B on p38 MAPK translates into a defective induction of the muscle-specific transcription factor myogenin and the antiproliferative factor p21(WAF1), while S100B's stimulatory effect on ERK1/2 results in stimulation of myoblast proliferation via cyclin D1 induction and Rb phosphorylation and protection against apoptosis via activation of NF-kappaB transcriptional activity. Also, the S100B's effects that are mediated by the Ras-MEK-ERK1/2 pathway that is, stimulation of proliferation and protection against apoptosis, depend on reactive oxygen species production, being inhibited by antioxidants, while the S100B inhibitory effect on the MKK6-p38 MAPK pathway is not. We propose that S100B might participate in the regulation of myoblast differentiation by stimulating myoblast proliferation, protecting myoblasts against apoptosis, and modulating myotube formation.
Collapse
Affiliation(s)
- Francesca Riuzzi
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy
| | | | | |
Collapse
|
43
|
Korenaga K, Micci MA, Taglialatela G, Pasricha PJ. Suppression of nNOS expression in rat enteric neurones by the receptor for advanced glycation end-products. Neurogastroenterol Motil 2006; 18:392-400. [PMID: 16629867 DOI: 10.1111/j.1365-2982.2006.00774.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Diabetes mellitus results in a loss of neuronal nitric oxide synthase (nNOS) expression in the myenteric plexus but the underlying mechanisms remain unknown. We hypothesized that this may be mediated by advanced glycation end-products (AGEs), a class of modified protein adducts formed by non-enzymatic glycation that interact with the receptor for AGE (RAGE) and which are important in the pathogenesis of other diabetic complications. Whole mount preparations of longitudinal muscles with adherent myenteric plexus (LM-MPs) from the duodenum of adult male rats were exposed to glycated bovines serum albumin (AGE-BSA) or BSA for 24 h. Western blotting, immunohistochemistry and real-time reverse transcriptase polymerase chain reaction (RT-PCR) for mRNA showed a significant reduction in nNOS expression in LM-MPs after exposure to AGE-BSA. NO release, as measured by the Griess reaction, was also significantly reduced by AGE-BSA. A neutralizing antibody against RAGE attenuated the reduction of nNOS protein caused by AGE-BSA. Immunohistochemistry revealed co-localization of RAGE expression with Hu, a marker for neuronal cells but not for S-100, a glial marker. Advanced glycation end-products reduce nNOS expression in the rat myenteric neurones acting via the receptor RAGE. Our results suggest novel pathways for disruption of the nitrergic phenotype in diabetes.
Collapse
Affiliation(s)
- K Korenaga
- Enteric Neuromuscular Disorders and Pain Group, Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555-0764, USA
| | | | | | | |
Collapse
|
44
|
Riuzzi F, Sorci G, Donato R. The amphoterin (HMGB1)/receptor for advanced glycation end products (RAGE) pair modulates myoblast proliferation, apoptosis, adhesiveness, migration, and invasiveness. Functional inactivation of RAGE in L6 myoblasts results in tumor formation in vivo. J Biol Chem 2006; 281:8242-53. [PMID: 16407300 DOI: 10.1074/jbc.m509436200] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
We reported that RAGE (receptor for advanced glycation end products), a multiligand receptor of the immunoglobulin superfamily expressed in myoblasts, when activated by its ligand amphoterin (HMGB1), stimulates rat L6 myoblast differentiation via a Cdc42-Rac-MKK6-p38 mitogen-activated protein kinase pathway, and that RAGE expression in skeletal muscle tissue is developmentally regulated. We show here that inhibition of RAGE function via overexpression of a signaling deficient RAGE mutant (RAGE delta cyto) results in increased myoblast proliferation, migration, and invasiveness, and decreased apoptosis and adhesiveness, whereas myoblasts overexpressing RAGE behave the opposite, compared with mock-transfected myoblasts. These effects are accompanied by a decreased induction of the proliferation inhibitor, p21(Waf1), and increased induction of cyclin D1 and extent of Rb, ERK1/2, and JNK phosphorylation in L6/RAGE delta cyto myoblasts, the opposite occurring in L6/RAGE myoblasts. Neutralization of culture medium amphoterin negates effects of RAGE activation, suggesting that amphoterin is the RAGE ligand involved in RAGE-dependent effects in myoblasts. Finally, mice injected with L6/RAGE delta cyto myoblasts develop tumors as opposed to mice injected with L6/RAGE or L6/mock myoblasts that do not. Thus, the amphoterin/RAGE pair stimulates myoblast differentiation by the combined effect of stimulation of differentiation and inhibition of proliferation, and deregulation of RAGE expression in myoblasts might contribute to their neoplastic transformation.
Collapse
Affiliation(s)
- Francesca Riuzzi
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Casella Postale 81 Succursale 3, 06122 Perugia, Italy
| | | | | |
Collapse
|
45
|
Esposito G, De Filippis D, Cirillo C, Sarnelli G, Cuomo R, Iuvone T. The astroglial-derived S100beta protein stimulates the expression of nitric oxide synthase in rodent macrophages through p38 MAP kinase activation. Life Sci 2005; 78:2707-15. [PMID: 16376947 DOI: 10.1016/j.lfs.2005.10.023] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2005] [Accepted: 10/22/2005] [Indexed: 10/25/2022]
Abstract
S100beta is an astroglial-derived Ca2+ -binding protein having neurotrophic role on neurons and glial cells. An aberrant S100beta production has been observed in neurodegenerative disease, as Alzheimer's disease and Down syndrome. S100beta is responsible to start up a gliotic reaction by the release of pro-inflammatory mediators, including nitric oxide (NO) and cytokines from microglia and astrocytes, which are, in turn, deleterious for neurons. Interestingly, pro-inflammatory effect of S100beta seems not be restricted into the brain. Macrophages play a pivotal role in inflammatory diseases, occurring both in the brain and in the periphery. In this study, we tested the hypothesis that S100beta may affect macrophage functions, amplifying thus the inflammatory process. Our results demonstrate that S100beta stimulates both NO production and iNOS protein transcription and expression in J774 and rat peritoneal macrophages. NO production was concentration and time-dependently inhibited by two iNOS inhibitors, L-NAME and SMT. We also demonstrated that S100beta induced oxidative stress by increasing H2O2 production and lipid peroxidation of cell membrane in both macrophage types. The pro-oxidant potential of S100beta activates p38 MAP kinase (MAPK), which has been described to directly activate NF-kappaB. In our study, SB203580, a p38 MAPK inhibitor, and two NF-kappaB inhibitors, TLCK and BAY 11-7082, decreased both NO production and iNOS protein transcription and expression in S100beta-stimulated J774 and peritoneal rat macrophages. Moreover, additional studies demonstrated that S100beta affected also TNF-alpha protein expression in J774 macrophages. In conclusion, our results highlight the potential role of S100beta during an inflammatory scenario identifying macrophages as a novel S100beta-responsive cell-type.
Collapse
Affiliation(s)
- Giuseppe Esposito
- Department of Experimental Pharmacology, Faculty of Pharmacy, University of Naples Federico II Naples, Italy
| | | | | | | | | | | |
Collapse
|
46
|
Tsoporis JN, Marks A, Haddad A, Dawood F, Liu PP, Parker TG. S100B expression modulates left ventricular remodeling after myocardial infarction in mice. Circulation 2005; 111:598-606. [PMID: 15699280 DOI: 10.1161/01.cir.0000154554.65287.f5] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND S100B, a 20-kDa, Ca2+-binding dimer, is a putative intrinsic negative regulator of myocardial hypertrophy expressed after myocardial infarction. S100B-overexpressing transgenic (TG) and S100B-knockout (KO) mice have been generated to assess the consequences of S100B expression and altered hypertrophy after infarction. METHODS AND RESULTS We compared 21 wild-type (WT), 20 TG, and 24 KO mice over 35 days after experimental myocardial infarction with sham-operated controls (n=56). Of those, 4 WT-infarcted mice, 7 TG-infarcted mice, and 1 KO-infarcted mouse and no sham-operated mice died during the observation period. Among survivors, echocardiography, hemodynamic studies, and postmortem examination indicated that the WT and KO groups of infarcted mice mounted a hypertrophic response that was augmented in KO mice. The S100B-overexpressing TG group did not develop hypertrophy but demonstrated increased apoptosis. The postinfarct end-diastolic pressure was lower in KO mice than in WT mice, in accordance with other structural, hemodynamic, and functional parameters, which suggests that abrogation of S100B expression augmented hypertrophy, decreased apoptosis, and was beneficial to preservation of cardiac function within this time frame. CONCLUSIONS S100B regulates the hypertrophic response and remodeling in the early postinfarct period and represents a potential novel therapeutic target.
Collapse
Affiliation(s)
- James N Tsoporis
- Division of Cardiology, St Michael's Hospital, Toronto, Ontario, Canada
| | | | | | | | | | | |
Collapse
|
47
|
Haslbeck KM, Friess U, Schleicher ED, Bierhaus A, Nawroth PP, Kirchner A, Pauli E, Neundörfer B, Heuss D. The RAGE pathway in inflammatory myopathies and limb girdle muscular dystrophy. Acta Neuropathol 2005; 110:247-54. [PMID: 15986224 DOI: 10.1007/s00401-005-1043-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2004] [Revised: 05/05/2005] [Accepted: 05/17/2005] [Indexed: 10/25/2022]
Abstract
Oxidative stress and nuclear factor-kappaB (NF-kappaB) activation are linked to the pathogenesis of many metabolic, degenerative, and chronic inflammatory diseases. Activation of the receptor for advanced glycation end products (RAGE) by its specific ligand N(epsilon)-carboxymethyllysine (CML) results in the activation of NF-kappaB and the production of proinflammatory cytokines. To determine whether engagement of RAGE contributes to the pathogenesis of inflammatory myopathies, we performed immunohistochemical studies on the presence of CML-modified proteins, RAGE and activated NF-kappaB in muscle biopsies of patients with polymyositis (PM, n=10), dermatomyositis (DM, n=10), limb girdle muscular dystrophy (LGMD, n=10) and in 10 controls with normal muscle biopsy results. In inflammatory myopathies CML, RAGE and NF-kappaB were detected in mononuclear cells and in regenerating muscle fibers. CML, NF-kappaB and, to a lesser extent, RAGE were also found in degenerating muscle fibers, but colocalization of CML, RAGE and NF-kappaB was only seen in infiltrating mononuclear cells and regenerating muscle fibers. Immunofluorescence double labeling demonstrated an expression of CML, RAGE and NF-kappaB in CD4-, CD8-, CD22- and CD68-positive mononuclear cells. Western blot analysis showed an increased immunoreactivity for CML-modified proteins in PM and DM. In LGMD, CML, RAGE and NF-kappaB were found in regenerating muscle fibers and less frequently in degenerating muscle fibers, and with lower staining intensities than in inflammatory myopathies. Our data suggests that the CML-RAGE-NF-kappaB pathway is an evident proinflammatory pathomechanism in mononuclear effector cells in PM and DM. RAGE-mediated NF-kappaB activation may be involved in muscle fiber regeneration in inflammatory myopathies and LGMD.
Collapse
MESH Headings
- Adult
- Aged
- Dermatomyositis/immunology
- Dermatomyositis/metabolism
- Dermatomyositis/physiopathology
- Glycation End Products, Advanced
- Humans
- Immunohistochemistry
- Leukocytes, Mononuclear/immunology
- Leukocytes, Mononuclear/metabolism
- Lysine/analogs & derivatives
- Lysine/metabolism
- Middle Aged
- Muscle Fibers, Skeletal/metabolism
- Muscle Fibers, Skeletal/pathology
- Muscle, Skeletal/immunology
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/physiopathology
- Muscular Dystrophies, Limb-Girdle/immunology
- Muscular Dystrophies, Limb-Girdle/metabolism
- Muscular Dystrophies, Limb-Girdle/physiopathology
- Myositis/immunology
- Myositis/metabolism
- Myositis/physiopathology
- NF-kappa B/metabolism
- Polymyositis/immunology
- Polymyositis/metabolism
- Polymyositis/physiopathology
- Receptor for Advanced Glycation End Products
- Receptors, Immunologic/immunology
- Receptors, Immunologic/metabolism
- Regeneration/physiology
- Signal Transduction
- Transcriptional Activation/physiology
Collapse
Affiliation(s)
- K M Haslbeck
- Department of Neurology, University Erlangen-Nürnberg, Schwabachanlage 6, 91054, Erlangen, Germany,
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Bierhaus A, Humpert PM, Morcos M, Wendt T, Chavakis T, Arnold B, Stern DM, Nawroth PP. Understanding RAGE, the receptor for advanced glycation end products. J Mol Med (Berl) 2005; 83:876-86. [PMID: 16133426 DOI: 10.1007/s00109-005-0688-7] [Citation(s) in RCA: 946] [Impact Index Per Article: 47.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2005] [Accepted: 05/20/2005] [Indexed: 12/11/2022]
Abstract
Advanced glycation end products (AGEs), S100/calgranulins, HMGB1-proteins, amyloid-beta peptides, and the family of beta-sheet fibrils have been shown to contribute to a number of chronic diseases such as diabetes, amyloidoses, inflammatory conditions, and tumors by promoting cellular dysfunction via binding to cellular surface receptors. The receptor for AGEs (RAGE) is a multiligand receptor of the immunoglobulin superfamily of cell surface molecules acting as counter-receptor for these diverse molecules. Engagement of RAGE converts a brief pulse of cellular activation to sustained cellular dysfunction and tissue destruction. The involvement of RAGE in pathophysiologic processes has been demonstrated in murine models of chronic disease using either a receptor decoy such as soluble RAGE (sRAGE), RAGE neutralizing antibodies, or a dominant-negative form of the receptor. Studies with RAGE-/- mice confirmed that RAGE contributes, at least in part, to the development of late diabetic complications, such as neuropathy and nephropathy, macrovascular disease, and chronic inflammation. Furthermore, deletion of RAGE provided protection from the lethal effects of septic shock caused by cecal ligation and puncture (CLP). In contrast, deletion of RAGE had no effect on the host response in delayed-type hypersensitivity (DTH). Despite the lack of effect seen in adaptive immunity by the deletion of RAGE, administration of the receptor decoy, sRAGE, still afforded a protective effect in RAGE-/- mice. Thus, sRAGE is likely to sequester ligands, thereby preventing their interaction with other receptors in addition to RAGE. These data suggest that, just as RAGE is a multiligand receptor, its ligands are also likely to recognize several receptors in mediating their biologic effects.
Collapse
|
49
|
Emberley ED, Murphy LC, Watson PH. S100 proteins and their influence on pro-survival pathways in cancer. Biochem Cell Biol 2005; 82:508-15. [PMID: 15284904 DOI: 10.1139/o04-052] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The S100 gene family is composed of at least 20 members that share a common structure defined in part by the Ca2+ binding EF-hand motif. These genes which are expressed in a discriminate fashion in specific cells and tissues, have been described to have either an intracellular or extracellular function, or both. S100 proteins are implicated in the immune response, differentiation, cytoskeleton dynamics, enzyme activity, Ca2+ homeostasis and growth. A potential role for S100 proteins in neoplasia stems from these activities and from the observation that several S100 proteins have altered levels of expression in different stages and types of cancer. While the precise role and importance of S100 proteins in the development and promotion of cancer is poorly understood, it appears that the binding of Ca2+ is essential for exposing amino acid residues that are important in forming protein-protein interactions with effector molecules. The identity of some of these effector molecules has also now begun to emerge, and with this the elucidation of the signaling pathways that are modulated by these proteins. Some of these interactions are consistent with the diverse functions noted above. Others suggest that, many S100s may also promote cancer progression through specific roles in cell survival and apoptosis pathways. This review summarizes these findings and their implications.
Collapse
|
50
|
Most P, Boerries M, Eicher C, Schweda C, Völkers M, Wedel T, Söllner S, Katus HA, Remppis A, Aebi U, Koch WJ, Schoenenberger CA. Distinct subcellular location of the Ca2+-binding protein S100A1 differentially modulates Ca2+-cycling in ventricular rat cardiomyocytes. J Cell Sci 2005; 118:421-31. [PMID: 15654019 DOI: 10.1242/jcs.01614] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Calcium is a key regulator of cardiac function and is modulated through the Ca2+-sensor protein S100A1. S100 proteins are considered to exert both intracellular and extracellular functions on their target cells. Here we report the impact of an increased intracellular S100A1 protein level on Ca2+-homeostasis in neonatal ventricular cardiomyocytes in vitro. Specifically, we compare the effects of exogenously added recombinant S100A1 to those resulting from the overexpression of a transduced S100A1 gene. Extracellularly added S100A1 enhanced the Ca2+-transient amplitude in neonatal ventricular cardiomyocytes (NVCMs) through a marked decrease in intracellular diastolic Ca2+-concentrations ([Ca2+]i). The decrease in [Ca2+]i was independent of sarcoplasmic reticulum Ca2+-ATPase (SERCA2a) activity and was probably the result of an increased sarcolemmal Ca2+-extrusion through the sodium-calcium exchanger (NCX). At the same time the Ca2+-content of the sarcoplasmic reticulum (SR) decreased. These effects were dependent on the uptake of extracellularly added S100A1 protein and its subsequent routing to the endosomal compartment. Phospholipase C and protein kinase C, which are tightly associated with this subcellular compartment, were found to be activated by endocytosed S100A1.
By contrast, adenoviral-mediated intracellular S100A1 overexpression enhanced the Ca2+-transient amplitude in NVCMs mainly through an increase in systolic [Ca2+]i. The increased Ca2+-load in the SR was based on an enhanced SERCA2a activity while NCX function was unaltered. Overexpressed S100A1 colocalized with SERCA2a and other Ca2+-regulatory proteins at the SR, whereas recombinant S100A1 protein that had been endocytosed did not colocalize with SR proteins. This study provides the first evidence that intracellular S100A1, depending on its subcellular location, modulates cardiac Ca2+-turnover via different Ca2+-regulatory proteins.
Collapse
Affiliation(s)
- Patrick Most
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|