Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Int J Mol Sci ; 24(23)2023 Nov 21.
Article in English | MEDLINE | ID: mdl-38068900

ABSTRACT

S100B, a homodimeric Ca2+-binding protein, is produced and secreted by astrocytes, and its extracellular levels have been used as a glial marker in brain damage and neurodegenerative and psychiatric diseases; however, its mechanism of secretion is elusive. We used primary astrocyte cultures and calcium measurements from real-time fluorescence microscopy to investigate the role of intracellular calcium in S100B secretion. In addition, the dimethyl sulfoxide (DMSO) effect on S100B was investigated in vitro and in vivo using Wistar rats. We found that DMSO, a widely used vehicle in biological assays, is a powerful S100B secretagogue, which caused a biphasic response of Ca2+ mobilization. Our data show that astroglial S100B secretion is triggered by the increase in intracellular Ca2+ and indicate that this increase is due to Ca2+ mobilization from the endoplasmic reticulum. Also, blocking plasma membrane Ca2+ channels involved in the Ca2+ replenishment of internal stores decreased S100B secretion. The DMSO-induced S100B secretion was confirmed in vivo and in ex vivo hippocampal slices. Our data support a nonclassic vesicular export of S100B modulated by Ca2+, and the results might contribute to understanding the mechanism underlying the astroglial release of S100B.


Subject(s)
Astrocytes , Dimethyl Sulfoxide , Rats , Animals , Rats, Wistar , Dimethyl Sulfoxide/pharmacology , Dimethyl Sulfoxide/metabolism , Astrocytes/metabolism , Colforsin/pharmacology , Secretagogues/pharmacology , Calcium/metabolism , Nerve Growth Factors/metabolism , S100 Calcium Binding Protein beta Subunit/metabolism , Endoplasmic Reticulum/metabolism , Cells, Cultured
2.
Cell Mol Life Sci ; 77(1): 129-147, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31363816

ABSTRACT

Obesity is an endemic pathophysiological condition and a comorbidity associated with hypercholesterolemia, hypertension, cardiovascular disease, type 2 diabetes mellitus, and cancer. The adipose tissue of obese subjects shows hypertrophic adipocytes, adipocyte hyperplasia, and chronic low-grade inflammation. S100 proteins are Ca2+-binding proteins exclusively expressed in vertebrates in a cell-specific manner. They have been implicated in the regulation of a variety of functions acting as intracellular Ca2+ sensors transducing the Ca2+ signal and extracellular factors affecting cellular activity via ligation of a battery of membrane receptors. Certain S100 proteins, namely S100A4, the S100A8/S100A9 heterodimer and S100B, have been implicated in the pathophysiology of obesity-promoting macrophage-based inflammation via toll-like receptor 4 and/or receptor for advanced glycation end-products ligation. Also, serum levels of S100A4, S100A8/S100A9, S100A12, and S100B correlate with insulin resistance/type 2 diabetes, metabolic risk score, and fat cell size. Yet, secreted S100B appears to exert neurotrophic effects on sympathetic fibers in brown adipose tissue contributing to the larger sympathetic innervation of this latter relative to white adipose tissue. In the present review we first briefly introduce S100 proteins and then critically examine their role(s) in adipose tissue and obesity.


Subject(s)
Adipose Tissue/metabolism , Obesity/metabolism , S100 Proteins/metabolism , Adipose Tissue/physiopathology , Animals , Cytokines/analysis , Cytokines/metabolism , Humans , Inflammation/complications , Inflammation/metabolism , Inflammation/physiopathology , Macrophages/metabolism , Macrophages/pathology , Obesity/complications , Obesity/physiopathology , S100 Proteins/analysis
3.
Cell Mol Life Sci ; 77(18): 3547-3565, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32072237

ABSTRACT

Reductive stress is defined as a condition of sustained increase in cellular glutathione/glutathione disulfide and NADH/NAD+ ratios. Reductive stress is emerging as an important pathophysiological event in several diseased states, being as detrimental as is oxidative stress. Occurrence of reductive stress has been documented in several cardiomyopathies and is an important pathophysiological factor particularly in coronary artery disease and myocardial infarction. Excess activation of the transcription factor, Nrf2-the master regulator of the antioxidant response-, consequent in most cases to defective autophagy, can lead to reductive stress. In addition, hyperglycemia-induced activation of the polyol pathway can lead to increased NADH/NAD+ ratio, which might translate into increased levels of hydrogen sulfide-via enhanced activity of cystathionine ß-synthase-that would fuel reductive stress through inhibition of mitochondrial complex I. Reductive stress may be either a potential weapon against cancer priming tumor cells to apoptosis or a cancer's ally promoting tumor cell proliferation and making tumor cells resistant to reactive oxygen species-inducing drugs. In non-cancer pathological states reductive stress is definitely harmful paradoxically leading to reactive oxygen species overproduction via excess NADPH oxidase 4 activity. In face of the documented occurrence of reductive stress in several heart diseases, there is much less information about the occurrence and effects of reductive stress in skeletal muscle tissue. In the present review we describe relevant results emerged from studies of reductive stress in the heart and review skeletal muscle conditions in which reductive stress has been experimentally documented and those in which reductive stress might have an as yet unrecognized pathophysiological role. Establishing whether reductive stress has a (patho)physiological role in skeletal muscle will hopefully contribute to answer the question whether antioxidant supplementation to the general population, athletes, and a large cohort of patients (e.g. heart, sarcopenic, dystrophic, myopathic, cancer, and bronco-pulmonary patients) is harmless or detrimental.


Subject(s)
Muscle Cells/metabolism , Oxidative Stress , Antioxidants/pharmacology , Autophagy , Glutathione/metabolism , Humans , Hyperglycemia/metabolism , Hyperglycemia/pathology , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Sequestosome-1 Protein/metabolism
4.
Hum Mol Genet ; 27(21): 3734-3746, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30085099

ABSTRACT

Duchenne muscular dystrophy (DMD) is a lethal X-linked disease affecting striated muscles, which undergo progressive degeneration and chronic inflammation. Receptor for advanced glycation end-products (RAGE), a multiligand receptor involved in myogenesis and inflammation, is absent in healthy adult muscles but is re-expressed in myoblasts, regenerating myofibers and activated immune cells upon acute muscle injury, and in certain myopathies. We show here that RAGE is expressed and chronically stimulated in muscles of mdx mice, an experimental model of DMD, which also release high amounts of the RAGE ligands, HMGB1 and S100B. We generated a double mutant, mdx/Ager-/- mouse lacking dystrophin and RAGE. Compared to mdx mice, muscles of mdx/Ager-/- mice show restrained inflammation, unaffected fibrosis and higher muscle strength. Mdx/Ager-/- macrophages are less responsive to proinflammatory stimuli and express lower levels of Ccr2, Ccl2 and Ccl7, which are involved in monocyte/macrophage chemotaxis and migration. In vivo treatment of dystrophic muscles with a RAGE blocking antibody results in reduced necrosis and inflammatory infiltrate. Our results suggest that RAGE sustains muscle inflammation and necrosis in DMD muscles and that reducing RAGE activity might represent a potential therapeutic tool to counteract muscle inflammation and rescue muscle morphology in DMD conditions.


Subject(s)
Inflammation , Muscle Strength , Muscular Dystrophy, Duchenne/metabolism , Receptor for Advanced Glycation End Products/physiology , Animals , Disease Models, Animal , Dystrophin/genetics , Fibrosis , Male , Mice , Mice, Inbred mdx , Mice, Knockout , Muscular Dystrophy, Duchenne/physiopathology , Receptor for Advanced Glycation End Products/genetics , Receptor for Advanced Glycation End Products/metabolism
5.
Biochim Biophys Acta Mol Cell Res ; 1865(5): 721-733, 2018 May.
Article in English | MEDLINE | ID: mdl-29499228

ABSTRACT

Nrf2 and its endogenous inhibitor, Keap1, function as a ubiquitous, evolutionarily conserved intracellular defense mechanism to counteract oxidative stress. Sequestered by cytoplasmic Keap1 and targeted to proteasomal degradation in basal conditions, in case of oxidative stress Nrf2 detaches from Keap1 and translocates to the nucleus, where it heterodimerizes with one of the small Maf proteins. The heterodimers recognize the AREs, that are enhancer sequences present in the regulatory regions of Nrf2 target genes, essential for the recruitment of key factors for transcription. In the present review we briefly introduce the Nrf2-Keap1 system and describe Nrf2 functions, illustrate the Nrf2-NF-κB cross-talk, and highlight the effects of the Nrf2-Keap1 system in the physiology and pathophysiology of striated muscle tissue taking into account its role(s) in oxidative stress and reductive stress.


Subject(s)
Kelch-Like ECH-Associated Protein 1/genetics , NF-E2-Related Factor 2/genetics , Oxidative Stress/genetics , Cell Nucleus/genetics , Humans , Maf Transcription Factors/genetics , NF-kappa B/genetics , Oxidation-Reduction , Signal Transduction
6.
Cell Mol Life Sci ; 74(15): 2749-2760, 2017 08.
Article in English | MEDLINE | ID: mdl-28417162

ABSTRACT

S100A6 protein belongs to the A group of the S100 protein family of Ca2+-binding proteins. It is expressed in a limited number of cell types in adult normal tissues and in several tumor cell types. As an intracellular protein, S100A6 has been implicated in the regulation of several cellular functions, such as proliferation, apoptosis, the cytoskeleton dynamics, and the cellular response to different stress factors. S100A6 can be secreted/released by certain cell types which points to extracellular effects of the protein. RAGE (receptor for advanced glycation endproducts) and integrin ß1 transduce some extracellular S100A6's effects. Dosage of serum S100A6 might aid in diagnosis in oncology.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Proliferation , Neoplasms/metabolism , S100 Proteins/metabolism , Animals , Apoptosis , Cell Cycle Proteins/blood , Cell Cycle Proteins/genetics , Cell Movement , Cytoskeleton/genetics , Cytoskeleton/metabolism , Gene Expression Regulation , Humans , Integrin beta1/metabolism , Neoplasms/blood , Neoplasms/genetics , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Receptor for Advanced Glycation End Products/metabolism , S100 Calcium Binding Protein A6 , S100 Proteins/blood , S100 Proteins/genetics , Signal Transduction , Stem Cells/metabolism
7.
Int J Mol Sci ; 19(1)2018 Jan 22.
Article in English | MEDLINE | ID: mdl-29361745

ABSTRACT

Depending on the species, microglial cells represent 5-20% of glial cells in the adult brain. As the innate immune effector of the brain, microglia are involved in several functions: regulation of inflammation, synaptic connectivity, programmed cell death, wiring and circuitry formation, phagocytosis of cell debris, and synaptic pruning and sculpting of postnatal neural circuits. Moreover, microglia contribute to some neurodevelopmental disorders such as Nasu-Hakola disease (NHD), and to aged-associated neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), and others. There is evidence that human and rodent microglia may become senescent. This event determines alterations in the microglia activation status, associated with a chronic inflammation phenotype and with the loss of neuroprotective functions that lead to a greater susceptibility to the neurodegenerative diseases of aging. In the central nervous system (CNS), Triggering Receptor Expressed on Myeloid Cells 2-DNAX activation protein 12 (TREM2-DAP12) is a signaling complex expressed exclusively in microglia. As a microglial surface receptor, TREM2 interacts with DAP12 to initiate signal transduction pathways that promote microglial cell activation, phagocytosis, and microglial cell survival. Defective TREM2-DAP12 functions play a central role in the pathogenesis of several diseases. The CX3CL1 (fractalkine)-CX3CR1 signaling represents the most important communication channel between neurons and microglia. The expression of CX3CL1 in neurons and of its receptor CX3CR1 in microglia determines a specific interaction, playing fundamental roles in the regulation of the maturation and function of these cells. Here, we review the role of the TREM2-DAP12 and CX3CL1-CX3CR1 axes in aged microglia and the involvement of these pathways in physiological CNS aging and in age-associated neurodegenerative diseases.


Subject(s)
Aging/physiology , Microglia/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Brain/metabolism , Brain/pathology , CX3C Chemokine Receptor 1/metabolism , Central Nervous System/metabolism , Central Nervous System/pathology , Chemokine CX3CL1/metabolism , Gene Expression Regulation , Humans , Membrane Glycoproteins/metabolism , Membrane Proteins/metabolism , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Receptors, Immunologic/metabolism , Signal Transduction
8.
J Cell Sci ; 127(Pt 8): 1699-711, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24554430

ABSTRACT

Embryonal rhabdomyosarcomas (ERMSs) show elevated levels of PAX7, a transcription factor that marks quiescent adult muscle stem (satellite) cells and is important for proliferation and survival of activated satellite cells and whose timely repression is required for myogenic differentiation. However, the mechanism of PAX7 accumulation in ERMSs and whether high PAX7 causes uncontrolled proliferation in ERMS remains to be elucidated. The receptor for advanced glycation end-products (RAGE, encoded by AGER) transduces a myogenic and anti-proliferative signal in myoblasts, and stable transfection of the ERMS cell line TE671, which does not express RAGE, with AGER results in reduced proliferation and formation of tumor masses in vivo, and enhanced apoptosis and myogenic differentiation. Herein, we show that RAGE expression is low or absent in human ERMSs. We also show that in ERMS cells (1) PAX7 accumulates owing to absent or low RAGE signaling; (2) elevated PAX7 levels reduce RAGE expression and levels of MyoD and myogenin, muscle-specific transcription factors required for myoblast proliferation arrest and differentiation, respectively; (3) PAX7 supports myoblast proliferation by reducing the levels of MyoD, primarily by promoting its degradation; and (4), when ectopically expressed in ERMS cells, that RAGE upregulates myogenin which upregulates MyoD and downregulates PAX7, with consequent inhibition of proliferation and stimulation of differentiation. Thus, failure to express RAGE and, hence, MyoD and myogenin above a critical level in ERMS cells might result in deregulated PAX7 expression leading to uncontrolled proliferation and, potentially, to rhabdomyosarcomagenesis.


Subject(s)
Cell Proliferation , PAX7 Transcription Factor/metabolism , Receptor for Advanced Glycation End Products/metabolism , Rhabdomyosarcoma, Embryonal/metabolism , Animals , Carcinogenesis/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Mice , MyoD Protein/metabolism , Myoblasts/metabolism , Myogenin/metabolism , Rhabdomyosarcoma, Embryonal/pathology , Signal Transduction , Up-Regulation
9.
Carcinogenesis ; 36(9): 1071-83, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26153023

ABSTRACT

Rhabdomyosarcoma represents about 50% of soft-tissue sarcomas and 10% of malignant solid tumors in childhood. Embryonal rhabdomyosarcoma (ERMS) is the most frequent subtype, suggested to have an origin in muscle precursor cells that fail to exit the cell cycle and terminally differentiate mainly because of overexpression of the transcription factor, PAX7, which sustains proliferation, migration and invasiveness in ERMS cells. Artesunate (ARS) is a semi-synthetic derivative of artemisinin (ART), a natural compound well known as an antimalarial drug. However, ART and its derivatives have been found efficacious even as anticancer drugs that induce cell cycle arrest and/or apoptosis in several kinds of cancer. Here, we show that ARS dose-dependently induces DNA damage and apoptosis in ERMS cell lines. Production of reactive oxygen species (ROS) and activation of p38 MAPK have a central role in triggering ARS-mediated apoptosis in ERMS cells; indeed either the antioxidant, N-acetylcysteine or the p38 MAPK inhibitor, SB203580, protects ERMS cells from ARS-induced apoptosis. Moreover, ARS treatment in ERMS cells ROS-dependently induces the expression of the myo-miRs, miR-133a and miR-206, which are down-regulated in RMS, and reduces PAX7 protein levels. Finally, ARS upregulates the expression of the adhesion molecules, NCAM and integrin ß1, and reduces migration and invasiveness of ERMS cells in vitro, and ARS treatment reduces of about 50% the growth of ERMS xenografts in vivo. Our results are the first evidence of efficacy of ART derivatives in restraining ERMS growth in vivo, and suggest ARS as a potential candidate for therapeutic treatment of ERMS.


Subject(s)
Apoptosis/drug effects , Artemisinins/pharmacology , Reactive Oxygen Species/metabolism , Rhabdomyosarcoma, Embryonal/drug therapy , Rhabdomyosarcoma, Embryonal/pathology , p38 Mitogen-Activated Protein Kinases/metabolism , Acetylcysteine/pharmacology , Animals , Antineoplastic Agents/pharmacology , Apoptosis/physiology , Artemisinins/therapeutic use , Artesunate , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , DNA Damage/drug effects , Enzyme Activation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Imidazoles/pharmacology , Integrin beta1/biosynthesis , Mice , MicroRNAs/biosynthesis , Neoplasm Invasiveness , Neoplasm Transplantation , Neural Cell Adhesion Molecules/biosynthesis , PAX7 Transcription Factor/biosynthesis , Pyridines/pharmacology , Transplantation, Heterologous , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
10.
Carcinogenesis ; 35(10): 2382-92, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25123133

ABSTRACT

Rhabdomyosarcoma is a muscle-derived malignant tumor mainly affecting children. The most frequent variant, embryonal rhabdomyosarcoma (ERMS) is characterized by overexpression of the transcription factor, PAX7 which prevents ERMS cells from exiting the cell cycle and terminally differentiating. However, a role for PAX7 in the invasive properties of ERMS cells has not been investigated in detail thus far. Here we show that ectopic expression of receptor for advanced glycation end-products (RAGE) in human ERMS cells results in the activation of a RAGE/myogenin axis which downregulates PAX7 by transcriptional and post-translational mechanisms, as in normal myoblasts, and reduces metastasis formation. High PAX7 sustains migration and invasiveness in ERMS cells by upregulating EPHA3 and EFNA1 and downregulating NCAM1 thus decreasing the neural cell adhesion molecule (NCAM)/polysialylated-NCAM ratio. Microarray gene expression analysis shows that compared with the RAGE(-ve) TE671/WT cells and similarly to primary human myoblasts, TE671/RAGE cells show upregulation of genes involved in muscle differentiation and cell adhesion, and downregulation of cell migration related and major histocompatibility complex class I genes. Our data reveal a link between PAX7 and metastasis occurrence in ERMSs, and support a role for the RAGE/myogenin axis in metastasis suppression. Thus, low RAGE expression in ERMS primary tumors may be predictive of metastatic behavior.


Subject(s)
PAX7 Transcription Factor/metabolism , Receptors, Immunologic/metabolism , Rhabdomyosarcoma, Embryonal/metabolism , Rhabdomyosarcoma, Embryonal/pathology , Animals , CD56 Antigen/genetics , Cell Line, Tumor/drug effects , Cell Movement/genetics , Ephrin-A1/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Leupeptins/pharmacology , Mice , Mice, Mutant Strains , Mice, Nude , Myoblasts/pathology , Myogenin/metabolism , PAX7 Transcription Factor/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptor for Advanced Glycation End Products , Receptor, EphA3 , Receptors, Immunologic/genetics , Rhabdomyosarcoma, Embryonal/drug therapy , Rhabdomyosarcoma, Embryonal/genetics , Xenograft Model Antitumor Assays
11.
Biochim Biophys Acta ; 1833(1): 101-9, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23103427

ABSTRACT

RAGE (receptor for advanced glycation end-products) is a multiligand receptor of the immunoglobulin superfamily involved in inflammation, diabetes, atherosclerosis, nephropathy, neurodegeneration, and cancer. Advanced glycation end-products, high mobility group box-1 (amphoterin), ß-amyloid fibrils, certain S100 proteins, and DNA and RNA are RAGE ligands. Upon RAGE ligation, adaptor proteins (i.e., diaphanous-1, TIRAP, MyD88 and/or other as yet unidentified adaptors) associate with RAGE cytoplasmic domain resulting in signaling. However, RAGE activation may not be restricted to pathological statuses, the receptor being involved in tissue homeostasis and regeneration/repair upon acute injury, and in resolution of inflammation. RAGE effects are strongly dependent on the cell type and the context, which may condition therapeutic strategies aimed at reducing RAGE signaling.


Subject(s)
Homeostasis/genetics , Receptors, Immunologic/physiology , Regeneration/genetics , Wound Healing/genetics , Animals , Glycation End Products, Advanced/metabolism , Glycation End Products, Advanced/pharmacology , Glycation End Products, Advanced/physiology , Homeostasis/drug effects , Homeostasis/physiology , Humans , Models, Biological , Receptor for Advanced Glycation End Products , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Regeneration/drug effects , Regeneration/physiology , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/physiology , Wound Healing/drug effects , Wound Healing/physiology
12.
J Cell Sci ; 125(Pt 6): 1440-54, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22328527

ABSTRACT

Expression of the paired-box 7 (PAX7) transcription factor is regulated during both myoblast proliferation and differentiation: high levels of PAX7 compromise myogenic differentiation because of excess and prolonged proliferation, whereas low levels of PAX7 result in precocious differentiation. We showed that myogenin repressed Pax7 transcription in differentiating myoblasts by binding to specific recognition sites in the Pax7 promoter, and that high-mobility group box 1 (HMGB1)-receptor for advanced glycation end-products (RAGE) signaling was required for myogenin induction and myogenin-dependent repression of Pax7 transcription. In addition, PAX7 negatively and myogenin positively regulated RAGE expression. RAGE, a multiligand receptor of the immunoglobulin superfamily, was not expressed in adult skeletal muscles, and was transiently expressed in activated, proliferating and differentiating satellite cells (SCs) in injured muscles. Compared with wild-type muscles, Rage(-/-) muscles exhibited increased numbers of basal SCs that were further increased in injured Rage(-/-) muscles following elevated myoblast asymmetric division; complete regeneration of injured Rage(-/-) muscles was found to be delayed by ~1 week. Thus, RAGE signaling physiologically repressed Pax7 transcription in SCs by upregulating myogenin, thereby accelerating muscle regeneration and limiting SC self-renewal.


Subject(s)
HMGB1 Protein/physiology , Homeostasis/physiology , Myogenin/physiology , PAX7 Transcription Factor/genetics , Receptors, Immunologic/genetics , Satellite Cells, Skeletal Muscle/metabolism , Animals , Mice , Mice, Inbred C57BL , Mice, Knockout , Myoblasts/cytology , Myoblasts/metabolism , PAX7 Transcription Factor/biosynthesis , Primary Cell Culture , Receptor for Advanced Glycation End Products , Receptors, Immunologic/deficiency , Regulatory Elements, Transcriptional/physiology , Repressor Proteins/physiology , Satellite Cells, Skeletal Muscle/cytology , p38 Mitogen-Activated Protein Kinases/physiology
13.
Am J Respir Crit Care Med ; 188(11): 1338-50, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24127697

ABSTRACT

RATIONALE: Hypoxia regulates the inflammatory-antiinflammatory balance by the receptor for advanced glycation end products (RAGE), a versatile sensor of damage-associated molecular patterns. The multiligand nature of RAGE places this receptor in the midst of chronic inflammatory diseases. OBJECTIVES: To characterize the impact of the hypoxia-RAGE pathway on pathogenic airway inflammation preventing effective pathogen clearance in cystic fibrosis (CF) and elucidate the potential role of this danger signal in pathogenesis and therapy of lung inflammation. METHODS: We used in vivo and in vitro models to study the impact of hypoxia on RAGE expression and activity in human and murine CF, the nature of the RAGE ligand, and the impact of RAGE on lung inflammation and antimicrobial resistance in fungal and bacterial pneumonia. MEASUREMENTS AND MAIN RESULTS: Sustained expression of RAGE and its ligand S100B was observed in murine lung and human epithelial cells and exerted a proximal role in promoting inflammation in murine and human CF, as revealed by functional studies and analysis of the genetic variability of AGER in patients with CF. Both hypoxia and infections contributed to the sustained activation of the S100B-RAGE pathway, being RAGE up-regulated by hypoxia and S100B by infection by Toll-like receptors. Inhibiting the RAGE pathway in vivo with soluble (s) RAGE reduced pathogen load and inflammation in experimental CF, whereas sRAGE production was defective in patients with CF. CONCLUSIONS: A causal link between hyperactivation of RAGE and inflammation in CF has been observed, such that targeting pathogenic inflammation alleviated inflammation in CF and measurement of sRAGE levels could be a useful biomarker for RAGE-dependent inflammation in patients with CF.


Subject(s)
Cystic Fibrosis/pathology , Hypoxia/pathology , Inflammation Mediators/physiology , Pneumonia/etiology , Receptors, Immunologic/immunology , Animals , Aspergillosis/microbiology , Biomarkers , Blotting, Western , Cystic Fibrosis/complications , Cystic Fibrosis/microbiology , Drug Resistance, Microbial , Enzyme-Linked Immunosorbent Assay , Female , Humans , Hypoxia/complications , Hypoxia/etiology , Italy , Male , Mice , Mice, Inbred C57BL , Middle Aged , Pneumonia/drug therapy , Pneumonia/microbiology , Pseudomonas Infections/microbiology , Receptor for Advanced Glycation End Products , Respiratory Mucosa , Tissue Culture Techniques , Up-Regulation
14.
J Cell Sci ; 124(Pt 14): 2389-400, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21693575

ABSTRACT

S100B protein has been shown to exert anti-myogenic and mitogenic effects in myoblast cultures through inhibition of the myogenic p38 MAPK and activation of the mitogenic ERK1/2. However, the receptor mediating these effects had not been identified. Here, we show that S100B increases and/or stabilizes the binding of basic fibroblast growth factor (bFGF) to bFGF receptor 1 (FGFR1) by interacting with bFGF, thereby enhancing FGFR1 activation and the mitogenic and anti-myogenic effects of FGFR1. S100B also binds to its canonical receptor RAGE (receptor for advanced glycation end-products), a multi-ligand receptor previously shown to transduce a pro-myogenic signal when activated by HMGB1, and recruits RAGE into a RAGE-S100B-bFGF-FGFR1 complex. However, when bound to S100B-bFGF-FGFR1, RAGE can no longer stimulate myogenic differentiation, whereas in the absence of either bFGF or FGFR1, binding of S100B to RAGE results in stimulation of RAGE anti-mitogenic and promyogenic signaling. An S100B-bFGF-FGFR1 complex also forms in Rage(-/-) myoblasts, leading to enhanced proliferation and reduced differentiation, which points to a dispensability of RAGE for the inhibitory effects of S100B on myoblasts under the present experimental conditions. These results reveal a new S100B-interacting protein - bFGF - in the extracellular milieu and suggest that S100B stimulates myoblast proliferation and inhibits myogenic differentiation by activating FGFR1 in a bFGF-dependent manner.


Subject(s)
Fibroblast Growth Factor 2/metabolism , Myoblasts/cytology , Myoblasts/metabolism , Nerve Growth Factors/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , S100 Proteins/metabolism , Animals , Cell Differentiation/physiology , Cell Growth Processes/physiology , Cell Line , Humans , Mice , Mice, Inbred C57BL , Phosphorylation , Receptor for Advanced Glycation End Products , Receptors, Immunologic/metabolism , S100 Calcium Binding Protein beta Subunit , Signal Transduction , Up-Regulation
15.
PLoS Pathog ; 7(3): e1001315, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21423669

ABSTRACT

Humans inhale hundreds of Aspergillus conidia without adverse consequences. Powerful protective mechanisms may ensure prompt control of the pathogen and inflammation. Here we reveal a previously unknown mechanism by which the danger molecule S100B integrates pathogen- and danger-sensing pathways to restrain inflammation. Upon forming complexes with TLR2 ligands, S100B inhibited TLR2 via RAGE, through a paracrine epithelial cells/neutrophil circuit that restrained pathogen-induced inflammation. However, upon binding to nucleic acids, S100B activated intracellular TLRs eventually resolve danger-induced inflammation via transcriptional inhibition of S100B. Thus, the spatiotemporal regulation of TLRs and RAGE by S100B provides evidence for an evolving braking circuit in infection whereby an endogenous danger protects against pathogen-induced inflammation and a pathogen-sensing mechanism resolves danger-induced inflammation.


Subject(s)
Aspergillus/physiology , Host-Pathogen Interactions/physiology , Nerve Growth Factors/metabolism , Receptors, Immunologic/antagonists & inhibitors , S100 Proteins/metabolism , Toll-Like Receptor 2/antagonists & inhibitors , Animals , Aspergillus/pathogenicity , Disease Models, Animal , Lung/metabolism , Lung/microbiology , Mice , Mice, Knockout , Pulmonary Aspergillosis/metabolism , Pulmonary Aspergillosis/microbiology , Receptor for Advanced Glycation End Products , Receptors, Immunologic/deficiency , Receptors, Immunologic/metabolism , S100 Calcium Binding Protein beta Subunit , Toll-Like Receptor 2/metabolism
16.
J Biol Chem ; 286(9): 7214-26, 2011 Mar 04.
Article in English | MEDLINE | ID: mdl-21209080

ABSTRACT

The Ca(2+)-binding protein of the EF-hand type, S100B, is abundantly expressed in and secreted by astrocytes, and release of S100B from damaged astrocytes occurs during the course of acute and chronic brain disorders. Thus, the concept has emerged that S100B might act an unconventional cytokine or a damage-associated molecular pattern protein playing a role in the pathophysiology of neurodegenerative disorders and inflammatory brain diseases. S100B proinflammatory effects require relatively high concentrations of the protein, whereas at physiological concentrations S100B exerts trophic effects on neurons. Most if not all of the extracellular (trophic and toxic) effects of S100B in the brain are mediated by the engagement of RAGE (receptor for advanced glycation end products). We show here that high S100B stimulates murine microglia migration in Boyden chambers via RAGE-dependent activation of Src kinase, Ras, PI3K, MEK/ERK1/2, RhoA/ROCK, Rac1/JNK/AP-1, Rac1/NF-κB, and, to a lesser extent, p38 MAPK. Recruitment of the adaptor protein, diaphanous-1, a member of the formin protein family, is also required for S100B/RAGE-induced migration of microglia. The S100B/RAGE-dependent activation of diaphanous-1/Rac1/JNK/AP-1, Ras/Rac1/NF-κB and Src/Ras/PI3K/RhoA/diaphanous-1 results in the up-regulation of expression of the chemokines, CCL3, CCL5, and CXCL12, whose release and activity are required for S100B to stimulate microglia migration. Lastly, RAGE engagement by S100B in microglia results in up-regulation of the chemokine receptors, CCR1 and CCR5. These results suggests that S100B might participate in the pathophysiology of brain inflammatory disorders via RAGE-dependent regulation of several inflammation-related events including activation and migration of microglia.


Subject(s)
Cell Movement/immunology , Chemokines/metabolism , Microglia , Nerve Growth Factors/metabolism , Receptors, Immunologic/metabolism , S100 Proteins/metabolism , Animals , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/immunology , Calcium-Binding Proteins/metabolism , Carrier Proteins/metabolism , Cattle , Cell Line , Chemokine CCL3/genetics , Chemokine CCL3/immunology , Chemokine CCL3/metabolism , Chemokine CCL5/genetics , Chemokine CCL5/immunology , Chemokine CCL5/metabolism , Chemokine CXCL12/genetics , Chemokine CXCL12/immunology , Chemokine CXCL12/metabolism , Chemokines/genetics , Chemokines/immunology , Cytoskeleton/metabolism , Encephalitis/immunology , Encephalitis/metabolism , Formins , Mice , Mice, Inbred C57BL , Microglia/cytology , Microglia/immunology , Microglia/metabolism , Nerve Growth Factors/genetics , Nerve Growth Factors/immunology , Rats , Receptor for Advanced Glycation End Products , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , S100 Calcium Binding Protein beta Subunit , S100 Proteins/genetics , S100 Proteins/immunology , Up-Regulation/immunology
17.
Biochim Biophys Acta ; 1813(5): 1092-104, 2011 May.
Article in English | MEDLINE | ID: mdl-21130124

ABSTRACT

S100B protein activates IKKß/NF-κB within myoblasts, thereby inhibiting the expression of MyoD and the MyoD-downstream effectors, myogenin and p21(WAF1), and myoblast differentiation. Herein we show that myoblasts downregulate S100B expression once transferred from proliferation medium to differentiation medium via a p38 MAPK-driven transcriptional mechanism as well as a post-translational, proteasome-dependent mechanism, and that myoblasts that have not been committed to differentiation resume expressing S100B once transferred back to proliferation medium. Likewise, myoblasts downregulate S100B expression once transferred to quiescence medium, and interference with S100B downregulation as obtained by stable overexpression of the protein results in reduced acquisition of quiescence and a faster proliferation upon transfer of the cells from quiescence medium to proliferation medium, compared to controls. These latter effects are dependent on S100B-induced activation of JNK. Moreover, S100B reduces myoblast apoptosis in an MEK-ERK1/2, Akt, JNK, and NF-κB-dependent manner. However, myogenin(+) myoblasts (i.e., myocytes) and myotubes abundantly express S100B likely induced by myogenin. Our results suggest that (1) a timely repression of S100B expression is required for efficient myogenic differentiation; (2) S100B plays an important role in the expansion of the activated (i.e., proliferating) myoblast population; (3) under conditions associated with enhanced expression of S100B, the transition from proliferation to quiescence and from quiescence to proliferation might be altered; and (4) S100B exerts different regulatory effects in myoblasts and myocytes/myotubes/myofibers. This article is part of a Special Issue entitled: 11th European Symposium on Calcium.


Subject(s)
Apoptosis , Cell Differentiation , Myoblasts/cytology , Myoblasts/metabolism , Nerve Growth Factors/metabolism , S100 Proteins/metabolism , Animals , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Culture Media/pharmacology , Cytoprotection/drug effects , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Hydrogen Peroxide/pharmacology , Intracellular Space/drug effects , Intracellular Space/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Models, Biological , Myoblasts/drug effects , Myoblasts/enzymology , Myogenin/metabolism , NF-kappa B/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , S100 Calcium Binding Protein beta Subunit
18.
J Biol Chem ; 285(14): 10385-96, 2010 Apr 02.
Article in English | MEDLINE | ID: mdl-20124407

ABSTRACT

Myogenesis is a process whereby myoblasts differentiate and fuse into multinucleated myotubes, the precursors of myofibers. Various signals and factors modulate this process, and glucocorticoids (GCs) are important regulators of skeletal muscle metabolism. We show that glucocorticoid-induced leucine zipper (GILZ), a GC-induced gene, and the newly identified isoform long GILZ (L-GILZ) are expressed in skeletal muscle tissue and in C2C12 myoblasts where GILZ/L-GILZ maximum expression occurs during the first few days in differentiation medium. Moreover, we observed that GC treatment of myoblasts, which increased GILZ/L-GILZ expression, resulted in reduced myotube formation, whereas GILZ and L-GILZ silencing dampened GC effects. Inhibition of differentiation caused by GILZ/L-GILZ overexpression correlated with inhibition of MyoD function and reduced expression of myogenin. Notably, results indicate that GILZ and L-GILZ bind and regulate MyoD/HDAC1 transcriptional activity, thus mediating the anti-myogenic effect of GCs.


Subject(s)
Cell Differentiation/drug effects , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Muscle Development/drug effects , Muscle, Skeletal/cytology , Myoblasts/cytology , Transcription Factors/metabolism , Alternative Splicing , Animals , Animals, Newborn , Blotting, Western , Cells, Cultured , Chromatin Immunoprecipitation , Electrophoretic Mobility Shift Assay , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Gene Expression Regulation, Developmental , Immunoenzyme Techniques , Immunoprecipitation , Luciferases/metabolism , Mice , Mice, Inbred C57BL , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , MyoD Protein/genetics , MyoD Protein/metabolism , Myoblasts/drug effects , Myoblasts/metabolism , Myogenin/metabolism , Promoter Regions, Genetic/genetics , Protein Isoforms , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcription, Genetic , Transfection
19.
Biomolecules ; 11(6)2021 06 12.
Article in English | MEDLINE | ID: mdl-34204735

ABSTRACT

The receptor for advanced glycation-end products (RAGE) is a multiligand receptor with a role in inflammatory and pulmonary pathologies. Hyperactivation of RAGE by its ligands has been reported to sustain inflammation and oxidative stress in common comorbidities of severe COVID-19. RAGE is essential to the deleterious effects of the renin-angiotensin system (RAS), which participates in infection and multiorgan injury in COVID-19 patients. Thus, RAGE might be a major player in severe COVID-19, and appears to be a useful therapeutic molecular target in infections by SARS-CoV-2. The role of RAGE gene polymorphisms in predisposing patients to severe COVID-19 is discussed. .


Subject(s)
COVID-19/metabolism , Inflammation/metabolism , Oxidative Stress , Receptor for Advanced Glycation End Products/metabolism , Renin-Angiotensin System , Animals , COVID-19/genetics , COVID-19/pathology , Humans , Inflammation/genetics , Inflammation/pathology , Polymorphism, Genetic , Receptor for Advanced Glycation End Products/genetics , Risk Factors , SARS-CoV-2/physiology , Severity of Illness Index
20.
Life Sci ; 272: 119251, 2021 May 01.
Article in English | MEDLINE | ID: mdl-33636175

ABSTRACT

A novel infectious disease (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), was detected in December 2019 and declared as a global pandemic by the World Health. Approximately 15% of patients with COVID-19 progress to severe pneumonia and eventually develop acute respiratory distress syndrome (ARDS), septic shock and/or multiple organ failure with high morbidity and mortality. Evidence points towards a determinant pathogenic role of members of the renin-angiotensin system (RAS) in mediating the susceptibility, infection, inflammatory response and parenchymal injury in lungs and other organs of COVID-19 patients. The receptor for advanced glycation end-products (RAGE), a member of the immunoglobulin superfamily, has important roles in pulmonary pathological states, including fibrosis, pneumonia and ARDS. RAGE overexpression/hyperactivation is essential to the deleterious effects of RAS in several pathological processes, including hypertension, chronic kidney and cardiovascular diseases, and diabetes, all of which are major comorbidities of SARS-CoV-2 infection. We propose RAGE as an additional molecular target in COVID-19 patients for ameliorating the multi-organ pathology induced by the virus and improving survival, also in the perspective of future infections by other coronaviruses.


Subject(s)
COVID-19/complications , Drug Discovery , Multiple Organ Failure/etiology , Multiple Organ Failure/prevention & control , Receptor for Advanced Glycation End Products/antagonists & inhibitors , SARS-CoV-2/physiology , Animals , COVID-19/metabolism , COVID-19/pathology , Humans , Molecular Targeted Therapy , Multiple Organ Failure/metabolism , Multiple Organ Failure/pathology , Receptor for Advanced Glycation End Products/metabolism , Renin-Angiotensin System/drug effects , SARS-CoV-2/drug effects , Signal Transduction/drug effects , COVID-19 Drug Treatment
SELECTION OF CITATIONS
SEARCH DETAIL