Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
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
2.
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
3.
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
4.
J Cell Mol Med ; 22(5): 2865-2883, 2018 05.
Article in English | MEDLINE | ID: mdl-29504694

ABSTRACT

Metastasis is the primary cause of death in prostate cancer (PCa) patients. Effective therapeutic intervention in metastatic PCa is undermined by our poor understanding of its molecular aetiology. Defining the mechanisms underlying PCa metastasis may lead to insights into how to decrease morbidity and mortality in this disease. Glyoxalase 1 (Glo1) is the detoxification enzyme of methylglyoxal (MG), a potent precursor of advanced glycation end products (AGEs). Hydroimidazolone (MG-H1) and argpyrimidine (AP) are AGEs originating from MG-mediated post-translational modification of proteins at arginine residues. AP is involved in the control of epithelial to mesenchymal transition (EMT), a crucial determinant of cancer metastasis and invasion, whose regulation mechanisms in malignant cells are still emerging. Here, we uncover a novel mechanism linking Glo1 to the maintenance of the metastatic phenotype of PCa cells by controlling EMT by engaging the tumour suppressor miR-101, MG-H1-AP and TGF-ß1/Smad signalling. Moreover, circulating levels of Glo1, miR-101, MG-H1-AP and TGF-ß1 in patients with metastatic compared with non-metastatic PCa support our in vitro results, demonstrating their clinical relevance. We suggest that Glo1, together with miR-101, might be potential therapeutic targets for metastatic PCa, possibly by metformin administration.


Subject(s)
Epithelial-Mesenchymal Transition , Lactoylglutathione Lyase/metabolism , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , 3' Untranslated Regions/genetics , Aged , Base Sequence , Cell Line, Tumor , Cell Movement , Gene Expression Regulation, Neoplastic/drug effects , Homoarginine/analogs & derivatives , Homoarginine/blood , Homoarginine/metabolism , Humans , Imidazoles/blood , Imidazoles/metabolism , Lactoylglutathione Lyase/blood , Male , Metformin/pharmacology , MicroRNAs/blood , MicroRNAs/metabolism , Middle Aged , Models, Biological , Neoplasm Invasiveness , Neoplasm Metastasis , Ornithine/analogs & derivatives , Ornithine/blood , Ornithine/metabolism , Phenotype , Prostatic Neoplasms/blood , Prostatic Neoplasms/genetics , Pyrimidines/blood , Pyrimidines/metabolism , Signal Transduction , Smad Proteins/metabolism , Thiolester Hydrolases/metabolism , Transforming Growth Factor beta1/blood , Transforming Growth Factor beta1/metabolism
5.
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
6.
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
7.
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
8.
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
9.
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
10.
Eur J Transl Myol ; 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38651523

ABSTRACT

The 2023 represented a milestone for the Interuniversity Institute of Myology (IIM) since it marked twenty years of IIM activity joined with the 20th annual meeting organized by the association. The 20th IIM meeting took place in the fascinating town of Assisi, in the heart of central Italy, from 12 to 15 October. The commemorative 20th edition of the meeting represented a success in terms of participation and contributions as it brought together 160 myologists, clinicians, pharmaceutical companies, and patient organization representatives from Italy, several European countries (especially France), the United Kingdom, Brazil, and the USA. Four main scientific sessions hosted 36 oral communications and 54 always-on-display posters reporting original and unpublished results. Four main lectures from internationally renowned invited speakers and talks from delegates of the Societé Française de Myologie gave particular interest and emphasis to the scientific discussion. In line with the traditional policy of the IIM to encourage the participation of young researchers, about 50% of the attendees were under 35 years old. Moreover, the 20th IIM meeting was part of the high-training course in "Advanced Myology Update 2023", reserved to young trainees and managed by the University of Perugia (Italy) in collaboration with the IIM. In addition to the meeting scientific sessions, the 29 attendees to the course had a dedicated round table and dedicated lessons with the IIM invited speakers as teachers. Awards for the best talk, best poster blitz, and best poster have been conferred to young attendees, who became part of the IIM Young Committee, involved in the scientific organization of the IIM meetings. To celebrate the 20th IIM anniversary, a special free-access educational convention on "Causes and mechanisms of muscle atrophy. From terrestrial disuse to Space flights" has been organized, in which IIM experts in the field have illustrated the current knowledge about the muscle atrophy process in several atrophying conditions, and the former Italian astronaut, Paolo Nespoli shared his incredible experience in Space fascinating the large audience attending both in presence and online live stream. The meeting was characterized by a vibrant, friendly, and inclusive atmosphere, and stimulated discussion on emerging areas of muscle research, fostering international collaborations, and confirming the IIM meeting as an ideal venue to discuss around muscle development, function, and diseases pointing to the development of efficacious therapeutic strategies. Here, the abstracts of the meeting illustrate the most recent results on basic, translational, and clinical research in the myology field. Some abstracts are missing as per authors' decision due to the patentability of the results.

11.
Cells ; 13(6)2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38534388

ABSTRACT

The Sertoli cells (SeCs) of the seminiferous tubules secrete a multitude of immunoregulatory and trophic factors to provide immune protection and assist in the orderly development of germ cells. Grafts of naked or encapsulated SeCs have been proved to represent an interesting therapeutic option in a plethora of experimental models of diseases. However, whether SeCs have immunosuppressive or immunomodulatory effects, which is imperative for their clinical translatability, has not been demonstrated. We directly assessed the immunopotential of intraperitoneally grafted microencapsulated porcine SeCs (MC-SeCs) in murine models of fungal infection (Aspergillus fumigatus or Candida albicans) or cancer (Lewis lung carcinoma/LLC or B16 melanoma cells). We found that MC-SeCs (i) provide antifungal resistance with minimum inflammatory pathology through the activation of the tolerogenic aryl hydrocarbon receptor/indoleamine 2,3-dioxygenase pathway; (ii) do not affect tumor growth in vivo; and (iii) reduce the LLC cell metastatic cancer spread associated with restricted Vegfr2 expression in primary tumors. Our results point to the fine immunoregulation of SeCs in the relative absence of overt immunosuppression in both infection and cancer conditions, providing additional support for the potential therapeutic use of SeC grafts in human patients.


Subject(s)
Carcinoma, Lewis Lung , Sertoli Cells , Male , Humans , Swine , Animals , Mice , Sertoli Cells/metabolism , Seminiferous Tubules/metabolism , Carcinoma, Lewis Lung/metabolism , Immunosuppressive Agents/therapeutic use , Immune Tolerance
12.
Biomed Pharmacother ; 174: 116517, 2024 May.
Article in English | MEDLINE | ID: mdl-38574619

ABSTRACT

Age-associated osteosarcopenia is an unresolved syndrome characterized by the concomitant loss of bone (osteopenia) and skeletal muscle (sarcopenia) tissues increasing falls, immobility, morbidity, and mortality. Unbalanced resorption of bone in the remodeling process and excessive protein breakdown, especially fast type II myosin heavy chain (MyHC-II) isoform and myofiber metabolic shift, are the leading causes of bone and muscle deterioration in the elderly, respectively. Equisetum arvense (EQ) is a plant traditionally recommended for many pathological conditions due to its anti-inflammatory properties. Thus, considering that a chronic low-grade inflammatory state predisposes to both osteoporosis and sarcopenia, we tested a standardized hydroalcoholic extract of EQ in in vitro models of muscle atrophy [C2C12 myotubes treated with proinflammatory cytokines (TNFα/IFNγ), excess glucocorticoids (dexamethasone), or the osteokine, receptor activator of nuclear factor kappa-B ligand (RANKL)] and osteoclastogenesis (RAW 264.7 cells treated with RANKL). We found that EQ counteracted myotube atrophy, blunting the activity of several pathways depending on the applied stimulus, and reduced osteoclast formation and activity. By in silico target fishing, IKKB-dependent nuclear factor kappa-B (NF-κB) inhibition emerges as a potential common mechanism underlying EQ's anti-atrophic effects. Consumption of EQ (500 mg/kg/day) by pre-geriatric C57BL/6 mice for 3 months translated into: i) maintenance of muscle mass and performance; ii) restrained myofiber oxidative shift; iii) slowed down age-related modifications in osteoporotic bone, significantly preserving trabecular connectivity density; iv) reduced muscle- and spleen-related inflammation. EQ can preserve muscle functionality and bone remodeling during aging, potentially valuable as a natural treatment for osteosarcopenia.


Subject(s)
Equisetum , Plant Extracts , Sarcopenia , Animals , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Mice , Sarcopenia/drug therapy , Sarcopenia/pathology , RAW 264.7 Cells , Equisetum/chemistry , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/pathology , Muscle Fibers, Skeletal/metabolism , Aging/drug effects , Aging/pathology , Muscular Atrophy/drug therapy , Muscular Atrophy/pathology , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteoclasts/pathology , RANK Ligand/metabolism , NF-kappa B/metabolism , Osteogenesis/drug effects , Anti-Inflammatory Agents/pharmacology
13.
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
14.
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
15.
Data Brief ; 40: 107744, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35141363

ABSTRACT

Sertoli cells (SeC) isolated from porcine testes have shown direct effects on muscle precursor cells sustaining C2C12 myoblasts proliferation and inhibiting oxidative stress and apoptosis in the early phase of the differentiation process, and stimulating myoblast fusion into myotubes and the expression of markers of myogenic differentiation in the late phase. This suggested that the cocktail of factors secreted by SeC stimulates proliferation in myoblasts without weakening their myogenic potential resulting in the formation of the critical myoblast amount necessary to rebuild the required muscle mass upon a damage. Here, we show that co-culturing C2C12 myoblasts with high doses of SeC microencapsulated in clinical grade alginate-based microcapsules (MC-SeC) for three days in differentiation medium (DM) translates into increased cell numbers and almost absence of myotube formation. However, after removal of MC-SeC, an intense fusion activity into myotubes was observed culminating in a fusion index similar to that of control after additional three days of culture in DM. These data definitely demonstrate that SeC-derived factors preserve the myogenic potential while sustaining cell proliferation in C2C12 myoblasts.

16.
Nutrients ; 14(15)2022 Jul 25.
Article in English | MEDLINE | ID: mdl-35893905

ABSTRACT

The imbalance in osteoblast (OB)-dependent bone formation in favor of osteoclast (OC)-dependent bone resorption is the main cause of loss of tissue mineral mass during bone remodeling leading to osteoporosis conditions. Thus, the suppression of OC activity together with the improvement in the OB activity has been proposed as an effective therapy for maintaining bone mass during aging. We tested the new dietary product, KYMASIN UP containing standardized Withania somnifera, Silybum marianum and Trigonella foenum-graecum herbal extracts or the single extracts in in vitro models mimicking osteoclastogenesis (i.e., RAW 264.7 cells treated with RANKL, receptor activator of nuclear factor kappa-Β ligand) and OB differentiation (i.e., C2C12 myoblasts treated with BMP2, bone morphogenetic protein 2). We found that the dietary product reduces RANKL-dependent TRAP (tartrate-resistant acid phosphatase)-positive cells (i.e., OCs) formation and TRAP activity, and down-regulates osteoclastogenic markers by reducing Src (non-receptor tyrosine kinase) and p38 MAPK (mitogen-activated protein kinase) activation. Withania somnifera appears as the main extract responsible for the anti-osteoclastogenic effect of the product. Moreover, KYMASIN UP maintains a physiological release of the soluble decoy receptor for RANKL, OPG (osteoprotegerin), in osteoporotic conditions and increases calcium mineralization in C2C12-derived OBs. Interestingly, KYMASIN UP induces differentiation in human primary OB-like cells derived from osteoporotic subjects. Based on our results, KYMASIN UP or Withania somnifera-based dietary supplements might be suggested to reverse the age-related functional decline of bone tissue by re-balancing the activity of OBs and OCs, thus improving the quality of life in the elderly and reducing social and health-care costs.


Subject(s)
Biological Products , Bone Resorption , Dietary Supplements , Osteogenesis , Animals , Biological Products/pharmacology , Bone Resorption/drug therapy , Cell Differentiation , Humans , Mice , Osteoblasts/metabolism , Osteoclasts , Osteogenesis/drug effects , RANK Ligand/metabolism , RAW 264.7 Cells , p38 Mitogen-Activated Protein Kinases/metabolism
17.
J Immunother Cancer ; 10(3)2022 03.
Article in English | MEDLINE | ID: mdl-35236743

ABSTRACT

BACKGROUND: Despite the great success, the therapeutic benefits of immune checkpoint inhibitors (ICIs) in cancer immunotherapy are limited by either various resistance mechanisms or ICI-associated toxic effects including gastrointestinal toxicity. Thus, novel therapeutic strategies that provide manageable side effects to existing ICIs would enhance and expand their therapeutic efficacy and application. Due to its proven role in cancer development and immune regulation, gut microbiome has gained increasing expectation as a potential armamentarium to optimize immunotherapy with ICI. However, much has to be learned to fully harness gut microbiome for clinical applicability. Here we have assessed whether microbial metabolites working at the interface between microbes and the host immune system may optimize ICI therapy. METHODS: To this purpose, we have tested indole-3-carboxaldehyde (3-IAld), a microbial tryptophan catabolite known to contribute to epithelial barrier function and immune homeostasis in the gut via the aryl hydrocarbon receptor (AhR), in different murine models of ICI-induced colitis. Epithelial barrier integrity, inflammation and changes in gut microbiome composition and function were analyzed. AhR, indoleamine 2,3-dioxygenase 1, interleukin (IL)-10 and IL-22 knockout mice were used to investigate the mechanism of 3-IAld activity. The function of the microbiome changes induced by 3-IAld was evaluated on fecal microbiome transplantation (FMT). Finally, murine tumor models were used to assess the effect of 3-IAld treatment on the antitumor activity of ICI. RESULTS: On administration to mice with ICI-induced colitis, 3-IAld protected mice from intestinal damage via a dual action on both the host and the microbes. Indeed, paralleling the activation of the host AhR/IL-22-dependent pathway, 3-IAld also affected the composition and function of the microbiota such that FMT from 3-IAld-treated mice protected against ICI-induced colitis with the contribution of butyrate-producing bacteria. Importantly, while preventing intestinal damage, 3-IAld did not impair the antitumor activity of ICI. CONCLUSIONS: This study provides a proof-of-concept demonstration that moving past bacterial phylogeny and focusing on bacterial metabolome may lead to a new class of discrete molecules, and that working at the interface between microbes and the host immune system may optimize ICI therapy.


Subject(s)
Colitis , Neoplasms , Animals , Colitis/chemically induced , Colitis/drug therapy , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Mice , Neoplasms/drug therapy , Treatment Outcome , Tryptophan/pharmacology
18.
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
19.
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
20.
Biomolecules ; 11(10)2021 10 12.
Article in English | MEDLINE | ID: mdl-34680138

ABSTRACT

Duchenne muscular dystrophy (DMD) is an X-linked disease caused by mutations in DMD gene translating in lack of functional dystrophin and resulting in susceptibility of myofibers to rupture during contraction. Inflammation and fibrosis are critical hallmarks of DMD muscles, which undergo progressive degeneration leading to loss of independent ambulation in childhood and death by early adulthood. We reported that intraperitoneal injection of microencapsulated Sertoli cells (SeC) in dystrophic mice translates into recovery of muscle morphology and performance thanks to anti-inflammatory effects and induction of the dystrophin paralogue, utrophin at the muscle level, opening new avenues in the treatment of DMD. The aim of this study is to obtain information about the direct effects of SeC on myoblasts/myotubes, as a necessary step in view of a translational application of SeC-based approaches to DMD. We show that (i) SeC-derived factors stimulate cell proliferation in the early phase of differentiation in C2C12, and human healthy and DMD myoblasts; (ii) SeC delay the expression of differentiation markers in the early phase nevertheless stimulating terminal differentiation in DMD myoblasts; (iii) SeC restrain the fibrogenic potential of fibroblasts, and inhibit myoblast-myofibroblast transdifferentiation; and, (iv) SeC provide functional replacement of dystrophin in preformed DMD myotubes regardless of the mutation by inducing heregulin ß1/ErbB2/ERK1/2-dependent utrophin expression. Altogether, these results show that SeC are endowed with promyogenic and antifibrotic effects on dystrophic myoblasts, further supporting their potential use in the treatment of DMD patients. Our data also suggest that SeC-based approaches might be useful in improving the early phase of muscle regeneration, during which myoblasts have to adequately proliferate to replace the damaged muscle mass.


Subject(s)
Muscular Dystrophy, Duchenne/genetics , Neuregulin-1/genetics , Receptor, ErbB-2/genetics , Sertoli Cells/metabolism , Utrophin/genetics , Animals , Cell Differentiation/genetics , Cell Proliferation/genetics , Cell Transdifferentiation/genetics , Disease Models, Animal , Dystrophin/genetics , Gene Expression Regulation/genetics , Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/metabolism , Genetic Diseases, X-Linked/pathology , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Injections, Intraperitoneal , MAP Kinase Signaling System/genetics , Male , Mice , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology , Myoblasts/metabolism , Regeneration/genetics , Sertoli Cells/pathology
SELECTION OF CITATIONS
SEARCH DETAIL