Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 9.214
Filtrer
1.
Chem Biol Drug Des ; 104(1): e14574, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38958121

RÉSUMÉ

To develop novel bovine lactoferrin (bLF) peptides targeting bLF-tumour necrosis factor (TNF) receptor-associated factor 6 (TRAF6) binding sites, we identified two peptides that could target bLF-TRAF6 binding sites using structural analysis. Moreover, another peptide that could bind to the TRAF6 dimerization area was selected from the bLF sequence. The effects of each peptide on cytokine expression in lipopolysaccharide (LPS)-stimulated osteoblasts (ST2) and on osteoclastogenesis were examined using an LPS-treated co-culture of primary bone marrow cells (BMCs) with ST2 cells and a single culture of osteoclast precursor cells (RAW-D) treated with soluble receptor activator of NF-κB ligand. Finally, the effectiveness of these peptides against LPS-induced alveolar bone destruction was assessed. Two of the three peptides significantly suppressed LPS-induced TNF-α and interleukin-1ß expression in ST2 cells. Additionally, these peptides inhibited and reversed LPS-induced receptor activator of NF-κB ligand (RANKL) upregulation and osteoprotegerin (OPG) downregulation, respectively. Furthermore, both peptides significantly reduced LPS-induced osteoclastogenesis in the BMC-ST2 co-culture and RANKL-induced osteoclastogenesis in RAW-D cells. In vivo, topical application of these peptides significantly reduced the osteoclast number by downregulating RANKL and upregulating OPG in the periodontal ligament. It is indicated that the novel bLF peptides can be used to treat periodontitis-associated bone destruction.


Sujet(s)
Lactoferrine , Lipopolysaccharides , Ostéoclastes , Peptides , Animaux , Lactoferrine/pharmacologie , Lactoferrine/composition chimique , Lactoferrine/métabolisme , Lipopolysaccharides/pharmacologie , Rats , Peptides/pharmacologie , Peptides/composition chimique , Ostéoclastes/effets des médicaments et des substances chimiques , Ostéoclastes/métabolisme , Ligand de RANK/métabolisme , Mâle , Résorption alvéolaire/traitement médicamenteux , Résorption alvéolaire/métabolisme , Résorption alvéolaire/anatomopathologie , Bovins , Souris , Ostéoblastes/effets des médicaments et des substances chimiques , Ostéoblastes/métabolisme , Ostéoblastes/cytologie , Rat Sprague-Dawley , Ostéogenèse/effets des médicaments et des substances chimiques , Facteur de nécrose tumorale alpha/métabolisme , Sites de fixation , Techniques de coculture , Ostéoprotégérine/métabolisme , Modèles animaux de maladie humaine
2.
FASEB J ; 38(13): e23779, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38967255

RÉSUMÉ

Epigenetic modifications affect cell differentiation via transcriptional regulation. G9a/EHMT2 is an important epigenetic modifier that catalyzes the methylation of histone 3 lysine 9 (H3K9) and interacts with various nuclear proteins. In this study, we investigated the role of G9a in osteoclast differentiation. When we deleted G9a by infection of Cre-expressing adenovirus into bone marrow macrophages (BMMs) from G9afl/fl (Ehmt2fl/fl) and induced osteoclastic differentiation by the addition of macrophage colony-stimulating factor (M-CSF) and receptor activator of NF-κB ligand (RANKL), the number of TRAP-positive multinucleated osteoclasts significantly increased compared with control. Furthermore, the mRNA expression of osteoclast markers, TRAP, and cathepsin K, and to a lesser extent, NFATc1, a critical transcription factor, increased in G9a KO cells. Infection of wild-type (WT) G9a-expressing adenovirus in G9a KO cells restored the number of TRAP-positive multinucleated cells. In G9a KO cells, increased nuclear accumulation of NFATc1 protein and decreased H3K9me2 accumulation were observed. Furthermore, ChIP experiments revealed that NFATc1 binding to its target, Ctsk promoter, was enhanced by G9a deletion. For in vivo experiments, we created G9a conditional knock-out (cKO) mice by crossing G9afl/fl mice with Rank Cre/+ (Tnfrsf11aCre/+) mice, in which G9a is deleted in osteoclast lineage cells. The trabecular bone volume was significantly reduced in female G9a cKO mice. The serum concentration of the C-terminal telopeptide of type I collagen (CTX), a bone-resorbing indicator, was higher in G9a cKO mice. In addition, osteoclasts differentiated from G9a cKO BMMs exhibited greater bone-resorbing activity. Our findings suggest that G9a plays a repressive role in osteoclastogenesis by modulating NFATc1 function.


Sujet(s)
Résorption osseuse , Différenciation cellulaire , Histone-lysine N-methyltransferase , Facteurs de transcription NFATC , Ostéoclastes , Ostéogenèse , Animaux , Facteurs de transcription NFATC/métabolisme , Facteurs de transcription NFATC/génétique , Histone-lysine N-methyltransferase/métabolisme , Histone-lysine N-methyltransferase/génétique , Souris , Ostéoclastes/métabolisme , Résorption osseuse/métabolisme , Ostéogenèse/physiologie , Souris knockout , Ligand de RANK/métabolisme , Souris de lignée C57BL , Cellules cultivées
3.
Stem Cell Res Ther ; 15(1): 203, 2024 Jul 06.
Article de Anglais | MEDLINE | ID: mdl-38971808

RÉSUMÉ

BACKGROUND: Skeletal Stem Cells (SSCs) are required for skeletal development, homeostasis, and repair. The perspective of their wide application in regenerative medicine approaches has supported research in this field, even though so far results in the clinic have not reached expectations, possibly due also to partial knowledge of intrinsic, potentially actionable SSC regulatory factors. Among them, the pleiotropic cytokine RANKL, with essential roles also in bone biology, is a candidate deserving deep investigation. METHODS: To dissect the role of the RANKL cytokine in SSC biology, we performed ex vivo characterization of SSCs and downstream progenitors (SSPCs) in mice lacking Rankl (Rankl-/-) by means of cytofluorimetric sorting and analysis of SSC populations from different skeletal compartments, gene expression analysis, and in vitro osteogenic differentiation. In addition, we assessed the effect of the pharmacological treatment with the anti-RANKL blocking antibody Denosumab (approved for therapy in patients with pathological bone loss) on the osteogenic potential of bone marrow-derived stromal cells from human healthy subjects (hBMSCs). RESULTS: We found that, regardless of the ossification type of bone, osteochondral SSCs had a higher frequency and impaired differentiation along the osteochondrogenic lineage in Rankl-/- mice as compared to wild-type. Rankl-/- mice also had increased frequency of committed osteochondrogenic and adipogenic progenitor cells deriving from perivascular SSCs. These changes were not due to the peculiar bone phenotype of increased density caused by lack of osteoclast resorption (defined osteopetrosis); indeed, they were not found in another osteopetrotic mouse model, i.e., the oc/oc mouse, and were therefore not due to osteopetrosis per se. In addition, Rankl-/- SSCs and primary osteoblasts showed reduced mineralization capacity. Of note, hBMSCs treated in vitro with Denosumab had reduced osteogenic capacity compared to control cultures. CONCLUSIONS: We provide for the first time the characterization of SSPCs from mouse models of severe recessive osteopetrosis. We demonstrate that Rankl genetic deficiency in murine SSCs and functional blockade in hBMSCs reduce their osteogenic potential. Therefore, we propose that RANKL is an important regulatory factor of SSC features with translational relevance.


Sujet(s)
Différenciation cellulaire , Ostéogenèse , Ligand de RANK , Animaux , Ligand de RANK/métabolisme , Ligand de RANK/génétique , Souris , Ostéogenèse/génétique , Humains , Cellules souches/métabolisme , Cellules souches/cytologie , Souris knockout , Dénosumab/pharmacologie , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/cytologie , Cellules cultivées , Souris de lignée C57BL
4.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 55(3): 777-782, 2024 May 20.
Article de Chinois | MEDLINE | ID: mdl-38948285

RÉSUMÉ

As a member of the tumor necrosis factor receptor family, osteoprotegerin (OPG) is highly expressed in adults in the lung, heart, kidney, liver, spleen, thymus, prostate, ovary, small intestines, thyroid gland, lymph nodes, trachea, adrenal gland, the testis, and bone marrow. Together with the receptor activator of nuclear factor-κB (RANK) and the receptor activator of nuclear factor-κB ligand (RANKL), it forms the RANK/RANKL/OPG pathway, which plays an important role in the molecular mechanism of the development of various diseases. MicroRNAs (miRNAs) are a class of endogenous non-coding RNAs performing regulatory functions in eukaryotes, with a size of about 20-25 nucleotides. miRNA genes are transcribed into primary transcripts by RNA polymerase, bind to RNA-induced silencing complexes, identify target mRNAs through complementary base pairing, with a single miRNA being capable of targeting hundreds of mRNAs, and influence the expression of many genes through pathways involved in functional interactions. In recent years, a large number of studies have been done to explore the mechanism of action of miRNA in diseases through miRNA isolation, miRNA quantification, miRNA spectrum analysis, miRNA target detection, in vitro and in vivo regulation of miRNA levels, and other technologies. It was found that miRNA can play a key role in the pathogenesis of osteoporosis, rheumatoid arthritis, and other diseases by targeting OPG. The purpose of this review is to explore the interaction between miRNA and OPG in various diseases, and to propose new ideas for studying the mechanism of action of OPG in diseases.


Sujet(s)
microARN , Ostéoprotégérine , Récepteur activateur du facteur nucléaire Kappa B , Ostéoprotégérine/métabolisme , Ostéoprotégérine/génétique , Humains , microARN/génétique , microARN/métabolisme , Récepteur activateur du facteur nucléaire Kappa B/métabolisme , Récepteur activateur du facteur nucléaire Kappa B/génétique , Ligand de RANK/métabolisme , Ligand de RANK/génétique , Tumeurs/génétique , Tumeurs/métabolisme , Animaux , Transduction du signal , Polyarthrite rhumatoïde/génétique , Polyarthrite rhumatoïde/métabolisme
5.
Appl Microbiol Biotechnol ; 108(1): 397, 2024 Jun 26.
Article de Anglais | MEDLINE | ID: mdl-38922350

RÉSUMÉ

Functional M cells are differentiated by receptor activator of NF-κB ligand (RANKL) and capture of luminal antigens to initiate immune responses. We aimed to use postbiotic-based recombinant chicken RANKL (cRANKL) to promote M cell differentiation and test the efficacy of oral vaccines. Chicks were divided into three groups that were administered phosphate-buffered saline (PBS), cell extracts of wild-type Lactococcus lactis subsp. lactis IL1403 (WT_CE), or cell extracts of recombinant L. lactis expressing cRANKL (cRANKL_CE). The expression of the M cell marker was measured, and the gut microbiome was profiled. The efficiency of the infectious bursal disease (IBD) vaccine was tested after 12 consecutive days of administering cRANKL_CE. The chickens that were administered cRANKL_CE (p = 0.038) had significantly higher Annexin A5 (ANXA5) mRNA expression levels than those in the PBS group (PBS vs. WT_CE, p = 0.657). In the gut microbiome analysis, no significant changes were observed. However, the relative abundance of Escherichia-Shigella was negatively correlated (r = - 0.43, p = 0.019) with ANXA5 mRNA expression in Peyer's patches. cRANKL_CE/IBD (p = 0.018) had significantly higher IBD-specific faecal IgA levels than PBS/IBD (PBS/IBD vs. WT_CE/IBD, p = 0.217). Postbiotic-based recombinant cRANKL effectively improved the expression of M cell markers and the efficiency of oral vaccines. No significant changes were observed in the gut microbiome after administration of postbiotic-based recombinant cRANKL. This strategy can be used for the development of feed additives and adjuvants. KEY POINTS: • Postbiotic-based recombinant cRANKL enhanced the expression of ANXA5 in chicken. • The relative abundance of Escherichia-Shigella was negatively correlated with ANXA5 expression. • Postbiotic-based recombinant cRANKL effectively improved the efficiency of oral vaccine.


Sujet(s)
Poulets , Microbiome gastro-intestinal , Lactococcus lactis , Ligand de RANK , Protéines recombinantes , Animaux , Poulets/immunologie , Administration par voie orale , Lactococcus lactis/génétique , Lactococcus lactis/métabolisme , Lactococcus lactis/immunologie , Ligand de RANK/immunologie , Ligand de RANK/génétique , Ligand de RANK/métabolisme , Protéines recombinantes/immunologie , Protéines recombinantes/génétique , Protéines recombinantes/administration et posologie , Infections à Birnaviridae/prévention et contrôle , Infections à Birnaviridae/immunologie , Infections à Birnaviridae/médecine vétérinaire , Maladies de la volaille/prévention et contrôle , Maladies de la volaille/immunologie , Maladies de la volaille/microbiologie , Virus de la bursite infectieuse/immunologie , Virus de la bursite infectieuse/génétique , Différenciation cellulaire , Plaques de Peyer/immunologie
6.
Front Endocrinol (Lausanne) ; 15: 1406248, 2024.
Article de Anglais | MEDLINE | ID: mdl-38904051

RÉSUMÉ

Magnesium (Mg), a nutritional element which is essential for bone development and mineralization, has a role in the progression of osteoporosis. Osteoporosis is a multifactorial disease characterized by significant deterioration of bone microstructure and bone loss. Mg deficiency can affect bone structure in an indirect way through the two main regulators of calcium homeostasis (parathyroid hormone and vitamin D). In human osteoblasts (OBs), parathyroid hormone regulates the expression of receptor activator of nuclear factor-κ B ligand (RANKL) and osteoprotegerin (OPG) to affect osteoclast (OC) formation. In addition, Mg may also affect the vitamin D3 -mediated bone remodeling activity. vitamin D3 usually coordinates the activation of the OB and OC. The unbalanced activation OC leads to bone resorption. The RANK/RANKL/OPG axis is considered to be a key factor in the molecular mechanism of osteoporosis. Mg participates in the pathogenesis of osteoporosis by affecting the regulation of parathyroid hormone and vitamin D levels to affect the RANK/RANKL/OPG axis. Different factors affecting the axis and enhancing OC function led to bone loss and bone tissue microstructure damage, which leads to the occurrence of osteoporosis. Clinical research has shown that Mg supplementation can alleviate the symptoms of osteoporosis to some extent.


Sujet(s)
Magnésium , Ostéoporose , Humains , Ostéoporose/étiologie , Ostéoporose/métabolisme , Magnésium/métabolisme , Animaux , Hormone parathyroïdienne/métabolisme , Ligand de RANK/métabolisme , Ostéoblastes/métabolisme , Remodelage osseux/physiologie , Vitamine D/métabolisme , Magnésium, carence/métabolisme , Magnésium, carence/complications , Ostéoclastes/métabolisme , Ostéoprotégérine/métabolisme
7.
Int J Mol Sci ; 25(11)2024 May 24.
Article de Anglais | MEDLINE | ID: mdl-38891922

RÉSUMÉ

Vascular calcification has a global health impact that is closely linked to bone loss. The Receptor Activator of Nuclear Factor Kappa B (RANK)/RANK ligand (RANKL)/osteoprotegerin (OPG) system, fundamental for bone metabolism, also plays an important role in vascular calcification. The Leucine-rich repeat-containing G-protein-coupled receptor 4 (LGR4), a novel receptor for RANKL, regulates bone remodeling, and it appears to be involved in vascular calcification. Besides RANKL, LGR4 interacts with R-spondins (RSPOs), which are known for their roles in bone but are less understood in vascular calcification. Studies were conducted in rats with chronic renal failure fed normal or high phosphorus diets for 18 weeks, with and without control of circulating parathormone (PTH) levels, resulting in different degrees of aortic calcification. Additionally, vascular smooth muscle cells (VSMCs) were cultured under non-calcifying (1 mM phosphate) and calcifying (3 mM phosphate) media with different concentrations of PTH. To explore the role of RANKL in VSMC calcification, increasing concentrations of soluble RANKL were added to non-calcifying and calcifying media. The effects mediated by RANKL binding to its receptor LGR4 were investigated by silencing the LGR4 receptor in VSMCs. Furthermore, the gene expression of the RANK/RANKL/OPG system and the ligands of LGR4 was assessed in human epigastric arteries obtained from kidney transplant recipients with calcification scores (Kauppila Index). Increased aortic calcium in rats coincided with elevated systolic blood pressure, upregulated Lgr4 and Rankl gene expression, downregulated Opg gene expression, and higher serum RANKL/OPG ratio without changes in Rspos gene expression. Elevated phosphate in vitro increased calcium content and expression of Rankl and Lgr4 while reducing Opg. Elevated PTH in the presence of high phosphate exacerbated the increase in calcium content. No changes in Rspos were observed under the conditions employed. The addition of soluble RANKL to VSMCs induced genotypic differentiation and calcification, partly prevented by LGR4 silencing. In the epigastric arteries of individuals presenting vascular calcification, the gene expression of RANKL was higher. While RSPOs show minimal impact on VSMC calcification, RANKL, interacting with LGR4, drives osteogenic differentiation in VSMCs, unveiling a novel mechanism beyond RANKL-RANK binding.


Sujet(s)
Muscles lisses vasculaires , Ligand de RANK , Récepteurs couplés aux protéines G , Calcification vasculaire , Ligand de RANK/métabolisme , Ligand de RANK/génétique , Animaux , Récepteurs couplés aux protéines G/métabolisme , Récepteurs couplés aux protéines G/génétique , Calcification vasculaire/métabolisme , Calcification vasculaire/anatomopathologie , Muscles lisses vasculaires/métabolisme , Muscles lisses vasculaires/anatomopathologie , Rats , Humains , Mâle , Myocytes du muscle lisse/métabolisme , Myocytes du muscle lisse/anatomopathologie , Ostéoprotégérine/métabolisme , Ostéoprotégérine/génétique , Hormone parathyroïdienne/métabolisme , Cellules cultivées , Rat Sprague-Dawley
8.
Prog Orthod ; 25(1): 23, 2024 Jun 10.
Article de Anglais | MEDLINE | ID: mdl-38853224

RÉSUMÉ

BACKGROUND: External apical root resorption (EARR) is a common undesirable outcome of orthodontic treatment, this study aimed to identify genetic polymorphisms associated with the susceptibility to extreme orthodontic-induced EARR in a Korean population using extreme phenotype analysis sampling. METHODS: Genomic DNA was isolated from the saliva of 77 patients who underwent orthodontic treatment involving two maxillary premolar extractions. The patients were divided into two groups based on EARR values measured on periapical radiographs: The significant resorption group (SG, EARR ≥ 4 mm) and the normal group (NG, EARR < 2 mm). In the NG group, patients with EARR < 1 mm were named the non-resorption group (NonG). Targeted next-generation sequencing was performed using the screened single nucleotide polymorphisms (SNPs), and firth logistic regression analysis was used to determine genetic associations with EARR. Haplotype-based association analysis was performed for specific SNPs. RESULTS: SNPs related to genes TNFSF11, TNFRSF11B, WNT3A, SFRP2, LRP6, P2RX7, and LRP1 were found to be significantly associated with severe EARR (p < 0.05, pre-Bonferroni correction p-values). Additionally, the haplotype CCA of rs17525809, rs208294, and rs1718119 P2RX7 had a higher frequency in the SG group. CONCLUSION: Extreme phenotype analysis has identified eleven SNPs related to genes TNFSF11, TNFRSF11B, WNT3A, SFRP2, LRP6, P2RX7, and LRP1 that are associated with severe root resorption in the Korean population. These findings will contribute to the development of predictive diagnostic tools for identifying severe root resorption that may occur during orthodontic treatment.


Sujet(s)
Polymorphisme de nucléotide simple , Rhizalyse , Humains , Rhizalyse/génétique , Rhizalyse/imagerie diagnostique , Femelle , Mâle , République de Corée , Haplotypes , Adolescent , Phénotype , Prédisposition génétique à une maladie , Récepteurs purinergiques P2X7/génétique , Ostéoprotégérine/génétique , Orthodontie correctrice , Asiatiques/génétique , Jeune adulte , Peuples d'Asie de l'Est , Ligand de RANK
9.
BMC Oral Health ; 24(1): 659, 2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38840172

RÉSUMÉ

BACKGROUND: Peri-implantitis (PI) is a frequent inflammatory disorder characterised by progressive loss of the supporting bone. Not all patients with recognised risk factors develop PI. The aim of this study is to evaluate the presence of single nucleotide polymorphisms (SNP) of inflammatory and bone metabolism related proteins in a population treated with dental implants from the Basque Country (Spain). METHODS: We included 80 patients with diagnosis of PI and 81 patients without PI, 91 women and 70 men, with a mean age of 60.90 years. SNPs of BMP-4, BRINP3, CD14, FGF-3, FGF-10, GBP-1, IL-1α, IL-1ß, IL-10, LTF, OPG and RANKL proteins were selected. We performed a univariate and bivariate analysis using IBM SPSS® v.28 statistical software. RESULTS: Presence of SNPs GBP1 rs7911 (p = 0.041) and BRINP3 rs1935881 (p = 0.012) was significantly more common in patients with PI. Patients with PI who smoked (> 10 cig/day) showed a higher presence of OPG rs2073617 SNP (p = 0.034). Also, BMP-4 rs17563 (p = 0.018) and FGF-3 rs1893047 (p = 0.014) SNPs were more frequent in patients with PI and Type II diabetes mellitus. CONCLUSIONS: Our findings suggest that PI could be favoured by an alteration in the osseointegration of dental implants, based on an abnormal immunological response to peri-implant infection in patients from the Basque Country (Spain).


Sujet(s)
Implants dentaires , Péri-implantite , Polymorphisme de nucléotide simple , Humains , Mâle , Femelle , Études cas-témoins , Adulte d'âge moyen , Espagne , Péri-implantite/génétique , Ostéoprotégérine/génétique , Sujet âgé , Protéine morphogénétique osseuse de type 4/génétique , Protéines G/génétique , Ligand de RANK/génétique , Interleukine-1 alpha/génétique , Phosphodiesterases , Pyrophosphatases
10.
Mol Biol Rep ; 51(1): 702, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38822942

RÉSUMÉ

BACKGROUND: The development of cost-effective, simple, environment-friendly biographene is an area of interest. To accomplish environmentally safe, benign culturing that has advantages over other methods to reduce the graphene oxide (GO), extracellular metabolites from actinobacteria associated with mushrooms were used for the first time. METHODS: Bactericidal effect of GO against methicillin-resistant Staphylococcus aureus, antioxidant activity, and hydroxyapatite-like bone layer formation, gene expression analysis and appropriate biodegradation of the microbe-mediated synthesis of graphene was studied. RESULTS: Isolated extracellular contents Streptomyces achromogenes sub sp rubradiris reduced nano-GO to graphene (rGO), which was further examined by spectrometry and suggested an efficient conversion and significant reduction in the intensity of all oxygen-containing moieties and shifted crystalline peaks. Electron microscopic results also suggested the reduction of GO layer. In addition, absence of significant toxicity in MG-63 cell line, intentional free radical scavenging prowess, liver and kidney histopathology, and Wistar rat bone regeneration through modulation of OPG/RANKL/RUNX2/ALP pathways show the feasibility of the prepared nano GO. CONCLUSIONS: The study demonstrates the successful synthesis of biographene from actinobacterial extracellular metabolites, its potential biomedical applications, and its promising role in addressing health and environmental concerns.


Sujet(s)
Régénération osseuse , Graphite , Ostéoprotégérine , Ligand de RANK , Rat Wistar , Graphite/pharmacologie , Animaux , Régénération osseuse/effets des médicaments et des substances chimiques , Rats , Ligand de RANK/métabolisme , Ostéoprotégérine/métabolisme , Humains , Matériaux biocompatibles/pharmacologie , Sous-unité alpha 1 du facteur CBF/métabolisme , Sous-unité alpha 1 du facteur CBF/génétique , Actinobacteria/métabolisme , Antibactériens/pharmacologie , Antioxydants/métabolisme , Antioxydants/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques
11.
Cell Death Dis ; 15(6): 437, 2024 Jun 20.
Article de Anglais | MEDLINE | ID: mdl-38902257

RÉSUMÉ

TNF receptor superfamily member 11a (TNFRSF11a, RANK) and its ligand TNF superfamily member 11 (TNFRSF11, RANKL) are overexpressed in many malignancies. However, the clinical importance of RANKL/RANK in colorectal cancer (CRC) is mainly unknown. We examined CRC samples and found that RANKL/RANK was elevated in CRC tissues compared with nearby normal tissues. A higher RANKL/RANK expression was associated with a worse survival rate. Furthermore, RANKL was mostly produced by regulatory T cells (Tregs), which were able to promote CRC advancement. Overexpression of RANK or addition of RANKL significantly increased the stemness and migration of CRC cells. Furthermore, RANKL/RANK signaling stimulated C-C motif chemokine ligand 20 (CCL20) production by CRC cells, leading to Treg recruitment and boosting tumor stemness and malignant progression. This recruitment process was accomplished by CCL20-CCR6 interaction, demonstrating a connection between CRC cells and immune cells. These findings suggest an important role of RANKL/RANK in CRC progression, offering a potential target for CRC prevention and therapy.


Sujet(s)
Chimiokine CCL20 , Tumeurs colorectales , Cellules souches tumorales , Ligand de RANK , Récepteur activateur du facteur nucléaire Kappa B , Récepteurs CCR6 , Transduction du signal , Lymphocytes T régulateurs , Humains , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/métabolisme , Tumeurs colorectales/génétique , Chimiokine CCL20/métabolisme , Chimiokine CCL20/génétique , Ligand de RANK/métabolisme , Récepteurs CCR6/métabolisme , Récepteurs CCR6/génétique , Récepteur activateur du facteur nucléaire Kappa B/métabolisme , Lymphocytes T régulateurs/immunologie , Lymphocytes T régulateurs/métabolisme , Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie , Animaux , Mâle , Souris , Femelle , Métastase tumorale , Lignée cellulaire tumorale , Adulte d'âge moyen , Souris nude , Mouvement cellulaire
12.
Cell Commun Signal ; 22(1): 322, 2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38863060

RÉSUMÉ

Bone resorption is driven through osteoclast differentiation by macrophage colony-stimulating factor (M-CSF) and receptor activator of nuclear factor kappa-Β ligand (RANKL). We noted that a disintegrin and metalloproteinase (ADAM) 10 and ADAM17 are downregulated at the expression level during osteoclast differentiation of the murine monocytic cell line RAW264.7 in response to RANKL. Both proteinases are well known to shed a variety of single-pass transmembrane molecules from the cell surface. We further showed that inhibitors of ADAM10 or ADAM17 promote osteoclastic differentiation and furthermore enhance the surface expression of receptors for RANKL and M-CSF on RAW264.7 cells. Using murine bone marrow-derived monocytic cells (BMDMCs), we demonstrated that a genetic deficiency of ADAM17 or its required regulator iRhom2 leads to increased osteoclast development in response to M-CSF and RANKL stimulation. Moreover, ADAM17-deficient osteoclast precursor cells express increased levels of the receptors for RANKL and M-CSF. Thus, ADAM17 negatively regulates osteoclast differentiation, most likely through shedding of these receptors. To assess the time-dependent contribution of ADAM10, we blocked this proteinase by adding a specific inhibitor on day 0 of BMDMC stimulation with M-CSF or on day 7 of subsequent stimulation with RANKL. Only ADAM10 inhibition beginning on day 7 increased the size of developing osteoclasts indicating that ADAM10 suppresses osteoclast differentiation at a later stage. Finally, we could confirm our findings in human peripheral blood mononuclear cells (PBMCs). Thus, downregulation of either ADAM10 or ADAM17 during osteoclast differentiation may represent a novel regulatory mechanism to enhance their differentiation process. Enhanced bone resorption is a critical issue in osteoporosis and is driven through osteoclast differentiation by specific osteogenic mediators. The present study demonstrated that the metalloproteinases ADAM17 and ADAM10 critically suppress osteoclast development. This was observed for a murine cell line, for isolated murine bone marrow cells and for human blood cells by either preferential inhibition of the proteinases or by gene knockout. As a possible mechanism, we studied the surface expression of critical receptors for osteogenic mediators on developing osteoclasts. Our findings revealed that the suppressive effects of ADAM17 and ADAM10 on osteoclastogenesis can be explained in part by the proteolytic cleavage of surface receptors by ADAM10 and ADAM17, which reduces the sensitivity of these cells to osteogenic mediators. We also observed that osteoclast differentiation was associated with the downregulation of ADAM10 and ADAM17, which reduced their suppressive effects. We therefore propose that this downregulation serves as a feedback loop for enhancing osteoclast development.


Sujet(s)
Protéine ADAM10 , Protéine ADAM17 , Amyloid precursor protein secretases , Différenciation cellulaire , Régulation négative , Protéines membranaires , Ostéoclastes , Ligand de RANK , Protéine ADAM17/métabolisme , Protéine ADAM17/génétique , Protéine ADAM10/métabolisme , Protéine ADAM10/génétique , Ostéoclastes/métabolisme , Ostéoclastes/cytologie , Animaux , Différenciation cellulaire/génétique , Souris , Régulation négative/génétique , Amyloid precursor protein secretases/métabolisme , Amyloid precursor protein secretases/génétique , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Humains , Ligand de RANK/métabolisme , Cellules RAW 264.7 , Facteur de stimulation des colonies de macrophages/pharmacologie , Facteur de stimulation des colonies de macrophages/métabolisme , Souris de lignée C57BL
13.
J Immunother Cancer ; 12(6)2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38908859

RÉSUMÉ

BACKGROUND: Receptor activator of nuclear factor kappa-B ligand (RANKL) can directly promote tumor growth and indirectly support tumor immune evasion by altering the tumor microenvironment and immune cell responses. This study aimed to assess the prognostic significance of soluble RANKL in patients with advanced non-small cell lung cancer (NSCLC) receiving programmed cell death 1 (PD1)/programmed death-ligand 1 (PDL1) checkpoint inhibitor therapy. METHODS: Plasma RANKL levels were measured in 100 patients with advanced NSCLC without bone metastases undergoing monotherapy with PD1/PDL1 checkpoint inhibitors. To establish the optimal cut-off value, we used the Cutoff Finder package in R. Survival curves for four distinct patient groups, according to their RANKL and PDL1 levels (high or low), were generated using the Kaplan-Meier method and compared with the log-rank test. The Cox regression model calculated HRs and 95% CIs for overall survival (OS) and progression-free survival (PFS). RESULTS: The optimal RANKL cut-off was established at 280.4 pg/mL, categorizing patients into groups with high or low RANKL levels. A significant association was observed between increased RANKL concentrations and decreased survival rates at 24 months, only within the subgroup expressing high levels of PDL1 (p=0.002). Additionally, low RANKL levels in conjunction with elevated PDL1 expression correlated with improved PFS (median 22 months, 95% CI 6.70 to 50 vs median 4 months, 95% CI 3.0 to 7.30, p=0.009) and OS (median 26 months, 95% CI 20 to not reached vs median 7 months, 95% CI 6 to 13, p=0.003), indicating RANKL's potential as an indicator of adverse prognosis in these patients. Multivariate analysis identified RANKL as an independent negative prognostic factor for both PFS and OS, regardless of other clinicopathological features. CONCLUSION: These results highlight the prognostic and predictive value of RANKL specifically in patients with high PDL1 expression.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Inhibiteurs de points de contrôle immunitaires , Tumeurs du poumon , Ligand de RANK , Humains , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/sang , Carcinome pulmonaire non à petites cellules/anatomopathologie , Mâle , Femelle , Ligand de RANK/sang , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/sang , Tumeurs du poumon/anatomopathologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Sujet âgé , Adulte d'âge moyen , Sujet âgé de 80 ans ou plus , Adulte , Antigène CD274/sang , Marqueurs biologiques tumoraux/sang , Pronostic
14.
BMC Nephrol ; 25(1): 205, 2024 Jun 23.
Article de Anglais | MEDLINE | ID: mdl-38910256

RÉSUMÉ

BACKGROUND: Sepsis-associated acute kidney injury (SA-AKI) has high mortality rates. The osteoprotegerin (OPG)/receptor activator of nuclear factor-κB ligand (RANKL)/receptor activator of nuclear factor-κB (RANK)/Toll-like receptor 4 (TLR4) pathway and its potential role in SA-AKI pathogenesis remain to be fully understood. Herein, we addressed this issue using mouse models. METHODS: An SA-AKI mouse model was established using the cecal ligation and puncture method (CLP). Mice were grouped into sham, CLP model, CLP + recombinant RANKL, and CLP + anti-RANKL groups. Serum creatinine (Scr) and blood urea nitrogen (BUN) levels were measured to assess kidney function. ELISA was used to detect serum IL-1ß, TNF-α, and IL-6 levels. Real-time quantitative PCR and Western blot were used to detect the mRNA and protein expression levels of OPG, RANKL, RANK, and TLR4 in kidney tissues. HE staining was performed to evaluate the pathological changes. RESULTS: The CLP model group showed higher levels of Scr and BUN, indicating impaired kidney function in SA-AKI, compared to the sham group. Treatment with recombinant RANKL in the CLP + recombinant RANKL group reduced Scr and BUN levels, while anti-RANKL treatment in the CLP + anti-RANKL group elevated their levels. Moreover, the CLP model group had significantly increased IL-1ß, TNF-α, and IL-6 than the sham group, indicating elevated inflammation in SA-AKI. The CLP + recombinant RANKL group demonstrated decreased cytokine levels, whereas the CLP + anti-RANKL group showed an increase. Additionally, the histopathological evaluation revealed distinct kidney tissue damage in the CLP model group. Recombinant RANKL treatment reduced this damage, while anti-RANKL treatment exacerbated it. Mechanically, the mRNA and protein expression of RANKL were significantly decreased, while those of OPG, RANK, and TLR4 were significantly increased in the CLP model group and the CLP + anti-RANKL group. Interestingly, treatment with recombinant RANKL reversed these changes, as evidenced by significantly increased RANKL but decreased OPG, RANK, and TLR4. CONCLUSION: The OPG/RANKL/RANK/TLR4 pathway is involved in SA-AKI pathogenesis. Recombinant RANKL treatment attenuates the inflammatory response and kidney tissue damage in SA-AKI, possibly via regulating this pathway. This pathway shows promise as a therapeutic target for SA-AKI.


Sujet(s)
Atteinte rénale aigüe , Ostéoprotégérine , Ligand de RANK , Récepteur activateur du facteur nucléaire Kappa B , Sepsie , Transduction du signal , Récepteur de type Toll-4 , Animaux , Atteinte rénale aigüe/métabolisme , Atteinte rénale aigüe/étiologie , Récepteur de type Toll-4/métabolisme , Ostéoprotégérine/métabolisme , Ligand de RANK/métabolisme , Souris , Sepsie/complications , Sepsie/métabolisme , Récepteur activateur du facteur nucléaire Kappa B/métabolisme , Mâle , Souris de lignée C57BL , Modèles animaux de maladie humaine
15.
Int J Mol Sci ; 25(12)2024 Jun 19.
Article de Anglais | MEDLINE | ID: mdl-38928460

RÉSUMÉ

Osteoporosis, a prevalent chronic health issue among the elderly, is a global bone metabolic disease. Flavonoids, natural active compounds widely present in vegetables, fruits, beans, and cereals, have been reported for their anti-osteoporotic properties. Onion is a commonly consumed vegetable rich in flavonoids with diverse pharmacological activities. In this study, the trabecular structure was enhanced and bone mineral density (BMD) exhibited a twofold increase following oral administration of onion flavonoid extract (OFE). The levels of estradiol (E2), calcium (Ca), and phosphorus (P) in serum were significantly increased in ovariectomized (OVX) rats, with effects equal to alendronate sodium (ALN). Alkaline phosphatase (ALP) and tartrate-resistant acid phosphatase (TRAP) levels in rat serum were reduced by 35.7% and 36.9%, respectively, compared to the OVX group. In addition, the effects of OFE on bone health were assessed using human osteoblast-like cells MG-63 and osteoclast precursor RAW 264.7 cells in vitro as well. Proliferation and mineralization of MG-63 cells were promoted by OFE treatment, along with increased ALP activity and mRNA expression of osteoprotegerin (OPG)/receptor activator of nuclear factor-kappaB ligand (RANKL). Additionally, RANKL-induced osteoclastogenesis and osteoclast activity were inhibited by OFE treatment through decreased TRAP activity and down-regulation of mRNA expression-related enzymes in RAW 264.7 cells. Overall findings suggest that OFE holds promise as a natural functional component for alleviating osteoporosis.


Sujet(s)
Différenciation cellulaire , Prolifération cellulaire , Flavonoïdes , Oignons , Ostéoblastes , Ostéogenèse , Ostéoporose , Extraits de plantes , Ligand de RANK , Animaux , Ostéoblastes/effets des médicaments et des substances chimiques , Ostéoblastes/métabolisme , Ligand de RANK/métabolisme , Ostéoporose/traitement médicamenteux , Ostéoporose/métabolisme , Ostéoporose/anatomopathologie , Flavonoïdes/pharmacologie , Souris , Oignons/composition chimique , Différenciation cellulaire/effets des médicaments et des substances chimiques , Extraits de plantes/pharmacologie , Extraits de plantes/composition chimique , Rats , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cellules RAW 264.7 , Ostéogenèse/effets des médicaments et des substances chimiques , Humains , Femelle , Ostéoclastes/effets des médicaments et des substances chimiques , Ostéoclastes/métabolisme , Densité osseuse/effets des médicaments et des substances chimiques , Ovariectomie , Rat Sprague-Dawley , Ostéoprotégérine/métabolisme , Ostéoprotégérine/génétique
16.
Biochim Biophys Acta Gen Subj ; 1868(8): 130635, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38788984

RÉSUMÉ

Gallein is known as an inhibitor of Gßγ subunits, but roles of gallein in bone metabolism have not been reported. Fibroblast growth factor 2 (FGF-2) increases angiogenesis and promotes bone regeneration during the early stages of fracture healing. Osteoprotegerin (OPG) secreted by osteoblasts, binds to the receptor activator of nuclear factor-κB (RANK) ligand (RANKL) as a decoy receptor and prevents RANKL from binding to RANK, resulting in the suppression of bone resorption. Our previous report demonstrated that FGF-2 activates the phosphorylation of p38 mitogen-activated protein kinase (MAPK), stress-activated protein kinase/c-Jun N-terminal kinase (JNK), and p44/p42 MAPK in osteoblast-like MC3T3-E1 cells. Additionally, FGF-2-activated phosphorylation of p38 MAPK and JNK but not p44/p42 MAPK is positively involved in OPG synthesis in these cells. This work aimed to investigate the effects of gallein on the FGF-2-elicited OPG synthesis in osteoblast-like MC3T3-E1 cells and the mechanism. Our findings demonstrated that gallein significantly increased the FGF-2-elicited OPG synthesis in MC3T3-E1 cells. By contrast, fluorescein, gallein-like compound that does not bind Gßγ, did not affect the FGF-2-elicited OPG synthesis. Gallein significantly enhanced the FGF-2-induced OPG mRNA expression levels. Gallein did not affect the FGF-2-activated phosphorylation of p38 MAPK and p44/p42 MAPK, but significantly increased the FGF-2-activated phosphorylation of JNK, while fluorescein did not affect JNK phosphorylation. SP600125, a specific JNK inhibitor, strongly inhibited gallein-induced enhancement of FGF-2-induced OPG synthesis and mRNA expression levels. Our results indicated that gallein increases the FGF-2-induced OPG synthesis due to the JNK activation in the osteoblast.


Sujet(s)
Facteur de croissance fibroblastique de type 2 , Ostéoblastes , Ostéoprotégérine , Ostéoblastes/métabolisme , Ostéoblastes/effets des médicaments et des substances chimiques , Ostéoprotégérine/métabolisme , Ostéoprotégérine/biosynthèse , Animaux , Facteur de croissance fibroblastique de type 2/métabolisme , Souris , Phosphorylation/effets des médicaments et des substances chimiques , p38 Mitogen-Activated Protein Kinases/métabolisme , Cellules 3T3 , Lignée cellulaire , Ligand de RANK/métabolisme
17.
Tissue Cell ; 88: 102412, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38776732

RÉSUMÉ

Tumor necrosis factor superfamily member 11 (TNFSF11), or receptor activator of nuclear factor-κB ligand (RANKL), is a crucial osteoclast-stimulating factor binding to RANK on osteoclast membranes. Mouse models are powerful tools for understanding the genetic mechanisms of related diseases. Here, we examined the utility of Tnfsf11 mutation in mice for understanding the mechanisms of bone remodeling and dysmorphology. The Tnfsf11gum mouse, discovered in 2011 at Jackson Laboratory, was used to study the genetic landscape associated with TNFSF11 inactivation in bone marrow tissues. Tnfsf11gum/+ and Tnfsf11+/+ mice were subjected to Micro-CT observation, ELISA analysis, histological evaluation, and massively-parallel mRNA sequencing (RNA-Seq) analysis. Tnfsf11gum/+ mice exhibited severe osteopetrotic changes in the bone marrow cavity, along with significantly lower serum RANKL levels and a reduced number of tartrate-resistant acid phosphatase (TRAP)-positive osteoclasts in the bone marrow compared to those in Tnfsf11+/+ mice. However, tooth eruption between Tnfsf11gum/+ and Tnfsf11+/+ mice did not differ. Furthermore, genes involved in osteoblast proliferation and differentiation, including Gli1, Slc35b2, Lrrc17, and Junb were differentially expressed. Heterozygous mutation of TNFSF11 was also associated with a slightly increased expression of genes involved in osteoclast proliferation and differentiation, including Tcirg1, Junb, Anxa2, and Atp6ap1. Overall, we demonstrate that single gene mutations in Tnfsf11 cause bone resorption instability without significantly altering the genes related to osteoblast and osteoclast activity in the bone marrow cavity, thus establishing an optimal resource as an experimental animal model for bone resorption in bone biology research.


Sujet(s)
Modèles animaux de maladie humaine , Ostéoclastes , Ostéopétrose , Ligand de RANK , Animaux , Ostéopétrose/génétique , Ostéopétrose/anatomopathologie , Ostéopétrose/métabolisme , Ligand de RANK/métabolisme , Ligand de RANK/génétique , Souris , Ostéoclastes/métabolisme , Ostéoclastes/anatomopathologie , Mutation
18.
Aging (Albany NY) ; 16(10): 9251-9263, 2024 05 27.
Article de Anglais | MEDLINE | ID: mdl-38809508

RÉSUMÉ

BACKGROUND: Senile osteoporosis may be caused by an imbalance in intestinal flora and oxidative stress. Trimethylamine-N-oxide (TMAO), a metabolite of dietary choline dependent on gut microbes, has been found to be significantly increased in osteoporosis. However, the role of TMAO in bone loss during osteoporosis remains poorly understood. In this study, we examined the impact of TMAO on osteoclast differentiation and bone resorption in an in vitro setting. METHODS: Osteoclast differentiation was induced by incubating RAW 264.7 cells in the presence of Receptor Activator for Nuclear Factor-κB Ligand (RANKL) and macrophage-stimulating factor (M-CSF). Flow cytometry, TRAP staining assay, CCK-8, and ELISA were employed to investigate the impact of TMAO on osteoclast differentiation and bone resorption activity in vitro. For mechanistic exploration, RT-PCR and Western blotting were utilized to assess the activation of the NF-κB pathway. Additionally, protein levels of secreted cytokines and growth factors were determined using suspension array technology. RESULTS: Our findings demonstrate that TMAO enhances RANKL and M-CSF-induced osteoclast formation and bone resorption in a dose-dependent manner. Mechanistically, TMAO triggers the upregulation of the NF-κB pathway and osteoclast-related genes (NFATc1, c-Fos, NF-κB p65, Traf6, and Cathepsin K). Furthermore, TMAO markedly elevated the levels of oxidative stress and inflammatory factors. CONCLUSIONS: In conclusion, TMAO enhances RANKL and M-CSF-induced osteoclast differentiation and inflammation in RAW 264.7 cells by activating the NF-κB signaling pathway. These findings offer a new rationale for further academic and clinical research on osteoporosis treatment.


Sujet(s)
Différenciation cellulaire , Méthylamines , Facteur de transcription NF-kappa B , Ostéoclastes , Stress oxydatif , Ligand de RANK , Transduction du signal , Animaux , Ostéoclastes/effets des médicaments et des substances chimiques , Ostéoclastes/métabolisme , Souris , Méthylamines/pharmacologie , Stress oxydatif/effets des médicaments et des substances chimiques , Différenciation cellulaire/effets des médicaments et des substances chimiques , Cellules RAW 264.7 , Facteur de transcription NF-kappa B/métabolisme , Ligand de RANK/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Facteur de stimulation des colonies de macrophages/métabolisme , Facteur de stimulation des colonies de macrophages/pharmacologie , Résorption osseuse/métabolisme
19.
Cell Rep Med ; 5(5): 101574, 2024 05 21.
Article de Anglais | MEDLINE | ID: mdl-38776873

RÉSUMÉ

The existing suite of therapies for bone diseases largely act to prevent further bone loss but fail to stimulate healthy bone formation and repair. We describe an endogenous osteopeptide (PEPITEM) with anabolic osteogenic activity, regulating bone remodeling in health and disease. PEPITEM acts directly on osteoblasts through NCAM-1 signaling to promote their maturation and formation of new bone, leading to enhanced trabecular bone growth and strength. Simultaneously, PEPITEM stimulates an inhibitory paracrine loop: promoting osteoblast release of the decoy receptor osteoprotegerin, which sequesters RANKL, thereby limiting osteoclast activity and bone resorption. In disease models, PEPITEM therapy halts osteoporosis-induced bone loss and arthritis-induced bone damage in mice and stimulates new bone formation in osteoblasts derived from patient samples. Thus, PEPITEM offers an alternative therapeutic option in the management of diseases with excessive bone loss, promoting an endogenous anabolic pathway to induce bone remodeling and redress the imbalance in bone turnover.


Sujet(s)
Résorption osseuse , Ostéoblastes , Ostéogenèse , Animaux , Humains , Ostéoblastes/métabolisme , Ostéoblastes/effets des médicaments et des substances chimiques , Ostéogenèse/effets des médicaments et des substances chimiques , Souris , Résorption osseuse/anatomopathologie , Résorption osseuse/métabolisme , Anabolisants/pharmacologie , Anabolisants/usage thérapeutique , Remodelage osseux/effets des médicaments et des substances chimiques , Ostéoporose/anatomopathologie , Ostéoporose/métabolisme , Ostéoporose/traitement médicamenteux , Ligand de RANK/métabolisme , Ostéoclastes/métabolisme , Ostéoclastes/effets des médicaments et des substances chimiques , Développement osseux/effets des médicaments et des substances chimiques , Ostéoprotégérine/métabolisme , Femelle , Transduction du signal/effets des médicaments et des substances chimiques , Peptides/pharmacologie , Mâle , Souris de lignée C57BL , Os et tissu osseux/effets des médicaments et des substances chimiques , Os et tissu osseux/métabolisme , Os et tissu osseux/anatomopathologie
20.
Mol Genet Genomic Med ; 12(5): e2471, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38803233

RÉSUMÉ

BACKGROUND: Bone tissue homeostasis relies on the coordinated activity of the bone-forming osteoblasts and bone-resorbing osteoclasts. Osteomesopyknosis is considered a distinctive rare sclerosing skeletal disorder of unelucidated pathophysiology and presumably autosomal dominant transmission. However, the causal genes are unknown. METHODS: We present a case report encompassing clinical assessments, imaging studies, and whole-exome sequencing analysis, complemented by functional in vitro experiments. RESULTS: This new case of osteomesopyknosis was associated with a missense ALOX5 variant predicted to induce protein misfolding and proteasomal degradation. Transfection experiments demonstrated that the variant was associated with reduced protein levels restored by proteasomal inhibition with bortezomib. Likewise, gene expression analysis showed that the mutated gene was associated with a decreased RANKL/OPG ratio, which is a critical driver of osteoclast precursor differentiation. CONCLUSION: Our data indicate impaired bone resorption as the underlying mechanism of this rare osteosclerosis, implicating ALOX5 pathogenic variants as potential etiological factors.


Sujet(s)
Arachidonate 5-lipoxygenase , Mutation faux-sens , Ligand de RANK , Femelle , Humains , Arachidonate 5-lipoxygenase/génétique , Arachidonate 5-lipoxygenase/métabolisme , Ostéoclastes/métabolisme , Ostéoclastes/anatomopathologie , Ostéosclérose/génétique , Ostéosclérose/anatomopathologie , Ostéosclérose/métabolisme , Ligand de RANK/métabolisme , Ligand de RANK/génétique , Transduction du signal , Adulte d'âge moyen
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...