Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.885
Filtrar
1.
Chem Biol Drug Des ; 104(1): e14574, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38958121

RESUMEN

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.


Asunto(s)
Lactoferrina , Lipopolisacáridos , Osteoclastos , Péptidos , Animales , Lactoferrina/farmacología , Lactoferrina/química , Lactoferrina/metabolismo , Lipopolisacáridos/farmacología , Ratas , Péptidos/farmacología , Péptidos/química , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Ligando RANK/metabolismo , Masculino , Pérdida de Hueso Alveolar/tratamiento farmacológico , Pérdida de Hueso Alveolar/metabolismo , Pérdida de Hueso Alveolar/patología , Bovinos , Ratones , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Osteoblastos/citología , Ratas Sprague-Dawley , Osteogénesis/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo , Sitios de Unión , Técnicas de Cocultivo , Osteoprotegerina/metabolismo , Modelos Animales de Enfermedad
2.
FASEB J ; 38(13): e23779, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-38967255

RESUMEN

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.


Asunto(s)
Resorción Ósea , Diferenciación Celular , N-Metiltransferasa de Histona-Lisina , Factores de Transcripción NFATC , Osteoclastos , Osteogénesis , Animales , Factores de Transcripción NFATC/metabolismo , Factores de Transcripción NFATC/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , Ratones , Osteoclastos/metabolismo , Resorción Ósea/metabolismo , Osteogénesis/fisiología , Ratones Noqueados , Ligando RANK/metabolismo , Ratones Endogámicos C57BL , Células Cultivadas
3.
Sci Rep ; 14(1): 15749, 2024 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-38977767

RESUMEN

Although bone dehiscence may occur during orthodontic tooth movement into the narrow alveolar ridge, a non-invasive prevention method is yet to be fully established. We show for the first time prevention of bone dehiscence associated with orthodontic tooth movement by prophylactic injection of bone anabolic agents in mice. In this study, we established a bone dehiscence mouse model by applying force application and used the granular type of scaffold materials encapsulated with bone morphogenetic protein (BMP)-2 and OP3-4, the receptor activator of NF-κB ligand (RANKL)-binding peptide, for the prophylactic injection to the alveolar bone. In vivo micro-computed tomography revealed bone dehiscence with decreased buccal alveolar bone thickness and height after force application, whereas no bone dehiscence was observed with the prophylactic injection after force application, and alveolar bone thickness and height were kept at similar levels as those in the control group. Bone histomorphometry analyses revealed that both bone formation and resorption parameters were significantly higher in the injection with force application group than in the force application without the prophylactic injection group. These findings suggest that the prophylactic local delivery of bone anabolic reagents can prevent bone dehiscence with increased bone remodelling activity.


Asunto(s)
Anabolizantes , Proteína Morfogenética Ósea 2 , Técnicas de Movimiento Dental , Microtomografía por Rayos X , Animales , Ratones , Técnicas de Movimiento Dental/efectos adversos , Anabolizantes/farmacología , Anabolizantes/administración & dosificación , Masculino , Osteogénesis/efectos de los fármacos , Remodelación Ósea/efectos de los fármacos , Ligando RANK/metabolismo , Proceso Alveolar/efectos de los fármacos , Proceso Alveolar/diagnóstico por imagen , Proceso Alveolar/patología , Modelos Animales de Enfermedad
4.
Cell Mol Biol Lett ; 29(1): 100, 2024 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-38977961

RESUMEN

OBJECTIVE: Osteoporosis is a global health issue characterized by decreased bone mass and microstructural degradation, leading to an increased risk of fractures. This study aims to explore the molecular mechanism by which P2X7 receptors influence osteoclast formation and bone resorption through the PI3K-Akt-GSK3ß signaling pathway. METHODS: An osteoporosis mouse model was generated through ovariectomy (OVX) in normal C57BL/6 and P2X7f/f; LysM-cre mice. Osteoclasts were isolated for transcriptomic analysis, and differentially expressed genes were selected for functional enrichment analysis. Metabolite analysis was performed using liquid chromatography-tandem mass spectrometry (LC-MS/MS), and multivariate statistical analysis and pattern recognition were used to identify differential lipid metabolism markers and their distribution. Bioinformatics analyses were conducted using the Encyclopedia of Genes and Genomes database and the MetaboAnalyst database to assess potential biomarkers and create a metabolic pathway map. Osteoclast precursor cells were used for in vitro cell experiments, evaluating cell viability and proliferation using the Cell Counting Kit 8 (CCK-8) assay. Osteoclast precursor cells were induced to differentiate into osteoclasts using macrophage colony-stimulating factor (M-CSF) and receptor activator of nuclear factor kappa-beta ligand (RANKL), and tartrate-resistant acid phosphatase (TRAP) staining was performed to compare differentiation morphology, size, and quantity between different groups. Western blot analysis was used to assess the expression of differentiation markers, fusion gene markers, and bone resorption ability markers in osteoclasts. Immunofluorescence staining was employed to examine the spatial distribution and quantity of osteoclast cell skeletons, P2X7 protein, and cell nuclei, while pit assay was used to evaluate osteoclast bone resorption ability. Finally, in vivo animal experiments, including micro computed tomography (micro-CT), hematoxylin and eosin (HE) staining, TRAP staining, and immunohistochemistry, were conducted to observe bone tissue morphology, osteoclast differentiation, and the phosphorylation level of the PI3K-Akt-GSK3ß signaling pathway. RESULTS: Transcriptomic and metabolomic data collectively reveal that the P2X7 receptor can impact the pathogenesis of osteoporosis through the PI3K-Akt-GSK3ß signaling pathway. Subsequent in vitro experiments showed that cells in the Sh-P2X7 + Recilisib group exhibited increased proliferative activity (1.15 versus 0.59), higher absorbance levels (0.68 versus 0.34), and a significant increase in resorption pit area (13.94 versus 3.50). Expression levels of osteoclast differentiation-related proteins MMP-9, CK, and NFATc1 were markedly elevated (MMP-9: 1.72 versus 0.96; CK: 2.54 versus 0.95; NFATc1: 3.05 versus 0.95), along with increased fluorescent intensity of F-actin rings. In contrast, the OE-P2X7 + LY294002 group showed decreased proliferative activity (0.64 versus 1.29), reduced absorbance (0.34 versus 0.82), and a significant decrease in resorption pit area (5.01 versus 14.96), accompanied by weakened expression of MMP-9, CK, and NFATc1 (MMP-9: 1.14 versus 1.79; CK: 1.26 versus 2.75; NFATc1: 1.17 versus 2.90) and decreased F-actin fluorescent intensity. Furthermore, in vivo animal experiments demonstrated that compared with the wild type (WT) + Sham group, mice in the WT + OVX group exhibited significantly increased levels of CTX and NTX in serum (CTX: 587.17 versus 129.33; NTX: 386.00 versus 98.83), a notable decrease in calcium deposition (19.67 versus 53.83), significant reduction in bone density, increased trabecular separation, and lowered bone mineral density (BMD). When compared with the KO + OVX group, mice in the KO + OVX + recilisib group showed a substantial increase in CTX and NTX levels in serum (CTX: 503.50 versus 209.83; NTX: 339.83 versus 127.00), further reduction in calcium deposition (29.67 versus 45.33), as well as decreased bone density, increased trabecular separation, and reduced BMD. CONCLUSION: P2X7 receptors positively regulate osteoclast formation and bone resorption by activating the PI3K-Akt-GSK3ß signaling pathway.


Asunto(s)
Resorción Ósea , Diferenciación Celular , Glucógeno Sintasa Quinasa 3 beta , Ratones Endogámicos C57BL , Osteoclastos , Fosfatidilinositol 3-Quinasas , Proteínas Proto-Oncogénicas c-akt , Receptores Purinérgicos P2X7 , Transducción de Señal , Animales , Osteoclastos/metabolismo , Resorción Ósea/metabolismo , Resorción Ósea/genética , Resorción Ósea/patología , Diferenciación Celular/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Glucógeno Sintasa Quinasa 3 beta/genética , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Receptores Purinérgicos P2X7/metabolismo , Receptores Purinérgicos P2X7/genética , Femenino , Osteoporosis/metabolismo , Osteoporosis/genética , Osteoporosis/patología , Ligando RANK/metabolismo , Ligando RANK/genética
5.
Bone Res ; 12(1): 40, 2024 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-38987568

RESUMEN

Efficient cellular fusion of mononuclear precursors is the prerequisite for the generation of fully functional multinucleated bone-resorbing osteoclasts. However, the exact molecular factors and mechanisms controlling osteoclast fusion remain incompletely understood. Here we identify RANKL-mediated activation of caspase-8 as early key event during osteoclast fusion. Single cell RNA sequencing-based analyses suggested that activation of parts of the apoptotic machinery accompanied the differentiation of osteoclast precursors into mature multinucleated osteoclasts. A subsequent characterization of osteoclast precursors confirmed that RANKL-mediated activation of caspase-8 promoted the non-apoptotic cleavage and activation of downstream effector caspases that translocated to the plasma membrane where they triggered activation of the phospholipid scramblase Xkr8. Xkr8-mediated exposure of phosphatidylserine, in turn, aided cellular fusion of osteoclast precursors and thereby allowed generation of functional multinucleated osteoclast syncytia and initiation of bone resorption. Pharmacological blockage or genetic deletion of caspase-8 accordingly interfered with fusion of osteoclasts and bone resorption resulting in increased bone mass in mice carrying a conditional deletion of caspase-8 in mononuclear osteoclast precursors. These data identify a novel pathway controlling osteoclast biology and bone turnover with the potential to serve as target for therapeutic intervention during diseases characterized by pathologic osteoclast-mediated bone loss. Proposed model of osteoclast fusion regulated by caspase-8 activation and PS exposure. RANK/RANK-L interaction. Activation of procaspase-8 into caspase-8. Caspase-8 activates caspase-3. Active capase-3 cleaves Xkr8. Local PS exposure is induced. Exposed PS is recognized by the fusion partner. FUSION. PS is re-internalized.


Asunto(s)
Caspasa 8 , Fusión Celular , Osteoclastos , Fosfatidilserinas , Proteínas de Transferencia de Fosfolípidos , Caspasa 8/metabolismo , Caspasa 8/genética , Animales , Osteoclastos/metabolismo , Fosfatidilserinas/metabolismo , Proteínas de Transferencia de Fosfolípidos/metabolismo , Proteínas de Transferencia de Fosfolípidos/genética , Ratones , Ratones Endogámicos C57BL , Resorción Ósea/metabolismo , Resorción Ósea/patología , Resorción Ósea/genética , Diferenciación Celular , Ligando RANK/metabolismo
6.
Stem Cell Res Ther ; 15(1): 203, 2024 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-38971808

RESUMEN

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.


Asunto(s)
Diferenciación Celular , Osteogénesis , Ligando RANK , Animales , Ligando RANK/metabolismo , Ligando RANK/genética , Ratones , Osteogénesis/genética , Humanos , Células Madre/metabolismo , Células Madre/citología , Ratones Noqueados , Denosumab/farmacología , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Células Cultivadas , Ratones Endogámicos C57BL
7.
Theriogenology ; 226: 277-285, 2024 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-38954996

RESUMEN

Tumour necrosis factor (TNF) superfamily member 11 (TNFSF11), also known as RANKL, plays a crucial role in regulating several physiological and pathological activities. Additionally, it is a vital factor in bone physiology, and the sex hormone progesterone regulates the expansion of stem cells and the proliferation of mammary epithelial cells. It is essential for animal growth and reproductive physiological processes. This study aimed to evaluate the tissue-specific expression characteristics and promoter activity of the TNFSF11 gene in pigs. As a result, the study examined the presence of TNFSF11 expression in the tissues of Xiangsu pigs at 0.6 and 12 months of age. Moreover, the core promoter region of TNFSF11 was also identified by utilizing a combination of bioinformatic prediction and dual-luciferase activity tests. Finally, the effect of transcription factors on the transcriptional activity of the core promoter region was determined using site-directed mutagenesis. TNFSF11 was uniformly expressed in all tissues; however, its expression in muscles was comparatively low. The core promoter region of TNFSF11 was located in the -555 to -1 region. The prediction of the transcription start site of TNFSF11 gene-2000 ∼ + 500bp showed that there was a CpG site in 17 ∼ + 487bp. Analysis of mutations in the transcription factor binding sites revealed that mutations in the Stat5b, Myog, Trl, and EN1 binding sites had significant effects on the transcriptional activity of the TNFSF11 gene, particularly following the EN1 binding site mutation (P < 0.001). This study provides insights into both the tissue-specific expression patterns of TNFSF11 in the tissues of Xiangsu pigs and the potential regulatory effects of transcription factors on its promoter activity. These results may be helpful for future research aimed at clarifying the expression and role of the porcine TNFSF11 gene.


Asunto(s)
Regulación de la Expresión Génica , Regiones Promotoras Genéticas , Ligando RANK , Animales , Porcinos/genética , Ligando RANK/genética , Ligando RANK/metabolismo
8.
Sci Adv ; 10(28): eadl4913, 2024 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-38985878

RESUMEN

The pathophysiology of silicosis is poorly understood, limiting development of therapies for those who have been exposed to the respirable particle. We explored mechanisms of silica-induced pulmonary fibrosis in human lung samples collected from patients with occupational exposure to silica and in a longitudinal mouse model of silicosis using multiple modalities including whole-lung single-cell RNA sequencing and histological, biochemical, and physiologic assessments. In addition to pulmonary inflammation and fibrosis, intratracheal silica challenge induced osteoclast-like differentiation of alveolar macrophages and recruited monocytes, driven by induction of the osteoclastogenic cytokine, receptor activator of nuclear factor κΒ ligand (RANKL) in pulmonary lymphocytes, and alveolar type II cells. Anti-RANKL monoclonal antibody treatment suppressed silica-induced osteoclast-like differentiation in the lung and attenuated pulmonary fibrosis. We conclude that silica induces differentiation of pulmonary osteoclast-like cells leading to progressive lung injury, likely due to sustained elaboration of bone-resorbing proteases and hydrochloric acid. Interrupting osteoclast-like differentiation may therefore constitute a promising avenue for moderating lung damage in silicosis.


Asunto(s)
Diferenciación Celular , Osteoclastos , Fibrosis Pulmonar , Dióxido de Silicio , Silicosis , Dióxido de Silicio/toxicidad , Animales , Humanos , Osteoclastos/metabolismo , Osteoclastos/efectos de los fármacos , Osteoclastos/patología , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/patología , Fibrosis Pulmonar/metabolismo , Ratones , Silicosis/patología , Silicosis/metabolismo , Silicosis/etiología , Diferenciación Celular/efectos de los fármacos , Ligando RANK/metabolismo , Modelos Animales de Enfermedad , Masculino , Pulmón/patología , Pulmón/metabolismo , Pulmón/efectos de los fármacos , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/patología , Macrófagos Alveolares/efectos de los fármacos , Femenino
9.
Lasers Med Sci ; 39(1): 187, 2024 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-39031220

RESUMEN

The purpose of this research was to investigate the effect of toluidine blue (TB) mediated photodynamic therapy (PDT) on the inhibition of lipopolysaccharide (LPS)-induced inflammation in rat gingival fibroblasts through in vitro experiments. Rat gingival fibroblasts were divided into five groups: (1) control, (2) LPS treatment, (3) laser treatment, (4) TB treatment (1.0 µg/mL), and (5) PDT treatment (TB plus laser irradiation at 320 mW/cm2 for 240 s). After 24 h, cell growth activity was measured using MTT assay. The levels of receptor activator for nuclear factor-κB ligand (RANKL) and osteoprotegerin (OPG) in the cell culture supernatant were measured using enzyme-linked immunosorbent assay (ELISA). Nuclear proteins were extracted and the phosphorylation levels of phosphorylated nuclear factor-κB/p65 (p-p65) and phosphorylated inhibitor of nuclear factor-κB (p-IκBα) were determined using Western Blot. MTT results showed no significant difference in cell viability between the groups (P > 0.05). After LPS induction, OPG expression decreased, RANKL expression increased, and the OPG/RANKL ratio decreased, which was different from the control group (P < 0.05). After PDT treatment, OPG expression increased, RANKL expression decreased (P < 0.05), and the OPG/RANKL ratio increased (P < 0.05). Compared to the control group, there was no significant difference in OPG and RANKL expression or the OPG/RANKL ratio (P > 0.05). The activation of NF-κB was closely related to the phosphorylation levels of p-p65 and p-IκBα. LPS significantly up-regulated p-p65 and p-IκBα expression (P < 0.05), while PDT treatment decreased their phosphorylation levels (P < 0.05). TB-PDT treatment can inhibit NF-κB signaling pathway activation, decrease RANKL and OPG expression, and reduce the OPG/RANKL ratio, thereby reducing inflammation and playing a role in periodontitis treatment.


Asunto(s)
Fibroblastos , Encía , Lipopolisacáridos , Osteoprotegerina , Fotoquimioterapia , Ligando RANK , Cloruro de Tolonio , Animales , Fotoquimioterapia/métodos , Ratas , Encía/efectos de los fármacos , Encía/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/efectos de la radiación , Fibroblastos/metabolismo , Ligando RANK/metabolismo , Osteoprotegerina/metabolismo , Células Cultivadas , Inflamación , FN-kappa B/metabolismo , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Fármacos Fotosensibilizantes/farmacología , Fosforilación
10.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 55(3): 777-782, 2024 May 20.
Artículo en Chino | MEDLINE | ID: mdl-38948285

RESUMEN

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.


Asunto(s)
MicroARNs , Osteoprotegerina , Receptor Activador del Factor Nuclear kappa-B , Osteoprotegerina/metabolismo , Osteoprotegerina/genética , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Receptor Activador del Factor Nuclear kappa-B/genética , Ligando RANK/metabolismo , Ligando RANK/genética , Neoplasias/genética , Neoplasias/metabolismo , Animales , Transducción de Señal , Artritis Reumatoide/genética , Artritis Reumatoide/metabolismo
11.
Front Endocrinol (Lausanne) ; 15: 1406248, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38904051

RESUMEN

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.


Asunto(s)
Magnesio , Osteoporosis , Humanos , Osteoporosis/etiología , Osteoporosis/metabolismo , Magnesio/metabolismo , Animales , Hormona Paratiroidea/metabolismo , Ligando RANK/metabolismo , Osteoblastos/metabolismo , Remodelación Ósea/fisiología , Vitamina D/metabolismo , Deficiencia de Magnesio/metabolismo , Deficiencia de Magnesio/complicaciones , Osteoclastos/metabolismo , Osteoprotegerina/metabolismo
12.
Appl Microbiol Biotechnol ; 108(1): 397, 2024 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-38922350

RESUMEN

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.


Asunto(s)
Pollos , Microbioma Gastrointestinal , Lactococcus lactis , Ligando RANK , Proteínas Recombinantes , Animales , Pollos/inmunología , Administración Oral , Lactococcus lactis/genética , Lactococcus lactis/metabolismo , Lactococcus lactis/inmunología , Ligando RANK/inmunología , Ligando RANK/genética , Ligando RANK/metabolismo , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/genética , Proteínas Recombinantes/administración & dosificación , Infecciones por Birnaviridae/prevención & control , Infecciones por Birnaviridae/inmunología , Infecciones por Birnaviridae/veterinaria , Enfermedades de las Aves de Corral/prevención & control , Enfermedades de las Aves de Corral/inmunología , Enfermedades de las Aves de Corral/microbiología , Virus de la Enfermedad Infecciosa de la Bolsa/inmunología , Virus de la Enfermedad Infecciosa de la Bolsa/genética , Diferenciación Celular , Ganglios Linfáticos Agregados/inmunología
13.
Cell Death Dis ; 15(6): 437, 2024 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-38902257

RESUMEN

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.


Asunto(s)
Quimiocina CCL20 , Neoplasias Colorrectales , Células Madre Neoplásicas , Ligando RANK , Receptor Activador del Factor Nuclear kappa-B , Receptores CCR6 , Transducción de Señal , Linfocitos T Reguladores , Humanos , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/genética , Quimiocina CCL20/metabolismo , Quimiocina CCL20/genética , Ligando RANK/metabolismo , Receptores CCR6/metabolismo , Receptores CCR6/genética , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Animales , Masculino , Ratones , Femenino , Metástasis de la Neoplasia , Línea Celular Tumoral , Persona de Mediana Edad , Ratones Desnudos , Movimiento Celular
14.
Cell Commun Signal ; 22(1): 322, 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38863060

RESUMEN

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.


Asunto(s)
Proteína ADAM10 , Proteína ADAM17 , Secretasas de la Proteína Precursora del Amiloide , Diferenciación Celular , Regulación hacia Abajo , Proteínas de la Membrana , Osteoclastos , Ligando RANK , Proteína ADAM17/metabolismo , Proteína ADAM17/genética , Proteína ADAM10/metabolismo , Proteína ADAM10/genética , Osteoclastos/metabolismo , Osteoclastos/citología , Animales , Diferenciación Celular/genética , Ratones , Regulación hacia Abajo/genética , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Secretasas de la Proteína Precursora del Amiloide/genética , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Humanos , Ligando RANK/metabolismo , Células RAW 264.7 , Factor Estimulante de Colonias de Macrófagos/farmacología , Factor Estimulante de Colonias de Macrófagos/metabolismo , Ratones Endogámicos C57BL
15.
Endocrinology ; 165(8)2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38935021

RESUMEN

Thyroid hormone (TH) plays a crucial role in regulating the functions of both bone and adipose tissue. Given that TH exerts its cholesterol-lowering effects in hepatic tissue through the TH receptor-ß (TRß), we hypothesized that TRß agonist therapy using MGL3196 (MGL) would be effective in treating increased adiposity and bone loss in response to a 12-week high-fat diet (HFD) in adult C57BL/6J mice. Transcriptional and serum profiling revealed that HFD-induced leptin promoted weight gain in both males and females, but MGL only suppressed leptin induction and weight gain in males. In vitro studies suggest that estrogen suppresses MGL activity in adipocytes, indicating that estrogen might interfere with MGL-TRß function. Compared to systemic adiposity, HFD reduced bone mass in male but not female mice. Paradoxically, MGL treatment reversed macroscopic bone mineral density loss in appendicular bones, but micro-CT revealed that MGL exacerbated HFD-induced trabecular bone loss, and reduced bone strength. In studies on the mechanisms for HFD effects on bone, we found that HFD induced Rankl expression in male femurs that was blocked by MGL. By ex vivo assays, we found that RANKL indirectly represses osteoblast lineage allocation of osteoprogenitors by induction of inflammatory cytokines TNFα, IL-1ß, and CCL2. Finally, we found that MGL functions in both systemic adiposity and bone by nongenomic TRß signaling, as HFD-mediated phenotypes were not rescued in TRß147F knockout mice with normal genomic but defective nongenomic TRß signaling. Our findings demonstrate that the negative effects of HFD on body fat and bone phenotypes are impacted by MGL in a gender-specific manner.


Asunto(s)
Dieta Alta en Grasa , Ratones Endogámicos C57BL , Transducción de Señal , Receptores beta de Hormona Tiroidea , Animales , Masculino , Femenino , Ratones , Transducción de Señal/efectos de los fármacos , Receptores beta de Hormona Tiroidea/metabolismo , Receptores beta de Hormona Tiroidea/genética , Densidad Ósea/efectos de los fármacos , Adiposidad/efectos de los fármacos , Caracteres Sexuales , Ligando RANK/metabolismo , Ligando RANK/genética , Leptina/metabolismo , Osteoblastos/metabolismo , Osteoblastos/efectos de los fármacos , Factores Sexuales , Adipocitos/metabolismo , Adipocitos/efectos de los fármacos , Aumento de Peso/efectos de los fármacos
16.
BMC Nephrol ; 25(1): 205, 2024 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-38910256

RESUMEN

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.


Asunto(s)
Lesión Renal Aguda , Osteoprotegerina , Ligando RANK , Receptor Activador del Factor Nuclear kappa-B , Sepsis , Transducción de Señal , Receptor Toll-Like 4 , Animales , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/etiología , Receptor Toll-Like 4/metabolismo , Osteoprotegerina/metabolismo , Ligando RANK/metabolismo , Ratones , Sepsis/complicaciones , Sepsis/metabolismo , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Masculino , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad
17.
J Endocrinol ; 262(3)2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-38913537

RESUMEN

Osteosarcopenia, which refers to the concomitant presence of osteoporosis and sarcopenia, is expected to increase in the rapidly progressive aging world, with serious clinical implications. However, the pathophysiology of osteosarcopenia has not been fully elucidated, and no optimal treatment specific to osteosarcopenia is available. The RANKL-RANK pathway is widely used as a therapeutic target for osteoporosis. Growing evidence supports the importance of the RANKL-RANK pathway, not only in bone, but also in muscle, and the therapeutic potential of targeting this pathway in muscle diseases has been noted. The muscles and bones closely communicate with each other through various secretory factors called myokines and osteokines. This review covers the roles of the RANKL-RANK pathway in the bone and muscle and their reciprocal interactions. Moreover, we will suggest future directions to move forward for the treatment of osteosarcopenia to prepare for an upcoming aging society.


Asunto(s)
Huesos , Osteoporosis , Ligando RANK , Sarcopenia , Transducción de Señal , Humanos , Sarcopenia/metabolismo , Ligando RANK/metabolismo , Huesos/metabolismo , Osteoporosis/metabolismo , Transducción de Señal/fisiología , Animales , Músculo Esquelético/metabolismo , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Envejecimiento/metabolismo , Envejecimiento/fisiología
18.
Int J Mol Sci ; 25(11)2024 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-38891922

RESUMEN

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.


Asunto(s)
Músculo Liso Vascular , Ligando RANK , Receptores Acoplados a Proteínas G , Calcificación Vascular , Ligando RANK/metabolismo , Ligando RANK/genética , Animales , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/genética , Calcificación Vascular/metabolismo , Calcificación Vascular/patología , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Ratas , Humanos , Masculino , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Osteoprotegerina/metabolismo , Osteoprotegerina/genética , Hormona Paratiroidea/metabolismo , Células Cultivadas , Ratas Sprague-Dawley
19.
Pflugers Arch ; 476(8): 1289-1302, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38833170

RESUMEN

Osteoclasts are multinucleated cells of hematopoietic origin, with a pivotal role in bone development and remodeling. Failure in osteoclast differentiation and activation leads to various bone disorders; thus, attention has focused on a search of molecules involved in osteoclast regulatory pathways. Caspase-8 appears to be an interesting candidate for further exploration, due to its potential function in bone development and homeostasis. Mouse bone marrow cells were differentiated into osteoclasts by RANKL stimulation. Increased activation of caspase-8 and its downstream executioner caspases (caspase-3 and caspase-6) was found during osteoclastogenesis. Subsequent inhibition of caspase-8, caspase-3, or caspase-6, respectively, during osteoclast differentiation showed distinct changes in the formation of TRAP-positive multinucleated cells and reduced expression of osteoclast markers including Acp5, Ctsk, Dcstamp, and Mmp9. Analysis of bone matrix resorption confirmed significantly reduced osteoclast function after caspase inhibition. The results clearly showed the role of caspases in the proper development of osteoclasts and contributed new knowledge about non-apoptotic function of caspases.


Asunto(s)
Células de la Médula Ósea , Inhibidores de Caspasas , Diferenciación Celular , Osteoclastos , Ligando RANK , Animales , Ratones , Células de la Médula Ósea/metabolismo , Resorción Ósea/metabolismo , Caspasa 3/metabolismo , Caspasa 6/metabolismo , Caspasa 8/metabolismo , Inhibidores de Caspasas/farmacología , Células Cultivadas , Ratones Endogámicos C57BL , Osteoclastos/metabolismo , Ligando RANK/metabolismo , Fosfatasa Ácida Tartratorresistente/metabolismo
20.
Arch Gynecol Obstet ; 310(2): 1223-1233, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38836929

RESUMEN

PURPOSE: The receptor activator of nuclear factor kappa B (RANK) and its ligand (RANKL) have been shown to promote proliferation of the breast and breast carcinogenesis. The objective of this analysis was to investigate whether tumor-specific RANK and RANKL expression in patients with primary breast cancer is associated with high percentage mammographic density (PMD), which is a known breast cancer risk factor. METHODS: Immunohistochemical staining of RANK and RANKL was performed in tissue microarrays (TMAs) from primary breast cancer samples of the Bavarian Breast Cancer Cases and Controls (BBCC) study. For RANK and RANKL expression, histochemical scores (H scores) with a cut-off value of > 0 vs 0 were established. PMD was measured in the contralateral, non-diseased breast. Linear regression models with PMD as outcome were calculated using common predictors of PMD (age at breast cancer diagnosis, body mass index (BMI) and parity) and RANK and RANKL H scores. Additionally, Spearman rank correlations (ρ) between PMD and RANK and RANKL H score were performed. RESULTS: In the final cohort of 412 patients, breast cancer-specific RANK and RANKL expression was not associated with PMD (P = 0.68). There was no correlation between PMD and RANK H score (Spearman's ρ = 0.01, P = 0.87) or RANKL H score (Spearman's ρ = 0.04, P = 0.41). RANK expression was highest in triple-negative tumors, followed by HER2-positive, luminal B-like and luminal A-like tumors, while no subtype-specific expression of RANKL was found. CONCLUSION: Results do not provide evidence for an association of RANK and RANKL expression in primary breast cancer with PMD.


Asunto(s)
Densidad de la Mama , Neoplasias de la Mama , Ligando RANK , Receptor Activador del Factor Nuclear kappa-B , Humanos , Ligando RANK/metabolismo , Ligando RANK/análisis , Femenino , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Neoplasias de la Mama/diagnóstico por imagen , Persona de Mediana Edad , Anciano , Adulto , Estudios de Casos y Controles , Inmunohistoquímica , Análisis de Matrices Tisulares , Mama/diagnóstico por imagen , Mama/patología , Mama/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...