Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25.355
Filter
3.
Bone Res ; 12(1): 29, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38744829

ABSTRACT

Mature osteoclasts degrade bone matrix by exocytosis of active proteases from secretory lysosomes through a ruffled border. However, the molecular mechanisms underlying lysosomal trafficking and secretion in osteoclasts remain largely unknown. Here, we show with GeneChip analysis that RUN and FYVE domain-containing protein 4 (RUFY4) is strongly upregulated during osteoclastogenesis. Mice lacking Rufy4 exhibited a high trabecular bone mass phenotype with abnormalities in osteoclast function in vivo. Furthermore, deleting Rufy4 did not affect osteoclast differentiation, but inhibited bone-resorbing activity due to disruption in the acidic maturation of secondary lysosomes, their trafficking to the membrane, and their secretion of cathepsin K into the extracellular space. Mechanistically, RUFY4 promotes late endosome-lysosome fusion by acting as an adaptor protein between Rab7 on late endosomes and LAMP2 on primary lysosomes. Consequently, Rufy4-deficient mice were highly protected from lipopolysaccharide- and ovariectomy-induced bone loss. Thus, RUFY4 plays as a new regulator in osteoclast activity by mediating endo-lysosomal trafficking and have a potential to be specific target for therapies against bone-loss diseases such as osteoporosis.


Subject(s)
Endosomes , Lysosomes , Osteoclasts , Animals , Osteoclasts/metabolism , Lysosomes/metabolism , Endosomes/metabolism , Mice , Mice, Knockout , Bone Resorption/metabolism , Bone Resorption/pathology , Bone Resorption/genetics , Protein Transport , Mice, Inbred C57BL , rab GTP-Binding Proteins/metabolism , rab GTP-Binding Proteins/genetics , Cell Differentiation , Gene Deletion , Cathepsin K/metabolism , Cathepsin K/genetics , Female , rab7 GTP-Binding Proteins
4.
J Med Chem ; 67(10): 8271-8295, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38717088

ABSTRACT

A series of heterocyclic ring-fused derivatives of bisnoralcohol (BA) were synthesized and evaluated for their inhibitory effects on RANKL-induced osteoclastogenesis. Most of these derivatives possessed potent antiosteoporosis activities in a dose-dependent manner. Among these compounds, 31 (SH442, IC50 = 0.052 µM) exhibited the highest potency, displaying 100% inhibition at 1.0 µM and 82.8% inhibition at an even lower concentration of 0.1 µM, which was much more potent than the lead compound BA (IC50 = 2.325 µM). Cytotoxicity tests suggested that the inhibitory effect of these compounds on RANKL-induced osteoclast differentiation did not result from their cytotoxicity. Mechanistic studies revealed that SH442 inhibited the expression of osteoclastogenesis-related marker genes and proteins, including TRAP, TRAF6, c-Fos, CTSK, and MMP9. Especially, SH442 could significantly attenuate bone loss of ovariectomy mouse in vivo. Therefore, these BA derivatives could be used as promising leads for the development of a new type of antiosteoporosis agent.


Subject(s)
Osteoclasts , Osteoporosis , Animals , Female , Mice , Bone Resorption/drug therapy , Cell Differentiation/drug effects , Coumarins/pharmacology , Coumarins/chemistry , Coumarins/chemical synthesis , Heterocyclic Compounds/pharmacology , Heterocyclic Compounds/chemistry , Heterocyclic Compounds/chemical synthesis , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteogenesis/drug effects , Osteoporosis/drug therapy , Ovariectomy , RANK Ligand/metabolism , RANK Ligand/antagonists & inhibitors , RAW 264.7 Cells , Small Molecule Libraries/pharmacology , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Structure-Activity Relationship
5.
Int J Mol Sci ; 25(10)2024 May 08.
Article in English | MEDLINE | ID: mdl-38791156

ABSTRACT

The deterioration of osteoblast-led bone formation and the upregulation of osteoclast-regulated bone resorption are the primary causes of bone diseases, including osteoporosis. Numerous circulating factors play a role in bone homeostasis by regulating osteoblast and osteoclast activity, including the sphingolipid-sphingosine-1-phosphate (S1P). However, to date no comprehensive studies have investigated the impact of S1P activity on human and murine osteoblasts and osteoclasts. We observed species-specific responses to S1P in both osteoblasts and osteoclasts, where S1P stimulated human osteoblast mineralisation and reduced human pre-osteoclast differentiation and mineral resorption, thereby favouring bone formation. The opposite was true for murine osteoblasts and osteoclasts, resulting in more mineral resorption and less mineral deposition. Species-specific differences in osteoblast responses to S1P were potentially explained by differential expression of S1P receptor 1. By contrast, human and murine osteoclasts expressed comparable levels of S1P receptors but showed differential expression patterns of the two sphingosine kinase enzymes responsible for S1P production. Ultimately, we reveal that murine models may not accurately represent how human bone cells will respond to S1P, and thus are not a suitable model for exploring S1P physiology or potential therapeutic agents.


Subject(s)
Cell Differentiation , Lysophospholipids , Osteoblasts , Osteoclasts , Species Specificity , Sphingosine , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Lysophospholipids/metabolism , Humans , Animals , Mice , Osteoclasts/metabolism , Osteoclasts/cytology , Osteoblasts/metabolism , Osteoblasts/drug effects , Osteogenesis/drug effects , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Sphingosine-1-Phosphate Receptors/metabolism , Bone and Bones/metabolism , Bone Resorption/metabolism , Cells, Cultured
6.
Eur J Pharmacol ; 974: 176630, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38692426

ABSTRACT

Osteoporosis is the most common bone disorder, in which an imbalance between osteoclastic bone resorption and osteoblastic bone formation disrupts bone homeostasis. Osteoporosis management using anti-osteoclastic agents is a promising strategy; however, this remains an unmet need. Sphingosine-1-phosphate (S1P) and its receptors (S1PRs) are essential for maintaining bone homeostasis. Here, we identified that Siponimod, a Food and Drug Administration-approved S1PR antagonist for the treatment of multiple sclerosis, shows promising therapeutic effects against osteoporosis by inhibiting osteoclast formation and function. We found that Siponimod inhibited osteoclast formation in a dose-dependent manner without causing cytotoxicity. Podosome belt staining and bone resorption assays indicated that Siponimod treatment impaired osteoclast function. Western blot and qPCR assays demonstrated that Siponimod suppressed the expression of osteoclast-specific markers, including C-Fos, Nftac1, and Ctsk. Mechanistically, we validated that Siponimod downregulated receptor activator of nuclear factor kappa B ligand (RANKL)-induced Mitogen-activated protein kinases (MAPKs) and nuclear factor kappa B (NF-κB) signaling pathways during osteoclastogenesis. Moreover, in a preclinical mouse model, Siponimod prevented ovariectomy-induced bone loss by suppressing osteoclast activity in vivo. Collectively, these results suggest that Siponimod could serve as an alternative therapeutic agent for the treatment of osteoporosis.


Subject(s)
Azetidines , Benzyl Compounds , Drug Repositioning , Multiple Sclerosis , Osteoclasts , Osteoporosis , Animals , Mice , Osteoporosis/drug therapy , Osteoporosis/metabolism , Osteoclasts/drug effects , Osteoclasts/metabolism , Benzyl Compounds/pharmacology , Benzyl Compounds/therapeutic use , Azetidines/pharmacology , Azetidines/therapeutic use , Multiple Sclerosis/drug therapy , Female , Sphingosine 1 Phosphate Receptor Modulators/pharmacology , Sphingosine 1 Phosphate Receptor Modulators/therapeutic use , Osteogenesis/drug effects , NF-kappa B/metabolism , Mice, Inbred C57BL , RAW 264.7 Cells , Bone Resorption/drug therapy , Signal Transduction/drug effects , RANK Ligand/metabolism , Humans
7.
Biomed Pharmacother ; 175: 116732, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38739990

ABSTRACT

Osteoporosis is a systemic bone disease characterized by decreased bone mass that is tightly regulated by the coordinated actions of osteoclasts and osteoblasts. Apoptosis as a precise programmed cell death involves a cascade of gene expression events which are mechanistically linked to the regulation of bone metabolism. Nevertheless, the critical biomolecules involved in regulating cell apoptosis in osteoporosis remain unknown. To gain a deeper insight into the relationship between apoptosis and osteoporosis, this study integrated the sequencing results of human samples and using a machine learning workflow to overcome the limitations of a single study. Among all immune cell populations, we assessed the apoptotic level and portrayed the distinct subtypes and lineage differentiation of monocytic cells in osteoporotic tissues. Osteoclasts expressed a higher level of Spermidine/spermine-N1-Acetyltransferase1 (SAT1) during osteoclastogenesis which prevented osteoclasts apoptosis and facilitate osteoporosis progression. In addition, Berenil, one potent SAT1 inhibitor, increased osteoclast apoptosis and reversed the bone loss in the femurs of a murine ovariectomy model. In summary, Berenil promotes osteoclast apoptosis, inhibits the bone resorption and improves the abnormal bone structure in vitro and in vivo models by targeting SAT1, demonstrating its potential as a precise therapeutic strategy for clinical osteoporosis treatment.


Subject(s)
Acetyltransferases , Apoptosis , Osteoclasts , Osteoporosis , Apoptosis/drug effects , Animals , Osteoclasts/metabolism , Osteoclasts/pathology , Osteoclasts/drug effects , Osteoporosis/pathology , Osteoporosis/prevention & control , Osteoporosis/metabolism , Humans , Female , Mice , Acetyltransferases/metabolism , Acetyltransferases/genetics , Mice, Inbred C57BL , Bone Resorption/metabolism , Bone Resorption/pathology , Bone Resorption/prevention & control , Ovariectomy , Osteogenesis/drug effects , Cell Differentiation , Disease Models, Animal
8.
Cell Rep Med ; 5(5): 101574, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38776873

ABSTRACT

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.


Subject(s)
Bone Resorption , Osteoblasts , Osteogenesis , Animals , Humans , Osteoblasts/metabolism , Osteoblasts/drug effects , Osteogenesis/drug effects , Mice , Bone Resorption/pathology , Bone Resorption/metabolism , Anabolic Agents/pharmacology , Anabolic Agents/therapeutic use , Bone Remodeling/drug effects , Osteoporosis/pathology , Osteoporosis/metabolism , Osteoporosis/drug therapy , RANK Ligand/metabolism , Osteoclasts/metabolism , Osteoclasts/drug effects , Bone Development/drug effects , Osteoprotegerin/metabolism , Female , Signal Transduction/drug effects , Peptides/pharmacology , Male , Mice, Inbred C57BL , Bone and Bones/drug effects , Bone and Bones/metabolism , Bone and Bones/pathology
9.
Aging (Albany NY) ; 16(10): 9251-9263, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38809508

ABSTRACT

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.


Subject(s)
Cell Differentiation , Methylamines , NF-kappa B , Osteoclasts , Oxidative Stress , RANK Ligand , Signal Transduction , Animals , Osteoclasts/drug effects , Osteoclasts/metabolism , Mice , Methylamines/pharmacology , Oxidative Stress/drug effects , Cell Differentiation/drug effects , RAW 264.7 Cells , NF-kappa B/metabolism , RANK Ligand/metabolism , Signal Transduction/drug effects , Macrophage Colony-Stimulating Factor/metabolism , Macrophage Colony-Stimulating Factor/pharmacology , Bone Resorption/metabolism
10.
Commun Biol ; 7(1): 548, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38719881

ABSTRACT

Hyperthyroidism is a well-known trigger of high bone turnover that can lead to the development of secondary osteoporosis. Previously, we have shown that blocking bone morphogenetic protein (BMP) signaling systemically with BMPR1A-Fc can prevent bone loss in hyperthyroid mice. To distinguish between bone cell type-specific effects, conditional knockout mice lacking Bmpr1a in either osteoclast precursors (LysM-Cre) or osteoprogenitors (Osx-Cre) were rendered hyperthyroid and their bone microarchitecture, strength and turnover were analyzed. While hyperthyroidism in osteoclast precursor-specific Bmpr1a knockout mice accelerated bone resorption leading to bone loss just as in wildtype mice, osteoprogenitor-specific Bmpr1a deletion prevented an increase of bone resorption and thus osteoporosis with hyperthyroidism. In vitro, wildtype but not Bmpr1a-deficient osteoblasts responded to thyroid hormone (TH) treatment with increased differentiation and activity. Furthermore, we found an elevated Rankl/Opg ratio with TH excess in osteoblasts and bone tissue from wildtype mice, but not in Bmpr1a knockouts. In line, expression of osteoclast marker genes increased when osteoclasts were treated with supernatants from TH-stimulated wildtype osteoblasts, in contrast to Bmpr1a-deficient cells. In conclusion, we identified the osteoblastic BMP receptor BMPR1A as a main driver of osteoporosis in hyperthyroid mice promoting TH-induced osteoblast activity and potentially its coupling to high osteoclastic resorption.


Subject(s)
Bone Morphogenetic Protein Receptors, Type I , Bone Resorption , Hyperthyroidism , Osteoblasts , Animals , Male , Mice , Bone Morphogenetic Protein Receptors, Type I/genetics , Bone Morphogenetic Protein Receptors, Type I/metabolism , Bone Resorption/metabolism , Bone Resorption/genetics , Cell Differentiation , Hyperthyroidism/metabolism , Hyperthyroidism/genetics , Hyperthyroidism/complications , Mice, Knockout , Osteoblasts/metabolism , Osteoclasts/metabolism , Osteoporosis/metabolism , Osteoporosis/genetics , Osteoporosis/etiology , Osteoporosis/pathology
11.
JCI Insight ; 9(10)2024 May 22.
Article in English | MEDLINE | ID: mdl-38713511

ABSTRACT

While sclerostin-neutralizing antibodies (Scl-Abs) transiently stimulate bone formation by activating Wnt signaling in osteoblast lineage cells, they exert sustained inhibition of bone resorption, suggesting an alternate signaling pathway by which Scl-Abs control osteoclast activity. Since sclerostin can activate platelet-derived growth factor receptors (PDGFRs) in osteoblast lineage cells in vitro and PDGFR signaling in these cells induces bone resorption through M-CSF secretion, we hypothesized that the prolonged anticatabolic effect of Scl-Abs could result from PDGFR inhibition. We show here that inhibition of PDGFR signaling in osteoblast lineage cells is sufficient and necessary to mediate prolonged Scl-Ab effects on M-CSF secretion and osteoclast activity in mice. Indeed, sclerostin coactivates PDGFRs independently of Wnt/ß-catenin signaling inhibition, by forming a ternary complex with LRP6 and PDGFRs in preosteoblasts. In turn, Scl-Ab prevents sclerostin-mediated coactivation of PDGFR signaling and consequent M-CSF upregulation in preosteoblast cultures, thereby inhibiting osteoclast activity in preosteoblast/osteoclast coculture assays. These results provide a potential mechanism explaining the dissociation between anabolic and antiresorptive effects of long-term Scl-Ab.


Subject(s)
Adaptor Proteins, Signal Transducing , Bone Resorption , Osteoblasts , Osteoclasts , Receptors, Platelet-Derived Growth Factor , Signal Transduction , Animals , Osteoblasts/metabolism , Mice , Adaptor Proteins, Signal Transducing/metabolism , Bone Resorption/metabolism , Osteoclasts/metabolism , Receptors, Platelet-Derived Growth Factor/metabolism , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Wnt Signaling Pathway/drug effects , Antibodies, Neutralizing/pharmacology , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Macrophage Colony-Stimulating Factor/metabolism , Cell Lineage , Osteogenesis/drug effects , Cell Differentiation
12.
Acta Odontol Scand ; 83: 132-139, 2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38597918

ABSTRACT

OBJECTIVE: Knowledge about oral hygiene, gingival bleeding, mineral density, and resorption of jaw bones in patients with hemophilia is limited. We evaluated the periodontal and bone status in such patients.  Material and methods: Forty-eight patients with severe type A/B hemophilia and 49 age- and sex-matched controls were included. Assessments included simplified oral hygiene index (OHI-S), calculus index, debris index, gingival index (GI), gingival bleeding time index (GBTI), and decayed, missing, and filled teeth index (DMFTI). Bone resorption was evaluated using panoramic mandibular index (PMI), mental index (MI), and alveolar crest ratio (ACR). Mineral density in the condyle, angulus, and premolar areas was assessed using fractal analysis, with fractal dimensions denoted as condyle fractal dimension (CFD) for the condyle, angulus fractal dimension (AFD) for angulus, and premolar fractal dimension (PFD) for premolar region. RESULTS: The mean scores were DMFTI = 11.77, OHI-S = 2.44, PMI = 0.268, MI = 5.822, GI = 3.02, GBTI = 2.64, ACR = 2.06, CFD = 1.31, AFD = 1.31, and PFD = 1.17 in the hemophilia group and DMFTI = 11.449, PMI = 0.494, MI = 7.43, GI = 0.67, GBTI = 0.98, OHI-S = 1.45, ACR = 2.87, CFD = 1.35, AFD = 1.35, and PDF = 1.23 in the control group. Differences were significant for all parameters (p < 0.005) except for the DMFTI index.  Conclusions: Because of poor oral hygiene, high bone resorption, and low bone mineral density in these patients, clinicians should consider potential bone changes when planning to treat these patients.


Subject(s)
Bone Resorption , Hemophilia A , Humans , Bone Density , Oral Health , Hemophilia A/complications , Case-Control Studies , Minerals
13.
Sci Rep ; 14(1): 8109, 2024 04 06.
Article in English | MEDLINE | ID: mdl-38582757

ABSTRACT

Bone resorption is highly dependent on the dynamic rearrangement of the osteoclast actin cytoskeleton to allow formation of actin rings and a functional ruffled border. Hem1 is a hematopoietic-specific subunit of the WAVE-complex which regulates actin polymerization and is crucial for lamellipodia formation in hematopoietic cell types. However, its role in osteoclast differentiation and function is still unknown. Here, we show that although the absence of Hem1 promotes osteoclastogenesis, the ability of Hem1-/- osteoclasts to degrade bone was severely impaired. Global as well as osteoclast-specific deletion of Hem1 in vivo revealed increased femoral trabecular bone mass despite elevated numbers of osteoclasts in vivo. We found that the resorption defect derived from the morphological distortion of the actin-rich sealing zone and ruffled border deformation in Hem1-deficient osteoclasts leading to impaired vesicle transport and increased intracellular acidification. Collectively, our data identify Hem1 as a yet unknown key player in bone remodeling by regulating ruffled border formation and consequently the resorptive capacity of osteoclasts.


Subject(s)
Bone Resorption , Osteoclasts , Humans , Osteoclasts/metabolism , Actins/metabolism , Bone Resorption/metabolism , Bone and Bones/metabolism , Osteogenesis
14.
J Nanobiotechnology ; 22(1): 153, 2024 Apr 05.
Article in English | MEDLINE | ID: mdl-38580995

ABSTRACT

BACKGROUND: Osteoporosis is characterized by an imbalance in bone homeostasis, resulting in the excessive dissolution of bone minerals due to the acidified microenvironment mediated by overactive osteoclasts. Oroxylin A (ORO), a natural flavonoid, has shown potential in reversing osteoporosis by inhibiting osteoclast-mediated bone resorption. The limited water solubility and lack of targeting specificity hinder the effective accumulation of Oroxylin A within the pathological environment of osteoporosis. RESULTS: Osteoclasts' microenvironment-responsive nanoparticles are prepared by incorporating Oroxylin A with amorphous calcium carbonate (ACC) and coated with glutamic acid hexapeptide-modified phospholipids, aiming at reinforcing the drug delivery efficiency as well as therapeutic effect. The obtained smart nanoparticles, coined as OAPLG, could instantly neutralize acid and release Oroxylin A in the extracellular microenvironment of osteoclasts. The combination of Oroxylin A and ACC synergistically inhibits osteoclast formation and activity, leading to a significant reversal of systemic bone loss in the ovariectomized mice model. CONCLUSION: The work highlights an intelligent nanoplatform based on ACC for spatiotemporally controlled release of lipophilic drugs, and illustrates prominent therapeutic promise against osteoporosis.


Subject(s)
Bone Resorption , Osteoporosis , Mice , Animals , Osteoclasts , Nanomedicine , Osteoporosis/drug therapy , Bone Resorption/drug therapy , Bone and Bones/pathology , Cell Differentiation
15.
Sci Rep ; 14(1): 8153, 2024 04 08.
Article in English | MEDLINE | ID: mdl-38589566

ABSTRACT

Osteoporosis is usually caused by excessive bone resorption and energy metabolism plays a critical role in the development of osteoporosis. However, little is known about the role of energy metabolism-related genes in osteoporosis. This study aimed to explore the important energy metabolism-related genes involved in the development of osteoporosis and develop a diagnosis signature for osteoporosis. The GSE56814, GSE62402, and GSE7158 datasets were downloaded from the NCBI Gene Expression Omnibus. The intersection of differentially expressed genes between high and low levels of body mineral density (BMD) and genes related to energy metabolism were screened as differentially expressed energy metabolism genes (DE-EMGs). Subsequently, a DE-EMG-based diagnostic model was constructed and differential expression of genes in the model was validated by RT-qPCR. Furthermore, a receiver operating characteristic curve and nomogram model were constructed to evaluate the predictive ability of the diagnostic model. Finally, the immune cell types in the merged samples and networks associated with the selected optimal DE-EMGs were constructed. A total of 72 overlapped genes were selected as DE-EMGs, and a five DE-EMG based diagnostic model consisting B4GALT4, ADH4, ACAD11, B4GALT2, and PPP1R3C was established. The areas under the curve of the five genes in the merged training dataset and B4GALT2 in the validation dataset were 0.784 and 0.790, respectively. Moreover, good prognostic prediction ability was observed using the nomogram model (C index = 0.9201; P = 5.507e-14). Significant differences were observed in five immune cell types between the high- and low-BMD groups. These included central memory, effector memory, and activated CD8 T cells, as well as regulatory T cells and activated B cells. A network related to DE-EMGs was constructed, including hsa-miR-23b-3p, DANCR, 17 small-molecule drugs, and two Kyoto Encyclopedia of Genes and Genomes pathways, including metabolic pathways and pyruvate metabolism. Our findings highlighted the important roles of DE-EMGs in the development of osteoporosis. Furthermore, the DANCR/hsa-miR-23b-3p/B4GALT4 axis might provide novel molecular insights into the process of osteoporosis development.


Subject(s)
Bone Resorption , MicroRNAs , Osteoporosis , Humans , B-Lymphocytes , Osteoporosis/diagnosis , Osteoporosis/genetics , Energy Metabolism/genetics
16.
J Orthop Surg Res ; 19(1): 243, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38622659

ABSTRACT

Inflammatory reactions are involved in the development of steroid-induced osteonecrosis of the femoral head(ONFH). Studies have explored the therapeutic efficacy of inhibiting inflammatory reactions in steroid-induced ONFH and revealed that inhibiting inflammation may be a new strategy for preventing the development of steroid-induced ONFH. Exosomes derived from M2 macrophages(M2-Exos) display anti-inflammatory properties. This study aimed to examine the preventive effect of M2-Exos on early-stage steroid-induced ONFH and explore the underlying mechanisms involved. In vitro, we explored the effect of M2-Exos on the proliferation and osteogenic differentiation of bone marrow-derived mesenchymal stem cells(BMMSCs). In vivo, we investigated the role of M2-Exos on inflammation, osteoclastogenesis, osteogenesis and angiogenesis in an early-stage rat model of steroid-induced ONFH. We found that M2-Exos promoted the proliferation and osteogenic differentiation of BMMSCs. Additionally, M2-Exos effectively attenuated the osteonecrotic changes, inhibited the expression of proinflammatory mediators, promoted osteogenesis and angiogenesis, reduced osteoclastogenesis, and regulated the polarization of M1/M2 macrophages in steroid-induced ONFH. Taken together, our data suggest that M2-Exos are effective at preventing steroid-induced ONFH. These findings may be helpful for providing a potential strategy to prevent the development of steroid-induced ONFH.


Subject(s)
Bone Resorption , Exosomes , Femur Head Necrosis , Osteonecrosis , Rats , Animals , Osteogenesis , Exosomes/metabolism , Femur Head/metabolism , Osteonecrosis/prevention & control , Inflammation/metabolism , Macrophages/metabolism , Steroids/adverse effects , Femur Head Necrosis/chemically induced , Femur Head Necrosis/prevention & control , Femur Head Necrosis/metabolism
17.
Proc Biol Sci ; 291(2021): 20232738, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38628118

ABSTRACT

Midfacial morphology varies between hominoids, in particular between great apes and humans for which the face is small and retracted. The underlying developmental processes for these morphological differences are still largely unknown. Here, we investigate the cellular mechanism of maxillary development (bone modelling, BM), and how potential changes in this process may have shaped facial evolution. We analysed cross-sectional developmental series of gibbons, orangutans, gorillas, chimpanzees and present-day humans (n = 183). Individuals were organized into five age groups according to their dental development. To visualize each species's BM pattern and corresponding morphology during ontogeny, maps based on microscopic data were mapped onto species-specific age group average shapes obtained using geometric morphometrics. The amount of bone resorption was quantified and compared between species. Great apes share a highly similar BM pattern, whereas gibbons have a distinctive resorption pattern. This suggests a change in cellular activity on the hominid branch. Humans possess most of the great ape pattern, but bone resorption is high in the canine area from birth on, suggesting a key role of canine reduction in facial evolution. We also observed that humans have high levels of bone resorption during childhood, a feature not shared with other apes.


Subject(s)
Bone Resorption , Hominidae , Animals , Humans , Hominidae/anatomy & histology , Hylobates , Cross-Sectional Studies , Gorilla gorilla , Pan troglodytes , Morphogenesis , Biological Evolution
18.
Hua Xi Kou Qiang Yi Xue Za Zhi ; 42(2): 163-171, 2024 Apr 01.
Article in English, Chinese | MEDLINE | ID: mdl-38597076

ABSTRACT

OBJECTIVES: To investigate the mechanism of circadian clock protein Bmal1 (Bmal1) on renal injury with chronic periodontitis, we established an experimental rat periodontitis model. METHODS: Twelve male Wistar rats were randomly divided into control and periodontitis groups (n=6, each group). The first maxillary molars on both sides of the upper jaw of rats with periodontitis were ligated by using orthodontic ligature wires, whereas the control group received no intervention measures. After 8 weeks, clinical periodontal parameters, including probing depth, bleeding index, and tooth mobility, were evaluated in both groups. Micro-CT scanning and three-dimensional image reconstruction were performed on the maxillary bones of the rats for the assessment of alveolar bone resorption. Histopatholo-gical observations of periodontal and renal tissues were conducted using hematoxylin-eosin (HE) and periodic acid-Schiff (PAS) staining. Renal function indicators, such as creatinine, albumin, and blood urea nitrogen levels, and oxidative stress markers, including superoxide dismutase, glutathione, and malondialdehyde levels, were measured using biochemical assay kits. MitoSOX red staining was used to detect reactive oxygen species (ROS) content in the kidneys. The gene and protein expression levels of Bmal1, nuclear factor erythroid 2-related factor 2 (Nrf2), and heme oxygenase-1 (HO-1) in rat renal tissues were assessed using real-time quantitative polymerase chain reaction (RT-qPCR) and immunohistochemical staining. RESULTS: Micro-CT and HE staining results showed significant bone resorption and attachment loss in the maxillary first molar region of the periodontitis group. Histological examination through HE and PAS staining revealed substantial histopathological damage to the renal tissues of the rats in the periodontitis group. The findings of the assessment of renal function and oxidative stress markers indicated that the periodontitis group exhibited abnormal levels of oxidative stress, whereas the renal function levels showed abnormalities without statistical significance. MitoSOX Red staining results showed that the content of ROS in the renal tissue of the periodontitis group was significantly higher than that of the control group, and RT-qPCR and immunohistochemistry results showed that the expression levels of Bmal1, Nrf2, and HO-1 in the renal tissues of the rats in the periodontitis group showed a decreasing trend. CONCLUSIONS: Circadian clock protein Bmal1 plays an important role in the oxidative damage process involved in the renal of rats with periodontitis.


Subject(s)
Bone Resorption , Circadian Clocks , Organophosphorus Compounds , Periodontitis , Phenanthridines , Animals , Male , Rats , Bone Resorption/metabolism , Kidney/metabolism , Kidney/pathology , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Periodontitis/metabolism , Rats, Wistar , Reactive Oxygen Species/metabolism
19.
Biochem Biophys Res Commun ; 710: 149860, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38604070

ABSTRACT

Schizophyllan (SPG), a ß-glucan from Schizophyllum commune, is recognized for its antioxidant, immunoregulatory, and anticancer activities. In this study, its effects on bone cells, particularly osteoclasts and osteoblasts, were examined. We demonstrated that SPG dose-dependently inhibited osteoclastogenesis and reduced gene expression associated with osteoclast differentiation. SPG also decreased bone resorption and F-actin ring formation. This inhibition could have been due to the downregulation of transcription factors c-Fos and nuclear factor of activated T cells 1 (NFATc1) via the MAPKs (JNK and p38), IκBα, and PGC1ß/PPARγ pathways. In coculture, SPG lowered osteoclastogenic activity in calvaria-derived osteoblasts by reducing macrophage colony-stimulating factor (M-CSF) and receptor activator of nuclear factor-κB ligand (RANKL) expression. In addition, SPG slightly enhanced osteoblast differentiation, as evidenced by increased differentiation marker gene expression and alizarin red staining. It also exhibited antiresorptive effects in a lipopolysaccharide-induced calvarial bone loss model. These results indicated a dual role of SPG in bone cell regulation by suppressing osteoclastogenesis and promoting osteoblast differentiation. Thus, SPG could be a therapeutic agent for bone resorption-related diseases such as osteoporosis, rheumatoid arthritis, and periodontitis.


Subject(s)
Bone Resorption , Sizofiran , Humans , Osteoclasts/metabolism , Sizofiran/metabolism , Sizofiran/pharmacology , NFATC Transcription Factors/metabolism , Osteoblasts/metabolism , Cell Differentiation , Bone Resorption/drug therapy , Bone Resorption/metabolism , Osteogenesis , RANK Ligand/metabolism
20.
Front Immunol ; 15: 1168323, 2024.
Article in English | MEDLINE | ID: mdl-38566990

ABSTRACT

Background: Myeloid-derived suppressor cells (MDSCs) are heterogeneous immature myeloid cells with immunosuppressive functions. It is known that MDSCs are expanded at inflammatory sites after migrating from bone marrow (BM) or spleen (Sp). In chronic inflammatory diseases such as rheumatoid arthritis (RA), previous reports indicate that MDSCs are increased in BM and Sp, but detailed analysis of MDSCs in inflamed joints is very limited. Objective: The purpose of this study is to characterize the MDSCs in the joints of mice with autoimmune arthritis. Methods: We sorted CD11b+Gr1+ cells from joints (Jo), bone marrow (BM) and spleen (Sp) of SKG mice with zymosan (Zym)-induced arthritis and investigated differentially expressed genes (DEGs) by microarray analysis. Based on the identified DEGs, we assessed the suppressive function of CD11b+Gr1+ cells from each organ and their ability to differentiate into osteoclasts. Results: We identified MDSCs as CD11b+Gr1+ cells by flow cytometry and morphological analysis. Microarray analysis revealed that Jo-CD11b+Gr1+ cells had different characteristics compared with BM-CD11b+Gr1+ cells or Sp-CD11b+Gr1+ cells. Microarray and qPCR analysis showed that Jo-CD11b+Gr1+ cells strongly expressed immunosuppressive DEGs (Pdl1, Arg1, Egr2 and Egr3). Jo-CD11b+Gr1+ cells significantly suppressed CD4+ T cell proliferation and differentiation in vitro, which confirmed Jo-CD11b+Gr1+ cells as MDSCs. Microarray analysis also revealed that Jo-MDSCs strongly expressed DEGs of the NF-κB non-canonical pathway (Nfkb2 and Relb), which is relevant for osteoclast differentiation. In fact, Jo-MDSCs differentiated into osteoclasts in vitro and they had bone resorptive function. In addition, intra-articular injection of Jo-MDSCs promoted bone destruction. Conclusions: Jo-MDSCs possess a potential to differentiate into osteoclasts which promote bone resorption in inflamed joints, while they are immunosuppressive in vitro.


Subject(s)
Arthritis , Bone Resorption , Myeloid-Derived Suppressor Cells , Mice , Animals , Osteoclasts , Myeloid Cells , Bone Resorption/metabolism , Arthritis/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...