Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.284
Filtrar
1.
Commun Biol ; 7(1): 548, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38719881

RESUMEN

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.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas de Tipo 1 , Resorción Ósea , Hipertiroidismo , Ratones Noqueados , Osteoblastos , Animales , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/genética , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/metabolismo , Osteoblastos/metabolismo , Hipertiroidismo/metabolismo , Hipertiroidismo/genética , Hipertiroidismo/complicaciones , Ratones , Resorción Ósea/metabolismo , Resorción Ósea/genética , Osteoporosis/metabolismo , Osteoporosis/genética , Osteoporosis/etiología , Osteoporosis/patología , Osteoclastos/metabolismo , Masculino , Diferenciación Celular
2.
Stem Cell Res Ther ; 15(1): 144, 2024 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-38764077

RESUMEN

BACKGROUND: The aim of this study was to evaluate potential synergistic effects of a single, local application of human umbilical cord MSC-derived sEVs in combination with a low dose of recombinant human rhBMP-2 to promote the regeneration of a metaphyseal femoral defect in an osteoporotic rat model. METHODS: 6 weeks after induction of osteoporosis by bilateral ventral ovariectomy and administration of a special diet, a total of 64 rats underwent a distal femoral metaphyseal osteotomy using a manual Gigli wire saw. Defects were stabilized with an adapted Y-shaped mini-locking plate and were subsequently treated with alginate only, or alginate loaded with hUC-MSC-sEVs (2 × 109), rhBMP-2 (1.5 µg), or a combination of sEVs and rhBMP-2 (n = 16 for each group). 6 weeks post-surgery, femora were evaluated by µCT, descriptive histology, and biomechanical testing. RESULTS: Native radiographs and µCT analysis confirmed superior bony union with callus formation after treatment with hUC-MSC-sEVs in combination with a low dose of rhBMP-2. This finding was further substantiated by histology, showing robust defect consolidation 6 weeks after treatment. Torsion testing of the explanted femora revealed increased stiffness after application of both, rhBMP-2 alone, or in combination with sEVs, whereas torque was only significantly increased after treatment with rhBMP-2 together with sEVs. CONCLUSION: The present study demonstrates that the co-application of hUC-MSC-sEVs can improve the efficacy of rhBMP-2 to promote the regeneration of osteoporotic bone defects.


Asunto(s)
Proteína Morfogenética Ósea 2 , Vesículas Extracelulares , Fémur , Osteoporosis , Proteínas Recombinantes , Cordón Umbilical , Animales , Proteína Morfogenética Ósea 2/farmacología , Proteína Morfogenética Ósea 2/genética , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/genética , Osteoporosis/patología , Ratas , Femenino , Humanos , Fémur/patología , Fémur/efectos de los fármacos , Fémur/diagnóstico por imagen , Cordón Umbilical/citología , Vesículas Extracelulares/metabolismo , Regeneración Ósea/efectos de los fármacos , Ratas Sprague-Dawley , Factor de Crecimiento Transformador beta/farmacología , Modelos Animales de Enfermedad , Microtomografía por Rayos X , Células Madre Mesenquimatosas/metabolismo
3.
Mol Biol Rep ; 51(1): 636, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38727863

RESUMEN

BACKGROUND: Osteoporosis (OP), characterized by compromised bone integrity and increased fracture risk, poses a significant health challenge. Circular RNAs (circRNAs) have emerged as crucial regulators in various pathophysiological processes, prompting investigation into their role in osteoporosis. This study aimed to elucidate the involvement of circCOX6A1 in OP progression and understand its underlying molecular mechanisms. The primary objective was to explore the impact of circCOX6A1 on bone marrow-derived mesenchymal stem cells (BMSCs) and its potential interactions with miR-512-3p and DYRK2. METHODS: GSE161361 microarray analysis was employed to assess circCOX6A1 expression in OP patients. We utilized in vitro and in vivo models, including BMSC cultures, osteogenic differentiation assays, and an OVX-induced mouse model of OP. Molecular techniques such as quantitative RT-PCR, western blotting, and functional assays like alizarin red staining (ARS) were employed to evaluate circCOX6A1 effects on BMSC proliferation, apoptosis, and osteogenic differentiation. The interaction between circCOX6A1, miR-512-3p, and DYRK2 was investigated through dual luciferase reporter assays, RNA immunoprecipitation, and RNA pull-down assays. RESULTS: CircCOX6A1 was found to be upregulated in osteoporosis patients, and its expression inversely correlated with osteogenic differentiation of BMSCs. CircCOX6A1 knockdown enhanced osteogenic differentiation, as evidenced by increased mineralized nodule formation and upregulation of osteogenic markers. In vivo, circCOX6A1 knockdown ameliorated osteoporosis progression in OVX mice. Mechanistically, circCOX6A1 acted as a sponge for miR-512-3p, subsequently regulating DYRK2 expression. CONCLUSION: This study provides compelling evidence for the role of circCOX6A1 in osteoporosis pathogenesis. CircCOX6A1 negatively regulates BMSC osteogenic differentiation through the miR-512-3p/DYRK2 axis, suggesting its potential as a therapeutic target for mitigating OP progression.


Asunto(s)
Diferenciación Celular , Quinasas DyrK , Células Madre Mesenquimatosas , MicroARNs , Osteogénesis , Osteoporosis , Proteínas Serina-Treonina Quinasas , Proteínas Tirosina Quinasas , ARN Circular , Osteoporosis/genética , Osteoporosis/metabolismo , Osteoporosis/patología , Osteogénesis/genética , MicroARNs/genética , MicroARNs/metabolismo , Animales , Diferenciación Celular/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Humanos , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Ratones , Células Madre Mesenquimatosas/metabolismo , ARN Circular/genética , ARN Circular/metabolismo , Femenino , Proliferación Celular/genética , Modelos Animales de Enfermedad , Apoptosis/genética , Persona de Mediana Edad
4.
FASEB J ; 38(9): e23657, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38713087

RESUMEN

The pathogenesis of osteoporosis (OP) is closely associated with the disrupted balance between osteogenesis and adipogenesis in bone marrow-derived mesenchymal stem cells (BMSCs). We analyzed published single-cell RNA sequencing (scRNA-seq) data to dissect the transcriptomic profiles of bone marrow-derived cells in OP, reviewing 56 377 cells across eight scRNA-seq datasets from femoral heads (osteoporosis or osteopenia n = 5, osteoarthritis n = 3). Seventeen genes, including carboxypeptidase M (CPM), were identified as key osteogenesis-adipogenesis regulators through comprehensive gene set enrichment, differential expression, regulon activity, and pseudotime analyses. In vitro, CPM knockdown reduced osteogenesis and promoted adipogenesis in BMSCs, while adenovirus-mediated CPM overexpression had the reverse effects. In vivo, intraosseous injection of CPM-overexpressing BMSCs mitigated bone loss in ovariectomized mice. Integrated scRNA-seq and bulk RNA sequencing analyses provided insight into the MAPK/ERK pathway's role in the CPM-mediated regulation of BMSC osteogenesis and adipogenesis; specifically, CPM overexpression enhanced MAPK/ERK signaling and osteogenesis. In contrast, the ERK1/2 inhibitor binimetinib negated the effects of CPM overexpression. Overall, our findings identify CPM as a pivotal regulator of BMSC differentiation, which provides new clues for the mechanistic study of OP.


Asunto(s)
Adipogénesis , Sistema de Señalización de MAP Quinasas , Células Madre Mesenquimatosas , Metaloendopeptidasas , Osteogénesis , Análisis de la Célula Individual , Animales , Osteogénesis/fisiología , Osteogénesis/genética , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Ratones , Femenino , Transcriptoma , Carboxipeptidasas/metabolismo , Carboxipeptidasas/genética , Humanos , Diferenciación Celular , Osteoporosis/genética , Osteoporosis/metabolismo , Osteoporosis/patología , Ratones Endogámicos C57BL , Proteínas Ligadas a GPI
5.
Acta Med Indones ; 56(1): 20-25, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38561882

RESUMEN

BACKGROUND: Osteoporosis is a major problem in transfusion-dependent thalassemia patients (TDT) patients. Osteoprotegerin (OPG) is one of several bone markers that are closely associated with osteoporosis in TDT patients. OPG is a glycoprotein that functions as a feedback receptor for the Receptor Activator of Nuclear Factor kappa B Ligand (RANKL), which is an alpha tumor necrosis factor receptor. One of the causes of decreased bone mass density is iron toxicity, which can be identified by showing elevated transferrin saturation. Bone mass dual X-ray absorptiometry (DEXA) is a gold standard for the diagnosis of osteoporosis, these procedures are not commonly available in Indonesia. This study was conducted to analyze the correlation between serum levels of OPG and transferrin saturation in TDT patients. METHODS: A correlational study with a cross-sectional approach analyzed data from TDT patients at Hemato-Oncology Medic Outpatient Clinic, Hasan Sadikin General Hospital, Bandung, Indonesia. Primary data were obtained through blood sampling and anthropometry measurement while secondary data were obtained from the patient's medical records. OPG and transferrin saturation levels were assessed using the ELISA method. Research data were analyzed using the rank Spearman correlation test. RESULTS: Data were collected from 51 research subjects (30 women dan 21 men). The median OPG level was 380 (170-1230) pg/mL and the median transferrin saturation level was 89.4 (66.7 - 96.2)%. Analysis of correlation showed a significant correlation between and transferrin saturation level with a coefficient value of r -0.539 and p-value <0.001. CONCLUSION: There was a significant inverse correlation between OPG with transferrin saturation in TDT patients.


Asunto(s)
Osteoporosis , Talasemia , Masculino , Humanos , Femenino , Osteoprotegerina , Densidad Ósea , Osteoporosis/etiología , Osteoporosis/patología , Talasemia/terapia , Talasemia/complicaciones , Transferrinas , Ligando RANK
6.
ACS Biomater Sci Eng ; 10(5): 3255-3267, 2024 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-38684056

RESUMEN

Osteoporosis is a metabolic disease characterized by bone density and trabecular bone loss. Bone loss may affect dental implant osseointegration in patients with osteoporosis. To promote implant osseointegration in osteoporotic patients, we further used a nonthermal atmospheric plasma (NTAP) treatment device previously developed by our research group. After the titanium implant (Ti) is placed into the device, the working gas flow and the electrode switches are turned on, and the treatment is completed in 30 s. Previous studies showed that this NTAP device can remove carbon contamination from the implant surface, increase the hydroxyl groups, and improve its wettability to promote osseointegration in normal conditions. In this study, we demonstrated the tremendous osteogenic enhancement effect of NTAP-Ti in osteoporotic conditions in rats for the first time. Compared to Ti, the proliferative potential of osteoporotic bone marrow mesenchymal stem cells on NTAP-Ti increased by 180% at 1 day (P = 0.004), while their osteogenic differentiation increased by 149% at 14 days (P < 0.001). In addition, the results indicated that NTAP-Ti significantly improved osseointegration in osteoporotic rats in vivo. Compared to the Ti, the bone volume fraction (BV/TV) and trabecular number (Tb.N) values of NTAP-Ti in osteoporotic rats, respectively, increased by 18% (P < 0.001) and 25% (P = 0.007) at 6 weeks and the trabecular separation (Tb.Sp) value decreased by 26% (P = 0.02) at 6 weeks. In conclusion, this study proved a novel NTAP irradiation titanium implant that can significantly promote osseointegration in osteoporotic conditions.


Asunto(s)
Células Madre Mesenquimatosas , Oseointegración , Osteogénesis , Osteoporosis , Gases em Plasma , Ratas Sprague-Dawley , Titanio , Titanio/farmacología , Animales , Osteogénesis/efectos de los fármacos , Osteoporosis/patología , Osteoporosis/terapia , Osteoporosis/tratamiento farmacológico , Gases em Plasma/farmacología , Gases em Plasma/uso terapéutico , Oseointegración/efectos de los fármacos , Femenino , Ratas , Células Madre Mesenquimatosas/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Prótesis e Implantes
7.
Acta Biomater ; 180: 82-103, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38621599

RESUMEN

The treatment of osteoporotic bone defect remains a big clinical challenge because osteoporosis (OP) is associated with oxidative stress and high levels of reactive oxygen species (ROS), a condition detrimental for bone formation. Anti-oxidative nanomaterials such as selenium nanoparticles (SeNPs) have positive effect on osteogenesis owing to their pleiotropic pharmacological activity which can exert anti-oxidative stress functions to prevent bone loss and facilitate bone regeneration in OP. In the current study a strategy of one-pot method by introducing Poly (lactic acid-carbonate) (PDT) and ß-Tricalcium Phosphate (ß-TCP) with SeNPs, is developed to prepare an injectable, anti-collapse, shape-adaptive and adhesive bone graft substitute material (PDT-TCP-SE). The PDT-TCP-SE bone graft substitute exhibits sufficient adhesion in biological microenvironments and osteoinductive activity, angiogenic effect and anti-inflammatory as well as anti-oxidative effect in vitro and in vivo. Moreover, the PDT-TCP-SE can protect BMSCs from erastin-induced ferroptosis through the Sirt1/Nrf2/GPX4 antioxidant pathway, which, in together, demonstrated the bone graft substitute material as an emerging biomaterial with potential clinical application for the future treatment of osteoporotic bone defect. STATEMENT OF SIGNIFICANCE: Injectable, anti-collapse, adhesive, plastic and bioactive bone graft substitute was successfully synthesized. Incorporation of SeNPs with PDT into ß-TCP regenerated new bone in-situ by moderating oxidative stress in osteoporotic bone defects area. The PDT-TCP-SE bone graft substitute reduced high ROS levels in osteoporotic bone defect microenvironment. The bone graft substitute could also moderate oxidative stress and inhibit ferroptosis via Sirt1/Nrf2/GPX4 pathway in vitro. Moreover, the PDT-TCP-SE bone graft substitute could alleviate the inflammatory environment and promote bone regeneration in osteoporotic bone defect in vivo. This biomaterial has the advantages of simple synthesis, biocompatibility, anti-collapse, injectable, and regulation of oxidative stress level, which has potential application value in bone tissue engineering.


Asunto(s)
Regeneración Ósea , Sustitutos de Huesos , Fosfatos de Calcio , Osteoporosis , Estrés Oxidativo , Estrés Oxidativo/efectos de los fármacos , Animales , Sustitutos de Huesos/química , Sustitutos de Huesos/farmacología , Regeneración Ósea/efectos de los fármacos , Osteoporosis/patología , Osteoporosis/terapia , Osteoporosis/tratamiento farmacológico , Fosfatos de Calcio/farmacología , Fosfatos de Calcio/química , Ratas Sprague-Dawley , Selenio/química , Selenio/farmacología , Femenino , Osteogénesis/efectos de los fármacos , Poliésteres/química , Poliésteres/farmacología , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/efectos de los fármacos , Ratas , Inyecciones
8.
Int J Mol Sci ; 25(8)2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38673956

RESUMEN

For a wide range of chronic autoimmune and inflammatory diseases in both adults and children, synthetic glucocorticoids (GCs) are one of the most effective treatments. However, besides other adverse effects, GCs inhibit bone mass at multiple levels, and at different ages, especially in puberty. Although extensive studies have investigated the mechanism of GC-induced osteoporosis, their target cell populations still be obscure. Here, our data show that the osteoblast subpopulation among Gli1+ metaphyseal mesenchymal progenitors (MMPs) is responsive to GCs as indicated by lineage tracing and single-cell RNA sequencing experiments. Furthermore, the proliferation and differentiation of Gli1+ MMPs are both decreased, which may be because GCs impair the oxidative phosphorylation(OXPHOS) and aerobic glycolysis of Gli1+ MMPs. Teriparatide, as one of the potential treatments for GCs in bone mass, is sought to increase bone volume by increasing the proliferation and differentiation of Gli1+ MMPs in vivo. Notably, our data demonstrate teriparatide ameliorates GC-caused bone defects by targeting Gli1+ MMPs. Thus, Gli1+ MMPs will be the potential mesenchymal progenitors in response to diverse pharmaceutical administrations in regulating bone formation.


Asunto(s)
Glucocorticoides , Células Madre Mesenquimatosas , Osteoporosis , Animales , Ratones , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Glucocorticoides/efectos adversos , Glucocorticoides/farmacología , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones Endogámicos C57BL , Osteoblastos/metabolismo , Osteoblastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Osteoporosis/inducido químicamente , Osteoporosis/metabolismo , Osteoporosis/patología , Teriparatido/farmacología , Proteína con Dedos de Zinc GLI1/metabolismo , Proteína con Dedos de Zinc GLI1/genética
9.
Biochem Biophys Res Commun ; 711: 149888, 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38603833

RESUMEN

OBJECTIVE: To investigate the effect of intermittent parathyroid hormone (iPTH) administration on pathological new bone formation during treatment of ankylosing spondylitis-related osteoporosis. METHODS: Animal models with pathological bone formation caused by hypothetical AS pathogenesis received treatment with iPTH. We determined the effects of iPTH on bone loss and the formation of pathological new bone with micro-computed tomography (micro-CT) and histological examination. In addition, the tamoxifen-inducible conditional knockout mice (CAGGCre-ERTM; PTHflox/flox, PTH-/-) was established to delete PTH and investigate the effect of endogenous PTH on pathological new bone formation. RESULTS: iPTH treatment significantly improved trabecular bone mass in the modified collagen-induced arthritis (m-CIA) model and unbalanced mechanical loading models. Meanwhile, iPTH treatment did not enhance pathological new bone formation in all types of animal models. Endogenous PTH deficiency had no effects on pathological new bone formation in unbalanced mechanical loading models. CONCLUSION: Experimental animal models of AS treated with iPTH show improvement in trabecular bone density, but not entheseal pathological bone formation,indicating it may be a potential treatment for inflammatory bone loss does in AS.


Asunto(s)
Osteogénesis , Hormona Paratiroidea , Animales , Hormona Paratiroidea/administración & dosificación , Hormona Paratiroidea/farmacología , Hormona Paratiroidea/uso terapéutico , Osteogénesis/efectos de los fármacos , Ratones , Osteoporosis/tratamiento farmacológico , Osteoporosis/patología , Ratones Noqueados , Masculino , Microtomografía por Rayos X , Espondilitis Anquilosante/tratamiento farmacológico , Espondilitis Anquilosante/patología , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Artritis Experimental/tratamiento farmacológico , Artritis Experimental/patología , Densidad Ósea/efectos de los fármacos
10.
Biochem Biophys Res Commun ; 711: 149858, 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38621345

RESUMEN

Systemic transplantation of mesenchymal stem cells (MSCs) and conditioned medium derived from MSCs have been reported to recover bone loss in animal models of osteoporosis; however, the underlying mechanisms remain unclear. We recently reported that extracellular vesicles released from human mesenchymal stem cells (hMSCs) prevent senescence of stem cells in bisphosphonate-related osteonecrosis of the jaw model. In this study, we aimed to compare the effects of conditioned medium (hMSCs-CM) from early and late passage hMSCs on cellular senescence and to verify the benefits of CM from early passage hMSCs in mitigating the progression of osteoporosis through the prevention of cellular senescence. We investigated the distinct endocrine effects of early (P5) and late (P17) passage hMSCs in vitro, as well as the preventive benefits of early passage hMSCs-CM in osteoporosis model triggered by ovariectomy. Our results indicate that long-term cultured hMSCs contributed to the progression of inflammatory transcriptional programs in P5 hMSCs, ultimately impairing their functionality and enhancing senescence-related characteristics. Conversely, early passage hMSCs reversed these alterations. Moreover, early passage hMSCs-CM infused intravenously in a postmenopausal osteoporosis mouse model suppressed bone degeneration and prevented osteoporosis by reducing ovariectomy-induced senescence in bone marrow MSCs and reducing the expression of senescence-associated secretory phenotype-related cytokines. Our findings highlight the high translational value of early passage hMSCs-CM in antiaging intervention and osteoporosis prevention.


Asunto(s)
Senescencia Celular , Células Madre Mesenquimatosas , Osteoporosis , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Humanos , Animales , Medios de Cultivo Condicionados/farmacología , Osteoporosis/patología , Osteoporosis/metabolismo , Femenino , Ratones , Células Cultivadas , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Ovariectomía
11.
Ecotoxicol Environ Saf ; 277: 116392, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38677065

RESUMEN

Smoking disrupts bone homeostasis and serves as an independent risk factor for the development and progression of osteoporosis. Tobacco toxins inhibit the proliferation and osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs), promote BMSCs aging and exhaustion, but the specific mechanisms are not yet fully understood. Herein, we successfully established a smoking-related osteoporosis (SROP) model in rats and mice through intraperitoneal injection of cigarette smoke extract (CSE), which significantly reduced bone density and induced aging and inhibited osteogenic differentiation of BMSCs both in vivo and in vitro. Bioinformatics analysis and in vitro experiments confirmed that CSE disrupts mitochondrial homeostasis through oxidative stress and inhibition of mitophagy. Furthermore, we discovered that CSE induced BMSCs aging by upregulating phosphorylated AKT, which in turn inhibited the expression of FOXO3a and the Pink1/Parkin pathway, leading to the suppression of mitophagy and the accumulation of damaged mitochondria. MitoQ, a mitochondrial-targeted antioxidant and mitophagy agonist, was effective in reducing CSE-induced mitochondrial oxidative stress, promoting mitophagy, significantly downregulating the expression of aging markers in BMSCs, restoring osteogenic differentiation, and alleviating bone loss and autophagy levels in CSE-exposed mice. In summary, our results suggest that BMSCs aging caused by the inhibition of mitophagy through the AKT/FOXO3a/Pink1/Parkin axis is a key mechanism in smoking-related osteoporosis.


Asunto(s)
Células Madre Mesenquimatosas , Mitofagia , Osteoporosis , Animales , Mitofagia/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones , Ratas , Osteoporosis/inducido químicamente , Osteoporosis/patología , Nicotiana/efectos adversos , Proteína Forkhead Box O3/metabolismo , Estrés Oxidativo/efectos de los fármacos , Masculino , Ratas Sprague-Dawley , Osteogénesis/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Humo/efectos adversos , Ubiquitina-Proteína Ligasas/metabolismo , Mitocondrias/efectos de los fármacos , Proteínas Quinasas/metabolismo , Ratones Endogámicos C57BL , Células de la Médula Ósea/efectos de los fármacos
12.
J Cell Mol Med ; 28(9): e18287, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38685675

RESUMEN

Single immobilization theory cannot fully account for the extensive bone loss observed after spinal cord injury (SCI). Bone marrow mesenchymal stem cells (BMSCs) are crucial in bone homeostasis because they possess self-renewal capabilities and various types of differentiation potential. This study aimed to explore the molecular mechanism of long non-coding RNA H19 in osteoporosis after SCI and provide new research directions for existing prevention strategies. We used small interfering RNA to knockdown H19 expression and regulated miR-29b-2p expression using miR-29b-3p mimetics and inhibitors. Western blotting, real-time fluorescence quantitative PCR, Alizarin red staining, alkaline phosphatase staining and double-luciferase reporter gene assays were used to assess gene expression, osteogenic ability and binding sites. lncRNA H19 was upregulated in BMSCs from the osteoporosis group, whereas miR-29b-3p was downregulated. We identified the binding sites between miR-29b-3p and lncRNAs H19 and DKK1. H19 knockdown promoted BMSCs' osteogenic differentiation, whereas miR-29b-3p inhibition attenuated this effect. We discovered potential binding sites for miR-29b-3p in lncRNAs H19 and DKK1. Our findings suggest that long non-coding RNA H19 mediates BMSCs' osteogenic differentiation in osteoporosis after SCI through the miR-29b-3p/DKK1 axis and by directly inhibiting the ß-catenin signalling pathway.


Asunto(s)
Diferenciación Celular , Péptidos y Proteínas de Señalización Intercelular , Células Madre Mesenquimatosas , MicroARNs , Osteogénesis , Osteoporosis , ARN Largo no Codificante , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , MicroARNs/genética , MicroARNs/metabolismo , Osteogénesis/genética , Diferenciación Celular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Animales , Osteoporosis/genética , Osteoporosis/patología , Osteoporosis/metabolismo , Humanos , Regulación de la Expresión Génica , Masculino , Ratas , Traumatismos de la Médula Espinal/genética , Traumatismos de la Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/patología
13.
Cell Death Differ ; 31(5): 605-617, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38431690

RESUMEN

Absent, small, or homeotic1-like (ASH1L) is a histone lysine methyltransferase that generally functions as a transcriptional activator in controlling cell fate. So far, its physiological relevance in bone homeostasis and osteoclast differentiation remains elusive. Here, by conditional deleting Ash1l in osteoclast progenitors of mice, we found ASH1L deficiency resulted in osteoporosis and potentiation of osteoclastogenesis in vivo and in vitro. Mechanistically, ASH1L binds the promoter of the Src homology 3 and cysteine-rich domain 2 (Stac2) and increases the gene's transcription via histone 3 lysine 4 (H3K4) trimethylation modification, thus augmenting the STAC2's protection against receptor activator of nuclear factor kB ligand (RANKL)-initiated inflammation during osteoclast formation. Collectively, we demonstrate the first piece of evidence to prove ASH1L as a critical checkpoint during osteoclastogenesis. The work sheds new light on our understanding about the biological function of ASH1L in bone homeostasis, therefore providing a valuable therapeutic target for the treatment of osteoporosis or inflammatory bone diseases.


Asunto(s)
N-Metiltransferasa de Histona-Lisina , Osteoclastos , Osteogénesis , Animales , N-Metiltransferasa de Histona-Lisina/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , Osteoclastos/metabolismo , Ratones , Diferenciación Celular , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Ligando RANK/metabolismo , Ratones Endogámicos C57BL , Osteoporosis/metabolismo , Osteoporosis/patología , Osteoporosis/genética , Ratones Noqueados , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Resorción Ósea/metabolismo , Resorción Ósea/patología , Histonas/metabolismo
14.
Environ Toxicol ; 39(6): 3381-3388, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38445413

RESUMEN

Osteoporosis is a common bone disease in aging populations, particularly in postmenopausal women. Anti-resorptive and anabolic drugs have been applied to prevent and cure osteoporosis and are linked with a variety of adverse effects. Antrodia cinnamomea extracts (ACE) are highly renowned for their anticancer, antioxidative, and anti-inflammatory properties. However, whether ACE-enriched anti-osteoporosis functions are largely unknown. In a preclinical animal model, we found that ovariectomy significantly decreased bone volume in the ovariectomized (OVX) rats. Administration of ACE antagonized OVX-induced bone loss. In addition, ACE reversed OVX-reduced biomechanical properties. The serum osteoclast marker also showed improvement in the ACE-treated group. In the cellular model, it was indicated that ACE inhibits RANKL-induced osteoclast formation. Taken together, ACE seems to be a hopeful candidate for the development of novel anti-osteoporosis treatment.


Asunto(s)
Osteoclastos , Osteoporosis , Ovariectomía , Ratas Sprague-Dawley , Animales , Femenino , Osteoclastos/efectos de los fármacos , Osteoporosis/prevención & control , Osteoporosis/tratamiento farmacológico , Osteoporosis/patología , Ratones , Ratas , Células RAW 264.7 , Polyporales/química , Resorción Ósea/prevención & control , Resorción Ósea/tratamiento farmacológico , Ligando RANK
15.
Ann Anat ; 254: 152244, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38492654

RESUMEN

The knowledge of bone biology has undergone major advances in recent decades. In bone, resorbing osteoclasts have classically been described as tissue-resident macrophages, however, it is currently known that a new subtype of macrophages, called OsteoMacs, are specialised bone-resident macrophages, which, depending on certain conditions, may play an important role not only in bone homeostasis, but also in promoting pro-anabolic functions or in creating an inflammatory environment. There is growing evidence that these osteal macrophages may influence the development of bone-loss diseases. It is essential to understand the biological bases underlying bone physiological processes to search for new therapeutic targets for bone-loss diseases, such as osteoporosis, rheumatoid arthritis, or even periodontal disease. This narrative review provides an update on the origin, characterisation, and possible roles of osteoMacs in bone biology. Finally, the potential clinical applications of this new cell in bone-loss disorders are discussed.


Asunto(s)
Huesos , Macrófagos , Humanos , Huesos/fisiología , Animales , Macrófagos/fisiología , Osteoclastos/fisiología , Enfermedades Óseas/patología , Osteoporosis/patología
16.
Life Sci ; 345: 122592, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38554947

RESUMEN

Osteoporosis, characterized by bone metabolism disruption leading to gradual bone loss and increased fracture susceptibility, is linked to the excessive activation of osteoclasts. Pseudolaric acid B (PAB), identified as an NF-κB signaling inhibitor crucial for osteoclastogenesis, is explored here for its protective effects in osteoporosis. Noncytotoxic PAB's impact on osteoclast differentiation was assessed through cell viability and osteoclast formation assays, with subsequent testing of osteoclast function via bone resorption assays. Quantitative real-time polymerase chain reaction evaluated PAB's genetic-level impact on osteoclastogenesis. Network pharmacology, western blot, and luciferase reporter gene assays were employed to elucidate PAB's regulatory mechanism. In an in vivo model of osteoporosis induced by ovariectomy (OVX) in mice, micro-CT, H&E staining, and TRAP staining facilitated histomorphometry analysis, while flow cytometry verified macrophage polarization. PAB demonstrated inhibitory effects on osteoclast formation and bone resorption in BMM and RAW264.7 cells, suppressing osteoclast-specific genes. Bioinformatic analysis, western blot, and luciferase assay results indicated PAB's inhibition of IκBα phosphorylation in the NF-κB signaling pathway and ERK in MAPKs, elucidating its mechanism. In vivo experiments confirmed PAB's attenuation of osteoporosis by reducing osteoclast formation in OVX mice. PAB further facilitated macrophage conversion from M1 to M2 and suppressed IL-1ß, TNF-α, and IL-6 synthesis. In conclusion, PAB prevents osteoporosis by inhibiting RANKL-induced osteoclastogenesis through NF-κB and ERK signaling pathway suppression, coupled with macrophage polarization. These findings indicate the potential therapeutic role of PAB in osteoporosis.


Asunto(s)
Resorción Ósea , Diterpenos , Osteoporosis , Animales , Femenino , Humanos , Ratones , Resorción Ósea/tratamiento farmacológico , Resorción Ósea/metabolismo , Diferenciación Celular , Diterpenos/farmacología , Luciferasas/metabolismo , Macrófagos/metabolismo , FN-kappa B/metabolismo , Osteoclastos , Osteogénesis/efectos de los fármacos , Osteoporosis/tratamiento farmacológico , Osteoporosis/metabolismo , Osteoporosis/patología , Ovariectomía , Ligando RANK/metabolismo , Transducción de Señal
17.
J Cell Physiol ; 239(5): e31214, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38358001

RESUMEN

Alleviating bone loss is an essential way to prevent osteoporotic fractures. Proper exercise improves bone density without the side effects of long-term medications, but the mechanism is unclear. Our study explored the role of Antxr1/LncRNA H19/Wnt/ß-catenin axis in the process of exercise-mediated alleviation of bone loss. Here we discovered that moderate-intensity treadmill exercise alleviates bone loss caused by ovariectomy and ameliorates bone strength accompanied by an increased lncRNA H19 expression. Concomitantly, Antxr1, a mechanosensitive protein was found downregulated by exercise but upregulated by ovariectomy. Interestingly, knockdown expression of Antxr1 increased lncRNA H19 expression and Wnt/ß-catenin signaling pathway in bone marrow mesenchymal stem cells, whereas overexpression of Antxr1 decreased lncRNA H19 expression and Wnt/ß-catenin signaling pathway. Hence, our study demonstrates the regulation of Antxr1/LncRNA H19/Wnt/ß-catenin axis in the process of mechanical strain-induced osteogenic differentiation, which provides further mechanistic insight into the role of mechanical regulation in bone metabolism.


Asunto(s)
Células Madre Mesenquimatosas , Osteogénesis , ARN Largo no Codificante , Estrés Mecánico , Vía de Señalización Wnt , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Osteogénesis/genética , Animales , Vía de Señalización Wnt/genética , Femenino , Células Madre Mesenquimatosas/metabolismo , beta Catenina/metabolismo , beta Catenina/genética , Diferenciación Celular , Ovariectomía/efectos adversos , Osteoporosis/genética , Osteoporosis/metabolismo , Osteoporosis/patología , Densidad Ósea/genética , Ratones
18.
Electromagn Biol Med ; 43(1-2): 46-60, 2024 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-38329038

RESUMEN

This study aimed to assess PEMF in a rat model of senile osteoporosis and its relationship with NLRP3-mediated low-grade inflammation in the bone marrow microenvironment. A total of 24 Sprague Dawley (SD) rats were included in this study. Sixteen of them were 24-month natural-aged male SD rats, which were randomly distributed into the Aged group and the PEMF group (n = 8 per group). The remaining 8 3-month -old rats were used as the Young positive control group (n = 8). Rats in the PEMF group received 12 weeks of PEMF with 40 min/day, five days per week, while the other rats received placebo PEMF intervention. Bone mineral density/microarchitecture, serum levels of CTX-1 and P1CP, and NLRP3-related signaling genes and proteins in rat bone marrow were then analyzed. The 12-week of PEMF showed significant mitigation of aging-induced bone loss and bone microarchitecture deterioration, i.e. PEMF increased the bone mineral density of the proximal femur and L5 vertebral body and improved parameters of the proximal tibia and L4 vertebral body. Further analysis showed that PEMF reversed aging-induced bone turnover, specifically, decreased serum CTX-1 and elevated serum P1CP. Furthermore, PEMF also dramatically inhibited NLRP3-mediated low-grade inflammation in the bone marrow, i.e. PEMF inhibited the levels of NLRP3, proCaspase1, cleaved Caspase1, IL-1ß, and GSDMD-N. The study demonstrated that PEMF could mitigate the aging-induced bone loss and reverses the deterioration of bone microarchitecture probably through inhibiting NLRP3-mediated low-grade chronic inflammation to improve the inflammatory bone microenvironment in aged rats.


Asunto(s)
Densidad Ósea , Campos Electromagnéticos , Inflamación , Proteína con Dominio Pirina 3 de la Familia NLR , Osteoporosis , Ratas Sprague-Dawley , Animales , Osteoporosis/terapia , Osteoporosis/prevención & control , Osteoporosis/sangre , Osteoporosis/metabolismo , Osteoporosis/patología , Masculino , Ratas , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Inflamación/terapia , Densidad Ósea/efectos de la radiación , Médula Ósea/efectos de la radiación , Médula Ósea/metabolismo , Microambiente Celular , Envejecimiento
19.
Phytother Res ; 38(4): 1971-1989, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38358727

RESUMEN

BACKGROUND AND AIM: Osteoporosis, a systemic metabolic bone disease, is characterized by the decline of bone mass and quality due to excessive osteoclast activity. Currently, drug-targeting osteoclasts show promising therapy for osteoporosis. In this study, we investigated the effect of cichoric acid (CA) on receptor activator of nuclear kappa-B ligand (RANKL)-induced osteoclastogenesis and the bone loss induced by ovariectomy in mice. EXPERIMENTAL PROCEDURE: Molecular docking technologies were employed to examine the interaction between CA and RANKL. CCK8 assay was used to evaluate the cell viability under CA treatment. TRAcP staining, podosome belt staining, and bone resorption assays were used to test the effect of CA on osteoclastogenesis and osteoclast function. Further, an OVX-induced osteoporosis mice model was employed to identify the effect of CA on bone loss using micro-CT scanning and histological examination. To investigate underlying mechanisms, network pharmacology was applied to predict the downstream signaling pathways, which were verified by Western blot and immunofluorescence staining. KEY RESULTS: The molecular docking analysis revealed that CA exhibited a specific binding affinity to RANKL, engaging multiple binding sites. CA inhibited RANKL-induced osteoclastogenesis and bone resorption without cytotoxic effects. Mechanistically, CA suppressed RANKL-induced intracellular reactive oxygen species, nuclear factor-kappa B, and mitogen-activated protein kinase pathways, followed by abrogated nuclear factor activated T-cells 1 activity. Consistent with this finding, CA attenuated post-ovariectomy-induced osteoporosis by ameliorating osteoclastogenesis. CONCLUSIONS AND IMPLICATIONS: CA inhibited osteoclast activity and bone loss by targeting RANKL. CA might represent a promising candidate for treating osteoclast-related diseases, such as osteoporosis.


Asunto(s)
Resorción Ósea , Ácidos Cafeicos , Osteoporosis , Succinatos , Animales , Femenino , Humanos , Ratones , Resorción Ósea/prevención & control , Diferenciación Celular , Ratones Endogámicos C57BL , Simulación del Acoplamiento Molecular , FN-kappa B/metabolismo , Osteoclastos , Osteogénesis , Osteoporosis/patología , Ovariectomía/efectos adversos , Ligando RANK/metabolismo
20.
Front Endocrinol (Lausanne) ; 15: 1325317, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38370357

RESUMEN

Osteoporosis (OP) is a systemic skeletal disorder characterized by reduced bone mass and structural deterioration of bone tissue, resulting in heightened vulnerability to fractures due to increased bone fragility. This condition primarily arises from an imbalance between the processes of bone resorption and formation. Mitochondrial dysfunction has been reported to potentially constitute one of the most crucial mechanisms influencing the pathogenesis of osteoporosis. In essence, mitochondria play a crucial role in maintaining the delicate equilibrium between bone formation and resorption, thereby ensuring optimal skeletal health. Nevertheless, disruption of this delicate balance can arise as a consequence of mitochondrial dysfunction. In dysfunctional mitochondria, the mitochondrial electron transport chain (ETC) becomes uncoupled, resulting in reduced ATP synthesis and increased generation of reactive oxygen species (ROS). Reinforcement of mitochondrial dysfunction is further exacerbated by the accumulation of aberrant mitochondria. In this review, we investigated and analyzed the correlation between mitochondrial dysfunction, encompassing mitochondrial DNA (mtDNA) alterations, oxidative phosphorylation (OXPHOS) impairment, mitophagy dysregulation, defects in mitochondrial biogenesis and dynamics, as well as excessive ROS accumulation, with regards to OP (Figure 1). Furthermore, we explore prospective strategies currently available for modulating mitochondria to ameliorate osteoporosis. Undoubtedly, certain therapeutic strategies still require further investigation to ensure their safety and efficacy as clinical treatments. However, from a mitochondrial perspective, the potential for establishing effective and safe therapeutic approaches for osteoporosis appears promising.


Asunto(s)
Enfermedades Mitocondriales , Osteoporosis , Humanos , Especies Reactivas de Oxígeno , Estudios Prospectivos , Mitocondrias/patología , ADN Mitocondrial/genética , Enfermedades Mitocondriales/genética , Osteoporosis/terapia , Osteoporosis/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA