Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34.981
Filtrar
1.
Pak J Pharm Sci ; 37(1(Special)): 223-229, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38747273

RESUMEN

In this study, the anti-osteogenic properties of the volatile oil extracted from Homalomena gigantea rhizome using ethyl acetate (EtOAc) and methanol (MeOH) were examined. Gas chromatography-mass spectrometry (GC-MS) was employed for the identification of volatile components. Following this, bioassays were performed to evaluate their effects on osteogenesis, encompassing parameters like cell viability, osteoblast differentiation, collagen synthesis and mineralization. The GC-MS analysis revealed 19 compounds in the EtOAc extract and 36 compounds in the MeOH extract. In the MeOH extract, major constituents included bis(2-ethylhexyl) terephthalate (13.83%), linalool (9.58%), palmitic acid (6.55%) and stearic acid (4.29%). The EtOAc extract contained bis(2-ethylhexyl) terephthalate (16.64%), palmitic acid (5.60%) and stearic acid (3.11%) as the predominant components. Both the EtOAc and MeOH extracts of H. gigantea exhibited promising potential for further investigation in anti-osteoporosis research. These findings contribute to the exploration of natural compounds with potential anti-osteoporotic properties, expanding our understanding of their therapeutic potential.


Asunto(s)
Cromatografía de Gases y Espectrometría de Masas , Aceites Volátiles , Osteogénesis , Extractos Vegetales , Rizoma , Osteogénesis/efectos de los fármacos , Rizoma/química , Extractos Vegetales/farmacología , Extractos Vegetales/química , Extractos Vegetales/aislamiento & purificación , Aceites Volátiles/farmacología , Aceites Volátiles/química , Aceites Volátiles/aislamiento & purificación , Animales , Supervivencia Celular/efectos de los fármacos , Osteoblastos/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Ratones , Ácido Palmítico/farmacología , Monoterpenos Acíclicos/farmacología
2.
J Physiol Pharmacol ; 75(2): 173-183, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38736264

RESUMEN

Quercetin is widely distributed in plants as a flavonol compound with multiple biological activities. It has been found that quercetin can regulate bone homeostasis through multiple pathways and targets. This study investigated the role and specific molecular mechanisms of quercetin in regulating osteoblast viability, proliferation, migration and osteogenic differentiation. A mouse model of traumatic fracture was established and then 100 mg/kg quercetin corn oil suspension was gavaged at the same time every day for 28 days. miR-6089 and E2F transcription factor 2 (E2F2) expression levels in mice were measured. Fracture healing in mice was observed. MC3T3-E1 cells were transfected with plasmids targeting miR-6089 and E2F2, and cell viability, proliferation, migration, apoptosis, and osteogenic differentiation were determined. The targeting relationship between miR-6089 and E2F2 was verified. In vivo experiments showed that quercetin significantly increased osteocalcin (OCN) expression (P<0.05) and promoted fracture healing in traumatic fracture (TF) mice. miR-6089 expression was down-regulated (P<0.05) and E2F2 expression was up-regulated (P<0.05) in TF mice. Quercetin promoted miR-6089 expression and inhibited E2F2 expression (both P<0.05). In vitro results showed that quercetin promoted miR-6089 expression and inhibited E2F2 expression in a dose-dependent manner (both P<0.05). Quercetin dose-dependently promoted MC3T3-E1 cell viability, proliferation, migration, and osteogenic differentiation, and inhibited MC3T3-E1 cell apoptosis (all P<0.05). Up-regulating miR-6089 further promoted MC3T3-E1 cell viability, proliferation, migration and osteogenic differentiation, and inhibited MC3T3-E1 cell apoptosis (all P<0.05). miR-6089 targeted and regulated E2F2 expression. Up-regulating E2F2 attenuated the promoting effect of up-regulated miR-6089 on MC3T3-E1 cell viability, proliferation, migration, osteogenic differentiation, and inhibition of apoptosis (all P<0.05). We conclude that quercetin enhances osteoblast viability, proliferation, migration, and osteogenic differentiation by modulating the miR-6089/E2F2 axis, thereby promoting fracture healing.


Asunto(s)
Factor de Transcripción E2F2 , Curación de Fractura , MicroARNs , Osteoblastos , Osteogénesis , Quercetina , Animales , Masculino , Ratones , Apoptosis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Línea Celular , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Factor de Transcripción E2F2/metabolismo , Factor de Transcripción E2F2/genética , Curación de Fractura/efectos de los fármacos , MicroARNs/genética , MicroARNs/metabolismo , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Osteogénesis/efectos de los fármacos , Quercetina/farmacología
3.
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
4.
Pak J Pharm Sci ; 37(1): 123-128, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38741408

RESUMEN

The study aimed to investigate the effects of aspirin on patients with metastatic colorectal cancer, focusing on circulating tumor DNA levels and bone tissue. Two groups (A and B) of ten patients with osteoporosis were selected for the study. Bone tissue samples were obtained from the patients and cultured under sterile conditions. The aspirin group showed a significant decrease in circulating tumor DNA levels and an increase in bone tissue density compared to the control group. Additionally, osteoblast apoptosis was reduced, while proliferation was enhanced in the aspirin group. The protein pAkt related to the PI3K/Akt signaling pathway was upregulated in the aspirin group. These results indicate that aspirin can effectively lower circulating tumor DNA levels, promote bone tissue proliferation, inhibit apoptosis, and activate the PI3K/Akt signaling pathway, thereby influencing bone cell function. These findings provide a basis for aspirin's potential application in treating metastatic colorectal cancer and encourage further research on its mechanism and clinical use.


Asunto(s)
Apoptosis , Aspirina , ADN Tumoral Circulante , Neoplasias Colorrectales , Humanos , Aspirina/farmacología , Aspirina/uso terapéutico , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Masculino , Femenino , Persona de Mediana Edad , Apoptosis/efectos de los fármacos , ADN Tumoral Circulante/sangre , ADN Tumoral Circulante/genética , Proliferación Celular/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Anciano , Transducción de Señal/efectos de los fármacos , Osteoblastos/efectos de los fármacos , Osteoblastos/patología , Osteoblastos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Densidad Ósea/efectos de los fármacos , Osteoporosis/tratamiento farmacológico
5.
Biol Open ; 13(5)2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38742438

RESUMEN

Bone is increasingly recognized as a target for diabetic complications. In order to evaluate the direct effects of high glucose on bone, we investigated the global transcriptional changes induced by hyperglycemia in osteoblasts in vitro. Rat bone marrow-derived mesenchymal stromal cells were differentiated into osteoblasts for 10 days, and prior to analysis, they were exposed to hyperglycemia (25 mM) for the short-term (1 or 3 days) or long-term (10 days). Genes and pathways regulated by hyperglycemia were identified using mRNA sequencing and verified with qPCR. Genes upregulated by 1-day hyperglycemia were, for example, related to extracellular matrix organization, collagen synthesis and bone formation. This stimulatory effect was attenuated by 3 days. Long-term exposure impaired osteoblast viability, and downregulated, for example, extracellular matrix organization and lysosomal pathways, and increased intracellular oxidative stress. Interestingly, transcriptional changes by different exposure times were mostly unique and only 89 common genes responding to glucose were identified. In conclusion, short-term hyperglycemia had a stimulatory effect on osteoblasts and bone formation, whereas long-term hyperglycemia had a negative effect on intracellular redox balance, osteoblast viability and function.


Asunto(s)
Regulación de la Expresión Génica , Glucosa , Osteoblastos , Osteoblastos/metabolismo , Osteoblastos/efectos de los fármacos , Animales , Glucosa/metabolismo , Ratas , Regulación de la Expresión Génica/efectos de los fármacos , Perfilación de la Expresión Génica , Hiperglucemia/metabolismo , Hiperglucemia/genética , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/efectos de los fármacos , Transcriptoma , Osteogénesis/efectos de los fármacos , Osteogénesis/genética , Supervivencia Celular/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Células Cultivadas , Estrés Oxidativo/efectos de los fármacos
6.
Cell Mol Life Sci ; 81(1): 204, 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38700532

RESUMEN

The silent information regulator T1 (SIRT1) is linked to longevity and is a crucial mediator of osteoblast function. We investigated the direct role of Sirt1 during bone modeling and remodeling stages in vivo using Tamoxifen-inducible osteoblast-specific Sirt1 conditional knockout (cKO) mice. cKO mice exhibited lower trabecular and cortical bone mass in the distal femur. These phenotypes were coupled with lower bone formation and bone resorption. Metabolomics analysis revealed that the metabolites involved in glycolysis were significantly decreased in cKO mice. Further analysis of the quantitative acetylome revealed 11 proteins with upregulated acetylation levels in both the femur and calvaria of cKO mice. Cross-analysis identified four proteins with the same upregulated lysine acetylation site in both the femur and calvaria of cKO mice. A combined analysis of the metabolome and acetylome, as well as immunoprecipitation, gene knockout, and site-mutation experiments, revealed that Sirt1 deletion inhibited glycolysis by directly binding to and increasing the acetylation level of Glutamine oxaloacetic transaminase 1 (GOT1). In conclusion, our study suggested that Sirt1 played a crucial role in regulating osteoblast metabolism to maintain bone homeostasis through its deacetylase activity on GOT1. These findings provided a novel insight into the potential targeting of osteoblast metabolism for the treatment of bone-related diseases.


Asunto(s)
Glucólisis , Homeostasis , Ratones Noqueados , Osteoblastos , Sirtuina 1 , Animales , Sirtuina 1/metabolismo , Sirtuina 1/genética , Osteoblastos/metabolismo , Ratones , Acetilación , Huesos/metabolismo , Osteogénesis , Fémur/metabolismo
7.
J Nanobiotechnology ; 22(1): 261, 2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38760744

RESUMEN

Delayed repair of fractures seriously impacts patients' health and significantly increases financial burdens. Consequently, there is a growing clinical demand for effective fracture treatment. While current materials used for fracture repair have partially addressed bone integrity issues, they still possess limitations. These challenges include issues associated with autologous material donor sites, intricate preparation procedures for artificial biomaterials, suboptimal biocompatibility, and extended degradation cycles, all of which are detrimental to bone regeneration. Hence, there is an urgent need to design a novel material with a straightforward preparation method that can substantially enhance bone regeneration. In this context, we developed a novel nanoparticle, mPPTMP195, to enhance the bioavailability of TMP195 for fracture treatment. Our results demonstrate that mPPTMP195 effectively promotes the differentiation of bone marrow mesenchymal stem cells into osteoblasts while inhibiting the differentiation of bone marrow mononuclear macrophages into osteoclasts. Moreover, in a mouse femur fracture model, mPPTMP195 nanoparticles exhibited superior therapeutic effects compared to free TMP195. Ultimately, our study highlights that mPPTMP195 accelerates fracture repair by preventing HDAC4 translocation from the cytoplasm to the nucleus, thereby activating the NRF2/HO-1 signaling pathway. In conclusion, our study not only proposes a new strategy for fracture treatment but also provides an efficient nano-delivery system for the widespread application of TMP195 in various other diseases.


Asunto(s)
Diferenciación Celular , Histona Desacetilasas , Células Madre Mesenquimatosas , Nanopartículas , Animales , Ratones , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Nanopartículas/química , Diferenciación Celular/efectos de los fármacos , Histona Desacetilasas/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Ratones Endogámicos C57BL , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Osteoblastos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Hemo-Oxigenasa 1/metabolismo , Masculino , Regeneración Ósea/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Núcleo Celular/metabolismo , Curación de Fractura/efectos de los fármacos , Humanos , Proteínas de la Membrana
8.
Int J Mol Sci ; 25(9)2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38732172

RESUMEN

Fibrous dysplasia (FD) poses a therapeutic challenge due to the dysregulated extracellular matrix (ECM) accumulation within affected bone tissues. In this study, we investigate the therapeutic potential of 1,25-dihydroxyvitamin D3 (1,25(OH)2D3) in managing FD by examining its effects on FD-derived cells in vitro. Our findings demonstrate that 1,25(OH)2D3 treatment attenuates the pro-fibrotic phenotype of FD-derived cells by suppressing the expression of key pro-fibrotic markers and inhibiting cell proliferation and migration. Moreover, 1,25(OH)2D3 enhances mineralization by attenuating pre-osteoblastic cellular hyperactivity and promoting maturation towards an osteocytic phenotype. These results offer valuable insights into potential treatments for FD, highlighting the role of 1,25(OH)2D3 in modulating the pathological properties of FD-derived cells.


Asunto(s)
Proliferación Celular , Displasia Fibrosa Ósea , Humanos , Proliferación Celular/efectos de los fármacos , Displasia Fibrosa Ósea/metabolismo , Displasia Fibrosa Ósea/patología , Displasia Fibrosa Ósea/tratamiento farmacológico , Fenotipo , Vitamina D/farmacología , Vitamina D/metabolismo , Fibrosis , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Movimiento Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Calcitriol/farmacología , Células Cultivadas
9.
Int J Mol Sci ; 25(9)2024 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-38732267

RESUMEN

Osteoporosis, characterized by reduced bone density and increased fracture risk, affects over 200 million people worldwide, predominantly older adults and postmenopausal women. The disruption of the balance between bone-forming osteoblasts and bone-resorbing osteoclasts underlies osteoporosis pathophysiology. Standard treatment includes lifestyle modifications, calcium and vitamin D supplementation and specific drugs that either inhibit osteoclasts or stimulate osteoblasts. However, these treatments have limitations, including side effects and compliance issues. Natural products have emerged as potential osteoporosis therapeutics, but their mechanisms of action remain poorly understood. In this study, we investigate the efficacy of natural compounds in modulating molecular targets relevant to osteoporosis, focusing on the Mitogen-Activated Protein Kinase (MAPK) pathway and the gut microbiome's influence on bone homeostasis. Using an in silico and in vitro methodology, we have identified quercetin as a promising candidate in modulating MAPK activity, offering a potential therapeutic perspective for osteoporosis treatment.


Asunto(s)
Productos Biológicos , Remodelación Ósea , Osteoporosis , Humanos , Remodelación Ósea/efectos de los fármacos , Osteoporosis/tratamiento farmacológico , Osteoporosis/metabolismo , Productos Biológicos/farmacología , Productos Biológicos/uso terapéutico , Quercetina/farmacología , Quercetina/uso terapéutico , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Huesos/metabolismo , Huesos/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Microbioma Gastrointestinal/efectos de los fármacos , Osteoclastos/metabolismo , Osteoclastos/efectos de los fármacos , Animales
10.
Int J Mol Sci ; 25(9)2024 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-38731934

RESUMEN

Adult bones are continuously remodeled by the balance between bone resorption by osteoclasts and subsequent bone formation by osteoblasts. Many studies have provided molecular evidence that bone remodeling is under the control of circadian rhythms. Circadian fluctuations have been reported in the serum and urine levels of bone turnover markers, such as digested collagen fragments and bone alkaline phosphatase. Additionally, the expressions of over a quarter of all transcripts in bones show circadian rhythmicity, including the genes encoding master transcription factors for osteoblastogenesis and osteoclastogenesis, osteogenic cytokines, and signaling pathway proteins. Serum levels of calcium, phosphate, parathyroid hormone, and calcitonin also display circadian rhythmicity. Finally, osteoblast- and osteoclast-specific knockout mice targeting the core circadian regulator gene Bmal1 show disrupted bone remodeling, although the results have not always been consistent. Despite these studies, however, establishing a direct link between circadian rhythms and bone remodeling in vivo remains a major challenge. It is nearly impossible to repeatedly collect bone materials from human subjects while following circadian changes. In addition, the differences in circadian gene regulation between diurnal humans and nocturnal mice, the main model organism, remain unclear. Filling the knowledge gap in the circadian regulation of bone remodeling could reveal novel regulatory mechanisms underlying many bone disorders including osteoporosis, genetic diseases, and fracture healing. This is also an important question for the basic understanding of how cell differentiation progresses under the influence of cyclically fluctuating environments.


Asunto(s)
Remodelación Ósea , Ritmo Circadiano , Remodelación Ósea/genética , Animales , Ritmo Circadiano/fisiología , Ritmo Circadiano/genética , Humanos , Osteoblastos/metabolismo , Osteogénesis/genética , Osteoclastos/metabolismo , Regulación de la Expresión Génica , Huesos/metabolismo
11.
Int J Mol Sci ; 25(9)2024 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-38731947

RESUMEN

Estrogen plays an important role in osteoporosis prevention. We herein report the possible novel signaling pathway of 17ß-estradiol (E2) in the matrix mineralization of MC3T3-E1, an osteoblast-like cell line. In the culture media-containing stripped serum, in which small lipophilic molecules such as steroid hormones including E2 were depleted, matrix mineralization was significantly reduced. However, the E2 treatment induced this. The E2 effects were suppressed by ICI182,780, the estrogen receptor (ER)α, and the ERß antagonist, as well as their mRNA knockdown, whereas Raloxifene, an inhibitor of estrogen-induced transcription, and G15, a G-protein-coupled estrogen receptor (GPER) 1 inhibitor, had little or no effect. Furthermore, the E2-activated matrix mineralization was disrupted by PMA, a PKC activator, and SB202190, a p38 MAPK inhibitor, but not by wortmannin, a PI3K inhibitor. Matrix mineralization was also induced by the culture media from the E2-stimulated cell culture. This effect was hindered by PMA or heat treatment, but not by SB202190. These results indicate that E2 activates the p38 MAPK pathway via ERs independently from actions in the nucleus. Such activation may cause the secretion of certain signaling molecule(s), which inhibit the PKC pathway. Our study provides a novel pathway of E2 action that could be a therapeutic target to activate matrix mineralization under various diseases, including osteoporosis.


Asunto(s)
Estradiol , Osteoblastos , Transducción de Señal , Animales , Ratones , Estradiol/farmacología , Osteoblastos/metabolismo , Osteoblastos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Calcificación Fisiológica/efectos de los fármacos , Línea Celular , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Receptores de Estrógenos/metabolismo , Receptores de Estrógenos/genética , Estrógenos/farmacología , Estrógenos/metabolismo , Receptor alfa de Estrógeno/metabolismo , Receptor alfa de Estrógeno/genética
12.
Biomed Eng Online ; 23(1): 44, 2024 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-38705993

RESUMEN

BACKGROUND: Osteocytes are critical mechanosensory cells in bone, and mechanically stimulated osteocytes produce exosomes that can induce osteogenesis. MicroRNAs (miRNAs) are important constituents of exosomes, and some miRNAs in osteocytes regulate osteogenic differentiation; previous studies have indicated that some differentially expressed miRNAs in mechanically strained osteocytes likely influence osteoblastic differentiation. Therefore, screening and selection of miRNAs that regulate osteogenic differentiation in exosomes of mechanically stimulated osteocytes are important. RESULTS: A mechanical tensile strain of 2500 µÎµ at 0.5 Hz 1 h per day for 3 days, elevated prostaglandin E2 (PGE2) and insulin-like growth factor-1 (IGF-1) levels and nitric oxide synthase (NOS) activity of MLO-Y4 osteocytes, and promoted osteogenic differentiation of MC3T3-E1 osteoblasts. Fourteen miRNAs differentially expressed only in MLO-Y4 osteocytes which were stimulated with mechanical tensile strain, were screened, and the miRNAs related to osteogenesis were identified. Four differentially expressed miRNAs (miR-1930-3p, miR-3110-5p, miR-3090-3p, and miR-3058-3p) were found only in mechanically strained osteocytes, and the four miRNAs, eight targeted mRNAs which were differentially expressed only in mechanically strained osteoblasts, were also identified. In addition, the mechanically strained osteocyte-derived exosomes promoted the osteoblastic differentiation of MC3T3-E1 cells in vitro, the exosomes were internalized by osteoblasts, and the up-regulated miR-3110-5p and miR-3058-3p in mechanically strained osteocytes, were both increased in the exosomes, which was verified via reverse transcription quantitative polymerase chain reaction (RT-qPCR). CONCLUSIONS: In osteocytes, a mechanical tensile strain of 2500 µÎµ at 0.5 Hz induced the fourteen differentially expressed miRNAs which probably were in exosomes of osteocytes and involved in osteogenesis. The mechanically strained osteocyte-derived exosomes which contained increased miR-3110-5p and miR-3058-3p (two of the 14 miRNAs), promoted osteoblastic differentiation.


Asunto(s)
Exosomas , MicroARNs , Osteocitos , Osteogénesis , Estrés Mecánico , Animales , Ratones , Línea Celular , Exosomas/metabolismo , Regulación de la Expresión Génica , MicroARNs/genética , MicroARNs/metabolismo , Osteoblastos/citología , Osteoblastos/metabolismo , Osteocitos/citología , Osteocitos/metabolismo , Osteogénesis/genética
13.
J Pharm Biomed Anal ; 245: 116192, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38703747

RESUMEN

Calcium sensing receptor (CaSR) has become the novel target of treating osteoporosis with herbal medicine Ligustri Lucidi Fructus (LLF), however, the bioactive compounds responsible for anti-osteoporosis are hard to clarify due to the complexity and diversity of chemical constituents in it. Herein, the immobilized CaSR column was packed with stationary phase materials, which were derived from integrating CLIP-tagged CaSR directly out of crude cell lysates onto the surface of silica gels (5.83 mg/g) in a site-specific covalent manner. The column had a great specificity of recognizing agonists and kept a good stability for at least 3 weeks. The two compounds from LLF extract were screened and identified as olenuezhenoside and ligustroflavone using the immobilized CaSR column in conjunction with mass spectrometry. Molecular docking predicted that both compounds were bound in venus flytrap (VFT) domain of CaSR by the formation of hydrogen bonds. Cellular results showed that both compounds exhibited the distinct osteogenic activity by enhancing the proliferation, differentiation and mineralization of osteoblastic cells. Our study demonstrated that, the immobilized protein column enables to screen the bioactive compounds rapidly from herbal extract, and the newly discovered natural product ligands towards CaSR, including olenuezhenoside and ligustroflavone, will be the candidates for the treatment of osteoporosis.


Asunto(s)
Ligustrum , Simulación del Acoplamiento Molecular , Osteogénesis , Extractos Vegetales , Receptores Sensibles al Calcio , Receptores Sensibles al Calcio/metabolismo , Receptores Sensibles al Calcio/antagonistas & inhibidores , Osteogénesis/efectos de los fármacos , Extractos Vegetales/farmacología , Extractos Vegetales/química , Ligustrum/química , Humanos , Osteoblastos/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Frutas/química , Animales , Osteoporosis/tratamiento farmacológico
14.
JCI Insight ; 9(10)2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38713511

RESUMEN

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.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Resorción Ósea , Osteoblastos , Osteoclastos , Receptores del Factor de Crecimiento Derivado de Plaquetas , Transducción de Señal , Animales , Osteoblastos/metabolismo , Ratones , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Resorción Ósea/metabolismo , Osteoclastos/metabolismo , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptores del Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Vía de Señalización Wnt/efectos de los fármacos , Anticuerpos Neutralizantes/farmacología , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/metabolismo , Factor Estimulante de Colonias de Macrófagos/metabolismo , Linaje de la Célula , Osteogénesis/efectos de los fármacos , Diferenciación Celular
15.
J Histochem Cytochem ; 72(5): 309-327, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38725403

RESUMEN

To clarify the cellular mechanism of cortical porosity induced by intermittent parathyroid hormone (PTH) administration, we examined the femoral cortical bone of mice that received 40 µg/kg/day (four times a day) human PTH (hPTH) (1-34). The PTH-driven cortical porosity initiated from the metaphyseal region and chronologically expanded toward the diaphysis. Alkaline phosphatase (ALP)-positive osteoblasts in the control mice covered the cortical surface, and endomucin-positive blood vessels were distant from these osteoblasts. In PTH-administered mice, endomucin-reactive blood vessels with TRAP-positive penetrated the ALP-positive osteoblast layer, invading the cortical bone. Statistically, the distance between endomucin-positive blood vessels and the cortical bone surface abated after PTH administration. Transmission electron microscopic observation demonstrated that vascular endothelial cells often pass through the flattened osteoblast layer and accompanied osteoclasts in the deep region of the cortical bone. The cell layers covering mature osteoblasts thickened with PTH administration and exhibited ALP, α-smooth muscle actin (αSMA), vascular cell adhesion molecule-1 (VCAM1), and receptor activator of NF-κB ligand (RANKL). Within these cell layers, osteoclasts were found near endomucin-reactive blood vessels. In PTH-administered femora, osteocytes secreted Dkk1, a Wnt inhibitor that affects angiogenesis, and blood vessels exhibited plasmalemma vesicle-associated protein, an angiogenic molecule. In summary, endomucin-positive blood vessels, when accompanied by osteoclasts in the ALP/αSMA/VCAM1/RANKL-reactive osteoblastic cell layers, invade the cortical bone, potentially due to the action of osteocyte-derived molecules such as DKK1.


Asunto(s)
Hueso Cortical , Células Endoteliales , Hormona Paratiroidea , Animales , Ratones , Hormona Paratiroidea/farmacología , Hormona Paratiroidea/administración & dosificación , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Hueso Cortical/efectos de los fármacos , Hueso Cortical/metabolismo , Porosidad , Masculino , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Inmunohistoquímica , Fémur/efectos de los fármacos , Fémur/irrigación sanguínea , Fémur/metabolismo , Humanos
16.
Biomed Mater ; 19(4)2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38740037

RESUMEN

The purpose of this study was to construct a rutin-controlled release system on the surface of Ti substrates and investigate its effects on osteogenesis and osseointegration on the surface of implants. The base layer, polyethylenimine (PEI), was immobilised on a titanium substrate. Then, hyaluronic acid (HA)/chitosan (CS)-rutin (RT) multilayer films were assembled on the PEI using layer-by-layer (LBL) assembly technology. We used scanning electron microscopy (SEM), Fourier transform infrared (FTIR) spectroscopy and contact angle measurements to examine all Ti samples. The drug release test of rutin was also carried out to detect the slow-release performance. The osteogenic abilities of the samples were evaluated by experiments on an osteoporosis rat model and MC3T3-E1 cells. The results (SEM, FTIR and contact angle measurements) all confirmed that the PEI substrate layer and HA/CS-RT multilayer film were effectively immobilised on titanium. The drug release test revealed that a rutin controlled release mechanism had been successfully established. Furthermore, thein vitrodata revealed that osteoblasts on the coated titanium matrix had greater adhesion, proliferation, and differentiation capacity than the osteoblasts on the pure titanium surface. When MC3T3-E1 cells were exposed to H2O2-induced oxidative stressin vitro, cell-based tests revealed great tolerance and increased osteogenic potential on HA/CS-RT substrates. We also found that the HA/CS-RT coating significantly increased the new bone mass around the implant. The LBL-deposited HA/CS-RT multilayer coating on the titanium base surface established an excellent rutin-controlled release system, which significantly improved osseointegration and promoted osteogenesis under oxidative stress conditions, suggesting a new implant therapy strategy for patients with osteoporosis.


Asunto(s)
Materiales Biocompatibles Revestidos , Ácido Hialurónico , Oseointegración , Osteoblastos , Osteogénesis , Osteoporosis , Prótesis e Implantes , Rutina , Propiedades de Superficie , Titanio , Animales , Titanio/química , Rutina/química , Rutina/farmacología , Osteogénesis/efectos de los fármacos , Ratas , Osteoporosis/tratamiento farmacológico , Ratones , Osteoblastos/efectos de los fármacos , Osteoblastos/citología , Osteoblastos/metabolismo , Oseointegración/efectos de los fármacos , Ácido Hialurónico/química , Materiales Biocompatibles Revestidos/química , Materiales Biocompatibles Revestidos/farmacología , Oxidación-Reducción , Quitosano/química , Femenino , Ratas Sprague-Dawley , Adhesión Celular/efectos de los fármacos , Espectroscopía Infrarroja por Transformada de Fourier , Diferenciación Celular/efectos de los fármacos , Microscopía Electrónica de Rastreo , Proliferación Celular/efectos de los fármacos , Polietileneimina/química , Células 3T3 , Estrés Oxidativo/efectos de los fármacos , Nanopartículas Capa por Capa
17.
Iran Biomed J ; 28(2&3): 82-9, 2024 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-38770885

RESUMEN

Background: Osteogenic, antioxidant and anti-inflammatory effects of Whey protein and M. oleifera gel prompted us to evaluate their role alone or in combination on osseointegration in rabbits. Methods: In this study, 24 titanium implants were inserted in the femurs of six rabbits. One implant was placed without treatment, and another one was coated with a mixture of whey protein and M. oleifera gel for each side. The animals were divided into two groups of 2- and 6-week intervals and evaluated using histopathological and immunohistochemical techniques. Results: Histological evaluation revealed a significant difference between the experimental and the control groups after two weeks in osteoblast and osteocyte counts. The experimental group had mature bone development after six weeks of implantation, while the control group had a woven bone. Immunohistochemical results showed that the experimental group, compared to the control group, exhibited early positive expression of osteoblast cells at two weeks after the experiment. Based on histopathological observations, the experimental group showed a tiny area of collagenous fiber in 6th week after the implantation. Conclusion: A mixture of whey protein and M. oleifera could accelerate osseointegration and healing processes.


Asunto(s)
Moringa oleifera , Oseointegración , Extractos Vegetales , Hojas de la Planta , Proteína de Suero de Leche , Animales , Proteína de Suero de Leche/farmacología , Conejos , Oseointegración/efectos de los fármacos , Moringa oleifera/química , Extractos Vegetales/farmacología , Hojas de la Planta/química , Masculino , Osteoblastos/efectos de los fármacos , Fémur/efectos de los fármacos , Osteogénesis/efectos de los fármacos
18.
Bull Exp Biol Med ; 176(5): 620-625, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38733480

RESUMEN

We studied the interaction of human buccal mesenchymal stem cells (MSCs) and osteoblasts differentiated from them with the surface of titanium samples. MSCs were isolated by enzymatic method from buccal fat pads. The obtained cell culture was presented by MSCs, which was confirmed by flow cytometry and differentiation into adipocytes and osteoblasts. Culturing of buccal MSCs on titanium samples was accompanied by an increase in the number of cells for 15 days and the formation of a developed network of F-actin fibers in the cells. The viability of buccal MSCs decreased by 8 days, but was restored by 15 days. Culturing of osteoblasts obtained as a result of buccal MSC differentiation on the surface of titanium samples was accompanied by a decrease in their viability and proliferation. Thus, MSCs from buccal fat pads can be used to coat implants to improve osseointegration during bone reconstruction in craniofacial surgery and dentistry. To improve the integration of osteoblasts, modification of the surface of titanium samples is required.


Asunto(s)
Diferenciación Celular , Células Madre Mesenquimatosas , Oseointegración , Osteoblastos , Titanio , Titanio/química , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/fisiología , Humanos , Oseointegración/fisiología , Osteoblastos/citología , Osteoblastos/fisiología , Células Cultivadas , Proliferación Celular , Implantes Dentales , Supervivencia Celular , Adipocitos/citología , Adipocitos/fisiología , Mucosa Bucal/citología , Osteogénesis/fisiología
19.
Front Immunol ; 15: 1397432, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38751427

RESUMEN

Introduction: The release of mature interleukin (IL-) 1ß from osteoblasts in response to danger signals is tightly regulated by the nucleotide-binding oligomerization domain leucine-rich repeat and pyrin-containing protein 3 (NLRP3) inflammasome. These danger signals include wear products resulting from aseptic loosening of joint arthroplasty. However, inflammasome activation requires two different signals: a nuclear factor-kappa B (NF-κB)-activating priming signal and an actual inflammasome-activating signal. Since human osteoblasts react to wear particles via Toll-like receptors (TLR), particles may represent an inflammasome activator that can induce both signals. Methods: Temporal gene expression profiles of TLRs and associated intracellular signaling pathways were determined to investigate the period when human osteoblasts take up metallic wear particles after initial contact and initiate a molecular response. For this purpose, human osteoblasts were treated with metallic particles derived from cobalt-chromium alloy (CoCr), lipopolysaccharides (LPS), and tumor necrosis factor-alpha (TNF) alone or in combination for incubation times ranging from one hour to three days. Shortly after adding the particles, their uptake was observed by the change in cell morphology and spectral data. Results: Exposure of osteoblasts to particles alone increased NLRP3 inflammasome-associated genes. The response was not significantly enhanced when cells were treated with CoCr + LPS or CoCr + TNF, whereas inflammation markers were induced. Despite an increase in genes related to the NLRP3 inflammasome, the release of IL-1ß was unaffected after contact with CoCr particles. Discussion: Although CoCr particles affect the expression of NLRP3 inflammasome-associated genes, a single stimulus was not sufficient to prime and activate the inflammasome. TNF was able to prime the NLRP3 inflammasome of human osteoblasts.


Asunto(s)
Regulación de la Expresión Génica , Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Osteoblastos , Factor de Necrosis Tumoral alfa , Humanos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Osteoblastos/metabolismo , Osteoblastos/efectos de los fármacos , Osteoblastos/inmunología , Inflamasomas/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Células Cultivadas , Transducción de Señal/efectos de los fármacos
20.
J Orthop Surg Res ; 19(1): 294, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38745231

RESUMEN

BACKGROUND: Osteonecrosis of the femoral head caused by glucocorticoids (GIONFH) is a significant issue resulting from prolonged or excessive clinical glucocorticoid use. Astaxanthin, an orange-red carotenoid present in marine organisms, has been the focus of this study to explore its impact and mechanism on osteoblast apoptosis induced by dexamethasone (Dex) and GIONFH. METHODS: In this experiment, bioinformatic prediction, molecular docking and dynamics simulation, cytotoxicity assay, osteogenic differentiation, qRT-PCR analysis, terminal uridine nickend labeling (TUNEL) assay, determination of intracellular ROS, mitochondrial function assay, immunofluorescence, GIONFH rat model construction, micro-computed tomography (micro-CT) scans were performed. RESULTS: Our research demonstrated that a low dose of astaxanthin was non-toxic to healthy osteoblasts and restored the osteogenic function of Dex-treated osteoblasts by reducing oxidative stress, mitochondrial dysfunction, and apoptosis. Furthermore, astaxanthin rescued the dysfunction in poor bone quality, bone metabolism and angiogenesis of GIONFH rats. The mechanism behind this involves astaxanthin counteracting Dex-induced osteogenic damage by activating the Nrf2 pathway. CONCLUSION: Astaxanthin shields osteoblasts from glucocorticoid-induced oxidative stress and mitochondrial dysfunction via Nrf2 pathway activation, making it a potential therapeutic agent for GIONFH treatment.


Asunto(s)
Necrosis de la Cabeza Femoral , Glucocorticoides , Mitocondrias , Factor 2 Relacionado con NF-E2 , Osteoblastos , Osteogénesis , Estrés Oxidativo , Xantófilas , Animales , Xantófilas/farmacología , Estrés Oxidativo/efectos de los fármacos , Factor 2 Relacionado con NF-E2/metabolismo , Glucocorticoides/efectos adversos , Glucocorticoides/toxicidad , Necrosis de la Cabeza Femoral/inducido químicamente , Necrosis de la Cabeza Femoral/metabolismo , Osteogénesis/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Ratas , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Masculino , Dexametasona/farmacología , Dexametasona/efectos adversos , Ratas Sprague-Dawley , Apoptosis/efectos de los fármacos , Modelos Animales de Enfermedad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA