Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nanoscale Adv ; 6(8): 1957-1973, 2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38633036

RESUMEN

With the development of biotechnology, biomaterials have been rapidly developed and shown great potential in bone regeneration therapy and bone tissue engineering. Nanoparticles have attracted the attention of researches and have applied in various fields especially in the biomedical field as the special physicochemical properties. Nanoparticles were found to regulate bone remodeling depending on their size, shape, composition, and charge. Therefore, in-depth research was necessary to provide the basic support to select the most suitable nanoparticles for bone relate diseases treatment. This article reviews the current development of nanoparticles in bone tissue engineering, focusing on drug delivery, gene delivery, and cell labeling. In addition, the research progress on the interaction of nanoparticles with bone cells, focusing on osteoblasts, osteoclasts, and bone marrow mesenchymal stem cells, and the underlying mechanism were also reviewed. Finally, the current challenges and future research directions are discussed. Thus, detailed study of nanoparticles may reveal new therapeutic strategies to improve the effectiveness of bone regeneration therapy or other bone diseases.

2.
Prog Biophys Mol Biol ; 188: 43-54, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38447710

RESUMEN

The emergence, evolution, and spread of life on Earth have all occurred in the geomagnetic field, and its extensive biological effects on living organisms have been documented. The charged characteristics of metal ions in biological fluids determine that they are affected by electromagnetic field forces, thus affecting life activities. Iron metabolism, as one of the important metal metabolic pathways, keeps iron absorption and excretion in a relatively balanced state, and this process is precisely and completely controlled. It is worth paying attention to how the iron metabolism process of living organisms is changed when exposed to electromagnetic fields. In this paper, the processes of iron absorption, storage and excretion in animals (mammals, fish, arthropods), plants and microorganisms exposed to electromagnetic field were summarized in detail as far as possible, in order to discover the regulation of iron metabolism by electromagnetic field. Studies and data on the effects of electromagnetic field exposure on iron metabolism in organisms show that exposure profiles vary widely across species and cell lines. This process involves a variety of factors, and the complexity of the results is not only related to the magnetic flux density/operating frequency/exposure time and the heterogeneity of the observed object. A systematic review of the biological regulation of iron metabolism by electromagnetic field exposure will not only contributes to a more comprehensive understanding of its biological effects and mechanism, but also is necessary to improve human awareness of the health related risks of electromagnetic field exposure.


Asunto(s)
Campos Electromagnéticos , Mamíferos , Humanos , Animales , Campos Electromagnéticos/efectos adversos , Mamíferos/metabolismo , Tiempo , Hierro/metabolismo
3.
iScience ; 26(8): 107365, 2023 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-37554458

RESUMEN

Osteocytes are the mechano-sensors of bones. Large gradient high-static magnetic fields (LG-HMFs) produce stable, high-precision, and non-attenuation mechanical forces. We discovered that magnetic forces opposite to gravity inhibited MLO-Y4 osteocyte proliferation and viability by inducing structural damage and apoptosis. In contrast, magnetic force loading in the same direction as that of gravity promoted the proliferation and inhibited apoptosis of MLO-Y4 osteocytes. Differentially expressed gene (DEG) analysis after magnetic force stimulation indicated that the ECM-integrin-CSK axis responded most significantly to mechanical signals. Wisp2 was the most significant DEG between the 12 T upward and downward groups, showing the highest correlation with the Wnt pathway according to the STRING protein interaction database. Explaining the cellular and molecular mechanisms by which mechanical stimuli influence bone remodeling is currently the focus of osteocyte-related research. Our findings provide insights into the effects of LG-HMFs on bone cells, which have further implications in clinical practice.

4.
FASEB J ; 37(7): e22985, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37249350

RESUMEN

Osteoporosis is one of the chronic complications of type 1 diabetes with high risk of fracture. The prevention of diabetic osteoporosis is of particular importance. Static magnetic fields (SMFs) exhibit advantages on improvement of diabetic complications. The biological effects and mechanism of SMFs on bone health of type 1 diabetic mice and functions of bone cells under high glucose have not been clearly clarified. In animal experiment, six-week-old male C57BL/6J mice were induced to type 1 diabetes and exposed to SMF of 0.4-0.7 T for 4 h/day lasting for 6 weeks. Bone mass, biomechanical strength, microarchitecture and metabolism were determined by DXA, three-point bending assay, micro-CT, histochemical and biochemical methods. Exposure to SMF increased BMD and BMC of femur, improved biomechanical strength with higher ultimate stress, stiffness and elastic modulus, and ameliorated the impaired bone microarchitecture in type 1 diabetic mice by decreasing Tb.Pf, Ct.Po and increasing Ct.Th. SMF enhanced bone turnover by increasing the level of markers for bone formation (OCN and Collagen I) as well as bone resorption (CTSK and NFAT2). In cellular experiment, MC3T3-E1 cells or primary osteoblasts and RAW264.7 cells were cultured in 25 mM high glucose-stimulated diabetic marrow microenvironment under differentiation induction and exposed to SMF. SMF promoted osteogenesis with higher ALP level and mineralization deposition in osteoblasts, and it also enhanced osteoclastogenesis with higher TRAP activity and bone resorption in osteoclasts under high glucose condition. Further, SMF increased iron content with higher FTH1 expression and regulated the redox level through activating HO-1/Nrf2 in tibial tissues, and lowered hepatic iron accumulation by BMP6-mediated regulation of hepcidin and lipid peroxidation in mice with type 1 diabetes. Thus, SMF may act as a potential therapy for improving bone health in type 1 diabetes with regulation on iron homeostasis metabolism and redox status.


Asunto(s)
Resorción Ósea , Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 1 , Osteoporosis , Ratones , Masculino , Animales , Diabetes Mellitus Tipo 1/terapia , Diabetes Mellitus Experimental/terapia , Ratones Endogámicos C57BL , Osteoblastos/metabolismo , Osteogénesis , Hierro/metabolismo , Oxidación-Reducción , Campos Magnéticos , Glucosa
5.
J Orthop Translat ; 38: 126-140, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36381248

RESUMEN

Objective: With the deepening of magnetic biomedical effects and electromagnetic technology, some medical instruments based on static magnetic field (SMF) have been used in orthopedic-related diseases treatment. Studies have shown SMF could combat osteoporosis by regulating the differentiation of mesenchymal stem cells (MSCs), osteoblast and osteoclast. With the development of nanotechnology, iron oxide nanoparticles (IONPs) have been reported to regulate the process of bone anabolism. As for SMF combined with IONPs, studies indicated osteogenic differentiation of MSCs were promoted by the combination of SMF and IONPs. However, there are few reports on the effects of SMF combined with IONPs on osteoclast. Herein, the purpose of this study was to investigate the effects of high static magnetic field (HiSMF) combined with IONPs on unloading-induced bone loss in vivo and osteoclastic formation in vitro, and elucidated the potential molecular mechanisms. Methods: In vivo, C57BL/6 â€‹J male mice were unloaded via tail suspension or housed normally. The hindlimb of mice were fixed and exposed to 1-2 â€‹T SMF for 1 â€‹h every day, 10 â€‹mg/kg of Ferumoxytol or saline were injected by tail vein once a week, last for 4 weeks. Bone microstructure, mechanical properties, and osteoclastogenesis were examined respectively. In vitro, the RAW264.7 â€‹cells were used to assess the effects of 1-2 â€‹T SMF combined with IONPs in osteoclastogenesis. The iron content was detected by atomic absorption spectrometry and Prussian blue staining. DCFH-DA and MitoSOX™ fluorescence staining were used to assess oxidative stress levels. NF-κB and MAPK signaling pathways were examined by western blot assay. Results: In vivo, the results showed 1-2 â€‹T SMF and IONPs prevented the damage to bone microstructure and improved the mechanical properties, diminished the number of osteoclasts in unloaded mice, 1-2 â€‹T SMF combined with IONPs was found more effective. The iron content in the liver and spleen was reduced by the combination of 1-2 â€‹T SMF and IONPs, enhancing iron levels in the femur. In vitro, osteoclast formation was inhibited by 1-2 â€‹T SMF and IONPs treatment, and 1-2 â€‹T SMF combined with IONPs had a more pronounced effect. Moreover, iron uptake of IONPs in osteoclast was reduced to 1-2 â€‹T SMF exposure. Oxidative stress levels were decreased in osteoclast differentiation under 1-2 â€‹T SMF combined with IONPs treatment. Molecularly, the expression of NF-κB and MAPK signaling pathways were inhibited under 1-2 â€‹T SMF combined with IONPs in osteoclastogenesis. Conclusions: Synthetically, our research illustrated 1-2 â€‹T SMF combined with IONPs prevented unloading-induced bone loss by regulating iron metabolism in osteoclastogenesis.Translational potential of this article: As a non-invasive alternative therapy, some medical instruments based on SMF have been used for orthopedic-related diseases treatment for their portability, cheapness and safety. Ferumoxytol (Feraheme™), the first FDA-approved IONP drug for the treatment of iron deficiency anemia, has been also adapted in translational research for osteoporosis. Based on the above-mentioned two points, we found the synergistic effects of SMF and Ferumoxytol for treatment of experimental osteoporosis. These results show translational potentials for clinical application.

6.
Cells ; 11(20)2022 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-36291164

RESUMEN

Iron oxide nanoparticles (IONPs) are extensively used in bone-related studies as biomaterials due to their unique magnetic properties and good biocompatibility. Through endocytosis, IONPs enter the cell where they promote osteogenic differentiation and inhibit osteoclastogenesis. Static magnetic fields (SMFs) were also found to enhance osteoblast differentiation and hinder osteoclastic differentiation. Once IONPs are exposed to an SMF, they become rapidly magnetized. IONPs and SMFs work together to synergistically enhance the effectiveness of their individual effects on the differentiation and function of osteoblasts and osteoclasts. This article reviewed the individual and combined effects of different types of IONPs and different intensities of SMFs on bone remodeling. We also discussed the mechanism underlying the synergistic effects of IONPs and SMFs on bone remodeling.


Asunto(s)
Campos Magnéticos , Osteogénesis , Remodelación Ósea , Materiales Biocompatibles/farmacología , Nanopartículas Magnéticas de Óxido de Hierro
7.
Front Endocrinol (Lausanne) ; 13: 1048818, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36818465

RESUMEN

PTH 1-34 (teriparatide) is approved by FDA for the treatment of postmenopausal osteoporosis. Iron overload is a major contributing factor for bone loss induced by unloading. Whether iron metabolism is involved in the regulation of PTH 1-34 on unloading-induced osteoporosis has not yet been reported. Here, we found that PTH 1-34 attenuated bone loss in unloading mice. PTH 1-34 regulated the disturbance of iron metabolism in unloading mice by activating Nrf2 and further promoting hepcidin expression in the liver. In addition, the Nrf2 inhibitor selectively blocked hepcidin expression in the liver of unloading mice, which neutralized the inhibitory effect of PTH 1-34 on bone loss and the recovery of iron metabolism in unloading mice. Finally, we found that PTH 1-34 promoted the differentiation and inhibited apoptosis of osteoblasts by regulating iron metabolism and maintaining redox balance under unloading conditions. Our results suggested that PTH 1-34 promoted bone formation by regulating iron metabolism under unloading conditions.


Asunto(s)
Hepcidinas , Osteogénesis , Ratones , Animales , Hepcidinas/farmacología , Factor 2 Relacionado con NF-E2 , Hormona Paratiroidea/farmacología , Hierro
8.
Cells ; 10(12)2021 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-34944025

RESUMEN

There are numerous studies that investigate the effects of static magnetic fields (SMFs) on osteoblasts and osteoclasts. However, although osteocytes are the most abundant cell type in bone tissue, there are few studies on the biological effects of osteocytes under magnetic fields. Iron is a necessary microelement that is involved in numerous life activities in cells. Studies have shown that high static magnetic fields (HiSMF) can regulate cellular iron metabolism. To illustrate the effect of HiSMF on activities of osteocytes, and whether iron is involved in this process, HiSMF of 16 tesla (T) was used, and the changes in cellular morphology, cytoskeleton, function-related protein expression, secretion of various cytokines, and iron metabolism in osteocytes under HiSMF were studied. In addition, the biological effects of HiSMF combined with iron preparation and iron chelator on osteocytes were also investigated. The results showed that HiSMF promoted cellular viability, decreased apoptosis, increased the fractal dimension of the cytoskeleton, altered the secretion of cytokines, and increased iron levels in osteocytes. Moreover, it was found that the biological effects of osteocytes under HiSMF are attenuated or enhanced by treatment with a certain concentration of iron. These data suggest that HiSMF-regulated cellular iron metabolism may be involved in altering the biological effects of osteocytes under HiSMF exposure.


Asunto(s)
Apoptosis/genética , Supervivencia Celular/genética , Hierro/metabolismo , Osteocitos/efectos de la radiación , Animales , Apoptosis/efectos de la radiación , Supervivencia Celular/efectos de la radiación , Citoesqueleto/genética , Citoesqueleto/metabolismo , Citoesqueleto/efectos de la radiación , Regulación de la Expresión Génica/efectos de la radiación , Hierro/efectos de la radiación , Campos Magnéticos/efectos adversos , Ratones , Microtúbulos/genética , Microtúbulos/efectos de la radiación , Osteoblastos/metabolismo , Osteoblastos/efectos de la radiación , Osteoclastos/metabolismo , Osteoclastos/efectos de la radiación , Osteocitos/metabolismo , Células RAW 264.7
9.
Int J Mol Sci ; 22(13)2021 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-34281233

RESUMEN

Osteosarcoma is a common malignant bone tumor in clinical orthopedics. Iron chelators have inhibitory effects on many cancers, but their effects and mechanisms in osteosarcoma are still uncertain. Our in vitro results show that deferoxamine (DFO) and deferasirox (DFX), two iron chelators, significantly inhibited the proliferation of osteosarcoma cells (MG-63, MNNG/HOS and K7M2). The viability of osteosarcoma cells was decreased by DFO and DFX in a concentration-dependent manner. DFO and DFX generated reactive oxygen species (ROS), altered iron metabolism and triggered apoptosis in osteosarcoma cells. Iron chelator-induced apoptosis was due to the activation of the MAPK signaling pathway, with increased phosphorylation levels of JNK, p38 and ERK, and ROS generation; in this process, the expression of C-caspase-3 and C-PARP increased. In an orthotopic osteosarcoma transplantation model, iron chelators (20 mg/kg every day, Ip, for 14 days) significantly inhibited the growth of the tumor. Immunohistochemical analysis showed that iron metabolism was altered, apoptosis was promoted, and malignant proliferation was reduced with iron chelators in the tumor tissues. In conclusion, we observed that iron chelators induced apoptosis in osteosarcoma by activating the ROS-related MAPK signaling pathway. Because iron is crucial for cell proliferation, iron chelators may provide a novel therapeutic strategy for osteosarcoma.


Asunto(s)
Deferasirox/uso terapéutico , Deferoxamina/uso terapéutico , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Osteosarcoma/tratamiento farmacológico , Sideróforos/uso terapéutico , Animales , Apoptosis/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Deferasirox/farmacología , Deferoxamina/farmacología , Humanos , Hierro/metabolismo , Ratones , Osteosarcoma/metabolismo , Sideróforos/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Bioelectromagnetics ; 42(3): 200-211, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33655538

RESUMEN

Static magnetic field (SMF), with constant magnetic field strength and direction, has a long history of basic and clinical research in bone biology. Numerous studies demonstrate that exposure to moderate SMF (1 mT-1 T) can increase bone mass and bone density. However, few studies pay attention to the effects of high SMF (>1 T) on the skeletal system. To investigate the physiological effects of high SMF on bone, mice were exposed to 2-4 T SMF for 28 days. Bone microstructure and mechanical properties were examined. The activity of osteoblasts and osteoclasts involved in bone remodeling was evaluated in vivo and in vitro. Compared with the unexposed group, 2-4 T significantly improved the femoral microstructure and tibial mechanical properties. For bone remodeling in vivo, the number of osteoblasts and bone formation was increased, and the osteoclastic number was decreased by 2-4 T. Moreover, the expression of marker proteins in the femur and concentrations of biochemical indicators in serum involved in bone formation were elevated and bone resorption was reduced under 2-4 T SMF. In vitro, osteoblast differentiation was promoted, and the osteoclastic formation and bone resorption ability were inhibited by 2 T SMF. Overall, these results demonstrate that 2-4 T SMF improved bone microarchitecture and strength by stimulating bone formation and restraining bone resorption, and imply that high SMF might become a potential biophysical treatment modality for bone diseases with abnormal bone remodeling. Bioelectromagnetics. © 2021 Bioelectromagnetics Society.


Asunto(s)
Osteoclastos , Osteogénesis , Animales , Diferenciación Celular , Campos Magnéticos , Ratones , Osteoblastos
11.
J Tissue Eng Regen Med ; 13(12): 2181-2190, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31622531

RESUMEN

High static magnetic fields (HiSMFs) are usually defined as those SMFs with intensities ≥1 T. Although many studies have indicated that SMFs have positive effects on bone tissue, there were limited studies that investigate the effects of cells, including osteoclasts, to illustrate the effect of HiSMF on osteoclast differentiation, and whether iron involve in the altered osteoclast formation and resorption ability under HiSMF. 16 T HiSMF generated from a superconducting magnet was used. Osteoclastogenesis, bone resorption, acting ring formation, messenger ribonucleic acid expression, and protein expression were determined by tartrate-resistant acid phosphatase staining, pits formation assay, rhodamine-conjugated phalloidine staining, quantitative real-time polymerase chain reaction, and western blot, respectively. The changes induced by HiSMF in the level of iron and the concentration of mitochondrial protein, adenosine triphosphate, reactive oxygen species, malonaldehyde, and glutathione were examined by atomic absorption spectrometry and corresponding commercial kits, respectively. The results showed that HiSMF significantly inhibited osteoclastic formation and resorption ability and reduced cellular iron content during osteoclast differentiation. Mitochondrial concentration and oxidative stress levels in osteoclasts were decreased under HiSMF. Mechanistically, HiSMF markedly blocked the expression of osteoclast-associated transcription factors and osteoclast marker genes and inhibited iron absorption and iron storage-related protein expression. These findings demonstrated that the effect of HiSMF on iron metabolism of osteoclasts was involved in the inhibition of HiSMF on osteoclast differentiation.


Asunto(s)
Diferenciación Celular , Campos Magnéticos , Osteoclastos/metabolismo , Ligando RANK/metabolismo , Animales , Ratones , Osteoclastos/citología , Células RAW 264.7
12.
J Cell Physiol ; 234(6): 8028-8039, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30362549

RESUMEN

Iron is an essential micronutrient in mammalian cells for basic processes such as DNA synthesis, cell cycle progression, and mitochondrial activity. Macrophages play a vital role in iron metabolism, which is tightly linked to their phagocytosis of senescent and death erythrocytes. It is now recognized that the polarization process of macrophages determines the expression profile of genes associated with iron metabolism. Although iron metabolism is strictly controlled by physiology, cancer has recently been connected with disordered iron metabolism. Moreover, in the environment of cancer, tumor-associated macrophages (TAMs) exhibit an iron release phenotype, which stimulates tumor cell survival and growth. Usually, the abundance of TAMs in the tumor is implicated in poor disease prognosis. Therefore, important attention has been drawn toward the development of tumor immunotherapies targeting these TAMs focussing on iron metabolism and reprogramming polarized phenotypes. Although further systematic research is still required, these efforts are almost certainly valuable in the search for new and effective cancer treatments.


Asunto(s)
Hierro/metabolismo , Macrófagos/metabolismo , Mitocondrias/metabolismo , Neoplasias/inmunología , Ciclo Celular/genética , Proliferación Celular/genética , Supervivencia Celular/genética , Reprogramación Celular/inmunología , Humanos , Inmunoterapia , Macrófagos/inmunología , Macrófagos/patología , Mitocondrias/inmunología , Mitocondrias/patología , Neoplasias/metabolismo , Neoplasias/patología , Neoplasias/terapia , Microambiente Tumoral/genética
13.
Int J Mol Sci ; 19(9)2018 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-30177626

RESUMEN

The space environment chiefly includes microgravity and radiation, which seriously threatens the health of astronauts. Bone loss and muscle atrophy are the two most significant changes in mammals after long-term residency in space. In this review, we summarized current understanding of the effects of microgravity and radiation on the musculoskeletal system and discussed the corresponding mechanisms that are related to iron overload and oxidative damage. Furthermore, we enumerated some countermeasures that have a therapeutic potential for bone loss and muscle atrophy through using iron chelators and antioxidants. Future studies for better understanding the mechanism of iron and redox homeostasis imbalance induced by the space environment and developing the countermeasures against iron overload and oxidative damage consequently may facilitate human to travel more safely in space.


Asunto(s)
Sobrecarga de Hierro/fisiopatología , Sistema Musculoesquelético/fisiopatología , Vuelo Espacial , Animales , Humanos , Sobrecarga de Hierro/metabolismo , Modelos Teóricos , Sistema Musculoesquelético/metabolismo , Estrés Oxidativo/fisiología , Ingravidez
14.
Bone ; 114: 235-245, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29929042

RESUMEN

During deep-space exploration missions, astronauts will be exposed to abnormal space environments including microgravity and hypomagnetic field (HyMF) that is 10,000 times weaker than geomagnetic field (GMF). It is well known that microgravity in space can induce bone loss; however, it is ill-defined whether HyMF involved in this process. Herein, we aimed to investigate the combined effects of HyMF and microgravity on bone loss. A mouse model of hindlimb suspension (HLU) was adopted to simulate microgravity-induced bone loss, that was exposed to a hypomagnetic field of <300 nanotesla (nT) generated by a geomagnetic field-shielding chamber. Besides, a recent study showed that HLU induced bone loss was orchestrated by iron overload. Therefore, the changes of iron content in unloading-induced bone loss under HyMF condition were detected simultaneously. The results showed HyMF exacerbated the loss of bone mineral content (BMC), induced more detrimental effects on microstructure of cancellous bone but not cortical bone and yielded greater negative effects on biomechanical characteristics in mice femur under unloading status. Concomitantly, there was more iron accumulation in serum, liver, spleen and bone in the combined treatment group than in the separate unloading group or HyMF exposure group. These results showed that HyMF promoted additional bone loss in mice femur during mechanical unloading, and the potential mechanism may be involved in inducing iron overload of mice.


Asunto(s)
Resorción Ósea/diagnóstico por imagen , Suspensión Trasera/efectos adversos , Sobrecarga de Hierro/diagnóstico por imagen , Campos Magnéticos/efectos adversos , Animales , Resorción Ósea/etiología , Resorción Ósea/metabolismo , Sobrecarga de Hierro/complicaciones , Sobrecarga de Hierro/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Soporte de Peso/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...