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

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Front Plant Sci ; 14: 1305069, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38126008

RESUMO

Biological effects of magnetic fields have been extensively studied in plants, microorganisms and animals, and applications of magnetic fields in regulation of plant growth and phytoprotection is a promising field in sustainable agriculture. However, the effect of magnetic fields especially ultra-high static magnetic field (UHSMF) on genomic stability is largely unclear. Here, we investigated the mutagenicity of 24.5, 30.5 and 33.0 T UHSMFs with the gradient of 150, 95 and 0 T/m, respectively, via whole genome sequencing. Our results showed that 1 h exposure of Arabidopsis dried seeds to UHSMFs has no significant effect on the average rate of DNA mutations including single nucleotide variations and InDels (insertions and deletions) in comparison with the control, but 33.0 T and 24.5 T treatments lead to a significant change in the rate of nucleotide transitions and InDels longer than 3 bp, respectively, suggesting that both strength and gradient of UHSMF impact molecular spectrum of DNA mutations. We also found that the decreased transition rate in UHSMF groups is correlated with the upstream flanking sequences of G and C mutation sites. Furthermore, the germination rate of seeds exposed to 24.5 T SMF with -150 T/m gradient showed a significant decrease at 24 hours after sowing. Overall, our data lay a basis for precisely assessing the potential risk of UHSMF on DNA stability, and for elucidating molecular mechanism underlying gradient SMF-regulated biological processes in the future.

2.
Cells ; 11(5)2022 02 27.
Artigo em Inglês | MEDLINE | ID: mdl-35269449

RESUMO

Magnetobiological effects on growth and virulence have been widely reported in Escherichia coli (E. coli). However, published results are quite varied and sometimes conflicting because the underlying mechanism remains unknown. Here, we reported that the application of 250 mT static magnetic field (SMF) significantly reduces the diameter of E. coli colony-forming units (CFUs) but has no impact on the number of CFUs. Transcriptomic analysis revealed that the inhibitory effect of SMF is attributed to differentially expressed genes (DEGs) primarily involved in carbon source utilization. Consistently, the addition of glycolate or glyoxylate to the culture media successfully restores the bacterial phenotype in SMF, and knockout mutants lacking glycolate oxidase are no longer sensitive to SMF. These results suggest that SMF treatment results in a decrease in glycolate oxidase activity. In addition, metabolomic assay showed that long-chain fatty acids (LCFA) accumulate while phosphatidylglycerol and middle-chain fatty acids decrease in the SMF-treated bacteria, suggesting that SMF inhibits LCFA degradation. Based on the published evidence together with ours derived from this study, we propose a model showing that free radicals generated by LCFA degradation are the primary target of SMF action, which triggers the bacterial oxidative stress response and ultimately leads to growth inhibition.


Assuntos
Escherichia coli , Campos Magnéticos , Carbono/metabolismo , Carbono/farmacologia , Escherichia coli/metabolismo , Ácidos Graxos/metabolismo , Estresse Oxidativo
3.
Bioelectromagnetics ; 42(3): 200-211, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33655538

RESUMO

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.


Assuntos
Osteoclastos , Osteogênese , Animais , Diferenciação Celular , Campos Magnéticos , Camundongos , Osteoblastos
4.
Stem Cell Res Ther ; 11(1): 487, 2020 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-33198804

RESUMO

BACKGROUND: Osteoporosis is a common metabolic bone disease without effective treatment. Bone marrow-derived mesenchymal stem cells (BMSCs) have the potential to differentiate into multiple cell types. Increased adipogenic differentiation or reduced osteogenic differentiation of BMSCs might lead to osteoporosis. Whether static magnetic fields (SMFs) might influence the adipo-osteogenic differentiation balance of BMSCs remains unknown. METHODS: The effects of SMFs on lineage differentiation of BMSCs and development of osteoporosis were determined by various biochemical (RT-PCR and Western blot), morphological (staining and optical microscopy), and micro-CT assays. Bioinformatics analysis was also used to explore the signaling pathways. RESULTS: In this study, we found that SMFs (0.2-0.6 T) inhibited the adipogenic differentiation of BMSCs but promoted their osteoblastic differentiation in an intensity-dependent manner. Whole genomic RNA-seq and bioinformatics analysis revealed that SMF (0.6 T) decreased the PPARγ-mediated gene expression but increased the RUNX2-mediated gene transcription in BMSCs. Moreover, SMFs markedly alleviated bone mass loss induced by either dexamethasone or all-trans retinoic acid in mice. CONCLUSIONS: Taken together, our results suggested that SMF-based magnetotherapy might serve as an adjunctive therapeutic option for patients with osteoporosis.


Assuntos
Células-Tronco Mesenquimais , Osteoporose , Animais , Diferenciação Celular , Células Cultivadas , Humanos , Campos Magnéticos , Camundongos , Osteogênese , Osteoporose/terapia
5.
Bioelectromagnetics ; 41(8): 598-610, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33179793

RESUMO

The purpose of this study was to investigate the effects of moderate-intensity static magnetic field (SMF) on diabetic mice. We studied the effects of SMF on blood glucose of normal mice by starch tolerance and glucose tolerance tests. Then, we evaluated the effects of SMF on blood glucose of diabetic mice by establishing alloxan-induced type 1 diabetic mice and high-fat diet + streptozotocin (STZ)-induced type 2 diabetic mice. The results showed that different magnetic field intensities and blank control did not affect the blood glucose of normal mice. After starch and glucose administration, different magnetic fields could improve the glucose tolerance of normal mice, and this was obvious in the 600 mT group. In the experiment of type 1 diabetic mice induced by alloxan, the results showed that different magnetic field intensities could improve the starch tolerance of mice, and that in the 400 mT group was obvious. In the experiment of type 2 diabetic mice induced by a high-fat diet + STZ, the 400 mT group could reduce food intake and water consumption in the later period. The 600 mT group could improve the starch tolerance of mice. The 400 and 600 mT groups could reduce fasting blood glucose. At the same time, total cholesterol and triglyceride decreased in different magnetic field intensities, and the 600 mT group could significantly increase the serum insulin content of mice. In summary, the results of this study suggest that SMF has a protective role in diabetic mice. Bioelectromagnetics. © 2020 Bioelectromagnetics Society.


Assuntos
Diabetes Mellitus Experimental/sangue , Campos Magnéticos , Animais , Glicemia/metabolismo , Teste de Tolerância a Glucose , Masculino , Camundongos
6.
Sci Rep ; 10(1): 14519, 2020 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-32884074

RESUMO

With the discovery of magnetoreceptor mechanisms in animals, it materialized the novel applications of controlling cell and animal behaviors using magnetic fields. T cells have shown to be sensitive to magnetic fields. Here, we reported that exposure to moderate SMFs (static magnetic fields) led to increased granule and cytokine secretion as well as ATP production and mitochondrial respiration from CD8+ T cells. These effects were inhibited by knocking down the Uqcrb and Ndufs6 genes of mitochondrial respiratory chain, whose transcriptions were regulated by candidate magnetoreceptor genes Isca1 and Cry1/Cry2. SMF exposure also promoted CD8+ T cell granule and cytokine secretion and repressed tumor growth in vivo. SMFs enhanced CD8+ T cell cytotoxicity, and the adoptive transfer into tumor-bearing mice resulted in enhanced antitumor effects. Collectively, our study suggests that moderate SMFs enhance CD8+ T cell cytotoxicity by promoting mitochondrial respiration and promoted the antitumor function of CD8+ T cells.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Campos Magnéticos , Mitocôndrias/metabolismo , Imunidade Adaptativa/fisiologia , Trifosfato de Adenosina/metabolismo , Animais , Linhagem Celular , Camundongos , Camundongos Endogâmicos C57BL
7.
J Bone Miner Res ; 35(6): 1163-1173, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31880821

RESUMO

Exposure of humans and animals to microgravity in spaceflight results in various deleterious effects on bone health. In addition to microgravity, the hypomagnetic field (HyMF) is also an extreme environment in space, such as on the Moon and Mars; magnetic intensity is far weaker than the geomagnetic field (GMF) on Earth. Recently, we showed that HyMF promoted additional bone loss in hindlimb unloading-induced bone loss, and the underlying mechanism probably involved an increase of body iron storage. Numerous studies have indicated that bone loss induced by mechanical unloading can be largely restored after skeletal reloading in GMF conditions. However, it is unknown whether this bone deficit can return to a healthy state under HyMF condition. Therefore, the purpose of this study is to examine the effects of HyMF on the recovery of microgravity-induced bone loss, and illustrates the changes of body iron storage in this process. Our results showed that there was lower bone mineral content (BMC) in the HyMF reloading group compared to the GMF reloading group. Reloaded mice in the HyMF condition had a worse microstructure of femur than in the GMF condition. Femoral mechanical properties, including elastic modulus, stiffness, and ultimate stress, were poorer and toughness was higher in the HyMF group compared with the GMF group. Simultaneously, more iron content in serum, the tibia, liver, and spleen was found under HyMF reloading than GMF reloading. The iron chelator deferoxamine mesylate (DFO) decreased the iron content in the bone, liver, and spleen, and significantly relieved unloading-induced bone loss under HyMF reloading. These results showed that HyMF inhibits the recovery of microgravity-induced bone loss, probably by suppressing the elevated iron levels' return to physiological level. © 2019 American Society for Bone and Mineral Research.


Assuntos
Doenças Ósseas Metabólicas , Elevação dos Membros Posteriores , Animais , Densidade Óssea , Doenças Ósseas Metabólicas/etiologia , Osso e Ossos , Ferro , Camundongos
8.
Bioelectromagnetics ; 41(1): 52-62, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31802523

RESUMO

We established three types of thrombosis models to explore the effects of the static magnetic field (SMF) on thrombosis in rats and mice with three different MF intensities. In the carrageenan-induced thrombosis model in rats, the SMF treatments reduced the black tail length of rats, extracorporeal thrombus, and the mass of wet and dry thrombus, and improved the coagulation index value. In FeCl3 -induced arterial thrombosis model in rats, the SMF treatment showed some anti-thrombotic effects. More specifically, the SMF treatment affected rodent blood pressure, plasma plasminogen activator inhibitor, tissue-type plasminogen activator, thrombus mass, and thrombus protein content. In the adrenaline-induced thrombosis model in mice, the SMF treatment had certain effects on the diameter and blood flow velocity of mouse auricle microcirculation in fine veins and arteries. Overall, the highest MF intensities we tested, 20-150 mT, showed a trend of anti-thrombotic effect, indicating that the moderate-intensity SMF might serve as a potential treatment for clot-related diseases in the future. Bioelectromagnetics. 2020;41:52-62 © 2019 Bioelectromagnetics Society.


Assuntos
Campos Magnéticos/efeitos adversos , Trombose/prevenção & controle , Animais , Velocidade do Fluxo Sanguíneo , Pressão Sanguínea , Carragenina/metabolismo , Epinefrina/metabolismo , Frequência Cardíaca , Compostos de Ferro/metabolismo , Masculino , Camundongos , Microcirculação , Ativadores de Plasminogênio/metabolismo , Ratos , Ratos Sprague-Dawley , Ativador de Plasminogênio Tecidual/metabolismo
9.
J Diabetes Res ; 2019: 5641271, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31886281

RESUMO

Impaired wound healing is commonly encountered in patients with diabetes mellitus, which may lead to severe outcomes such as amputation, if untreated timely. Macrophage plays a critical role in the healing process including the resolution phase. Although magnetic therapy is known to improve microcirculation, its effect on wound healing remains uncertain. In the present study, we found that 0.6 T static magnetic field (SMF) significantly accelerated wound closure and elevated reepithelialization and revascularization in diabetic mice. Notably, SMF promoted the wound healing by skewing the macrophage polarization towards M2 phenotype, thus facilitating the resolution of inflammation. In addition, SMF upregulated anti-inflammatory gene expression via activating STAT6 and suppressing STAT1 in macrophage. Taken together, our results indicate that SMF may be a promising adjuvant therapeutic tool for treating diabetic wounds.


Assuntos
Angiopatias Diabéticas/terapia , Inflamação/terapia , Magnetoterapia , Pele/patologia , Cicatrização , Animais , Células Cultivadas , Angiopatias Diabéticas/genética , Angiopatias Diabéticas/metabolismo , Angiopatias Diabéticas/patologia , Modelos Animais de Doenças , Inflamação/metabolismo , Inflamação/patologia , Mediadores da Inflamação/metabolismo , Macrófagos Peritoneais/metabolismo , Macrófagos Peritoneais/patologia , Masculino , Camundongos Endogâmicos , Fenótipo , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT6/genética , Fator de Transcrição STAT6/metabolismo , Transdução de Sinais , Pele/metabolismo , Fatores de Tempo
10.
Sci Rep ; 9(1): 14384, 2019 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-31591431

RESUMO

Static magnetic field (SMF) plays important roles in biological processes of many living organisms. In plants, however, biological significance of SMF and molecular mechanisms underlying SMF action remain largely unknown. To address these questions, we treated Arabidopsis young seedlings with different SMF intensities and directions. Magnetic direction from the north to south pole was adjusted in parallel (N0) with, opposite (N180) and perpendicular to the gravity vector. We discovered that root growth is significantly inhanced by 600 mT treatments except for N180, but not by any 300 mT treatments. N0 treatments lead to more active cell division of the meristem, and higher auxin content that is regulated by coordinated expression of PIN3 and AUX1 in root tips. Consistently, N0-promoted root growth disappears in pin3 and aux1 mutants. Transcriptomic and gene ontology analyses revealed that in roots 85% of the total genes significantly down-regulated by N0 compared to untreatment are enriched in plastid biological processes, such as metabolism and chloroplast development. Lastly, no difference in root length is observed between N0-treated and untreated roots of the double cryptochrome mutant cry1 cry2. Taken together, our data suggest that SMF-regulated root growth is mediated by CRY and auxin signaling pathways in Arabidopsis.


Assuntos
Arabidopsis/citologia , Arabidopsis/crescimento & desenvolvimento , Ácidos Indolacéticos/metabolismo , Campos Magnéticos , Raízes de Plantas/crescimento & desenvolvimento , Transdução de Sinais , Folhas de Planta/crescimento & desenvolvimento
11.
Eur Radiol ; 29(11): 6029-6037, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31115627

RESUMO

OBJECTIVES: We aimed to evaluate the biological effects of high static magnetic field (HiSMF, 2-12 Tesla [T]) exposure on mice in a stable and effective breeding environment in the chamber of a superconducting magnet. METHODS: C57BL/6 mice were bred in the geomagnetic field and HiSMF with different magnetic field strengths (2-4 T, 6-8 T, and 10-12 T) for 28 days. The body weight, blood indices, organ coefficients, and histomorphology of major organs were analyzed. RESULTS: The results showed that the HiSMF had no significant effect on the body weight, organ coefficients, or histomorphology of major organs in mice. The HiSMF had no effect on most routine blood and biochemical indices, but the value of the mean corpuscular hemoglobin (MCH) was increased in the 2-4 T group compared with that of the other groups, and the uric acid level (UA) was decreased in the three HiSMF groups compared with that of the control group. CONCLUSION: The C57BL/6 mice were not affected when they were exposed to different HiSMF environments for 28 days. KEY POINTS: • No physiological problems were observed in mice with long-term whole-body exposure to HiSMF.


Assuntos
Campos Magnéticos , Imageamento por Ressonância Magnética/métodos , Exposição à Radiação , Animais , Peso Corporal , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais
12.
Am J Cancer Res ; 8(10): 1933-1946, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30416846

RESUMO

Ferroptosis is an iron depend cell death which caused by lipid peroxidation. Abnormal iron metabolism and high intracellular iron content are the characteristics of most cancer cells. Iron is a promoter of cell growth and proliferation. However, iron also could take part in Fenton reaction to produce reactive oxygen species (ROS). The intercellular ROS could induce lipid peroxidation, which is necessary for ferroptosis. Iron metabolism mainly includes three parts: iron uptake, storage and efflux. Therefore, iron metabolism-related genes could regulate intercellular iron content and status, which can be involved ferroptosis. In recent years, the application of nanoparticles in cancer therapy research has become more and more extensive. The iron-based nanoparticles (iron-based NPs) can release ferrous (Fe2+) or ferric (Fe3+) in acidic lysosomes and inducing ferroptosis. Magnetic field is widely used in the targeted concentration of iron-based NPs related disease therapy. Furthermore, multiple studies showed that magnetic fields can inhibit cancer cell proliferation by promoting intracellular ROS production. Herein, we focus on the relationship of between ferroptosis and iron metabolism in cancer cells, the application of nanoparticles and magnetic field in inducing ferroptosis of cancer cells, and trying to provide new ideas for cancer treatment research.

13.
Electromagn Biol Med ; 37(4): 192-201, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30142006

RESUMO

Moderate intensity low frequency rotating magnetic field (LF-RMF) has been shown to inhibit melanoma, liver and lung cancer growth in mice. However, its effects on other types of cancers have not been investigated in vivo. Here, we show that 0-0.15T moderate intensity 4.2 Hz LF-RMF can inhibit tumor growth in mice bearing MDA-MB231 and MCF7 human breast cancer cells by over 30%. In contrast, the human gastrointestinal stromal tumor GIST-T1 growth was not inhibited by LF-RMF. In all RMF treatments, there were no apparent adverse effects on mice organs, body weight or water/food consumptions. However, the alanine aminotransferase (ALT) level was decreased in LF-RMF-treated mice bearing MCF7 and GIST-T1 cells, which indicated alleviated liver damage. Therefore, our study shows that moderate intensity LF-RMF might be a safe physical method that has clinical potentials to be used to inhibit breast cancer growth in the future.


Assuntos
Magnetoterapia/métodos , Neoplasias Mamárias Experimentais/patologia , Neoplasias Mamárias Experimentais/terapia , Rotação , Animais , Transformação Celular Neoplásica , Feminino , Humanos , Células MCF-7 , Magnetoterapia/instrumentação , Camundongos
14.
Sheng Wu Yi Xue Gong Cheng Xue Za Zhi ; 30(1): 176-90, 2013 Feb.
Artigo em Chinês | MEDLINE | ID: mdl-23488161

RESUMO

Nowadays, health care products based on static magnetic fields (SMF) and merchandise of magnetic therapy are popular around the world. But the biomedical effects of SMF to animals or human beings remain a widely concerned controversy. In this paper, the recent researches in China and abroad about the biomedical effects of SMF were reviewed in three levels: the cellular, animal and human levels. Nevertheless, these data were not consistent with each other and even some contradicts others' researches. So, it is necessary to do more and further studies on SMF dosing regiman, sham control magnetic device and blinding procedures to obtain the optimal magnetic intensity, the desired therapeutic effects in practical cases and prepare for applying the SMF in biomedical fields more effectively in the future.


Assuntos
Magnetoterapia/métodos , Campos Magnéticos , Animais , Humanos , Neoplasias/terapia , Dor/prevenção & controle
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA