Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 22
1.
Nanoscale Adv ; 6(8): 1957-1973, 2024 Apr 16.
Article En | MEDLINE | ID: mdl-38633036

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.
Article En | MEDLINE | ID: mdl-38447710

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.


Electromagnetic Fields , Mammals , Humans , Animals , Electromagnetic Fields/adverse effects , Mammals/metabolism , Time , Iron/metabolism
3.
Biomed Eng Online ; 22(1): 107, 2023 Nov 15.
Article En | MEDLINE | ID: mdl-37968671

BACKGROUND: Fractures are the most common orthopedic diseases. It is known that static magnetic fields (SMFs) can contribute to the maintenance of bone health. However, the effect and mechanism of SMFs on fracture is still unclear. This study is aim to investigate the effect of moderate static magnetic fields (MMFs) on bone structure and metabolism during fracture healing. METHODS: Eight-week-old male C57BL/6J mice were subjected to a unilateral open transverse tibial fracture, and following treatment under geomagnetic field (GMF) or MMF. The micro-computed tomography (Micro-CT) and three-point bending were employed to evaluate the microarchitecture and mechanical properties. Endochondral ossification and bone remodeling were evaluated by bone histomorphometric and serum biochemical assay. In addition, the atomic absorption spectroscopy and ELISA were utilized to examine the influence of MMF exposure on iron metabolism in mice. RESULTS: MMF exposure increased bone mineral density (BMD), bone volume per tissue volume (BV/TV), mechanical properties, and proportion of mineralized bone matrix of the callus during fracture healing. MMF exposure reduced the proportion of cartilage in the callus area during fracture healing. Meanwhile, MMF exposure increased the number of osteoblasts in callus on the 14th day, and reduced the number of osteoclasts on the 28th day of fracture healing. Furthermore, MMF exposure increased PINP and OCN levels, and reduced the TRAP-5b and ß-CTX levels in serum. It was also observed that MMF exposure reduced the iron content in the liver and callus, as well as serum ferritin levels while elevating the serum hepcidin concentration. CONCLUSIONS: MMF exposure could accelerate fracture healing via promote the endochondral ossification and bone formation while regulating systemic iron metabolism during fracture healing. This study suggests that MMF may have the potential to become a form of physical therapy for fractures.


Fracture Healing , Fractures, Bone , Male , Animals , Mice , Fracture Healing/physiology , X-Ray Microtomography , Mice, Inbred C57BL , Bony Callus/diagnostic imaging , Bony Callus/physiology , Magnetic Fields , Iron
4.
iScience ; 26(8): 107365, 2023 Aug 18.
Article En | MEDLINE | ID: mdl-37554458

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.

5.
FASEB J ; 37(7): e22985, 2023 07.
Article En | MEDLINE | ID: mdl-37249350

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.


Bone Resorption , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1 , Osteoporosis , Mice , Male , Animals , Diabetes Mellitus, Type 1/therapy , Diabetes Mellitus, Experimental/therapy , Mice, Inbred C57BL , Osteoblasts/metabolism , Osteogenesis , Iron/metabolism , Oxidation-Reduction , Magnetic Fields , Glucose
6.
Prog Biophys Mol Biol ; 177: 14-23, 2023 01.
Article En | MEDLINE | ID: mdl-36240898

With the widespread use of static magnetic fields (SMFs) in medicine, it is imperative to explore the biological effects of SMFs and the mechanisms underlying their effects on biological systems. The presence of magnetic materials within cells and organisms could affect various biological metabolism and processes, including stress responses, proliferation, and structural alignment. SMFs were generally found to be safe at the organ and organism levels. However. human subjects exposed to strong SMFs have reported side effects. In this review, we combined the magnetic properties of biological samples to illustrate the mechanism of action of SMFs on biological systems from a biophysical point of view. We suggest that the mechanisms of action of SMFs on biological systems mainly include the induction of electric fields and currents, generation of magnetic effects, and influence of electron spins. An electrolyte flowing in a static magnetic field generates an induced current and an electric field. Magnetomechanical effects include orientation effects upon subjecting biological samples to SMFs and movement of biological samples in strong field gradients. SMFs are thought to affect biochemical reaction rates and yields by influencing electron spin. This paper helps people how can harness the favorable biological effects of SMFs.


Magnetic Fields , Humans , Biophysics
7.
Exp Cell Res ; 417(2): 113223, 2022 08 15.
Article En | MEDLINE | ID: mdl-35643180

Many studies indicated that static magnetic fields (SMFs) have anti-cancer effects. However, effect of SMFs on cancer cells with strength exceeding 12 T are rarely reported. The intracellular iron could participate in the reactive oxygen species (ROS) production and affect cell proliferation. This study aimed to investigate the effect of 12 T high static magnetic field (HiSMF) on osteosarcoma cells and the relationship with intracellular iron. The 12 T HiSMF was generated by a superconducting magnet. The proliferation was evaluated by CCK-8 assays and cell counting. The apoptosis, cell cycle distribution, and ROS were evaluated by flow cytometry. Intracellular iron status was evaluated by atomic absorption spectroscopy and Calcein-AM/2,2'-bipyridyl. The expression of cell cycle and iron metabolism-related genes were analyzed by Western Blot. The result showed that 12 T HiSMF exposure suppressed the proliferation of osteosarcoma cell lines MNNG/HOS, U-2 OS, and MG63 via cell cycle arrest in S and G2/M. Meanwhile, 12 T HiSMF increasing intracellular ROS, and its antitumor effect was reduced by antioxidant. Furthermore, the intracellular total and free iron levels, the expression of FTH1 and DMT1 were increased by 12 HiSMF. The iron chelator (DFO) could reduce the cytotoxicity of 12 T HiSMF on osteosarcoma cells. Moreover, 12 T HiSMF could enhance the cytotoxicity of cisplatin and sorafenib in osteosarcoma cells. In Conclusion, 12 T HiSMF could suppress osteosarcoma cells proliferation via intracellular iron and ROS related cell cycle arrest, and have application potential in osteosarcoma therapy combined with sorafenib and cisplatin.


Bone Neoplasms , Osteosarcoma , Apoptosis , Bone Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Cisplatin/pharmacology , Humans , Iron , Magnetic Fields , Osteosarcoma/genetics , Reactive Oxygen Species/metabolism , Sorafenib/pharmacology
8.
Ecotoxicol Environ Saf ; 230: 113125, 2022 Jan 15.
Article En | MEDLINE | ID: mdl-34971997

OBJECTIVES: This study evaluated the associated biological effects of radio-frequency (RF) exposure at 16 T magnetic resonance imaging (MRI) on mice health. MATERIAL AND METHODS: A total of 48 healthy 8-week-old male C57BL/6 mice were investigated. A 16 T high static magnetic field (HiSMF) was generated by a superconducting magnet, and a radiofrequency (RF) electromagnetic field for hydrogen resonance at 16 T (700 MHz) was transmitted via a homemade RF system. The mice were exposed inside the 16 T HiSMF with the 700 MHz RF field for 60 min, and the body weight, organ coefficients, histomorphology of major organs, and blood indices were analyzed for the basal state of the mice on day 0 and day 14. The Heat Shock Protein 70 (HSP70), cyclooxygenase 2 (COX2), and interleukin- 6 (IL-6) were used to evaluate the thermal effects on the brain. Locomotor activity, the open field test, tail suspension test, forced swimming test, and grip strength test were used to assess the behavioral characteristics of the mice. RESULTS: The 16 T HiSMF with 700 MHz RF electromagnetic field exposure had no significant effects on body weight, organ coefficients, or histomorphology of major organs in the mice. On day 0, the expressions of HSP70 and COX2 in the brain were increased by 16 T HiSMF with 700 MHz RF electromagnetic field exposure. However, the expression of HSP70, COX2, and IL-6 had no significant difference compared with the sham group on day 14. Compared with the sham groups, the meancorpuscularvolume (MCV) on day 0 and the total protein (TP) on day 14 were increased significantly, whereas the other blood indices did not change significantly. The 16 T HiSMF with 700 MHz RF electromagnetic field exposure caused the mice to briefly circle tightly but had no effect on other behavioral indicators. CONCLUSIONS: In summary, 16 T HiSMF with 700 MHz RF electromagnetic field exposure for 60 min did not have severe effects on mice.

9.
Sci Rep ; 11(1): 14915, 2021 07 21.
Article En | MEDLINE | ID: mdl-34290353

Droplet-based transport driven by surface tension has been explored as an automated pumping source for several biomedical applications. This paper presented a simple and fast superhydrophobic modify and patterning approach to fabricate various open-surface platforms to manipulate droplets to achieve transport, mixing, concentration, and rebounding control. Several commercial reagents were tested in our approach, and the Glaco reagent was selected to create a superhydrophobic layer; laser cutters are utilized to scan on these superhydrophobic surface to create gradient hydrophilic micro-patterns. Implementing back-and-forth vibrations on the predetermined parallel patterns, droplets can be transported and mixed successfully. Colorimetry of horseradish peroxidase (HRP) mixing with substrates also reduced the reaction time by more than 5-times with the help of superhydrophobic patterned chips. Besides, patterned superhydrophobic chips can significantly improve the sensitivity of colorimetric glucose-sensing by more than 10 times. Moreover, all bioassays were distributed homogeneously within the region of hydrophilic micropatterns without the coffee-ring effect. In addition, to discuss further applications of the surface wettability, the way of controlling the droplet impacting and rebounding phenomenon was also demonstrated. This work reports a rapid approach to modify and patterning superhydrophobic films to perform droplet-based manipulations with a lower technical barrier, higher efficiency, and easier operation. It holds the potential to broaden the applications of open microfluidics in the future.

10.
Int J Mol Sci ; 22(13)2021 Jul 02.
Article En | MEDLINE | ID: mdl-34281233

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.


Deferasirox/therapeutic use , Deferoxamine/therapeutic use , MAP Kinase Signaling System/drug effects , Osteosarcoma/drug therapy , Siderophores/therapeutic use , Animals , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Deferasirox/pharmacology , Deferoxamine/pharmacology , Humans , Iron/metabolism , Mice , Osteosarcoma/metabolism , Siderophores/pharmacology , Xenograft Model Antitumor Assays
11.
Bioelectromagnetics ; 42(5): 371-383, 2021 Jul.
Article En | MEDLINE | ID: mdl-34082485

Static magnetic field (SMF) can alter cell fate decisions in many ways. However, the effects of SMF on cancer stem cells (CSCs) are little-known. In this particular study, we evaluate the biological effect of moderate-intensity SMF on osteosarcoma stem cells (OSCs) and try to clarify the underlying mechanisms of action. First, we demonstrated that prolonged exposure to SMF induced the proliferation and tumorsphere formation in K7M2 and MG63 OSCs. Moreover, SMF promoted the release of ferrous iron (Fe2+ ) and provoked reactive oxygen species (ROS) in OSCs. Interestingly, SMF evidently triggered the autophagic degradation of ferritin, which is characterized by the activation of microtubule-associated protein 1 light chain 3 (LC3) and nuclear receptor co-activator 4 (NCOA4), and downregulation of ferritin heavy chain 1 (FTH1) in OSCs. Particularly, the colony-forming ability of K7M2 OSCs promoted by SMF was obviously abolished by using a small interfering RNA (siRNA) against NCOA4. Finally, treatment of the tumor-bearing mice with SMF did not affect the tumor volume or tumor mass, nor pulmonary metastasis of K7M2 OSCs, but the SMF-treated K7M2 OSCs caused a preference of pulmonary metastasis in a mouse model, which suggested that SMF might induce the metastatic characteristic of OSCs. Consequently, this paper demonstrates for the first time that the cumulative SMF exposure promoted the self-renewal ability of OSCs via autophagic degradation of ferritin, implying that ferritinophagy may be a potential molecular target for cancer. © 2021 Bioelectromagnetics Society.


Bone Neoplasms , Osteosarcoma , Animals , Ferritins , Magnetic Fields , Mice , Stem Cells
12.
Bioelectromagnetics ; 42(3): 200-211, 2021 Apr.
Article En | MEDLINE | ID: mdl-33655538

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.


Osteoclasts , Osteogenesis , Animals , Cell Differentiation , Magnetic Fields , Mice , Osteoblasts
13.
Biol Trace Elem Res ; 199(9): 3416-3422, 2021 Sep.
Article En | MEDLINE | ID: mdl-33411150

Relative stability of mineral elements in tissues is necessary for health. High static magnetic fields (HiSMFs) have been widely used in biomedical research and industry. However, the bioeffect of HiSMFs on animals is still unclear. In this study, we investigated the effects of HiSMF exposure on the levels of Mg, Fe, Zn, Ca, and Cu in the main organs of mice. The 8-week male C57BL/6 mice were treated by 2-4 T, 6-8 T, 10-12 T HiSMFs for 28 days. The mass fractions of Mg, Fe, Zn, Ca, and Cu in the liver, brain, kidney, and heart in mice were respectively measured by atomic absorption spectroscopy, and used to evaluate mineral element content in tissues. The 2-4 T HiSMF exposure has increased the Mg, Fe, and Ca content in the kidney, as well as the Zn content in the brain. The 6-8 T HiSMF exposure has increased the Zn level in the liver; Mg, Fe, and Ca levels in the kidney; and Fe level in the heart, while the Zn in the kidney, and Zn and Ca in the heart was decreased by 6-8 T HiSMF exposure. For the 10-12 T HiSMF exposure, the Mg in the kidney, the Fe in the liver and kidney, and Cu in the brain have been increased significantly. However, the Zn in the kidney and the Ca in the brain and the heart were reduced by 10-12 T HiSMF exposure. The HiSMF exposure for 28 days can alter the Mg, Fe, Zn, Ca, and Cu content in mice, and change with the different magnetic flux density of HiSMFs (2-4 T, 6-8 T, 10-12 T), elements, and organ types.


Minerals , Trace Elements , Animals , Copper , Kidney , Liver , Magnetic Fields , Male , Mice , Mice, Inbred C57BL
14.
Free Radic Res ; 54(6): 385-396, 2020 Jun.
Article En | MEDLINE | ID: mdl-32183598

Vitamin C and iron are both important nutrients for humans and involved in several physiological processes. The biological activities of vitamin C and iron are based on their abilities to accept or donate electrons. Although vitamin C is well known as an excellent electron donor in physiological conditions, it also has pro-oxidant properties, especially with catalytic metal iron. Cancer cells have a higher iron requirement than normal cells, which allows pharmacological ascorbate to kill cancer cells selectively. In this study, we demonstrated that the levels of H2O2 in cells were significantly raised after treated with pharmacological ascorbate, and intracellular labile iron could increase pharmacological ascorbate-mediated oxidative stress by Fenton reaction. Catalytic metal iron plays opposite roles in and outside cells. Intracellular excess labile iron improved ascorbate-induced toxicity, while the excess labile iron in the medium abolished ascorbate-induced toxicity. Fe3+ and Fe2+ have the same effect on ascorbate-induced toxicity, but Fe3+ chelator deferoxamine (DFO) has a profound inhibition effect than Fe2+ chelator 2,2'-bipyridyl (BIP) on ascorbate-induced toxicity. The influence of intracellular labile iron and ascorbate on the ferritin expression may cause selective sensitivity in osteosarcoma cell lines on pharmacological ascorbate. High iron requirement of many cancer cells facilitates pharmacological ascorbate on cancer treatment. In addition, increasing iron content in tumour tissue may be effective strategies to improve the effects of pharmacological ascorbate.


Ascorbic Acid/toxicity , Bone Neoplasms/drug therapy , Iron/therapeutic use , Osteosarcoma/drug therapy , Oxidative Stress/drug effects , Bone Neoplasms/pathology , Cell Line, Tumor , Humans , Iron/metabolism , Osteosarcoma/pathology
15.
Biochim Biophys Acta Gen Subj ; 1864(4): 129539, 2020 04.
Article En | MEDLINE | ID: mdl-31958545

BACKGROUND: Cancer is one of the major threats to human health and current cancer therapies have been unsuccessful in eradicating it. Ferroptosis is characterized by iron-dependence and lipid hydroperoxides accumulation, and its primary mechanism involves the suppression of system Xc--GSH (glutathione)-GPX4 (glutathione peroxidase 4) axis. Co-incidentally, cancer cells are also metabolically characterized by iron addiction and ROS tolerance, which makes them vulnerable to ferroptosis. This may provide a new tactic for cancer therapy. SCOPE OF REVIEW: The general features and mechanisms of ferroptosis, and the basis that makes cancer cells vulnerable to ferroptosis are described. Further, we emphatically discussed that disrupting GSH may not be ideal for triggering ferroptosis of cancer cells in vivo, but directly inhibiting GPX4 and its compensatory members could be more effective. Finally, the various approaches to directly inhibit GPX4 without disturbing GSH were described. MAJOR CONCLUSIONS: Targeting system Xc- or GSH may not effectively trigger cancer cells' ferroptosis in vivo the existence of other compensatory pathways. However, directly targeting GPX4 and its compensatory members without disrupting GSH may be more effective to induce ferroptosis in cancer cells in vivo, as GPX4 is essential in preventing ferroptosis. GENERAL SIGNIFICANCE: Cancer is a severe threat to human health. Ferroptosis-based cancer therapy strategies are promising, but how to effectively induce ferroptosis in cancer cells in vivo is still a question without clear answers. Thus, the viewpoints raised in this review may provide some references and different perspectives for researchers working on ferroptosis-based cancer therapy.


Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Ferroptosis/drug effects , Glutathione/antagonists & inhibitors , Neoplasms/drug therapy , Phospholipid Hydroperoxide Glutathione Peroxidase/antagonists & inhibitors , Animals , Drug Screening Assays, Antitumor , Glutathione/metabolism , Humans , Neoplasms/metabolism , Neoplasms/pathology , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Reactive Oxygen Species/metabolism
16.
J Exp Clin Cancer Res ; 38(1): 406, 2019 Sep 13.
Article En | MEDLINE | ID: mdl-31519186

Iron, an indispensable element for life, is involved in all kinds of important physiological activities. Iron promotes cell growth and proliferation, but it also causes oxidative stress damage. The body has a strict regulation mechanism of iron metabolism due to its potential toxicity. As a cancer of the bone marrow and blood cells, leukemia threatens human health seriously. Current studies suggest that dysregulation of iron metabolism and subsequent accumulation of excess iron are closely associated with the occurrence and progress of leukemia. Specifically, excess iron promotes the development of leukemia due to the pro-oxidative nature of iron and its damaging effects on DNA. On the other hand, leukemia cells acquire large amounts of iron to maintain rapid growth and proliferation. Therefore, targeting iron metabolism may provide new insights for approaches to the treatment of leukemia. This review summarizes physiologic iron metabolism, alternations of iron metabolism in leukemia and therapeutic opportunities of targeting the altered iron metabolism in leukemia, with a focus on acute leukemia.


Iron/metabolism , Leukemia/etiology , Leukemia/metabolism , Animals , Disease Management , Disease Susceptibility , Drug Carriers , Drug Delivery Systems , Humans , Iron/chemistry , Leukemia/therapy , Metabolic Networks and Pathways/drug effects , Metal Nanoparticles/chemistry , Molecular Targeted Therapy , Oxidation-Reduction/drug effects , Oxidative Stress
17.
Eur Radiol ; 29(11): 6029-6037, 2019 Nov.
Article En | MEDLINE | ID: mdl-31115627

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.


Magnetic Fields , Magnetic Resonance Imaging/methods , Radiation Exposure , Animals , Body Weight , Male , Mice , Mice, Inbred C57BL , Models, Animal
18.
J Cell Physiol ; 234(6): 8028-8039, 2019 06.
Article En | MEDLINE | ID: mdl-30362549

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.


Iron/metabolism , Macrophages/metabolism , Mitochondria/metabolism , Neoplasms/immunology , Cell Cycle/genetics , Cell Proliferation/genetics , Cell Survival/genetics , Cellular Reprogramming/immunology , Humans , Immunotherapy , Macrophages/immunology , Macrophages/pathology , Mitochondria/immunology , Mitochondria/pathology , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/therapy , Tumor Microenvironment/genetics
19.
Am J Cancer Res ; 8(10): 1933-1946, 2018.
Article En | MEDLINE | ID: mdl-30416846

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.

20.
J Cell Biochem ; 119(11): 9178-9204, 2018 11.
Article En | MEDLINE | ID: mdl-30076742

Iron metabolism is crucial to hepatocellular carcinoma progression and is a key determinant of prognosis. Protein-protein interactions within the iron metabolism gene network were analyzed using the European Molecular Biology Laboratory's Search Tool for Recurring Instances of Neighbouring Genes/Proteins database. We obtained 423 liver hepatocellular carcinoma gene expression profiles from the Cancer Genome Atlas database. The expression and pathway enrichment of representative iron intake genes (TFRC and DMT1), utilization genes (FTH1, FTL, HIF1A, HMOX1, SLC25A37, and SLC25A38), and efflux genes (FLVCR1 and SLC40A1) was investigated in tumor and adjacent tissues. We determined the relationship between iron metabolism and the prognostic features of liver hepatocellular carcinoma. The liver metabolism genes TFRC and FLVCR1 were related to survival, disease status, and prognosis in patients with hepatocellular carcinoma. Our results provide novel insight into liver cancer therapy.


Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Iron/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/physiology , Gene Regulatory Networks/genetics , Gene Regulatory Networks/physiology , Humans , Male , Prognosis , Transcriptome
...