Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Environ Pollut ; 335: 122296, 2023 Oct 15.
Article in English | MEDLINE | ID: mdl-37536476

ABSTRACT

Uranium is a contaminate in the underground water in many regions of the world, which poses health risks to the local populations through drinking water. Although the health hazards of natural uranium have been concerned for decades, the controversies about its detrimental effects continue at present since it is still unclear how uranium interacts with molecular regulatory networks to generate toxicity. Here, we integrate transcriptomic and metabolomic methods to unveil the molecular mechanism of lipid metabolism disorder induced by uranium. Following exposure to uranium in drinking water for twenty-eight days, aberrant lipid metabolism and lipogenesis were found in the liver, accompanied with aggravated lipid peroxidation and an increase in dead cells. Multi-omics analysis reveals that uranium can promote the biosynthesis of unsaturated fatty acids through dysregulating the metabolism of arachidonic acid (AA), linoleic acid, and glycerophospholipid. Most notably, the disordered metabolism of polyunsaturated fatty acids (PUFAs) like AA may contribute to lipid peroxidation induced by uranium, which in turn triggers ferroptosis in hepatocytes. Our findings highlight disorder of lipid metabolism as an essential toxicological mechanism of uranium in the liver, offering insight into the health risks of uranium in drinking water.


Subject(s)
Drinking Water , Uranium , Mice , Animals , Uranium/toxicity , Uranium/metabolism , Transcriptome , Liver/metabolism , Fatty Acids, Unsaturated/metabolism , Metabolomics
2.
Metallomics ; 15(3)2023 03 06.
Article in English | MEDLINE | ID: mdl-36869799

ABSTRACT

Understanding of how mercury species cause cellular impairments at the molecular level is critical for explaining the detrimental effects of mercury exposure on the human body. Previous studies have reported that inorganic and organic mercury compounds can induce apoptosis and necrosis in a variety of cell types, but more recent advances reveal that mercuric mercury (Hg2+) and methylmercury (CH3Hg+) may result in ferroptosis, a distinct form of programmed cell death. However, it is still unclear which protein targets are responsible for ferroptosis induced by Hg2+ and CH3Hg+. In this study, human embryonic kidney 293T cells were used to investigate how Hg2+ and CH3Hg+ trigger ferroptosis, given their nephrotoxicity. Our results demonstrate that glutathione peroxidase 4 (GPx4) plays a key role in lipid peroxidation and ferroptosis in renal cells induced by Hg2+ and CH3Hg+. The expression of GPx4, the only lipid repair enzyme in mammal cells, was downregulated in response to Hg2+ and CH3Hg+ stress. More importantly, the activity of GPx4 could be markedly inhibited by CH3Hg+, owing to the direct binding of the selenol group (-SeH) in GPx4 to CH3Hg+. Selenite supplementation was demonstrated to enhance the expression and activity of GPx4 in renal cells, and consequently relieve the cytotoxicity of CH3Hg+, suggesting that GPx4 is a crucial modulator implicated in the Hg-Se antagonism. These findings highlight the importance of GPx4 in mercury-induced ferroptosis, and provide an alternative explanation for how Hg2+ and CH3Hg+ induce cell death.


Subject(s)
Ferroptosis , Mercury , Selenium , Animals , Humans , Mercury/toxicity , Mercury/metabolism , Selenium/pharmacology , Phospholipid Hydroperoxide Glutathione Peroxidase , Kidney/metabolism , Glutathione Peroxidase/metabolism , Mammals/metabolism
3.
Biol Trace Elem Res ; 201(2): 627-635, 2023 Feb.
Article in English | MEDLINE | ID: mdl-35305538

ABSTRACT

The relationship between trace elements and neurological development is an emerging research focus. We performed a case-control study to explore (1) the differences of 13 trace elements chromium (Cr), manganese (Mn), cobalt (Co), zinc (Zn), arsenic (As), selenium (Se), molybdenum (Mo), cadmium (Cd), stannum (Sn), stibium (Sb), mercury (Hg), titanium (TI), and plumbum (Pb) concentration in whole blood and urine between autism spectrum disorder (ASD) children and their typical development peers, and (2) the association between the 13 trace elements and core behaviors of ASD. Thirty ASD subjects (cases) and 30 age-sex-matched healthy subjects from Baise City, Guangxi Zhuang Autonomous Region, China, were recruited. Element analysis was carried out by inductively coupled plasma-optical emission spectrometry. Autistic behaviors were assessed using Autism Behavior Checklist (ABC), Childhood Autism Rating Scale (CARS), and Children Neuropsychological and Behavior Scale (CNBS). The whole blood concentrations of Mo (p = 0.004), Cd (0.007), Sn (p = 0.003), and Pb (p = 0.037) were significantly higher in the ASD cases than in the controls. Moreover, Se (0.393), Hg (0.408), and Mn (- 0.373) concentrations were significantly correlated between whole blood and urine levels in ASD case subjects. There were significant correlations between whole blood Sb (0.406), Tl (0.365), Mo (- 0.4237), Mn (- 0.389), Zn (0.476), and Se (0.375) levels and core behaviors of ASD. Although the mechanism of trace element imbalance in ASD is unclear, these data demonstrate that core behaviors of ASD may be affected by certain trace elements. Further studies are recommended for exploring the mechanism of element imbalance and providing corresponding clinical treatment measures.


Subject(s)
Autism Spectrum Disorder , Autistic Disorder , Mercury , Selenium , Trace Elements , Humans , Child , Trace Elements/analysis , Cadmium/analysis , Case-Control Studies , Lead/analysis , China , Selenium/analysis , Manganese/analysis , Molybdenum/analysis , Tin/analysis , Mercury/analysis
4.
Adv Sci (Weinh) ; 9(29): e2202679, 2022 10.
Article in English | MEDLINE | ID: mdl-36031399

ABSTRACT

Excess iron accumulation occurs in organs of patients with certain genetic disorders or after repeated transfusions. No physiological mechanism is available to excrete excess iron and iron overload to promote lipid peroxidation to induce ferroptosis, thus iron chelation becomes critical for preventing ion toxicity in these patients. To date, several iron chelators have been approved for iron chelation therapy, such as deferiprone and deferoxamine, but the current iron chelators suffer from significant limitations. In this context, new agents are continuously sought. Here, a library of new deferric amine compounds (DFAs) with adjustable skeleton and flexibility is synthesized by adopting the beneficial properties of conventional chelators. After careful evaluations, compound DFA1 is found to have greater efficacy in binding iron through two molecular oxygens in the phenolic hydroxyl group and the nitrogen atom in the amine with a 2:1 stoichiometry. This compound remarkably ameliorates iron overload in diverse murine models through both oral and intravenous administration, including hemochromatosis, high iron diet-induced, and iron dextran-stimulated iron accumulation. Strikingly, this compound is found to suppress iron-induced ferroptosis by modulating the intracellular signaling that drives lipid peroxidation. This study opens a new approach for the development of iron chelators to treat iron overload.


Subject(s)
Ferroptosis , Hemochromatosis , Iron Overload , Amines , Animals , Deferiprone , Deferoxamine/pharmacology , Deferoxamine/therapeutic use , Dextrans , Humans , Iron/metabolism , Iron Chelating Agents/pharmacology , Iron Chelating Agents/therapeutic use , Iron Overload/drug therapy , Mice , Nitrogen , Pyridones/pharmacology , Pyridones/therapeutic use
5.
J Nutr Biochem ; 107: 109034, 2022 09.
Article in English | MEDLINE | ID: mdl-35500829

ABSTRACT

Autism spectrum disorder (ASD) refers to a group of neurodevelopmental disorders. The etiology and pathological mechanisms of ASD are still unknown, and its prognosis is poor. This study investigated the effects of selenium (Se) supplementation on abnormal behavior and cognitive function in ASD model mice, as well as the potential action pathways. BTBR mice were randomly assigned to either a model group (BTBR group), a model selenium supplement group (BTBR+Se group), a normal control group (B6 group) or a normal selenium supplement group (B6+Se group). Sodium selenite, at a dosage of 1 mg/kg/day, was administered to the selenium supplementation groups by gavage. The mice in the BTBR group and the B6 group received the same amount of 0.9% saline by gavage. After 4 weeks of continuous intervention, the social functions and cognitive behaviors of the mice and the selenium concentration in hippocampal tissue were assessed. Hippocampal tissue structures were observed. Changes in neurotransmitter levels, oxidative stress and neuroinflammatory indicators were detected. SelP protein expression was significantly lower in hippocampal tissue from BTBR mice than in hippocampal tissue from B6 mice. The administration of sodium selenite in BTBR mice: (1) increased the expression of SelP; (2) attenuated spatial learning, memory impairment and improved social behaviors; (3) changed the serum levels of 5-HT, DA and Glu; (4) decreased the levels of inflammatory cytokines IL-6, IL-1ß, and TNF-α in serum and hippocampal tissue; (5) reduced the ROS and MDA contents and significantly increased SOD activity, CAT activity, GSH-px activity, and antioxidant GSH levels; and (6) protected against neuronal loss in the hippocampus. Se supplementation significantly improved the social functioning, repetitive stereotyped behavior and cognitive function in BTBR mice. Se may play a protective role in the hippocampus of BTBR mice by regulating neurotransmitter levels, reducing oxidative stress, alleviating neuroinflammation and rescuing neural cell damage.


Subject(s)
Autism Spectrum Disorder , Autistic Disorder , Selenium , Animals , Autism Spectrum Disorder/drug therapy , Autistic Disorder/drug therapy , Autistic Disorder/etiology , Dietary Supplements , Disease Models, Animal , Gene Expression , Inflammation/metabolism , Mice , Mice, Inbred Strains , Oxidative Stress , Selenium/pharmacology , Social Behavior , Sodium Selenite
6.
Brain Res Bull ; 183: 38-48, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35227767

ABSTRACT

BACKGROUND: Autism spectrum disorder (ASD) is a group of extensive neurodevelopmental disorders for which few efficacious drugs are available. Sodium selenite (Se), the most common inorganic form of selenium given to humans and animals, has antioxidant, anti-inflammatory, and neuroprotective effects in several psychiatric and neurological disorders. However, the effect of Se on ASD is unclear. METHODS: Using the BTBR T + tf/J (BTBR) mouse model of ASD, we investigated the therapeutic effects and underlying mechanism of action of Se on ASD. BTBR mice were randomly divided into four groups: BTBR, BTBR+Se, BTBR+Se+ML385, and BTBR+Se+RSL3. The normal control group was composed of C57BL/6 (B6) mice. Se, Nuclear factor erythroid 2-related factor 2 (Nrf2), and glutathione peroxidase 4 (GPx4) inhibitors were administered separately for 28 days using oral gavage. After 28 days, social behavior, ferroptosis indices, and gene and protein expression levels for components of the Nrf2/GPx4 pathway were assessed to explore the correlation between Se levels and ASD. RESULTS: We demonstrated that Se significantly mitigated impairments in learning and memory, improved social functions, reduced repetitive behaviors, and inhibited ferroptosis in the CA1 area of the hippocampus. We also found that the Nrf2/GPX4 pathway was a target for Se. Treatment with Se increased levels of Nrf2 and GPX4. The Nrf2 inhibitor ML385 reduced the effect of Se on ferroptosis and abnormal behaviors in BTBR mice. In addition, the GPx4 inhibitor RSL3 revealed similar efficacy to ML385 CONCLUSION: We determined that Se exhibited a beneficial effect on autism-relevant behaviors and inhibited ferroptosis in the BTBR mouse model of ASD, possibly through modulation of the Nrf2/GPX4 signaling pathway.


Subject(s)
Autism Spectrum Disorder , Autistic Disorder , Ferroptosis , Selenium , Animals , Autism Spectrum Disorder/drug therapy , Autistic Disorder/drug therapy , Disease Models, Animal , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase , Selenium/pharmacology , Selenium/therapeutic use
7.
Environ Int ; 145: 106107, 2020 12.
Article in English | MEDLINE | ID: mdl-32932066

ABSTRACT

Uranium contamination is a global health concern. Regarding natural or anthropogenic uranium contamination, the major sources of concern are groundwater, mining, phosphate fertilizers, nuclear facilities, and military activities. Many epidemiological and laboratory studies have demonstrated that environmental and occupational uranium exposure can induce multifarious health problems. Uranium exposure may cause health risks because of its chemotoxicity and radiotoxicity in natural or anthropogenic scenarios: the former is generally thought to play a more significant role with regard to the natural uranium exposure, and the latter is more relevant to enriched uranium exposure. The understanding of the health risks and underlying toxicological mechanisms of uranium remains at a preliminary stage, and many controversial findings require further research. In order to present state-of-the-art status in this field, this review will primarily focus on the chemotoxicity of uranium, rather than its radiotoxicity, as well as the involved toxicological mechanisms. First, the natural or anthropogenic uranium contamination scenarios will be briefly summarized. Second, the health risks upon natural uranium exposure, for example, nephrotoxicity, bone toxicity, reproductive toxicity, hepatotoxicity, neurotoxicity, and pulmonary toxicity, will be discussed based on the reported epidemiological cases and laboratory studies. Third, the recent advances regarding the toxicological mechanisms of uranium-induced chemotoxicity will be highlighted, including oxidative stress, genetic damage, protein impairment, inflammation, and metabolic disorder. Finally, the gaps and challenges in the knowledge of uranium-induced chemotoxicity and underlying mechanisms will be discussed.


Subject(s)
Groundwater , Occupational Exposure , Uranium , Fertilizers , Mining , Uranium/analysis , Uranium/toxicity
8.
Theranostics ; 10(14): 6384-6398, 2020.
Article in English | MEDLINE | ID: mdl-32483459

ABSTRACT

To circumvent the huge cost, long R&D time and the difficulty to identify the targets of new drugs, repurposing the ones that have been clinically approved has been considered as a viable strategy to treat different diseases. In the current study, we outlined the rationale for repurposing disulfiram (DSF, an old alcohol-aversion drug) to treat primary breast cancer and its metastases. Methods: To overcome a few shortcomings of the individual administration of DSF, such as the dependence on copper ions (Cu2+) and limited capability in selective targeting, we here artificially synthesized the active form of DSF, diethyldithiocarbamate (DTC)-Cu complex (CuET) for cancer therapeutics. To achieve a greater efficacy in vivo, smart nanomedicines were devised through a one-step self-assembly of three functional components including a chemically stable and biocompatible phase-change material (PCM), the robust anticancer drug (CuET) and a near-infrared (NIR) dye (DIR), namely CuET/DIR NPs. A number of in vitro assays were performed including the photothermal efficacy, light-triggered drug release behavior, nuclear localization, DNA damage and induction of apoptosis of CuET/DIR NPs and molecular mechanisms underlying CuET-induced repression on cancer metastatic behaviors. Meanwhile, the mice bearing 4T1-LG12-drived orthotopic tumors were employed to evaluate in vivo biodistribution and anti-tumor effect of CuET/DIR NPs. The intravenous injection model was employed to reflect the changes of the intrinsic metastatic propensity of 4T1-LG12 cells responding to CuET/DIR NPs. Results: The rationally designed nanomedicines have self-traceability for bioimaging, long blood circulation time for enhanced drug accumulation in the tumor site and photo-responsive release of the anticancer drugs. Moreover, our data unearthed that CuET/DIR nanomedicines behave like "Trojan horse" to transport CuET into the cytoplasm, realizing substantial intracellular accumulation. Upon NIR laser irradiation, massive CuET would be triggered to release from the nanomedicines and reach a high local concentration towards the nucleus, where the pro-apoptotic effects were conducted. Importantly, our CuET/DIR nanomedicines revealed a pronounced capability to leash breast cancer metastases through inhibition on EMT. Additionally, these nanomedicines showed great biocompatibility in animals. Conclusion: These combined data unearthed a remarkably enhanced tumor-killing efficacy of our CuET nanomedicines through nuclear targeting. This work may open a new research area of repurposing DSF as innovative therapeutic agents to treat breast cancer and its metastases.


Subject(s)
Antineoplastic Agents/pharmacology , Copper , Disulfiram , Ditiocarb , Nanoparticles , Animals , Antineoplastic Agents/chemistry , Breast Neoplasms/drug therapy , Cell Line, Tumor/drug effects , Cell Nucleus , Copper/chemistry , Copper/pharmacology , Disulfiram/chemistry , Disulfiram/pharmacology , Ditiocarb/chemistry , Ditiocarb/pharmacology , Drug Delivery Systems , Drug Liberation , Drug Repositioning , Female , Humans , Low-Level Light Therapy , Mice , Nanomedicine , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Neoplasm Metastasis/drug therapy , Neoplasms/drug therapy , Theranostic Nanomedicine/methods
9.
Curr Clin Microbiol Rep ; 7(2): 13-19, 2020.
Article in English | MEDLINE | ID: mdl-32318324

ABSTRACT

PURPOSE OF THE REVIEW: The ongoing outbreak of novel coronavirus pneumonia (COVID-19) caused by the 2019 novel coronavirus (SARS-CoV-2) in China is lifting widespread concerns. Thus, therapeutic options are urgently needed, and will be discussed in this review. RECENT FINDINGS: Iron-containing enzymes are required for viruses most likely including coronaviruses (CoVs) to complete their replication process. Moreover, poor prognosis occurred in the conditions of iron overload for patients upon infections of viruses. Thus, limiting iron represents a promising adjuvant strategy in treating viral infection through oral uptake or venous injection of iron chelators, or through the manipulation of the key iron regulators. For example, treatment with iron chelator deferiprone has been shown to prolong the survival of acquired immunodeficiency syndrome (AIDS) patients. Increasing intracellular iron efflux via increasing iron exporter ferroportin expression also exhibits antiviral effect on human immunodeficiency virus (HIV). The implications of other metals besides iron are also briefly discussed. SUMMARY: For even though we know little about iron regulation in COVID-19 patients thus far, it could be deduced from other viral infections that iron chelation might be an alternative beneficial adjuvant in treating COVID-19.

10.
Oxid Med Cell Longev ; 2019: 5738368, 2019.
Article in English | MEDLINE | ID: mdl-31485296

ABSTRACT

Diabetic foot ulcers (DFUs), the most serious complication of diabetes mellitus, can induce high morbidity, the need to amputate lower extremities, and even death. Although many adjunctive strategies have been applied for the treatment of DFUs, the low treatment efficiency, potential side effects, and high cost are still huge challenges. Recently, nanomaterial-based drug delivery systems (NDDSs) have achieved targeted drug delivery and controlled drug release, offering great promises in various therapeutics for diverse disorders. Additionally, the radial extracorporeal shock wave (rESW) has been shown to function as a robust trigger source for the NDDS to release its contents, as the rESW harbors a potent capability in generating pressure waves and in creating the cavitation effect. Here, we explored the performance of oxygen-loaded nanoperfluorocarbon (Nano-PFC) combined with the rESW as a treatment for DFUs. Prior to in vivo assessment, we first demonstrated the high oxygen affinity in vitro and great biocompatibility of Nano-PFC. Moreover, the rESW-responsive oxygen release behavior from oxygen-saturated Nano-PFC was also successfully verified in vitro and in vivo. Importantly, the wound healing of DFUs was significantly accelerated due to improved blood microcirculation, which was a result of rESW therapy (rESWT), and the targeted release of oxygen into the wound from oxygen-loaded Nano-PFC, which was triggered by the rESW. Collectively, the oxygen-saturated Nano-PFC and rESW provide a completely new approach to treat DFUs, and this study highlights the advantages of combining nanotechnology with rESW in therapeutics.


Subject(s)
Diabetes Complications/therapy , Diabetic Foot/therapy , Extracorporeal Shockwave Therapy/methods , Hyperbaric Oxygenation/methods , Oxygen/therapeutic use , Wound Healing/physiology , Animals , Humans , Oxygen/administration & dosage , Oxygen/pharmacology , Rats
11.
Nano Lett ; 19(8): 5587-5594, 2019 08 14.
Article in English | MEDLINE | ID: mdl-31260628

ABSTRACT

In breast cancer chemophotothermal therapy, it is a great challenge for the development of multifunctional nanoagents for precision targeting and the effective treatment of tumors, especially for metastasis. Herein, we successfully design and synthesize a multifunctional black phosphorus (BP)-based nanoagent, BP/DTX@PLGA, to address this challenge. In this composite nanoagent, BP quantum dots (BPQDs) are loaded into poly(lactic-co-glycolic acid) (PLGA) with additional conjugation of a chemotherapeutic agent, docetaxel (DTX). The in vivo distribution results demonstrate that BP/DTX@PLGA shows striking tropism for targeting both primary tumors and lung metastatic tumors. Moreover, BP/DTX@PLGA exhibits outstanding controllable chemophotothermal combinatory therapeutics, which dramatically improves the efficacy of photothermal tumor ablation when combined with near-light irradiation. Mechanistically, accelerated DTX release from the nanocomplex upon heating and thermal treatment per se synergistically incurs apoptosis-dependent cell death, resulting in the elimination of lung metastasis. Meanwhile, in vitro and in vivo results further confirm that BP/DTX@PLGA possesses good biocompatibility. This study provides a promising BP-based multimodal nanoagent to constrain cancer metastasis.


Subject(s)
Antineoplastic Agents/therapeutic use , Docetaxel/therapeutic use , Mammary Neoplasms, Animal/therapy , Nanoconjugates/therapeutic use , Phosphorus/therapeutic use , Animals , Antineoplastic Agents/pharmacokinetics , Docetaxel/pharmacokinetics , Female , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Mammary Neoplasms, Animal/pathology , Mice , Neoplasm Metastasis/pathology , Neoplasm Metastasis/therapy , Phosphorus/pharmacokinetics , Polylactic Acid-Polyglycolic Acid Copolymer/pharmacokinetics , Polylactic Acid-Polyglycolic Acid Copolymer/therapeutic use
12.
Angew Chem Int Ed Engl ; 56(46): 14488-14493, 2017 11 13.
Article in English | MEDLINE | ID: mdl-28892587

ABSTRACT

Black phosphorus nanosheets (BPs) show great potential for various applications including biomedicine, thus their potential side effects and corresponding improvement strategy deserve investigation. Here, in vitro and in vivo biological effects of BPs with and without titanium sulfonate ligand (TiL4 ) modification are investigated. Compared to bare BPs, BPs with TiL4 modification (TiL4 @BPs) can efficiently escape from macrophages uptake, and reduce cytotoxicity and proinflammation. The corresponding mechanisms are also discussed. These findings may not only guide the applications of BPs, but also propose an efficient strategy to further improve the biocompatibility of BPs.


Subject(s)
Biocompatible Materials/metabolism , Nanostructures/chemistry , Phosphorus/metabolism , Animals , Cell Line , Ligands , Macrophages/metabolism , Mice , Microscopy, Atomic Force , Microscopy, Electron, Transmission , Phosphorus/chemistry , Photoelectron Spectroscopy , Spectrum Analysis, Raman , Sulfonic Acids/chemistry , Sulfonic Acids/metabolism , Titanium/chemistry , Titanium/metabolism
13.
Adv Mater ; 28(38): 8379-8387, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27461987

ABSTRACT

Mesoporous carbon nanospheres containing porphyrin-like metal centers (denoted as "PMCS") are successfully synthesized by the pyrolysis of an imidazolate framework using a mesoporous-silica protection strategy. The PMCS allow infrared and photoacoustic imaging and synergetic photothermal therapy/photodynamic therapy derived from the porphyrin-like moieties, offering the possibility of real-time monitoring of therapeutic processes and image-guided precise conformal phototherapy. PMCS thus represent a novel multifunctional theranostic platform for improved treatment efficiencies.


Subject(s)
Nanospheres , Carbon , Metals , Phototherapy , Porphyrins
14.
Int J Mol Med ; 37(5): 1379-88, 2016 May.
Article in English | MEDLINE | ID: mdl-27035325

ABSTRACT

Systemic iron homeostasis is strictly controlled under normal conditions to ensure a balance between the absorption, utilization, storage and recycling of iron. The hepcidin-ferroportin (FPN) axis is of critical importance in the maintenance of iron homeostasis. Hepcidin deficiency gives rise to enhanced dietary iron absorption, as well as to increased iron release from macrophages, and this in turn results in iron accumulation in the plasma and organs, and is associated with a range of tissue pathologies. Low hepcidin levels have been demonstrated in most forms of hereditary hemochromatosis (HH), as well as in ß-thalassemia. Therapies that increase hepcidin concentrations may potentially play a role in the treatment of these iron overload-related diseases. To date, natural compounds have not been extensively investigated for this purpose, to the best of our knowledge. Thus, in the present study, we screened natural compounds that have the potential to regulate hepcidin expression. By performing hepcidin promoter-luciferase assay, RT-qPCR and animal experiments, we demonstrated that icariin and berberine were potent stimulators of hepcidin transcription. Mechanistic experiments indicated that icariin and berberine increased hepcidin expression by activating the signal transducer and activator of transcription 3 (Stat3) and Smad1/5/8 signaling pathways. The induction of hepcidin was confirmed in mice following icariin administration, coupled with associated changes in serum and tissue iron concentrations. In support of these findings, the icariin analogues, epimedin A, B and C, also increased hepatic hepcidin expression. However, these changes were not observed in hepcidin-deficient [Hamp1-/- or Hamp1­knockout (KO)] mice following icariin administration, thereby verifying hepatic hepcidin as the target of icariin. Although berberine exhibited a robust capacity to promote hepcidin expression in vitro, it failed to alter hepcidin expression in mice. Taken together, the findings of the present study suggest that icariin exhibits a robust capacity to increase hepatic hepcidin expression and to modulate systemic iron homeostasis. The present study therefore highlights the significance of using natural compounds to ameliorate iron disorders through the regulation of hepcidin expression.


Subject(s)
Flavonoids/pharmacology , Gene Expression Regulation/drug effects , Hepcidins/genetics , Iron/metabolism , Liver/drug effects , Liver/metabolism , STAT3 Transcription Factor/metabolism , Smad Proteins/metabolism , Animals , Berberine/chemistry , Berberine/pharmacology , Cell Line, Tumor , Drug Discovery , Drug Evaluation, Preclinical , Flavonoids/chemistry , Hepcidins/deficiency , Hepcidins/metabolism , Humans , Mice , Mice, Knockout , Reproducibility of Results , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL