Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
Gynecol Oncol ; 166(1): 126-137, 2022 07.
Article in English | MEDLINE | ID: mdl-35688655

ABSTRACT

OBJECTIVE: Iron depletion may be a novel therapeutic strategy for cancer. This study aimed to assess the inhibition effects of deferasirox (DFX), an oral iron chelator, on cervical cancer. METHODS: In this study, we performed immunohistochemical analysis, enzyme-linked immunoassay, cell viability and invasive ability assay, cell cycle and apoptosis analysis, protein expression investigation, molecular mechanism investigation, and in vivo murine xenograft model to evaluate the impact of DFX on cervical cancer. RESULTS: The cervical cancer cell lines viability decreased and cell apoptosis was induced after DFX incubation. Additionally, DFX promoted cell cycle arrest by regulating the expression of cell cycle regulators cyclin D1, cyclin E and proliferating cell nuclear antigen (PCNA) in cervical cancer cell lines. DFX also decreased cell invasion by upregulating the expression of NDRG1 and downregulating c-Myc. The activation of Akt and the MEK/ERK signaling pathway was inhibited by DFX. DFX also significantly suppressed xenograft tumor growth, decreased the levels of ferritin in serum and tumor tissue, reduced iron deposits and reactive oxygen species (ROS) levels in xenografts of DFX-treated group compared with the control group, with no serious side effects. CONCLUSION: Present study demonstrated the inhibitory effect of DFX against cervical cancer, and provided a potential therapeutic agent for cervical cancer.


Subject(s)
Iron Chelating Agents , Uterine Cervical Neoplasms , Animals , Benzoates/pharmacology , Benzoates/therapeutic use , Deferasirox/pharmacology , Female , Humans , Iron , Iron Chelating Agents/pharmacology , Iron Chelating Agents/therapeutic use , Mice , Triazoles/pharmacology , Triazoles/therapeutic use , Uterine Cervical Neoplasms/drug therapy
2.
Inflammation ; 45(3): 1089-1100, 2022 Jun.
Article in English | MEDLINE | ID: mdl-34837126

ABSTRACT

Ferritin, which is composed of a heavy chain and a light chain, plays a critical role in maintaining iron homeostasis by sequestering iron. The ferritin light chain (FTL) is responsible for the stability of the ferritin complex. We have previously shown that overexpression of FTL decreases the levels of the labile iron pool (LIP) and reactive oxygen species (ROS) in lipopolysaccharide (LPS)-treated murine macrophage cells. The protein level of FTL was downregulated by LPS within a short treatment period. However, the mechanism underlying the LPS-induced changes in the FTL levels is not known. In the present study, we report that LPS induces the ubiquitin-proteasome-dependent degradation of FTL and that the mechanism of LPS-induced FTL degradation involves the JNK/Itch axis. We found that LPS downregulates the protein and mRNA levels of FTL in a time-dependent manner. The proteasome inhibitor MG-132 significantly reverses the LPS-induced decrease in FTL. Furthermore, we observed that LPS treatment cannot cause ubiquitination of the lysine site (K105 and K144) mutant of FTL. Interestingly, LPS-mediated ubiquitin-dependent degradation of FTL is significantly inhibited by the JNK-specific inhibitor SP600125. Moreover, LPS could upregulate the protein level of E3 ubiquitin ligase Itch, a substrate of JNK kinases. Immunoprecipitation analyses revealed an increase in the association of FTL with Itch, a substrate of JNK kinases, in response to LPS stimulation. SP600125 decreased LPS-induced Itch upregulation. Taken together, these results suggest that LPS stimulation leads to the degradation of FTL through the ubiquitin-proteasome proteolytic pathway, and this FTL degradation is mediated by the JNK/Itch axis in murine macrophage cells.


Subject(s)
Apoferritins , Macrophages , Proteasome Endopeptidase Complex , Animals , Apoferritins/genetics , Apoferritins/metabolism , Iron , Lipopolysaccharides/pharmacology , MAP Kinase Signaling System , Macrophages/metabolism , Mice , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/genetics
3.
J Trace Elem Med Biol ; 68: 126796, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34098241

ABSTRACT

BACKGROUND: Osteoporosis is frequently accompanied by iron disorders. Calcitonin (CT) was approved as a clinical drug to treat osteoporosis. Hepcidin is a peptide hormone that is secreted by the liver and controls body iron homeostasis. Hepcidin deficiency leads to iron overload diseases. This study was aimed at investigating the effect of CT on hepatic hepcidin and the mechanism by which CT modulates hepatic hepcidin pathways and iron metabolism. METHOD: RT-PCR, Western blot, ELISA and siRNA were used to detect the effect of CT on iron metabolism in vivo and in vitro. In addition, the regulatory signal molecules of hepcidin were measured to explore the molecular mechanism of its regulation. RESULTS: The results showed that CT strongly increased hepcidin expression and altered iron homeostasis, after mice were intraperitoneal injection of CT. In response to CT administration, BMP6 level in kidney and the serum BMP6 was increased significantly. The phosphorylation of Smad1/5/8 proteins in liver was increased at 3 h and 6 h. Moreover, the Bmp inhibitor LDN-193,189 pretreatment significantly attenuated the CT-mediated increases in phosphorylated Smad1/5/8 and Hamp1 mRNA levels. Calcitonin receptor (CTR) siRNA transfection significant suppressed the role of CT on BMP6 expression in Caki-1 cells. CONCLUSION: Our results suggest that CT strongly induces hepcidin expression and affected iron metabolism. It will provide a new strategy for the treatment of calcium iron related diseases.


Subject(s)
Calcitonin , Hepcidins , Osteoporosis , Peptide Hormones , Animals , Bone Morphogenetic Protein 6 , Iron , Kidney , Liver , Mice , RNA, Small Interfering
4.
Am J Respir Cell Mol Biol ; 65(2): 189-200, 2021 08.
Article in English | MEDLINE | ID: mdl-33861690

ABSTRACT

Strict control of iron homeostasis is critical for the maintenance of normal lung function. Iron accumulates in the lungs of patients with idiopathic pulmonary fibrosis (PF), but the characteristics of iron metabolism in the pathogenesis of PF and related targeting therapeutics are not well studied. In this study, we investigated the cellular and molecular characteristics of iron metabolism in fibrotic lungs and further explored the efficacy of clioquinol (CQ) for the treatment of PF as well as its functional mechanism. Iron aggregates accumulated in the lungs of patients with idiopathic PF, and FTL (ferritin light chain) transcripts were increased in their pulmonary fibroblasts. In the bleomycin (BLM)-induced PF (BLM-PF) mouse model, pulmonary iron accumulation is a very early and concomitant event of PF. Labile iron pool levels in both fibroblasts and macrophages from the BLM-PF model were elevated, and iron metabolism was dysregulated. CQ attenuated PF induced by BLM and FITC, and iron-saturated CQ did not alleviate BLM-PF. Furthermore, CQ inhibited the activation of fibroblasts, including proliferation, fibrotic differentiation, proinflammatory cytokine secretion, and migration. In conclusion, our study demonstrated that CQ, acting as an iron chelator, attenuates experimental PF through inactivation of fibroblasts, providing support for targeting iron metabolism as a basis for PF treatment.


Subject(s)
Chelating Agents/pharmacology , Clioquinol/pharmacology , Fibroblasts/metabolism , Idiopathic Pulmonary Fibrosis/metabolism , Iron/metabolism , Animals , Bleomycin/adverse effects , Bleomycin/pharmacology , Disease Models, Animal , Female , Fibroblasts/pathology , Humans , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/drug therapy , Idiopathic Pulmonary Fibrosis/pathology , Male , Mice
5.
Br J Pharmacol ; 178(5): 1182-1199, 2021 03.
Article in English | MEDLINE | ID: mdl-33450067

ABSTRACT

BACKGROUND AND PURPOSE: Acute kidney injury is a common clinical problem with no definitive or specific treatment. Therefore, the molecular mechanisms of acute kidney injury must be fully understood to develop novel treatments. Nuciferine, a major bioactive compound isolated from the lotus leaf, possesses extensive pharmacological activities. Its effect on folic acid-induced acute kidney injury, however, remains unknown. Here, we aimed to clarify the pharmacological effects of nuciferine and its mechanisms of action in acute kidney injury. EXPERIMENTAL APPROACH: The effects of nuciferine on folic acid-induced acute kidney injury in mice were investigated. HK-2 human proximal tubular epithelial cells and HEK293T HEK cells were used to evaluate the protective effect of nuciferine on RSL3-induced ferroptosis. KEY RESULTS: Nuciferine treatment mitigated the pathological alterations, ameliorated inflammatory cell infiltration and improved kidney dysfunction in mice with folic acid-induced acute kidney injury. In HK-2 and HEK293T cells, nuciferine significantly prevented RSL3-induced ferroptotic cell death. Mechanistically, nuciferine significantly inhibited ferroptosis by preventing iron accumulation and lipid peroxidation in vitro and in vivo. Moreover, knockdown of glutathione (GSH) peroxidase 4 (GPX4) abolished the protective effect of nuciferine against ferroptosis. CONCLUSION AND IMPLICATIONS: Nuciferine ameliorated renal injury in mice with acute kidney injury, perhaps by inhibiting the ferroptosis. Nuciferine may represent a novel treatment that improves recovery from acute kidney injury by targeting ferroptosis.


Subject(s)
Acute Kidney Injury , Ferroptosis , Acute Kidney Injury/chemically induced , Acute Kidney Injury/drug therapy , Acute Kidney Injury/prevention & control , Animals , Aporphines , Folic Acid , HEK293 Cells , Humans , Mice
6.
Can J Infect Dis Med Microbiol ; 2020: 4783062, 2020.
Article in English | MEDLINE | ID: mdl-33062082

ABSTRACT

OBJECTIVE: To explore the clinical characteristics of patients with different severity in the early outbreak of COVID-19, hoping to provide reference for clinical diagnosis and treatment. METHODS: We retrospectively analyzed the clinical data of 95 COVID-19 patients in Wuhan Red Cross Hospital of China from January 17 to February 13, 2020. All patients were investigated with epidemiological questionnaires. Outcomes were followed up until April 1, 2020. RESULTS: There were 53 males and 42 females, aged 22-84 years (mean 57.3 years). Clinical classification included 54 cases of common type, 27 cases of severe type, and 14 cases of critical type. Six patients had been exposed to the local Huanan seafood market. There were 38 clusters of COVID-19, including 27 family clusters and 11 work unit clusters. Common symptoms included fever (86 (90.5%) of 95), cough (73 (76.8%)), and fatigue (50 (52.6%)). Laboratory findings showed that the most common abnormalities were lymphopenia (75 (78.9%)), elevated D-dimer (60 (63.2%)), and elevated C-reactive protein (56 (58.9%)) on admission. All patients had abnormal chest computed tomography, showing patchy shadows or ground-glass opacities. Severe and critical cases were older, more likely to have shortness of breath, more likely to have underlying comorbidities, and more likely to have abnormal laboratory findings than common cases. The prognosis of patients with different degrees of severity was significantly different. All common and severe patients (100%) were cured and discharged from the hospital, while 10 (71.4%) of 14 critical patients died. CONCLUSIONS: COVID-19 has fast transmission speed and high pathogenicity. We must assess the severity of the disease and take corresponding treatment measures as early as possible.

7.
Zhongguo Xiu Fu Chong Jian Wai Ke Za Zhi ; 34(8): 1041-1043, 2020 Aug 15.
Article in Chinese | MEDLINE | ID: mdl-32794676

ABSTRACT

A 49-year-old male patient with compartment syndrome of the right leg caused by acute carbon monoxide poisoning was admitted on December 30, 2019. The patient had a 10-year history of chronic nephritis and began dialysis treatment due to renal failure 1 month ago. Emergency surgical decompression for compartment syndrome was performed after admission. Two weeks later, the patient was diagnosed as the novel coronavirus pneumonia caused by 2019 novel coronavirus (2019-nCoV) infection. Then, the patient was transferred to the isolation ward, where he was given anti-infection, anti-virus, expectorant, heat-clearing and detoxifying drugs, bedside dialysis, and nutrition support symptomatic treatment. After 2 weeks of treatment, the patient is getting better, with no fever, cough, wheezing, and other discomfort. Meanwhile, the sensory and motor functions of right lower limb recovered gradually. This case is rare, severe, and difficult to diagnose and treat. It is the first reported case of novel coronavirus pneumonia after orthopedic surgery.


Subject(s)
Compartment Syndromes/complications , Compartment Syndromes/surgery , Coronavirus Infections/complications , Coronavirus Infections/therapy , Pneumonia, Viral/complications , Pneumonia, Viral/therapy , Betacoronavirus , COVID-19 , Carbon Monoxide Poisoning/complications , Decompression, Surgical , Humans , Male , Middle Aged , Pandemics , SARS-CoV-2
8.
Biochem Biophys Res Commun ; 524(3): 736-743, 2020 04 09.
Article in English | MEDLINE | ID: mdl-32035614

ABSTRACT

Deferasirox (DFX) is an iron chelator approved for the treatment of iron overload diseases. However, the role of DFX in oxidative stress-induced cell apoptosis and the exact molecular mechanisms underlying these processes remain poorly understood and require further investigation. In this study, we found that DFX rendered resistant to H2O2-induced apoptosis in HEK293T cells, reduced the intracellular levels of the labile iron pool (LIP) and oxidative stress induced by H2O2. Furthermore, DFX inhibited the ubiquitination and degradation of the cyclin-dependent kinase inhibitor p21WAF1/CIP1 (p21) via modulation of the interaction of p21 with SCF-Skp2. DFX also showed the inhibition effect on the activation of c-Jun N-terminal kinase (JNK), pro-caspase-3 and related mitochondrial apoptosis pathway induced by H2O2. These results provide novel insights into the molecular mechanism underpinning iron-mediated oxidative stress and apoptosis, and they may represent a promising target for therapeutic interventions in related pathological conditions.


Subject(s)
Apoptosis/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cytoprotection/drug effects , Deferasirox/pharmacology , Proteolysis/drug effects , Ubiquitination/drug effects , Caspase 3/metabolism , HEK293 Cells , Humans , Hydrogen Peroxide , Iron/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Oxidative Stress/drug effects , Signal Transduction/drug effects , Ubiquitin/metabolism , Up-Regulation/drug effects
9.
Biochim Biophys Acta Gene Regul Mech ; 1862(8): 846-857, 2019 08.
Article in English | MEDLINE | ID: mdl-31054961

ABSTRACT

Heat shock factor 1 (HSF1) is a transcription factor essential for tumorigenesis, and targeting HSF1 may be effective in combined therapeutics for cervical cancer. Cyclosporin A (CsA) is an immunosuppressant that has revolutionized organ transplantation. However, the roles and regulatory mechanisms by which CsA modulates HSP expression remain largely unknown. In this study, we found that CsA pretreatment prevented induction of HSPs during heat shock by enhancing the phosphorylation of Ser303 and Ser307 on HSF1 and thus inhibiting its transcriptional activity. Suppression of ERK1/2, GSK3ß and CK2 activities attenuated CsA-induced down-regulation of HSP expression and up-regulation of HSF1 phosphorylation. CsA interfered with HSF1-SSBP1 complex formation and HSF1 nuclear translocation and recruitment to the HSP70 promoter. CsA clearly caused HeLa cell death during proteotoxic stress through reduced expression of HSPs. These results indicate that CsA suppresses HSP induction during heat shock by regulating the phosphorylation and nuclear translocation of HSF1. Our results provide a conceptual framework for the development of novel therapeutic strategies for cervical cancer through application of CsA during hyperthermia or chemotherapy.


Subject(s)
Cyclosporine/pharmacology , Heat Shock Transcription Factors/metabolism , Hyperthermia, Induced/methods , Uterine Cervical Neoplasms/metabolism , Combined Modality Therapy , Female , HSP70 Heat-Shock Proteins/genetics , HeLa Cells , Heat-Shock Response , Humans , MAP Kinase Signaling System/drug effects , Phosphorylation/drug effects , Serine/metabolism , Uterine Cervical Neoplasms/therapy
10.
Int J Biochem Cell Biol ; 109: 33-39, 2019 04.
Article in English | MEDLINE | ID: mdl-30716474

ABSTRACT

Erythropoietin (EPO) is a secreted hormone that stimulates the production of red blood cells, and the level of EPO is increased under hypoxia. The expression of EPO is regulated not only by the hypoxia-inducible factor (HIF) but also partly through epigenetic modifications, including histone acetylation and methylation. In this study, we report that histone H3K9 demethylase JMJD1 A is regulated by HIF-2α in HepG2 cells under hypoxia. Knockdown or over-expression of JMJD1 A can decrease or increase EPO expression, respectively. JMJD1 A can interact with HIF-2α to form a co-activator complex, which binds to the hypoxia response elements of EPO and increases EPO expression by catalyzing demethylation of H3K9me2, a transcription suppression marker. The results demonstrate that JMJD1 A is a co-activator of EPO expression.


Subject(s)
Erythropoietin/metabolism , Gene Expression Regulation , Histones/chemistry , Histones/metabolism , Hypoxia/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Biocatalysis , Hep G2 Cells , Humans , Hypoxia/enzymology , Hypoxia/genetics , Jumonji Domain-Containing Histone Demethylases/genetics , Lysine/metabolism , Male , Methylation , Mice , Mice, Inbred C57BL , Transcription, Genetic
11.
Aging Dis ; 8(4): 458-470, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28840060

ABSTRACT

Oxidative stress and iron accumulation are tightly associated with neurodegenerative diseases. Mitochondrial ferritin (FtMt) is identified as an iron-storage protein located in the mitochondria, and its role in regulation of iron hemeostasis in neurodegenerative diseases has been reported. However, the role of FtMt in hydrogen peroxide (H2O2)-induced oxidative stress and iron accumulation in neuronal cells has not been studied. Here, we overexpressed FtMt in neuroblastoma SH-SY5Y cells and induced oxidative stress by treating with extracellular H2O2. We found that overexpression of FtMt significantly prevented cell death induced by H2O2, particularly the apoptosis-dependent cell death. The protective effects involved inhibiting the generation of cellular reactive oxygen species, sustaining mitochondrial membrane potential, maintaining the level of anti-apoptotic protein Bcl-2, and inhibiting the activation of pro-apoptotic protein caspase 3. We further explored the mechanism of these protective effects and found that FtMt expression markedly altered iron homeostasis of the H2O2 treated cells as compared to that of controls. The FtMt overexpression significantly reduced cellular labile iron pool (LIP) and protected H2O2-induced elevation on LIP. While in H2O2 treated SH-SY5Y cells, the increased iron uptake and reduced iron release, in correlation with levels of DMT1(-IRE) and ferroportin 1, resulted in heavy iron accumulation, the FtMt overexpressing cells didn't show any significant changes in levels of iron transport proteins and in the level of LIP. These results implicate a neuroprotective role of FtMt on H2O2-induced oxidative stress, which may provide insights into the treatment of iron accumulation associated neurodegenerative diseases.

12.
Cell Death Dis ; 7(11): e2475, 2016 11 17.
Article in English | MEDLINE | ID: mdl-27853170

ABSTRACT

Mitochondrial ferritin (FtMt) is a mitochondrially localized protein possessing ferroxidase activity and the ability to store iron. FtMt overexpression in cultured cells protects against oxidative damage by sequestering redox-active, intracellular iron. Here, we found that acute exhaustive exercise significantly increases FtMt expression in the murine heart. FtMt gene disruption decreased the exhaustion exercise time and altered heart morphology with severe cardiac mitochondrial injury and fibril disorganization. The number of apoptotic cells as well as the levels of apoptosis-related proteins was increased in the FtMt-/- mice, though the ATP levels did not change significantly. Concomitant to the above was a high 'uncommitted' iron level found in the FtMt-/- group when exposed to acute exhaustion exercise. As a result of the increase in catalytic metal, reactive oxygen species were generated, leading to oxidative damage of cellular components. Taken together, our results show that the absence of FtMt, which is highly expressed in the heart, increases the sensitivity of mitochondria to cardiac injury via oxidative stress.


Subject(s)
Cardiotonic Agents/metabolism , Ferritins/metabolism , Mitochondria/metabolism , Myocardium/metabolism , Physical Conditioning, Animal , Acute Disease , Animals , Apoptosis , Calcium/metabolism , Gene Deletion , Immunoblotting , Iron/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/ultrastructure , Myocardium/pathology , Myocardium/ultrastructure , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Oxidative Stress
13.
Nanomedicine ; 12(7): 1747-1755, 2016 10.
Article in English | MEDLINE | ID: mdl-27112306

ABSTRACT

The absorption mechanism of heme iron remains unclear due to the limit of labeling techniques. Quantum dots (QDs) are powerful fluorescent probes resistant to photobleaching, however, there is no data about the application of QDs in heme iron absorption. Herein, we prepared hemin-coated CdSe/ZnS (QDs-hemin), and studied their absorption in vitro and in vivo. Results showed that QDs-hemin had uniform particle sizes, physiological stability and high joint efficiency. Moreover, QDs-hemin could be successfully absorbed gradually into the duodenum with the time using synchrotron radiation micro X-ray fluorescence and confocal laser scanning microscopy. Furthermore, QDs-hemin were observed to degrade in lysosomes, and their absorption was blocked by Heme Carrier Protein 1 (HCP1) antibody and HCP1 siRNA. All the results demonstrate that QDs can be a good tracer for heme iron and that HCP1 pathway is critical and predominant over the endocytosis pathway in the absorption mechanism.


Subject(s)
Hemin/pharmacokinetics , Quantum Dots , Animals , Duodenum , Fluorescent Dyes , Iron , Mice , Particle Size , Protein-Arginine N-Methyltransferases
14.
Brain Res ; 1642: 33-42, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27017962

ABSTRACT

Our previous work showed that mitochondrial ferritin (MtFt) played an important role in preventing neuronal damage in 6-OHDA-induced Parkinson's disease (PD). However, the role of MtFt in a PD model induced by MPTP is not clear. Here, we found that methyl-4-phenyl-1, 2, 3, 6-tetra-pyridine (MPTP) significantly upregulated MtFt in the mouse hippocampus, substantia nigra (SN) and striatum. To explore the effect of MtFt upregulation on the MPTP-mediated injury to neural cells, MtFt-/- mice and MtFt-overexpressing cells were used to construct models of PD induced by MPTP. Our results showed that MPTP dramatically downregulated expression of transferrin receptor 1 (TfR1) and tyrosine hydroxylase and upregulated L-ferritin expression in the mouse striatum and SN. Interestingly, MPTP induced high levels of MtFt in these tissues, indicating that MtFt was involved in iron metabolism and influenced dopamine synthesis induced by MPTP. Meanwhile, the Bcl2/Bax ratio was decreased significantly by MPTP in the striatum and SN of MtFt knockout (MtFt-/-) mice compared with controls. Overexpression of MtFt increased TfR1 and decreased ferroportin 1 induced by 1-methyl-4-phenylpyridinium ions (MPP+). MtFt strongly inhibited mitochondrial damage through maintaining the mitochondrial membrane potential and protecting the integrity of the mitochondrial membrane. It also suppressed the increase of the labile iron pool, decreased production of reactive oxygen species and dramatically rescued the apoptosis induced by MPP+. In conclusion, this study demonstrates that MtFt plays an important role in preventing neuronal damage in the MPTP-induced parkinsonian phenotype by inhibiting cellular iron accumulation and subsequent oxidative stress.


Subject(s)
Brain/metabolism , Ferritins/metabolism , Iron/metabolism , MPTP Poisoning/metabolism , Mitochondria/metabolism , Oxidative Stress , Parkinson Disease/metabolism , 1-Methyl-4-phenylpyridinium/administration & dosage , Animals , Apoferritins/metabolism , Apoptosis/drug effects , Brain/drug effects , Cation Transport Proteins/metabolism , Cell Survival/drug effects , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Disease Models, Animal , Ferritins/genetics , Hippocampus/drug effects , Hippocampus/metabolism , Mice , Mice, Knockout , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Receptors, Transferrin/metabolism , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Tyrosine 3-Monooxygenase/metabolism
15.
Nanoscale ; 8(9): 5126-38, 2016 Mar 07.
Article in English | MEDLINE | ID: mdl-26875690

ABSTRACT

Prostate cancer is highly prevalent and has become the second leading cause of cancer-related death in men. Its treatment remains a challenge in the clinic, particularly in patients who have advanced to "castration-resistant prostate cancer" (CRPC). Thus, more effective therapeutic strategies are required. Quercetin (QCT) is a natural flavonoid compound that has attracted increasing interest due to its anticancer activity. However, the clinical application of quercetin is largely hampered by its poor water solubility and low bioavailability. The objective of this study was to evaluate the therapeutic potential of novel QCT-loaded nanomicelles (M-QCTs) assembled from DSPE-PEG2000 for prostate cancer treatment. Our results indicated that QCT was efficiently encapsulated into micelles up to 1 mg mL(-1), which corresponds to a 450-fold increase of its water solubility. In vitro studies showed that the half-maximal inhibitory concentration (IC50) value (20.2 µM) of M-QCTs was much lower than free QCT (>200 µM). Thus, M-QCTs were considerably more effective than free QCT in proliferation inhibition and apoptosis induction of human androgen-independent PC-3 cells. Furthermore, M-QCTs showed superior antitumor efficacy and the tumor proliferation rate reduced by 52.03% compared to the control group in the PC-3 xenograft mouse model, possibly due to increased accumulation of M-QCTs at the tumor site by the enhanced permeability and retention (EPR) effect. Collectively, our studies demonstrated that M-QCTs significantly increase drug accumulation at the tumor site and exhibit superior anticancer activity in prostate cancer. Thus, our nanomicelle-based drug delivery system constitutes a promising and effective therapeutic strategy for clinical treatment.


Subject(s)
Drug Carriers , Micelles , Nanoparticles/chemistry , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/metabolism , Quercetin , Animals , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Drug Carriers/pharmacology , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Quercetin/chemistry , Quercetin/pharmacokinetics , Quercetin/pharmacology , Xenograft Model Antitumor Assays/methods
16.
Biochem Biophys Res Commun ; 465(2): 305-11, 2015 Sep 18.
Article in English | MEDLINE | ID: mdl-26277391

ABSTRACT

To examine the role of the intracellular labile iron pool (LIP) in the induction of inflammatory responses, we investigated the anti-inflammatory effect of the iron chelator deferoxamine (DFO) on lipopolysaccharide (LPS)-induced inflammatory responses in RAW264.7 macrophage cells and endotoxic shock in mice in the present study. Our data showed that DFO significantly decreased LPS-induced LIP and ROS upregulation. We then found that DFO inhibited phosphorylation of MAP kinases such as ERK and p38 and also inhibited the activation of NF-κB induced by LPS. Furthermore, the production of tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß), nitric oxide (NO) and prostaglandin E2 (PGE2) induced by LPS was inhibited by DFO in RAW264.7 macrophages. Administration of DFO significantly decreased the mortality and improved the survival of septic mice with lethal endotoxemia in LPS-injected mice. These results demonstrate that iron plays a pivotal role in the induction of inflammatory responses and against septic shock. DFO has effective inhibitory effect on the production of inflammatory mediators via suppressing activation of MAPKs and NF-κB signaling pathways; it also has a protective effect on LPS-induced endotoxic shock in mice. Our findings open doors to further studies directed at exploring a new class of drugs against septic shock or other inflammatory diseases by modulating cellular chelatable iron.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Deferoxamine/pharmacology , Iron Chelating Agents/pharmacology , Iron/metabolism , Shock, Septic/drug therapy , Animals , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Cell Line , Deferoxamine/metabolism , Dinoprostone/antagonists & inhibitors , Dinoprostone/metabolism , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Iron Chelating Agents/metabolism , Lipopolysaccharides , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Male , Mice , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , NF-kappa B/metabolism , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/metabolism , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Shock, Septic/chemically induced , Shock, Septic/metabolism , Shock, Septic/mortality , Signal Transduction , Survival Analysis , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
17.
Cell Mol Life Sci ; 72(5): 983-97, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25213357

ABSTRACT

Mitochondrial ferritin (FtMt) has a significant effect on the regulation of cytosolic and mitochondrial iron levels. However, because of the deficiency of iron regulatory elements (IRE) in FtMt's gene sequence, the exact function of FtMt remains unclear. In the present study, we found that FtMt dramatically inhibited SH-SY5Y cell proliferation and tumor growth in nude mice. Interestingly, excess FtMt did not adversely affect the development of drosophila. Additionally, we found that the expression of FtMt in human normal brain tissue was significantly higher than that of neuroblastoma, but not higher than that of neurospongioma. However, the expression of transferrin receptor 1 is completely opposite. We therefore hypothesized that increased expression of FtMt may negatively affect the vitality of neuronal tumor cells. Therefore, we further investigated the underlying mechanisms of FtMt's inhibitory effects on neuronal tumor cell proliferation. As expected, FtMt overexpression disturbed the iron homeostasis of tumor cells and significantly downregulated the expression of proliferating cell nuclear antigen. Moreover, FtMt affected cell cycle, causing G1/S arrest by modifying the expression of cyclinD1, cyclinE, Cdk2, Cdk4 and p21. Remarkably, FtMt strongly upregulated the expression of the tumor suppressors, p53 and N-myc downstream-regulated gene-1 (NDRG1), but dramatically decreased C-myc, N-myc and p-Rb levels. This study demonstrates for the first time a new role and mechanism for FtMt in the regulation of cell cycle. We thus propose FtMt as a new candidate target for inhibiting neuronal tumor cell proliferation. Appropriate regulation of FtMt expression may prevent tumor cell growth. Our study may provide a new strategy for neuronal cancer therapy.


Subject(s)
Ferritins/metabolism , Mitochondria/metabolism , Animals , Apoptosis , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation , Cyclin D1/metabolism , Cyclin E/metabolism , Cyclin-Dependent Kinase 2/metabolism , Ferritins/genetics , G1 Phase Cell Cycle Checkpoints , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neuroblastoma/metabolism , Neuroblastoma/pathology , Proto-Oncogene Proteins c-myc/metabolism , Retinoblastoma Protein/metabolism , Tumor Suppressor Protein p53/metabolism
18.
J Gastroenterol Hepatol ; 30(3): 513-20, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25318588

ABSTRACT

BACKGROUND AND AIM: The body's requirement for iron is different at different developmental stages. However, the molecular mechanisms of age-dependent iron metabolism are poorly understood. In the present study, we investigated the expression of iron transport proteins in the duodenum of Sprague-Dawley rats at five different age stages. METHODS: Male Sprague-Dawley rats at postnatal week (PNW) 1, 3, 12, 44, and 88 were employed in the study. Serum iron status and tissue non-heme iron concentrations in the spleen, liver, bone marrow, heart, kidney, duodenal epithelium, and gastrocnemius were examined at each age stage. The expression of duodenal cytochrome b (DcytB), divalent metal transporter 1 (DMT1), ferroportin 1 (FPN1), hephaestin, and hepcidin were measured by real-time polymerase chain reaction or Western blot. RESULTS: The levels of serum iron and transferrin saturation were higher in the rats at PNW1 and 3 than in those at PNW12, 44, and 88. Non-heme iron contents decreased from PNW1 to PNW3 and then increased thereafter. Duodenal DcytB, DMT1, and FPN1 increased to the highest level at PNW3 and then decreased from PNW12 to 88. The hepatic hepcidin mRNA level decreased to the lowest level at PNW3 and then increased with age. CONCLUSION: Our findings showed that age had a significant effect on body iron status. The increased duodenal DcytB, DMT1, and FPN1 expression can enhance intestinal iron absorption to meet the high iron requirements in infants. Hepcidin or enterocyte iron levels may be involved in the regulation of age-dependent FPN1, DMT1, and DcytB expression in the duodenum.


Subject(s)
Aging/genetics , Aging/metabolism , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cytochromes b/genetics , Cytochromes b/metabolism , Duodenum/metabolism , Gene Expression Regulation, Developmental/genetics , Gene Expression , Iron/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Animals , Blotting, Western , Enterocytes/metabolism , Hepcidins/metabolism , Intestinal Absorption/genetics , Male , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Tissue Distribution , Transferrin/metabolism
19.
Biomed Res Int ; 2015: 105325, 2015.
Article in English | MEDLINE | ID: mdl-26788496

ABSTRACT

In female, inadequate iron supply is a highly prevalent problem that often leads to iron-deficiency anemia. This study aimed to understand the effects of pregnancy and lactation on iron metabolism. Rats with different days of gestation and lactation were used to determine the variations in iron stores and serum iron level and the changes in expression of iron metabolism-related proteins, including ferritin, ferroportin 1 (FPN1), ceruloplasmin (Cp), divalent metal transporter 1 (DMT1), transferrin receptor 1 (TfR1), and the major iron-regulatory molecule-hepcidin. We found that iron stores decline dramatically at late-pregnancy period, and the low iron store status persists throughout the lactation period. The significantly increased FPN1 level in small intestine facilitates digestive iron absorption, which maintains the serum iron concentration at a near-normal level to meet the increase of iron requirements. Moreover, a significant decrease of hepcidin expression is observed during late-pregnancy and early-lactation stages, suggesting the important regulatory role that hepcidin plays in iron metabolism during pregnancy and lactation. These results are fundamental to the understanding of iron homeostasis during pregnancy and lactation and may provide experimental bases for future studies to identify key molecules expressed during these special periods that regulate the expression of hepcidin, to eventually improve the iron-deficiency status.


Subject(s)
Anemia, Iron-Deficiency/genetics , Cation Transport Proteins/blood , Hepcidins/blood , Iron/blood , Lactation/metabolism , Anemia, Iron-Deficiency/blood , Anemia, Iron-Deficiency/pathology , Animals , Antigens, CD/blood , Ceruloplasmin/biosynthesis , Ceruloplasmin/metabolism , Female , Ferritins/blood , Gene Expression , Humans , Pregnancy , Rats , Receptors, Transferrin/blood
20.
Acta Biochim Biophys Sin (Shanghai) ; 47(2): 106-13, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25520177

ABSTRACT

Lysine (K)-specific demethylase 6B (KDM6B) is a histone H3K27 demethylase, which specifically catalyzes the demethylation of H3 lysine-27 tri/dimethylation (H3K27me3/2). KDM6B can activate gene transcription by promoting transcriptional elongation which is associated with RNA polymerase II and related elongation factors. So KDM6B is important for the regulation of gene expression. Previous studies have indicated that several histone demethylases such as KDM3A, KDM4B, and KDM4C are regulated by hypoxia-inducible factor (HIF). But, the effect of hypoxia on KDM6B is not fully understood. In this study, we found that the expression levels of KDM6B mRNA and protein are modestly up-regulated under hypoxia (1% O2) or mimic hypoxia (desferrioxamine mesylate or CoCl2 treatment) (P<0.05). The result of RNAi shows that the up-regulation of KDM6B is dependent on HIF-2α, but not on HIF-1α. The result of chromatin immunoprecipitation assay indicates that there is a hypoxia response element in KDM6B promoter (-4041 to -4037). The result of Co-IP assay indicates that KDM6B can form complex with HIF-2α or HIF-1α. The knockdown experiment implies that KDM6B is a potential regulator for HIF-2α target genes. These data demonstrate that KDM6B is a new hypoxia response gene regulated by HIF-2α. Our results also show that KDM6B is a potential co-activator of HIF-α, which is important for the activation of hypoxia response genes.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Gene Expression Regulation, Enzymologic , Jumonji Domain-Containing Histone Demethylases/metabolism , Active Transport, Cell Nucleus , Catalysis , Cell Hypoxia , Cell Nucleus/metabolism , Chromatin Immunoprecipitation , Epigenesis, Genetic , HEK293 Cells , Hep G2 Cells , Humans , Promoter Regions, Genetic , RNA Interference , RNA, Messenger/metabolism , Response Elements , Signal Transduction , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL
...