Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 113
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Semin Cell Dev Biol ; 115: 45-53, 2021 07.
Article in English | MEDLINE | ID: mdl-33419608

ABSTRACT

Manganese serves as an indispensable catalytic center and the structural core of various enzymes that participate in a plethora of biological processes, including oxidative phosphorylation, glycosylation, and signal transduction. In pathogenic microorganisms, manganese is required for survival by maintaining basic biochemical activity and virulence; in contrast, the host utilizes a process known as nutritional immunity to sequester manganese from invading pathogens. Recent epidemiological and animal studies have shown that manganese increases the immune response in a wide range of vertebrates, including humans, rodents, birds, and fish. On the other hand, excess manganese can cause neurotoxicity and other detrimental effects. Here, we review recent data illustrating the essential role of manganese homeostasis at the host-pathogen interface and in the host immune system. We also discuss the accumulating body of evidence that manganese modulates various signaling pathways in immune processes. Finally, we discuss the key molecular players involved in manganese's immune regulatory function, as well as the clinical implications with respect to cancer immunotherapy.


Subject(s)
Host-Pathogen Interactions/immunology , Immune System/immunology , Manganese/metabolism , Homeostasis , Humans
2.
Blood ; 138(8): 689-705, 2021 08 26.
Article in English | MEDLINE | ID: mdl-33895792

ABSTRACT

Ferroportin (FPN), the body's sole iron exporter, is essential for maintaining systemic iron homeostasis. In response to either increased iron or inflammation, hepatocyte-secreted hepcidin binds to FPN, inducing its internalization and subsequent degradation. However, the E3 ubiquitin ligase that underlies FPN degradation has not been identified. Here, we report the identification and characterization of a novel mechanism involving the RNF217-mediated degradation of FPN. A combination of 2 different E3 screens revealed that the Rnf217 gene is a target of Tet1, mediating the ubiquitination and subsequent degradation of FPN. Interestingly, loss of Tet1 expression causes an accumulation of FPN and an impaired response to iron overload, manifested by increased iron accumulation in the liver together with decreased iron in the spleen and duodenum. Moreover, we found that the degradation and ubiquitination of FPN could be attenuated by mutating RNF217. Finally, using 2 conditional knockout mouse lines, we found that knocking out Rnf217 in macrophages increases splenic iron export by stabilizing FPN, whereas knocking out Rnf217 in intestinal cells appears to increase iron absorption. These findings suggest that the Tet1-RNF217-FPN axis regulates iron homeostasis, revealing new therapeutic targets for FPN-related diseases.


Subject(s)
Carrier Proteins/metabolism , Cation Transport Proteins/metabolism , Iron/metabolism , Proteolysis , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Animals , Carrier Proteins/genetics , Cation Transport Proteins/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Iron Overload/genetics , Iron Overload/metabolism , Mice , Mice, Knockout , Organ Specificity/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Ubiquitin-Protein Ligases/genetics
3.
Nucleic Acids Res ; 49(10): 5537-5552, 2021 06 04.
Article in English | MEDLINE | ID: mdl-33999206

ABSTRACT

The intestinal invasion of pathogenic microorganisms can have serious health consequences. Recent evidence has shown that the N6-methyladenosine (m6A) mRNA modification is closely associated with innate immunity; however, the underlying mechanism is poorly understood. Here, we examined the function and mechanism of m6A mRNA modification and the YTH domain-containing protein YTHDF1 (YTH N6-methyladenosine RNA-binding protein 1) in the innate immune response against bacterial pathogens in the intestine. Ribo-seq and m6A-seq analyses revealed that YTHDF1 directs the translation of Traf6 mRNA, which encodes tumor necrosis factor receptor-associated factor 6, thereby regulating the immune response via the m6A modification near the transcript's stop codon. Furthermore, we identified a unique mechanism by which the P/Q/N-rich domain in YTHDF1 interacts with the DEAD domain in the host factor DDX60, thereby regulating the intestinal immune response to bacterial infection by recognizing the target Traf6 transcript. These results provide novel insights into the mechanism by which YTHDF1 recognizes its target and reveal YTHDF1 as an important driver of the intestinal immune response, opening new avenues for developing therapeutic strategies designed to modulate the intestinal immune response to bacterial infection.


Subject(s)
Escherichia coli Infections/immunology , Immunity, Innate , Intracellular Signaling Peptides and Proteins/immunology , RNA-Binding Proteins/immunology , Animals , Caco-2 Cells , Enterotoxigenic Escherichia coli/immunology , Epithelial Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Swine , TNF Receptor-Associated Factor 6/immunology
4.
J Mol Cell Cardiol ; 173: 141-153, 2022 12.
Article in English | MEDLINE | ID: mdl-36273661

ABSTRACT

With its complicated pathobiology and pathophysiology, heart failure (HF) remains an increasingly prevalent epidemic that threatens global human health. Ferroptosis is a form of regulated cell death characterized by the iron-dependent lethal accumulation of lipid peroxides in the membrane system and is different from other types of cell death such as apoptosis and necrosis. Mounting evidence supports the claim that ferroptosis is mainly regulated by several biological pathways including iron handling, redox homeostasis, and lipid metabolism. Recently, ferroptosis has been identified to play an important role in HF induced by different stimuli such as myocardial infarction, myocardial ischemia reperfusion, chemotherapy, and others. Thus, it is of great significance to deeply explore the role of ferroptosis in HF, which might be a prerequisite to precise drug targets and novel therapeutic strategies based on ferroptosis-related medicine. Here, we review current knowledge on the link between ferroptosis and HF, followed by critical perspectives on the development and progression of ferroptotic signals and cardiac remodeling in HF.


Subject(s)
Ferroptosis , Heart Failure , Humans , Cell Death , Apoptosis , Iron/metabolism , Lipid Peroxidation
5.
Blood ; 136(6): 726-739, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32374849

ABSTRACT

Although the serum-abundant metal-binding protein transferrin (encoded by the Trf gene) is synthesized primarily in the liver, its function in the liver is largely unknown. Here, we generated hepatocyte-specific Trf knockout mice (Trf-LKO), which are viable and fertile but have impaired erythropoiesis and altered iron metabolism. Moreover, feeding Trf-LKO mice a high-iron diet increased their susceptibility to developing ferroptosis-induced liver fibrosis. Importantly, we found that treating Trf-LKO mice with the ferroptosis inhibitor ferrostatin-1 potently rescued liver fibrosis induced by either high dietary iron or carbon tetrachloride (CCl4) injections. In addition, deleting hepatic Slc39a14 expression in Trf-LKO mice significantly reduced hepatic iron accumulation, thereby reducing ferroptosis-mediated liver fibrosis induced by either a high-iron diet or CCl4 injections. Finally, we found that patients with liver cirrhosis have significantly lower levels of serum transferrin and hepatic transferrin, as well as higher levels of hepatic iron and lipid peroxidation, compared with healthy control subjects. Taken together, these data indicate that hepatic transferrin plays a protective role in maintaining liver function, providing a possible therapeutic target for preventing ferroptosis-induced liver fibrosis.


Subject(s)
Ferroptosis/physiology , Iron/metabolism , Liver Cirrhosis/metabolism , Liver/metabolism , Transferrin/physiology , Animals , Carbon Tetrachloride Poisoning/drug therapy , Carbon Tetrachloride Poisoning/metabolism , Carbon Tetrachloride Poisoning/pathology , Cation Transport Proteins/deficiency , Cation Transport Proteins/genetics , Cyclohexylamines/pharmacology , Cytokines/analysis , Erythropoiesis/physiology , Erythropoietin/analysis , Female , Ferroptosis/drug effects , Hepatocytes/metabolism , Homeostasis , Iron Overload/complications , Iron, Dietary/toxicity , Lipid Peroxidation , Liver Cirrhosis/chemically induced , Liver Cirrhosis/drug therapy , Liver Cirrhosis/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Proteins/analysis , Phenylenediamines/pharmacology , Transferrin/analysis
6.
Circ Res ; 127(4): 486-501, 2020 07 31.
Article in English | MEDLINE | ID: mdl-32349646

ABSTRACT

RATIONALE: Maintaining iron homeostasis is essential for proper cardiac function. Both iron deficiency and iron overload are associated with cardiomyopathy and heart failure via complex mechanisms. Although ferritin plays a central role in iron metabolism by storing excess cellular iron, the molecular function of ferritin in cardiomyocytes remains unknown. OBJECTIVE: To characterize the functional role of Fth (ferritin H) in mediating cardiac iron homeostasis and heart disease. METHODS AND RESULTS: Mice expressing a conditional Fth knockout allele were crossed with 2 distinct Cre recombinase-expressing mouse lines, resulting in offspring that lack Fth expression specifically in myocytes (MCK-Cre) or cardiomyocytes (Myh6-Cre). Mice lacking Fth in cardiomyocytes had decreased cardiac iron levels and increased oxidative stress, resulting in mild cardiac injury upon aging. However, feeding these mice a high-iron diet caused severe cardiac injury and hypertrophic cardiomyopathy, with molecular features typical of ferroptosis, including reduced glutathione (GSH) levels and increased lipid peroxidation. Ferrostatin-1, a specific inhibitor of ferroptosis, rescued this phenotype, supporting the notion that ferroptosis plays a pathophysiological role in the heart. Finally, we found that Fth-deficient cardiomyocytes have reduced expression of the ferroptosis regulator Slc7a11, and overexpressing Slc7a11 selectively in cardiomyocytes increased GSH levels and prevented cardiac ferroptosis. CONCLUSIONS: Our findings provide compelling evidence that ferritin plays a major role in protecting against cardiac ferroptosis and subsequent heart failure, thereby providing a possible new therapeutic target for patients at risk of developing cardiomyopathy.


Subject(s)
Amino Acid Transport System y+/metabolism , Apoferritins/deficiency , Cardiomyopathies/etiology , Ferroptosis/physiology , Iron/metabolism , Myocardium/metabolism , Aging , Alleles , Animals , Apoferritins/adverse effects , Apoferritins/genetics , Cardiomyopathies/metabolism , Cardiomyopathies/prevention & control , Cardiomyopathy, Hypertrophic/etiology , Cardiomyopathy, Hypertrophic/prevention & control , Crosses, Genetic , Cyclohexylamines/administration & dosage , Glutathione/metabolism , Heart Failure/etiology , Homeostasis , Hypertrophy, Left Ventricular/etiology , Iron Deficiencies , Iron Overload , Iron, Dietary/adverse effects , Lipid Peroxidation , Male , Mice , Mice, Transgenic , Myocytes, Cardiac/metabolism , Oxidative Stress , Phenylenediamines/administration & dosage , Reactive Oxygen Species/metabolism
7.
Proc Natl Acad Sci U S A ; 116(7): 2672-2680, 2019 02 12.
Article in English | MEDLINE | ID: mdl-30692261

ABSTRACT

Heart disease is the leading cause of death worldwide. A key pathogenic factor in the development of lethal heart failure is loss of terminally differentiated cardiomyocytes. However, mechanisms of cardiomyocyte death remain unclear. Here, we discovered and demonstrated that ferroptosis, a programmed iron-dependent cell death, as a mechanism in murine models of doxorubicin (DOX)- and ischemia/reperfusion (I/R)-induced cardiomyopathy. In canonical apoptosis and/or necroptosis-defective Ripk3-/-, Mlkl-/-, or Fadd-/-Mlkl-/- mice, DOX-treated cardiomyocytes showed features of typical ferroptotic cell death. Consistently, compared with dexrazoxane, the only FDA-approved drug for treating DOX-induced cardiotoxicity, inhibition of ferroptosis by ferrostatin-1 significantly reduced DOX cardiomyopathy. RNA-sequencing results revealed that heme oxygenase-1 (Hmox1) was significantly up-regulated in DOX-treated murine hearts. Administering DOX to mice induced cardiomyopathy with a rapid, systemic accumulation of nonheme iron via heme degradation by Nrf2-mediated up-regulation of Hmox1, which effect was abolished in Nrf2-deficent mice. Conversely, zinc protoporphyrin IX, an Hmox1 antagonist, protected the DOX-treated mice, suggesting free iron released on heme degradation is necessary and sufficient to induce cardiac injury. Given that ferroptosis is driven by damage to lipid membranes, we further investigated and found that excess free iron accumulated in mitochondria and caused lipid peroxidation on its membrane. Mitochondria-targeted antioxidant MitoTEMPO significantly rescued DOX cardiomyopathy, supporting oxidative damage of mitochondria as a major mechanism in ferroptosis-induced heart damage. Importantly, ferrostatin-1 and iron chelation also ameliorated heart failure induced by both acute and chronic I/R in mice. These findings highlight that targeting ferroptosis serves as a cardioprotective strategy for cardiomyopathy prevention.


Subject(s)
Apoptosis , Cardiomyopathies/prevention & control , Iron/metabolism , Animals , Cardiomyopathies/chemically induced , Doxorubicin/pharmacology , Doxorubicin/toxicity , Heme/metabolism , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Lipid Peroxidation , Mice , Mice, Knockout , Mitochondria, Heart/drug effects , Mitochondria, Heart/enzymology , Mitochondria, Heart/metabolism , Myocytes, Cardiac/metabolism , NF-E2-Related Factor 2/genetics , Reperfusion Injury/prevention & control , Up-Regulation
8.
J Pineal Res ; 70(2): e12704, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33206394

ABSTRACT

Accumulating evidence demonstrates that ferroptosis may be important in the pathophysiological process of traumatic brain injury (TBI). As a major hormone of the pineal gland, melatonin exerts many beneficial effects on TBI, but there is no information regarding the effects of melatonin on ferroptosis after TBI. As expected, TBI resulted in the time-course changes of ferroptosis-related molecules expression and iron accumulation in the ipsilateral cortex. Importantly, we found that treating with melatonin potently rescued TBI induced the changes mentioned above and improved functional deficits versus vehicle. Similar results were obtained with a ferroptosis inhibitor, liproxstatin-1. Moreover, the protective effect of melatonin is likely dependent on melatonin receptor 1B (MT2). Although ferritin plays a vital role in iron metabolism by storing excess cellular iron, its precise function in the brain, and whether it involves melatonin's neuroprotection remain unexplored. Considering ferritin H (Fth) is expressed predominantly in the neurons and global loss of Fth in mice induces early embryonic lethality, we then generated neuron-specific Fth conditional knockout (Fth-KO) mice, which are viable and fertile but have altered iron metabolism. In addition, Fth-KO mice were more susceptible to ferroptosis after TBI, and the neuroprotection by melatonin was largely abolished in Fth-KO mice. In vitro siFth experiments further confirmed the results mentioned above. Taken together, these data indicate that melatonin produces cerebroprotection, at least partly by inhibiting neuronal Fth-mediated ferroptosis following TBI, supporting the notion that melatonin is an excellent ferroptosis inhibitor and its anti-ferroptosis provides a potential therapeutic target for treating TBI.


Subject(s)
Apoferritins/metabolism , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/metabolism , Melatonin/therapeutic use , Animals , Apoferritins/genetics , Blotting, Western , Ferroptosis/drug effects , Immunohistochemistry , Iron/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Real-Time Polymerase Chain Reaction
9.
J Cell Mol Med ; 24(7): 4118-4126, 2020 04.
Article in English | MEDLINE | ID: mdl-32108988

ABSTRACT

The GNPAT variant rs11558492 (p.D519G) was identified as a novel genetic factor that modifies the iron-overload phenotype in homozygous carriers of the HFE p.C282Y variant. However, the reported effects of the GNPAT p.D519G variant vary among study populations. Here, we investigated the role of GNPAT in iron metabolism using Gnpat-knockout (Gnpat-/- ), Gnpat/Hfe double-knockout (Gnpat-/- Hfe-/- or DKO) mice and hepatocyte-specific Gnpat-knockout mice (Gnpatfl/fl ;Alb-Cre). Our analysis revealed no significant difference between wild-type (Gnpat+/+ ) and Gnpat-/- mice, between Hfe-/- and DKO mice, or between Gnpatfl/fl and Gnpatfl/fl ;Alb-Cre with respect to serum iron and tissue iron. In addition, the expression of hepcidin was not affected by deleting Gnpat expression in the presence or absence of Hfe. Feeding Gnpat-/- and DKO mice a high-iron diet had no effect on tissue iron levels compared with wild-type and Hfe-/- mice, respectively. Gnpat knockdown in primary hepatocytes from wild-type or Hfe-/- mice did not alter hepcidin expression, but it repressed BMP6-induced hepcidin expression. Taken together, these results support the hypothesis that deleting Gnpat expression has no effect on either systemic iron metabolism or the iron-overload phenotype that develops in Hfe-/- mice, suggesting that GNPAT does not directly mediate iron homeostasis under normal or high-iron dietary conditions.


Subject(s)
Acyltransferases/genetics , Hemochromatosis Protein/genetics , Hepcidins/genetics , Iron Overload/genetics , Iron/metabolism , Animals , Disease Models, Animal , Ferritins/genetics , Hepatocytes/metabolism , Homeostasis/genetics , Homozygote , Humans , Iron/blood , Iron Overload/metabolism , Iron Overload/pathology , Mice , Mice, Knockout , Phenotype
10.
Haematologica ; 105(8): 2071-2082, 2020 08.
Article in English | MEDLINE | ID: mdl-31601687

ABSTRACT

Transferrin receptor 1 (Tfr1) mediates the endocytosis of diferric transferrin in order to transport iron, and Tfr1 has been suggested to play an important role in hematopoiesis. To study the role of Tfr1 in hematopoiesis, we generated hematopoietic stem cell (HSC) specific Tfr1 knockout mice. We found that Tfr1 conditional knockout mice reached full term but died within one week of birth. Further analyses revealed that Tfr1-deficient HSC had impaired development of all hematopoietic progenitors except thrombocytes and B lymphocytes. In addition, Tfr1-deficient cells had cellular iron deficiency, which blocked the proliferation and differentiation of hematopoietic precursor cells, attenuated the commitment of hematopoietic lineages, and reduced the regeneration potential of HSC. Notably, hemin rescued the colony-forming capacity of Tfr1-deficient HSC, whereas expressing a mutant Tfr1 that lacks the protein's iron-transporting capacity failed to rescue hematopoiesis. These findings provide direct evidence that Tfr1 is essential for hematopoiesis through binding diferric transferrin to supply iron to cells.


Subject(s)
Iron , Receptors, Transferrin , Animals , Biological Transport , Hematopoiesis/genetics , Iron/metabolism , Mice , Mice, Knockout , Receptors, Transferrin/genetics , Transferrin/metabolism
11.
Environ Health ; 19(1): 104, 2020 10 02.
Article in English | MEDLINE | ID: mdl-33008482

ABSTRACT

BACKGROUND: Although prior studies showed a correlation between environmental manganese (Mn) exposure and neurodevelopmental disorders in children, the results have been inconclusive. There has yet been no consistent biomarker of environmental Mn exposure. Here, we summarized studies that investigated associations between manganese in biomarkers and childhood neurodevelopment and suggest a reliable biomarker. METHODS: We searched PubMed and Web of Science for potentially relevant articles published until December 31th 2019 in English. We also conducted a meta-analysis to quantify the effects of manganese exposure on Intelligence Quotient (IQ) and the correlations of manganese in different indicators. RESULTS: Of 1754 citations identified, 55 studies with 13,388 subjects were included. Evidence from cohort studies found that higher manganese exposure had a negative effect on neurodevelopment, mostly influencing cognitive and motor skills in children under 6 years of age, as indicated by various metrics. Results from cross-sectional studies revealed that elevated Mn in hair (H-Mn) and drinking water (W-Mn), but not blood (B-Mn) or teeth (T-Mn), were associated with poorer cognitive and behavioral performance in children aged 6-18 years old. Of these cross-sectional studies, most papers reported that the mean of H-Mn was more than 0.55 µg/g. The meta-analysis concerning H-Mn suggested that a 10-fold increase in hair manganese was associated with a decrease of 2.51 points (95% confidence interval (CI), - 4.58, - 0.45) in Full Scale IQ, while the meta-analysis of B-Mn and W-Mn generated no such significant effects. The pooled correlation analysis revealed that H-Mn showed a more consistent correlation with W-Mn than B-Mn. Results regarding sex differences of manganese associations were inconsistent, although the preliminary meta-analysis found that higher W-Mn was associated with better Performance IQ only in boys, at a relatively low water manganese concentrations (most below 50 µg/L). CONCLUSIONS: Higher manganese exposure is adversely associated with childhood neurodevelopment. Hair is the most reliable indicator of manganese exposure for children at 6-18 years of age. Analysis of the publications demonstrated sex differences in neurodevelopment upon manganese exposure, although a clear pattern has not yet been elucidated for this facet of our study.


Subject(s)
Child Development/drug effects , Environmental Exposure/adverse effects , Environmental Pollutants/adverse effects , Manganese/adverse effects , Neurodevelopmental Disorders/chemically induced , Biomarkers/analysis , Child , Hair/chemistry , Humans , Intelligence/drug effects
12.
Proc Natl Acad Sci U S A ; 114(49): 12940-12945, 2017 12 05.
Article in English | MEDLINE | ID: mdl-29180421

ABSTRACT

Zn plays a key role in controlling macrophage function during an inflammatory event. Cellular Zn homeostasis is regulated by two families of metal transporters, the SLC39A family of importers and the SLC30A family of exporters; however, the precise role of these transporters in maintaining macrophage function is poorly understood. Using macrophage-specific Slc39a10-knockout (Slc39a10fl/fl;LysM-Cre+ ) mice, we found that Slc39a10 plays an essential role in macrophage survival by mediating Zn homeostasis in response to LPS stimulation. Compared with Slc39a10fl/fl mice, Slc39a10fl/fl;LysM-Cre+ mice had significantly lower mortality following LPS stimulation as well as reduced liver damage and lower levels of circulating inflammatory cytokines. Moreover, reduced intracellular Zn concentration in Slc39a10fl/fl;LysM-Cre+ macrophages led to the stabilization of p53, which increased apoptosis upon LPS stimulation. Concomitant knockout of p53 largely rescued the phenotype of Slc39a10fl/fl;LysM-Cre+ mice. Finally, the phenotype in Slc39a10fl/fl;LysM-Cre+ mice was mimicked in wild-type mice using the Zn chelator TPEN and was reversed with Zn supplementation. Taken together, these results suggest that Slc39a10 plays a role in promoting the survival of macrophages through a Zn/p53-dependent axis in response to inflammatory stimuli.


Subject(s)
Cation Transport Proteins/physiology , Macrophages/physiology , Sepsis/immunology , Animals , Apoptosis/immunology , Cell Survival , Cytokines/blood , Humans , Lipopolysaccharides/pharmacology , Mice , Mice, Knockout , Sepsis/metabolism , Survivors , Tumor Suppressor Protein p53/metabolism , Zinc/metabolism
13.
PLoS Genet ; 13(7): e1006892, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28692648

ABSTRACT

Recent studies found that mutations in the human SLC30A10 gene, which encodes a manganese (Mn) efflux transporter, are associated with hypermanganesemia with dystonia, polycythemia, and cirrhosis (HMDPC). However, the relationship between Mn metabolism and HMDPC is poorly understood, and no specific treatments are available for this disorder. Here, we generated two zebrafish slc30a10 mutant lines using the CRISPR/Cas9 system. Compared to wild-type animals, mutant adult animals developed significantly higher systemic Mn levels, and Mn accumulated in the brain and liver of mutant embryos in response to exogenous Mn. Interestingly, slc30a10 mutants developed neurological deficits in adulthood, as well as environmental Mn-induced manganism in the embryonic stage; moreover, mutant animals had impaired dopaminergic and GABAergic signaling. Finally, mutant animals developed steatosis, liver fibrosis, and polycythemia accompanied by increased epo expression. This phenotype was rescued partially by EDTA- CaNa2 chelation therapy and iron supplementation. Interestingly, prior to the onset of slc30a10 expression, expressing ATP2C1 (ATPase secretory pathway Ca2+ transporting 1) protected mutant embryos from Mn exposure, suggesting a compensatory role for Atp2c1 in the absence of Slc30a10. Notably, expressing either wild-type or mutant forms of SLC30A10 was sufficient to inhibit the effect of ATP2C1 in response to Mn challenge in both zebrafish embryos and HeLa cells. These findings suggest that either activating ATP2C1 or restoring the Mn-induced trafficking of ATP2C1 can reduce Mn accumulation, providing a possible target for treating HMDPC.


Subject(s)
Calcium-Transporting ATPases/genetics , Cation Transport Proteins/genetics , Homeostasis/genetics , Manganese/metabolism , Metabolic Diseases/genetics , Animals , Brain/metabolism , Brain/pathology , CRISPR-Cas Systems , Cation Transport Proteins/deficiency , Genotype , HeLa Cells , Humans , Metabolic Diseases/metabolism , Metabolic Diseases/pathology , Mutation , Zebrafish/genetics , Zinc Transporter 8
14.
Zhejiang Da Xue Xue Bao Yi Xue Ban ; 49(1): 44-57, 2020 05 25.
Article in Zh | MEDLINE | ID: mdl-32621416

ABSTRACT

Recently, ferroptosis, an iron-dependent novel type of cell death, has been characterized as an excessive accumulation of lipid peroxides and reactive oxygen species. Emerging studies demonstrate that ferroptosis not only plays an important role in the pathogenesis and progression of chronic diseases, but also functions differently in the different disease context. Notably, it is shown that activation of ferroptosis could potently inhibit tumor growth and increase sensitivity to chemotherapy and immunotherapy in various cancer settings. As a result, the development of more efficacious ferroptosis agonists remains the mainstay of ferroptosis-targeting strategy for cancer therapeutics. By contrast, in non-cancerous chronic diseases, including cardiovascular & cerebrovascular diseases and neurodegenerative diseases, ferroptosis functions as a risk factor to promote these diseases progression through triggering or accelerating tissue injury. As a matter of fact, blocking ferroptosis has been demonstrated to effectively prevent ischemia-reperfusion heart disease in preclinical animal models. Therefore, it is a promising field to develope potent ferroptosis inhibitors for preventing and treating cardiovascular & cerebrovascular diseases and neurodegenerative diseases. In this article, we summarize the most recent progress on ferroptosis in chronic diseases, and draw attention to the possible clinical impact of this recently emerged ferroptosis modalities.


Subject(s)
Chronic Disease , Ferroptosis , Iron , Animals , Ferroptosis/physiology , Iron/metabolism , Reactive Oxygen Species
15.
Zhejiang Da Xue Xue Bao Yi Xue Ban ; 49(1): 58-70, 2020 05 25.
Article in Zh | MEDLINE | ID: mdl-32621410

ABSTRACT

Iron homeostasis plays an important role for the maintenance of human health. It is known that iron metabolism is tightly regulated by several key genes, including divalent metal transport-1(DMT1), transferrin receptor 1(TFR1), transferrin receptor 2(TFR2), ferroportin(FPN), hepcidin(HAMP), hemojuvelin(HJV) and Ferritin H. Recently, it is reported that DNA methylation, histone acetylation, and microRNA (miRNA) epigenetically regulated iron homeostasis. Among these epigenetic regulators, DNA hypermethylation of the promoter region of FPN, TFR2, HAMP, HJV and bone morphogenetic protein 6 (BMP6) genes result in inhibitory effect on the expression of these iron-related gene. In addition, histone deacetylase (HADC) suppresses HAMP gene expression. On the contrary, HADC inhibitor upregulates HAMP gene expression. Additional reports showed that miRNA can also modulate iron absorption, transport, storage and utilization via downregulation of DMT1, FPN, TFR1, TFR2, Ferritin H and other genes. It is noteworthy that some key epigenetic regulatory enzymes, such as DNA demethylase TET2 and histone lysine demethylase JmjC KDMs, require iron for the enzymatic activities. In this review, we summarize the recent progress of DNA methylation, histone acetylation and miRNA in regulating iron metabolism and also discuss the future research directions.


Subject(s)
Epigenesis, Genetic , Homeostasis , Iron , Gene Expression Regulation/genetics , Humans , Iron/metabolism , Receptors, Transferrin
16.
J Cell Mol Med ; 22(6): 3035-3044, 2018 06.
Article in English | MEDLINE | ID: mdl-29575577

ABSTRACT

To maintain iron homoeostasis, the iron regulatory hormone hepcidin is tightly controlled by BMP-Smad signalling pathway, but the physiological role of Smad7 in hepcidin regulation remains elusive. We generated and characterized hepatocyte-specific Smad7 knockout mice (Smad7Alb/Alb ), which showed decreased serum iron, tissue iron, haemoglobin concentration, up-regulated hepcidin and increased phosphor-Smad1/5/8 levels in both isolated primary hepatocytes and liver tissues. Increased levels of hepcidin lead to reduced expression of intestinal ferroportin and mild iron deficiency anaemia. Interestingly, we found no difference in hepcidin expression or phosphor-Smad1/5/8 levels between iron-challenged Smad7Alb/Alb and Smad7flox/flox , suggesting other factors assume the role of iron-induced hepcidin regulation in Smad7 deletion. We performed RNA-seq to identify differentially expressed genes in the liver. Significantly up-regulated genes were then mapped to pathways, revealing TGF-ß signalling as one of the most relevant pathways, including the up-regulated genes Smad6, Bambi and Fst (Follistatin). We found that Smad6 and Bambi-but not Follistatin-are controlled by the iron-BMP-Smad pathway. Overexpressing Smad6, Bambi or Follistatin in cells significantly reduced hepcidin expression. Smad7 functions as a key regulator of iron homoeostasis by negatively controlling hepcidin expression, and Smad6 and Smad7 have non-redundant roles. Smad6, Bambi and Follistatin serve as additional inhibitors of hepcidin in the liver.


Subject(s)
Hepcidins/genetics , Liver/metabolism , Smad7 Protein/genetics , Transforming Growth Factor beta/genetics , Animals , Follistatin/genetics , Gene Expression Regulation/genetics , Hemoglobins/genetics , Hemoglobins/metabolism , Hepatocytes/metabolism , Hepcidins/metabolism , Humans , Iron/metabolism , Membrane Proteins/genetics , Mice , Mice, Knockout , Sequence Analysis, RNA , Signal Transduction , Smad6 Protein/genetics , Smad7 Protein/deficiency
17.
J Cell Physiol ; 233(9): 7268-7278, 2018 09.
Article in English | MEDLINE | ID: mdl-29600572

ABSTRACT

Hemochromatosis is prevalent and often associated with high rates of morbidity and mortality worldwide. The safe alternative iron-reducing approaches are urgently needed in order to better control iron overload. Our unbiased vitamin screen for modulators of hepcidin, a master iron regulatory hormone, identifies adenine (vitamin B4) as a potent hepcidin agonist. Adenine significantly induced hepcidin mRNA level and promoter activity activation in human cell lines, possibly through BMP/SMAD pathway. Further studies in mice validated the effect of adenine on hepcidin upregulation. Consistently, adenine dietary supplement in mice led to an increase of hepatic hepcidin expression compared with normal diet-fed mice via BMP/SMAD pathway. Notably, adenine-rich diet significantly ameliorated iron overload accompanied by the enhanced hepcidin expression in both high iron-fed mice and in Hfe-/- mice, a murine model of hereditary hemochromatosis. To further validate this finding, we selected pharmacological inhibitors against BMP (LDN193189). We found LDN193189 strongly blocked the hepcidin induction by adenine. Moreover, we uncovered an essential role of cAMP/PKA-dependent axis in triggering adenine-induced hepcidin expression in primary hepatocytes by using 8 br cAMP, a cAMP analog, and H89, a potent inhibitor for PKA signaling. These findings suggest a potential therapeutic role of adenine for hereditary hemochromatosis.


Subject(s)
Adenine/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Hepcidins/metabolism , Iron Overload/metabolism , Liver/metabolism , Animals , Bone Morphogenetic Proteins/metabolism , Cell Line, Tumor , Diet , Disease Models, Animal , Hemochromatosis Protein/deficiency , Hemochromatosis Protein/metabolism , Humans , Iron/metabolism , Male , Mice, Inbred C57BL , Models, Biological , Signal Transduction , Smad Proteins/metabolism , Time Factors , Up-Regulation/drug effects , Vitamins/metabolism
19.
Int J Cancer ; 143(7): 1595-1603, 2018 10 01.
Article in English | MEDLINE | ID: mdl-29696630

ABSTRACT

Numerous studies have suggested that excess body weight is associated with increased cancer risk. To examine this putative association, we performed a systematic review and quantitative meta-analysis of cohort studies reporting body mass index (BMI) and the risk of 23 cancer types. PubMed, Embase, and Web of Science were searched for cohort studies, yielding 325 articles with 1,525,052 cases. Strong positive associations were observed between BMI and endometrial cancer (RR: 1.48), esophageal adenocarcinoma (RR: 1.45), and kidney cancer (RR: 1.20); weaker associations (RR < 1.20) were also found for several other cancer types. Interestingly, we found significant inverse associations between BMI and oral cavity (RR: 0.93), lung (RR: 0.91), premenopausal breast (RR: 0.95), and localized prostate (RR: 0.97) cancers. A male-specific association was found for colorectal cancer (p = 0.023), and a female-specific association was found for cancer in brain (p = 0.025) or kidney (p = 0.035). With respect to geography, the strongest positive association was found for total cancer in North America (p = 0.038). This comprehensive meta-analysis provides epidemiological evidence supporting the association between BMI and cancer risk. These findings can be used to drive public policies and to help guide personalized medicine in order to better manage body weight, thereby reducing the risk of developing obesity-related cancer.


Subject(s)
Body Mass Index , Body Weight/physiology , Neoplasms/epidemiology , Adult , Female , Humans , Life Style , Male , North America/epidemiology , Obesity/epidemiology , Risk Factors
20.
Hepatology ; 66(2): 449-465, 2017 08.
Article in English | MEDLINE | ID: mdl-28195347

ABSTRACT

Ferroptosis is a recently identified iron-dependent form of nonapoptotic cell death implicated in brain, kidney, and heart pathology. However, the biological roles of iron and iron metabolism in ferroptosis remain poorly understood. Here, we studied the functional role of iron and iron metabolism in the pathogenesis of ferroptosis. We found that ferric citrate potently induces ferroptosis in murine primary hepatocytes and bone marrow-derived macrophages. Next, we screened for ferroptosis in mice fed a high-iron diet and in mouse models of hereditary hemochromatosis with iron overload. We found that ferroptosis occurred in mice fed a high-iron diet and in two knockout mouse lines that develop severe iron overload (Hjv-/- and Smad4Alb/Alb mice) but not in a third line that develops only mild iron overload (Hfe-/- mice). Moreover, we found that iron overload-induced liver damage was rescued by the ferroptosis inhibitor ferrostatin-1. To identify the genes involved in iron-induced ferroptosis, we performed microarray analyses of iron-treated bone marrow-derived macrophages. Interestingly, solute carrier family 7, member 11 (Slc7a11), a known ferroptosis-related gene, was significantly up-regulated in iron-treated cells compared with untreated cells. However, genetically deleting Slc7a11 expression was not sufficient to induce ferroptosis in mice. Next, we studied iron-treated hepatocytes and bone marrow-derived macrophages isolated from Slc7a11-/- mice fed a high-iron diet. CONCLUSION: We found that iron treatment induced ferroptosis in Slc7a11-/- cells, indicating that deleting Slc7a11 facilitates the onset of ferroptosis specifically under high-iron conditions; these results provide compelling evidence that iron plays a key role in triggering Slc7a11-mediated ferroptosis and suggest that ferroptosis may be a promising target for treating hemochromatosis-related tissue damage. (Hepatology 2017;66:449-465).


Subject(s)
Apoptosis/physiology , Ferric Compounds/pharmacology , Hemochromatosis/pathology , Iron/metabolism , Receptors, Somatostatin/metabolism , Analysis of Variance , Animals , Blotting, Western/methods , Cell Survival , Cells, Cultured , Disease Models, Animal , Hepatocytes/metabolism , Iron Chelating Agents/pharmacology , Lipid Peroxidation , Macrophages/metabolism , Male , Mice , Mice, Knockout , Oxidative Stress , Random Allocation , Real-Time Polymerase Chain Reaction/methods , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL