Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Arterioscler Thromb Vasc Biol ; 43(8): 1494-1509, 2023 08.
Article in English | MEDLINE | ID: mdl-37381987

ABSTRACT

BACKGROUND: MAGT1 (magnesium transporter 1) is a subunit of the oligosaccharide protein complex with thiol-disulfide oxidoreductase activity, supporting the process of N-glycosylation. MAGT1 deficiency was detected in human patients with X-linked immunodeficiency with magnesium defect syndrome and congenital disorders of glycosylation, resulting in decreased cation responses in lymphocytes, thereby inhibiting the immune response against viral infections. Curative hematopoietic stem cell transplantation of patients with X-linked immunodeficiency with magnesium defect causes fatal bleeding and thrombotic complications. METHODS: We studied the role of MAGT1 deficiency in platelet function in relation to arterial thrombosis and hemostasis using several in vitro experimental settings and in vivo models of arterial thrombosis and transient middle cerebral artery occlusion model of ischemic stroke. RESULTS: MAGT1-deficient mice (Magt1-/y) displayed accelerated occlusive arterial thrombus formation in vivo, a shortened bleeding time, and profound brain damage upon focal cerebral ischemia. These defects resulted in increased calcium influx and enhanced second wave mediator release, which further reinforced platelet reactivity and aggregation responses. Supplementation of MgCl2 or pharmacological blockade of TRPC6 (transient receptor potential cation channel, subfamily C, member 6) channel, but not inhibition of store-operated calcium entry, normalized the aggregation responses of Magt1-/y platelets to the control level. GP (glycoprotein) VI activation of Magt1-/y platelets resulted in hyperphosphorylation of Syk (spleen tyrosine kinase), LAT (linker for activation of T cells), and PLC (phospholipase C) γ2, whereas the inhibitory loop regulated by PKC (protein kinase C) was impaired. A hyperaggregation response to the GPVI agonist was confirmed in human platelets isolated from a MAGT1-deficient (X-linked immunodeficiency with magnesium defect) patient. Haploinsufficiency of TRPC6 in Magt1-/y mice could normalize GPVI signaling, platelet aggregation, and thrombus formation in vivo. CONCLUSIONS: These results suggest that MAGT1 and TRPC6 are functionally linked. Therefore, deficiency or impaired functionality of MAGT1 could be a potential risk factor for arterial thrombosis and stroke.


Subject(s)
Cation Transport Proteins , Homeostasis , Infarction, Middle Cerebral Artery , Ischemic Stroke , Thrombosis , Animals , Humans , Mice , Blood Platelets/metabolism , Calcium/metabolism , Cations/metabolism , Ischemic Stroke/genetics , Ischemic Stroke/complications , Ischemic Stroke/metabolism , Magnesium/metabolism , Platelet Activation , Platelet Aggregation , Platelet Membrane Glycoproteins/metabolism , Thrombosis/genetics , Thrombosis/metabolism , TRPC6 Cation Channel/metabolism , Cation Transport Proteins/deficiency
2.
Cell Death Dis ; 13(1): 11, 2021 12 20.
Article in English | MEDLINE | ID: mdl-34930890

ABSTRACT

TMEM165 deficiency leads to skeletal disorder characterized by major skeletal dysplasia and pronounced dwarfism. However, the molecular mechanisms involved have not been fully understood. Here, we uncover that TMEM165 deficiency impairs the synthesis of proteoglycans by producing a blockage in the elongation of chondroitin-and heparan-sulfate glycosaminoglycan chains leading to the synthesis of proteoglycans with shorter glycosaminoglycan chains. We demonstrated that the blockage in elongation of glycosaminoglycan chains is not due to defect in the Golgi elongating enzymes but rather to availability of the co-factor Mn2+. Supplementation of cell with Mn2+ rescue the elongation process, confirming a role of TMEM165 in Mn2+ Golgi homeostasis. Additionally, we showed that TMEM165 deficiency functionally impairs TGFß and BMP signaling pathways in chondrocytes and in fibroblast cells of TMEM165 deficient patients. Finally, we found that loss of TMEM165 impairs chondrogenic differentiation by accelerating the timing of Ihh expression and promoting early chondrocyte maturation and hypertrophy. Collectively, our results indicate that TMEM165 plays an important role in proteoglycan synthesis and underline the critical role of glycosaminoglycan chains structure in the regulation of chondrogenesis. Our data also suggest that Mn2+ supplementation may be a promising therapeutic strategy in the treatment of TMEM165 deficient patients.


Subject(s)
Antiporters/deficiency , Antiporters/metabolism , Cation Transport Proteins/deficiency , Cation Transport Proteins/metabolism , Cell Differentiation/genetics , Chondrocytes/metabolism , Chondrocytes/pathology , Chondroitin Sulfates/biosynthesis , Dwarfism/metabolism , Heparan Sulfate Proteoglycans/biosynthesis , Signal Transduction/genetics , Animals , Antiporters/genetics , Case-Control Studies , Cation Transport Proteins/genetics , Cell Line, Tumor , Chondrogenesis/genetics , Dwarfism/pathology , Fibroblasts/metabolism , Gene Knockout Techniques/methods , Glycosylation , HEK293 Cells , Humans , Hypertrophy/metabolism , Mice , Transfection
3.
FASEB J ; 33(12): 14625-14635, 2019 12.
Article in English | MEDLINE | ID: mdl-31690120

ABSTRACT

Ferroportin 1 (FPN1) is a major facilitator superfamily transporter that is essential for proper maintenance of human iron homeostasis at the systemic and cellular level. FPN1 dysfunction leads to the progressive accumulation of iron in reticuloendothelial cells, causing hemochromatosis type 4A (or ferroportin disease), an autosomal dominant disorder that displays large phenotypic heterogeneity. Although crystal structures have unveiled the outward- and inward-facing conformations of the bacterial homolog Bdellovibrio bacteriovorus Fpn (or Bd2019) and calcium has recently been identified as an essential cofactor, our molecular understanding of the iron transport mechanism remains incomplete. Here, we used a combination of molecular modeling, molecular dynamics simulations, and Ala site-directed mutagenesis, followed by complementary in vitro functional analyses, to explore the structural architecture of the human FPN1 intracellular gate. We reveal an interdomain network that involves 5 key amino acids and is likely very important for stability of the iron exporter facing the extracellular milieu. We also identify inter- and intradomain interactions that rely on the 2 Asp84 and Asn174 critical residues and do not exist in the bacterial homolog. These interactions are thought to play an important role in the modulation of conformational changes during the transport cycle. We interpret these results in the context of hemochromatosis type 4A, reinforcing the idea that different categories of loss-of-function mutations exist. Our findings provide an unprecedented view of the human FPN1 outward-facing structure and the particular function of the so-called "gating residues" in the mechanism of iron export.-Guellec, J., Elbahnsi, A., Le Tertre, M., Uguen, K., Gourlaouen, I., Férec, C., Ka, C., Callebaut, I., Le Gac, G. Molecular model of the ferroportin intracellular gate and implications for the human iron transport cycle and hemochromatosis type 4A.


Subject(s)
Cation Transport Proteins/deficiency , Hemochromatosis/genetics , Molecular Dynamics Simulation , Mutation , Cation Transport Proteins/chemistry , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , HEK293 Cells , Humans , Iron/metabolism , Protein Domains
4.
Cell Death Differ ; 26(6): 1138-1155, 2019 06.
Article in English | MEDLINE | ID: mdl-30237509

ABSTRACT

Regulation of cell and tissue homeostasis by programmed cell death is a fundamental process with wide physiological and pathological implications. The advent of scalable somatic cell genetic technologies creates the opportunity to functionally map such essential pathways, thereby identifying potential disease-relevant components. We investigated the genetic basis underlying necroptotic cell death by performing a complementary set of loss-of-function and gain-of-function genetic screens. To this end, we established FADD-deficient haploid human KBM7 cells, which specifically and efficiently undergo necroptosis after a single treatment with either TNFα or the SMAC mimetic compound birinapant. A series of unbiased gene-trap screens identified key signaling mediators, such as TNFR1, RIPK1, RIPK3, and MLKL. Among the novel components, we focused on the zinc transporter SLC39A7, whose knock-out led to necroptosis resistance by affecting TNF receptor surface levels. Orthogonal, solute carrier (SLC)-focused CRISPR/Cas9-based genetic screens revealed the exquisite specificity of SLC39A7, among ~400 SLC genes, for TNFR1-mediated and FAS-mediated but not TRAIL-R1-mediated responses. Mechanistically, we demonstrate that loss of SLC39A7 resulted in augmented ER stress and impaired receptor trafficking, thereby globally affecting downstream signaling. The newly established cellular model also allowed genome-wide gain-of-function screening for genes conferring resistance to necroptosis via the CRISPR/Cas9-based synergistic activation mediator approach. Among these, we found cIAP1 and cIAP2, and characterized the role of TNIP1, which prevented pathway activation in a ubiquitin-binding dependent manner. Altogether, the gain-of-function and loss-of-function screens described here provide a global genetic chart of the molecular factors involved in necroptosis and death receptor signaling, prompting further investigation of their individual contribution and potential role in pathological conditions.


Subject(s)
Cation Transport Proteins/genetics , Chromosome Mapping , Necroptosis/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , CRISPR-Cas Systems/genetics , Cation Transport Proteins/deficiency , Cation Transport Proteins/metabolism , Cell Death , Cell Line , Cell Survival , HEK293 Cells , Humans , Receptors, Tumor Necrosis Factor, Type I/genetics , Tumor Necrosis Factor-alpha/metabolism
5.
Am J Physiol Gastrointest Liver Physiol ; 315(4): G569-G579, 2018 10 01.
Article in English | MEDLINE | ID: mdl-29927321

ABSTRACT

Zrt/Irt-like protein 8 (ZIP8) (encoded by Slc39a8) is a multifunctional membrane transporter that influxes essential metal cations Zn2+, Mn2+, Fe2+, and nonmetal inorganic selenite (HSeO3-). Physiological roles of ZIP8 in different cell types and tissues remain to be elucidated. We aimed to investigate ZIP8 functions in liver. Two mouse models were used in this study: 1) 13- to 21-mo-old Slc39a8(+/neo) hypomorphs having diminished ZIP8 levels and 2) a liver-specific ZIP8 acute knockdown mouse (Ad-shZip8). Histology, immunohistochemistry, and Western blotting were used to investigate ZIP8-deficiency effects on hepatic injury, inflammatory changes, and oxidative stress. Selenium (Se) and zinc (Zn) were quantified in tissues by inductively coupled plasma-mass spectrophotometry. We found that ZIP8 is required to maintain normal liver function; moderate or acute decreases in ZIP8 activity resulted in hepatic pathology. Spontaneous liver neoplastic nodules appeared in ~50% of Slc39a8(+/neo) between 13 and 21 mo of age, exhibiting features of inflammation, fibrosis, and liver injury. In Ad-shZip8 mice, significant hepatomegaly was observed; histology showed ZIP8 deficiency was associated with hepatocyte injury, inflammation, and proliferation. Significant decreases in Se, but not Zn, were found in Ad-shZip8 liver. Consistent with this Se deficit, liver expression of selenoproteins glutathione peroxidases 1 and 2 was downregulated, along with decreases in antioxidant superoxide dismutases 1 and 2, consistent with increased oxidative stress. Thus, ZIP8 plays an important role in maintaining normal hepatic function, likely through regulating Se homeostasis and redox balance. Hepatic ZIP8 deficiency is associated with liver pathology, including oxidative stress, inflammation, proliferation, and hepatocellular injury. NEW & NOTEWORTHY Zrt/Irt-like protein 8 (ZIP8) is a multifunctional membrane transporter that facilitates biometal and mineral uptake. The role of ZIP8 in liver physiology has not been previously investigated. Liu et al. discovered unique ZIP8 functions, i.e., regulation of hepatic selenium content and association of ZIP8 deficiency in mouse liver with liver defects.


Subject(s)
Cation Transport Proteins/deficiency , Hepatocytes/metabolism , Homeostasis , Liver Neoplasms/metabolism , Selenium/metabolism , Animals , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cell Line , Cells, Cultured , Glutathione Peroxidase/metabolism , Hepatocytes/pathology , Male , Mice , Mice, Inbred C57BL , Oxidative Stress , Superoxide Dismutase/metabolism , Zinc/metabolism
6.
Genet Med ; 20(2): 259-268, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28749473

ABSTRACT

PurposeSLC39A8 deficiency is a severe inborn error of metabolism that is caused by impaired function of manganese metabolism in humans. Mutations in SLC39A8 lead to impaired function of the manganese transporter ZIP8 and thus manganese deficiency. Due to the important role of Mn2+ as a cofactor for a variety of enzymes, the resulting phenotype is complex and severe. The manganese-dependence of ß-1,4-galactosyltransferases leads to secondary hypoglycosylation, making SLC39A8 deficiency both a disorder of trace element metabolism and a congenital disorder of glycosylation. Some hypoglycosylation disorders have previously been treated with galactose administration. The development of an effective treatment of the disorder by high-dose manganese substitution aims at correcting biochemical, and hopefully, clinical abnormalities.MethodsTwo SCL39A8 deficient patients were treated with 15 and 20 mg MnSO4/kg bodyweight per day. Glycosylation and blood manganese were monitored closely. In addition, magnetic resonance imaging was performed to detect potential toxic effects of manganese.ResultsAll measured enzyme dysfunctions resolved completely and considerable clinical improvement regarding motor abilities, hearing, and other neurological manifestations was observed.ConclusionHigh-dose manganese substitution was effective in two patients with SLC39A8 deficiency. Close therapy monitoring by glycosylation assays and blood manganese measurements is necessary to prevent manganese toxicity.


Subject(s)
Cation Transport Proteins/deficiency , Genetic Association Studies , Genetic Predisposition to Disease , Alleles , Biomarkers , Dietary Supplements , Electroencephalography , Female , Genetic Association Studies/methods , Glycosylation/drug effects , Humans , Magnetic Resonance Imaging , Manganese/administration & dosage , Manganese/adverse effects , Manganese/therapeutic use , Mutation , Phenotype , Treatment Outcome
7.
Neuromolecular Med ; 19(2-3): 375-386, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28695462

ABSTRACT

Exposure to divalent metals such as iron and manganese is thought to increase the risk for Parkinson's disease (PD). Under normal circumstances, cellular iron and manganese uptake is regulated by the divalent metal transporter 1 (DMT1). Accordingly, alterations in DMT1 levels may underlie the abnormal accumulation of metal ions and thereby disease pathogenesis. Here, we have generated transgenic mice overexpressing DMT1 under the direction of a mouse prion promoter and demonstrated its robust expression in several regions of the brain. When fed with iron-supplemented diet, DMT1-expressing mice exhibit rather selective accumulation of iron in the substantia nigra, which is the principal region affected in human PD cases, but otherwise appear normal. Alongside this, the expression of Parkin is also enhanced, likely as a neuroprotective response, which may explain the lack of phenotype in these mice. When DMT1 is overexpressed against a Parkin null background, the double-mutant mice similarly resisted a disease phenotype even when fed with iron- or manganese-supplemented diet. However, these mice exhibit greater vulnerability toward 6-hydroxydopamine-induced neurotoxicity. Taken together, our results suggest that iron accumulation alone is not sufficient to cause neurodegeneration and that multiple hits are required to promote PD.


Subject(s)
Cation Transport Proteins/physiology , Iron/metabolism , Parkinsonian Disorders/metabolism , Substantia Nigra/metabolism , Ubiquitin-Protein Ligases/biosynthesis , Animals , Cation Transport Proteins/deficiency , Cation Transport Proteins/genetics , Gene Expression Regulation , Iron/toxicity , Macaca fascicularis/genetics , Manganese/toxicity , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oxidopamine/toxicity , Parkinsonian Disorders/chemically induced , Prions/genetics , Promoter Regions, Genetic , Recombinant Proteins/metabolism , Rotarod Performance Test , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics
8.
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
9.
J Inherit Metab Dis ; 40(4): 519-529, 2017 07.
Article in English | MEDLINE | ID: mdl-28303424

ABSTRACT

Elements with a biological role include six trace transition metals: manganese, iron, cobalt, copper, zinc and molybdenum. Transition metals participate in group transfer reactions such as glycosylation and phosphorylation and those that can transfer an electron by alternating between two redox states such as iron (3+/2+) and copper (2+/1+) are also very important in biological redox reactions including the reduction of molecular oxygen and the transport of oxygen. However, these trace metals are also potentially toxic, generating reactive oxygen species through Fenton chemistry. Recently, a role of trace metals in host defence ("nutritional immunity") has been recognized. The host can deprive the pathogen of a trace metal or poison it with a toxic concentration. Disorders leading to low concentrations of a trace metal can often be treated by supplementing that metal; disorders leading to excessively high concentrations can often be treated with chelating agents such as penicillamine and disodium calcium edetate. This update will address: i) the manganese/zinc transporters (because two new treatable disorders were described in 2016 - SLC39A8 deficiency and SLC39A14 deficiency); ii) copper transporter disorders because we need to improve the treatment of patients with neurological symptoms due to Wilson's disease; and iii) iron homeostasis because recent progress in research into the metabolism of iron and its regulation helps us better understand several inborn errors affecting these pathways.


Subject(s)
Metabolism, Inborn Errors/genetics , Metals/metabolism , Trace Elements/metabolism , Animals , Cation Transport Proteins/deficiency , Cation Transport Proteins/genetics , Chelating Agents , Copper/metabolism , Electrons , Glycosylation , Hepatolenticular Degeneration/therapy , Homeostasis , Humans , Iron/metabolism , Manganese/chemistry , Mice , Mutation , Neurotoxins/chemistry , Oxidation-Reduction , Phosphorylation , Zinc
10.
Birth Defects Res ; 109(2): 81-91, 2017 01 30.
Article in English | MEDLINE | ID: mdl-28008752

ABSTRACT

BACKGROUND: Periconception maternal nutrition and folate in particular are important factors influencing the incidence of neural tube defects (NTDs). Many but not all NTDs are prevented by folic acid supplementation and there is a pressing need for additional strategies to prevent these birth defects. Other micronutrients such as iron are potential candidates, yet a clear role for iron deficiency in contributing to NTDs is lacking. Our previous studies with the flatiron (ffe) mouse model of Ferroportin1 (Fpn1) deficiency suggest that iron is required for neural tube closure and forebrain development raising the possibility that iron supplementation could prevent NTDs. METHODS: We determined the effect of periconception iron and/or folic acid supplementation on the penetrance of NTDs in the Fpn1ffe mouse model. Concurrently, measurements of folate and iron were made to ensure supplementation had the intended effects. RESULTS: High levels of iron supplementation significantly reduced the incidence of NTDs in Fpn1ffe mutants. Fpn1 deficiency resulted in reduced folate levels in both pregnant dams and embryos. Yet folic acid supplementation did not prevent NTDs in the Fpn1ffe model. Similarly, forebrain truncations were rescued with iron. Surprisingly, the high levels of iron supplementation used in this study caused folate deficiency in wild-type dams and embryos. CONCLUSION: Our results demonstrate that iron supplementation can prevent NTDs and forebrain truncations in the Fpn1ffe model. Surprisingly, high levels of iron supplementation and iron overload can cause folate deficiency. If iron is essential for neural tube closure, it is possible that iron deficiency might contribute to NTDs. Birth Defects Research 109:81-91, 2017. © 2016 The Authors Birth Defects Research Published by Wiley Periodicals, Inc.


Subject(s)
Cation Transport Proteins/genetics , Dietary Supplements , Folic Acid Deficiency/diet therapy , Folic Acid/administration & dosage , Iron/administration & dosage , Neural Tube Defects/prevention & control , Animals , Cation Transport Proteins/deficiency , Crosses, Genetic , Disease Models, Animal , Embryo, Mammalian , Female , Folic Acid Deficiency/genetics , Folic Acid Deficiency/metabolism , Folic Acid Deficiency/pathology , Gene Deletion , Humans , Maternal Nutritional Physiological Phenomena , Mice , Mice, Transgenic , Neural Tube/abnormalities , Neural Tube/drug effects , Neural Tube/metabolism , Neural Tube Defects/genetics , Neural Tube Defects/metabolism , Neural Tube Defects/pathology , Penetrance , Pregnancy , Prosencephalon/abnormalities , Prosencephalon/drug effects , Prosencephalon/metabolism
11.
Am J Hum Genet ; 97(6): 894-903, 2015 Dec 03.
Article in English | MEDLINE | ID: mdl-26637979

ABSTRACT

SLC39A8 is a membrane transporter responsible for manganese uptake into the cell. Via whole-exome sequencing, we studied a child that presented with cranial asymmetry, severe infantile spasms with hypsarrhythmia, and dysproportionate dwarfism. Analysis of transferrin glycosylation revealed severe dysglycosylation corresponding to a type II congenital disorder of glycosylation (CDG) and the blood manganese levels were below the detection limit. The variants c.112G>C (p.Gly38Arg) and c.1019T>A (p.Ile340Asn) were identified in SLC39A8. A second individual with the variants c.97G>A (p.Val33Met) and c.1004G>C (p.Ser335Thr) on the paternal allele and c.610G>T (p.Gly204Cys) on the maternal allele was identified among a group of unresolved case subjects with CDG. These data demonstrate that variants in SLC39A8 impair the function of manganese-dependent enzymes, most notably ß-1,4-galactosyltransferase, a Golgi enzyme essential for biosynthesis of the carbohydrate part of glycoproteins. Impaired galactosylation leads to a severe disorder with deformed skull, severe seizures, short limbs, profound psychomotor retardation, and hearing loss. Oral galactose supplementation is a treatment option and results in complete normalization of glycosylation. SLC39A8 deficiency links a trace element deficiency with inherited glycosylation disorders.


Subject(s)
Cation Transport Proteins/genetics , Congenital Disorders of Glycosylation/genetics , Dwarfism/genetics , Manganese/blood , Spasms, Infantile/genetics , Amino Acid Sequence , Carbohydrate Sequence , Cation Transport Proteins/deficiency , Cations, Divalent , Congenital Disorders of Glycosylation/blood , Congenital Disorders of Glycosylation/complications , Congenital Disorders of Glycosylation/diet therapy , Dwarfism/blood , Dwarfism/complications , Dwarfism/diet therapy , Female , Galactose/therapeutic use , Gene Expression , High-Throughput Nucleotide Sequencing , Humans , Infant , Ion Transport , Manganese/deficiency , Molecular Sequence Data , Mutation , Pedigree , Sequence Alignment , Spasms, Infantile/blood , Spasms, Infantile/complications , Spasms, Infantile/diet therapy
12.
Am J Physiol Gastrointest Liver Physiol ; 309(9): G768-78, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26272258

ABSTRACT

Integrity of the immune system is particularly dependent on the availability of zinc. Recent data suggest that zinc is involved in the development of sepsis, a life-threatening systemic inflammation with high death rates, but with limited therapeutic options. Altered cell zinc transport mechanisms could contribute to the inflammatory effects of sepsis. Zip14, a zinc importer induced by proinflammatory stimuli, could influence zinc metabolism during sepsis and serve as a target for therapy. Using cecal ligation-and-puncture (CLP) to model polymicrobial sepsis, we narrowed the function of ZIP14 to regulation of zinc homeostasis in hepatocytes, while hepatic leukocytes were mostly responsible for driving inflammation, as shown by higher expression of IL-1ß, TNFα, S100A8, and matrix metalloproteinase-8. Using Zip14 knockout (KO) mice as a novel approach, we found that ablation of Zip14 produced a delay in development of leukocytosis, prevented zinc accumulation in the liver, altered the kinetics of hypozincemia, and drastically increased serum IL-6, TNFα, and IL-10 concentrations following CLP. Hence, this model revealed that the zinc transporter ZIP14 is a component of the pathway for zinc redistribution that contributes to zinc dyshomeostasis during polymicrobial sepsis. In contrast, using the identical CLP model, we found that supplemental dietary zinc reduced the severity of sepsis, as shown by amelioration of cytokines, calprotectins, and blood bacterial loads. We conclude that the zinc transporter ZIP14 influences aspects of the pathophysiology of nonlethal polymicrobial murine sepsis induced by CLP through zinc delivery. The results are promising for the use of zinc and its transporters as targets for future sepsis therapy.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Cation Transport Proteins/metabolism , Dietary Supplements , Liver/drug effects , Sepsis/prevention & control , Zinc/pharmacology , Animals , Bacterial Load , Biomarkers/blood , Cation Transport Proteins/deficiency , Cation Transport Proteins/genetics , Cecum/microbiology , Cecum/surgery , Cytokines/blood , Disease Models, Animal , Disease Progression , Female , Hepatocytes/drug effects , Hepatocytes/immunology , Hepatocytes/metabolism , Homeostasis , Inflammation Mediators/blood , Leukocytes/drug effects , Leukocytes/immunology , Leukocytes/metabolism , Ligation , Liver/immunology , Liver/metabolism , Liver/microbiology , Male , Mice, Inbred C57BL , Mice, Knockout , Punctures , Sepsis/blood , Sepsis/genetics , Sepsis/immunology , Sepsis/microbiology , Severity of Illness Index , Time Factors , Zinc/metabolism
13.
Transplantation ; 99(11): 2317-24, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26018347

ABSTRACT

BACKGROUND: Liver transplantation (LT) is the only option of treatment for Wilson disease (WD) when chelation therapy fails, but it is limited due to the shortage of donor. Auxiliary partial orthotopic LT (APOLT) has been performed successfully in end-stage WD patients, which expands the donor pool. METHODS: Atp7bmice were used as experimental model of WD. Eight- and 20-week-old mice were used as different timepoints to perform APOLT. Serum copper, tissue copper, serum ceruloplasmin (CP), and liver histological examination were observed after operation. RESULTS: Hepatic and serum copper levels in Atp7b mice decreased after APOLT, and copper metabolism disorder of WD mice was relieved at both early and late stages. The progression of pathology in the native liver was delayed only when transplantation was performed at an early stage. CONCLUSIONS: Auxiliary partial orthotopic LT can significantly improve copper metabolism disorder in the Atp7b mice, and early transplantation may prevent the disease progression.


Subject(s)
Hepatolenticular Degeneration/surgery , Liver Transplantation/methods , Liver/surgery , Adenosine Triphosphatases/deficiency , Adenosine Triphosphatases/genetics , Animals , Biomarkers/blood , Cation Transport Proteins/deficiency , Cation Transport Proteins/genetics , Ceruloplasmin/metabolism , Copper/blood , Copper-Transporting ATPases , Disease Models, Animal , Disease Progression , Feasibility Studies , Hepatolenticular Degeneration/blood , Hepatolenticular Degeneration/genetics , Hepatolenticular Degeneration/pathology , Liver/metabolism , Liver/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Time Factors
14.
J Antimicrob Chemother ; 69(10): 2634-43, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25028451

ABSTRACT

BACKGROUND: Metronidazole is the most commonly used antimicrobial for Bacteroides fragilis infections and is recommended for prophylaxis of colorectal surgery. Metronidazole resistance is increasing and the mechanisms of resistance are not clear. METHODS: A transposon mutant library was generated in B. fragilis 638R (BF638R) to identify the genetic loci associated with resistance to metronidazole. RESULTS: Thirty-two independently isolated metronidazole-resistant mutants had a transposon insertion in BF638R_1421 that encodes the ferrous transport fusion protein (feoAB). Deletion of feoAB resulted in a 10-fold increased MIC of metronidazole for the strain. The metronidazole MIC for the feoAB mutant was similar to that for the parent strain when grown on media supplemented with excess iron, suggesting that the increase seen in the MIC of metronidazole was due to reduced cellular iron transport in the feoAB mutant. The furA gene repressed feoAB transcription in an iron-dependent manner and disruption of furA resulted in constitutive transcription of feoAB, regardless of whether or not iron was present. However, disruption of feoAB also diminished the capacity of BF638R to grow in a mouse intraperitoneal abscess model, suggesting that inorganic ferrous iron assimilation is essential for B. fragilis survival in vivo. CONCLUSIONS: Selection for feoAB mutations as a result of metronidazole treatment will disable the pathogenic potential of B. fragilis and could contribute to the clinical efficacy of metronidazole. While mutations in feoAB are probably not a direct cause of clinical resistance, this study provides a key insight into intracellular metronidazole activity and the link with intracellular iron homeostasis.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteroides fragilis/drug effects , Bacteroides fragilis/genetics , Cation Transport Proteins/deficiency , Drug Resistance, Bacterial/genetics , Metronidazole/pharmacology , Bacteroides fragilis/metabolism , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , DNA Transposable Elements , Ferrous Compounds/metabolism , Gene Deletion , Gene Expression , Gene Expression Profiling , Gene Expression Regulation, Bacterial , Gene Library , Gene Order , Genotype , Microbial Sensitivity Tests , Microbial Viability/genetics , Mutation , Transcription, Genetic , Transcriptome
15.
Cell Mol Life Sci ; 71(17): 3281-95, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24710731

ABSTRACT

Zinc transporters, the Zrt-, Irt-like protein (ZIP) family and the Zn transporter (ZnT) family transporters, are found in all aspects of life. Increasing evidence has clarified the molecular mechanism, in which both transporters play critical roles in cellular and physiological functions via mobilizing zinc across the cellular membrane. In the last decade, mutations in ZIP and ZnT transporter genes have been shown to be implicated in a number of inherited human diseases. Moreover, dysregulation of expression and activity of both transporters has been suggested to be involved in the pathogenesis and progression of chronic diseases including cancer, immunological impairment, and neurodegenerative diseases, although comprehensive understanding is far from complete. The diverse phenotypes of diseases related to ZIP and ZnT transporters reflect the multifarious biological functions of both transporters. The present review summarizes the current understanding of ZIP and ZnT transporter functions from the standpoint of human health and diseases. The study of zinc transporters is currently of great clinical interest.


Subject(s)
Cation Transport Proteins/physiology , Zinc/metabolism , Brain/metabolism , Cation Transport Proteins/classification , Cation Transport Proteins/deficiency , Cation Transport Proteins/genetics , Diet , Dietary Supplements , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/metabolism , Homeostasis , Humans , Immune System/metabolism , Intestinal Absorption , Models, Molecular , Neoplasms/metabolism , Neurodegenerative Diseases/metabolism , Polymorphism, Single Nucleotide , Zinc/deficiency , Zinc/pharmacokinetics
16.
FASEB J ; 28(8): 3671-8, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24776745

ABSTRACT

The ferritin core is composed of fine nanoparticulate Fe(3+) oxohydroxide, and we have developed a synthetic mimetic, nanoparticulate Fe(3+) polyoxohydroxide (nanoFe(3+)). The aim of this study was to determine how dietary iron derived in this fashion is absorbed in the duodenum. Following a 4 wk run-in on an Fe-deficient diet, mice with intestinal-specific disruption of the Fpn-1 gene (Fpn-KO), or littermate wild-type (WT) controls, were supplemented with Fe(2+) sulfate (FeSO4), nanoFe(3+), or no added Fe for a further 4 wk. A control group was Fe sufficient throughout. Direct intestinal absorption of nanoFe(3+) was investigated using isolated duodenal loops. Our data show that FeSO4 and nanoFe(3+) are equally bioavailable in WT mice, and at wk 8 the mean ± SEM hemoglobin increase was 18 ± 7 g/L in the FeSO4 group and 30 ± 5 g/L in the nanoFe(3+) group. Oral iron failed to be utilized by Fpn-KO mice and was retained in enterocytes, irrespective of the iron source. In summary, although nanoFe(3+) is taken up directly by the duodenum its homeostasis is under the normal regulatory control of dietary iron absorption, namely via ferroportin-dependent efflux from enterocytes, and thus offers potential as a novel oral iron supplement.


Subject(s)
Cation Transport Proteins/physiology , Duodenum/metabolism , Enterocytes/metabolism , Ferric Compounds/pharmacokinetics , Intestinal Absorption/physiology , Iron, Dietary/pharmacokinetics , Nanoparticles , Administration, Oral , Anemia, Iron-Deficiency/metabolism , Animals , Biological Availability , Cation Transport Proteins/biosynthesis , Cation Transport Proteins/deficiency , Cation Transport Proteins/genetics , Ferrous Compounds/pharmacokinetics , Gene Expression Regulation , Hemoglobins/analysis , Hepcidins/biosynthesis , Hepcidins/genetics , Homeostasis , Iron Deficiencies , Mice , Mice, Knockout , Spleen/metabolism
17.
Nature ; 509(7501): 492-6, 2014 May 22.
Article in English | MEDLINE | ID: mdl-24717435

ABSTRACT

The BRAF kinase is mutated, typically Val 600→Glu (V600E), to induce an active oncogenic state in a large fraction of melanomas, thyroid cancers, hairy cell leukaemias and, to a smaller extent, a wide spectrum of other cancers. BRAF(V600E) phosphorylates and activates the MEK1 and MEK2 kinases, which in turn phosphorylate and activate the ERK1 and ERK2 kinases, stimulating the mitogen-activated protein kinase (MAPK) pathway to promote cancer. Targeting MEK1/2 is proving to be an important therapeutic strategy, given that a MEK1/2 inhibitor provides a survival advantage in metastatic melanoma, an effect that is increased when administered together with a BRAF(V600E) inhibitor. We previously found that copper (Cu) influx enhances MEK1 phosphorylation of ERK1/2 through a Cu-MEK1 interaction. Here we show decreasing the levels of CTR1 (Cu transporter 1), or mutations in MEK1 that disrupt Cu binding, decreased BRAF(V600E)-driven signalling and tumorigenesis in mice and human cell settings. Conversely, a MEK1-MEK5 chimaera that phosphorylated ERK1/2 independently of Cu or an active ERK2 restored the tumour growth of murine cells lacking Ctr1. Cu chelators used in the treatment of Wilson disease decreased tumour growth of human or murine cells transformed by BRAF(V600E) or engineered to be resistant to BRAF inhibition. Taken together, these results suggest that Cu-chelation therapy could be repurposed to treat cancers containing the BRAF(V600E) mutation.


Subject(s)
Cell Transformation, Neoplastic , Copper/metabolism , MAP Kinase Signaling System , Proto-Oncogene Proteins B-raf/metabolism , Animals , Cation Transport Proteins/deficiency , Cation Transport Proteins/genetics , Cell Line, Tumor , Cell Transformation, Neoplastic/drug effects , Chelating Agents/pharmacology , Chelating Agents/therapeutic use , Copper/pharmacology , Copper Transporter 1 , Disease Models, Animal , Drug Repositioning , Drug Resistance, Neoplasm/drug effects , Female , Hepatolenticular Degeneration/drug therapy , Humans , Indoles/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , MAP Kinase Signaling System/drug effects , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Sulfonamides/pharmacology , Survival Analysis , Vemurafenib
18.
PLoS One ; 8(10): e77445, 2013.
Article in English | MEDLINE | ID: mdl-24204829

ABSTRACT

The activation process of secretory or membrane-bound zinc enzymes is thought to be a highly coordinated process involving zinc transport, trafficking, transfer and coordination. We have previously shown that secretory and membrane-bound zinc enzymes are activated in the early secretory pathway (ESP) via zinc-loading by the zinc transporter 5 (ZnT5)-ZnT6 hetero-complex and ZnT7 homo-complex (zinc transport complexes). However, how other proteins conducting zinc metabolism affect the activation of these enzymes remains unknown. Here, we investigated this issue by disruption and re-expression of genes known to be involved in cytoplasmic zinc metabolism, using a zinc enzyme, tissue non-specific alkaline phosphatase (TNAP), as a reporter. We found that TNAP activity was significantly reduced in cells deficient in ZnT1, Metallothionein (MT) and ZnT4 genes (ZnT1(-/-) MT(-/-) ZnT4(-/-) cells), in spite of increased cytosolic zinc levels. The reduced TNAP activity in ZnT1(-/-) MT(-/-) ZnT4(-/-) cells was not restored when cytosolic zinc levels were normalized to levels comparable with those of wild-type cells, but was reversely restored by extreme zinc supplementation via zinc-loading by the zinc transport complexes. Moreover, the reduced TNAP activity was adequately restored by re-expression of mammalian counterparts of ZnT1, MT and ZnT4, but not by zinc transport-incompetent mutants of ZnT1 and ZnT4. In ZnT1(-/-) MT(-/-) ZnT4(-/-) cells, the secretory pathway normally operates. These findings suggest that cooperative zinc handling of ZnT1, MT and ZnT4 in the cytoplasm is required for full activation of TNAP in the ESP, and present clear evidence that the activation process of zinc enzymes is elaborately controlled.


Subject(s)
Alkaline Phosphatase/genetics , Avian Proteins/genetics , Cation Transport Proteins/genetics , Metallothionein/genetics , Secretory Pathway/genetics , Signal Transduction/genetics , Zinc/metabolism , Alkaline Phosphatase/metabolism , Animals , Avian Proteins/metabolism , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Cation Transport Proteins/deficiency , Cell Line, Transformed , Chickens/genetics , Chickens/metabolism , Cytoplasm/metabolism , Enzyme Activation , Gene Expression Regulation , Metallothionein/deficiency , Protein Isoforms/deficiency , Protein Isoforms/genetics , Protein Transport
19.
Am J Physiol Gastrointest Liver Physiol ; 303(11): G1236-44, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-23064757

ABSTRACT

The essential requirement for copper in early development is dramatically illustrated by Menkes disease, a fatal neurodegenerative disorder of early childhood caused by loss-of-function mutations in the gene encoding the copper transporting ATPase ATP7A. In this study, we generated mice with enterocyte-specific knockout of the murine ATP7A gene (Atp7a) to test its importance in dietary copper acquisition. Although mice lacking Atp7a protein within intestinal enterocytes appeared normal at birth, they exhibited profound growth impairment and neurological deterioration as a consequence of copper deficiency, resulting in excessive mortality prior to weaning. Copper supplementation of lactating females or parenteral copper injection of the affected offspring markedly attenuated this rapid demise. Enterocyte-specific deletion of Atp7a in rescued pregnant females did not restrict embryogenesis; however, copper accumulation in the late-term fetus was severely reduced, resulting in early postnatal mortality. Taken together, these data demonstrate unique and specific requirements for enterocyte Atp7a in neonatal and maternofetal copper acquisition that are dependent on dietary copper availability, thus providing new insights into the mechanisms of gene-nutrient interaction essential for early human development.


Subject(s)
Adenosine Triphosphatases/deficiency , Cation Transport Proteins/deficiency , Copper/metabolism , Enterocytes/metabolism , Menkes Kinky Hair Syndrome/genetics , Animals , Animals, Newborn , Copper/deficiency , Copper/therapeutic use , Copper-Transporting ATPases , Duodenum/metabolism , Female , Growth Disorders/diet therapy , Lactation , Mice , Nutritional Requirements , Pregnancy
20.
Development ; 137(18): 3079-88, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20702562

ABSTRACT

Neural tube defects (NTDs) are some of the most common birth defects observed in humans. The incidence of NTDs can be reduced by peri-conceptional folic acid supplementation alone and reduced even further by supplementation with folic acid plus a multivitamin. Here, we present evidence that iron maybe an important nutrient necessary for normal development of the neural tube. Following implantation of the mouse embryo, ferroportin 1 (Fpn1) is essential for the transport of iron from the mother to the fetus and is expressed in the visceral endoderm, yolk sac and placenta. The flatiron (ffe) mutant mouse line harbors a hypomorphic mutation in Fpn1 and we have created an allelic series of Fpn1 mutations that result in graded developmental defects. A null mutation in the Fpn1 gene is embryonic lethal before gastrulation, hypomorphic Fpn1(ffe/ffe) mutants exhibit NTDs consisting of exencephaly, spina bifida and forebrain truncations, while Fpn1(ffe/KI) mutants exhibit even more severe NTDs. We show that Fpn1 is not required in the embryo proper but rather in the extra-embryonic visceral endoderm. Our data indicate that loss of Fpn1 results in abnormal morphogenesis of the anterior visceral endoderm (AVE). Defects in the development of the forebrain in Fpn1 mutants are compounded by defects in multiple signaling centers required for maintenance of the forebrain, including the anterior definitive endoderm (ADE), anterior mesendoderm (AME) and anterior neural ridge (ANR). Finally, we demonstrate that this loss of forebrain maintenance is due in part to the iron deficiency that results from the absence of fully functional Fpn1.


Subject(s)
Body Patterning , Cation Transport Proteins/metabolism , Neural Tube Defects/embryology , Neural Tube Defects/metabolism , Prosencephalon/embryology , Prosencephalon/metabolism , Alleles , Animals , Cation Transport Proteins/deficiency , Cation Transport Proteins/genetics , Embryo Culture Techniques , Endoderm/metabolism , Iron Deficiencies , Mice , Mutation , Neural Tube Defects/genetics
SELECTION OF CITATIONS
SEARCH DETAIL