Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Am J Physiol Gastrointest Liver Physiol ; 324(3): G159-G176, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36537699

ABSTRACT

Zinc has anti-inflammatory properties using mechanisms that are unclear. Zip14 (Slc39a14) is a zinc transporter induced by proinflammatory stimuli and is highly expressed at the basolateral membrane of intestinal epithelial cells (IECs). Enterocyte-specific Zip14 ablation (Zip14ΔIEC) in mice was developed to study the functions of this transporter in enterocytes. This gene deletion led to increased intestinal permeability, increased IL-6 and IFNγ expression, mild endotoxemia, and intestinal dysbiosis. RNA sequencing was used for transcriptome profiling. These analyses revealed differential expression of specific intestinal proinflammatory and tight junction (TJ) genes. Binding of transcription factors, including NF-κß, STAT3, and CDX2, to appropriate promoter sites of these genes supports the differential expression shown with chromatin immunoprecipitation assays. Total histone deacetylase (HDAC), and specifically HDAC3, activities were markedly reduced with Zip14 ablation. Intestinal organoids derived from ΔIEC mice display TJ and cytokine gene dysregulation compared with control mice. Differential expression of specific genes was reversed with zinc supplementation of the organoids. We conclude that zinc-dependent HDAC enzymes acquire zinc ions via Zip14-mediated transport and that intestinal integrity is controlled in part through epigenetic modifications.NEW & NOTEWORTHY We show that enterocyte-specific ablation of zinc transporter Zip14 (Slc39a14) results in selective dysbiosis and differential expression of tight junction proteins, claudin 1 and 2, and specific cytokines associated with intestinal inflammation. HDAC activity and zinc uptake are reduced with Zip14 ablation. Using intestinal organoids, the expression defects of claudin 1 and 2 are resolved through zinc supplementation. These novel results suggest that zinc, an essential micronutrient, influences gene expression through epigenetic mechanisms.


Subject(s)
Cation Transport Proteins , Enterocytes , Mice , Animals , Enterocytes/metabolism , Claudin-1/genetics , Claudin-1/metabolism , Dysbiosis , Mice, Knockout , Zinc/metabolism , Homeostasis , Epigenesis, Genetic , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism
2.
J Neurosci ; 40(30): 5871-5891, 2020 07 22.
Article in English | MEDLINE | ID: mdl-32576620

ABSTRACT

Manganese exposure produces Parkinson's-like neurologic symptoms, suggesting a selective dysregulation of dopamine transmission. It is unknown, however, how manganese accumulates in dopaminergic brain regions or how it regulates the activity of dopamine neurons. Our in vivo studies in male C57BLJ mice suggest that manganese accumulates in dopamine neurons of the VTA and substantia nigra via nifedipine-sensitive Ca2+ channels. Manganese produces a Ca2+ channel-mediated current, which increases neurotransmitter release and rhythmic firing activity of dopamine neurons. These increases are prevented by blockade of Ca2+ channels and depend on downstream recruitment of Ca2+-activated potassium channels to the plasma membrane. These findings demonstrate the mechanism of manganese-induced dysfunction of dopamine neurons, and reveal a potential therapeutic target to attenuate manganese-induced impairment of dopamine transmission.SIGNIFICANCE STATEMENT Manganese is a trace element critical to many physiological processes. Overexposure to manganese is an environmental risk factor for neurologic disorders, such as a Parkinson's disease-like syndrome known as manganism. We found that manganese concentration-dependently increased the excitability of dopamine neurons, decreased the amplitude of action potentials, and narrowed action potential width. Blockade of Ca2+ channels prevented these effects as well as manganese accumulation in the mouse midbrain in vivo Our data provide a potential mechanism for manganese regulation of dopaminergic neurons.


Subject(s)
Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Manganese/metabolism , Manganese/toxicity , Action Potentials/drug effects , Action Potentials/physiology , Animals , Cells, Cultured , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Organ Culture Techniques
3.
PLoS Genet ; 14(4): e1007321, 2018 04.
Article in English | MEDLINE | ID: mdl-29621230

ABSTRACT

Hyperostosis Cranialis Interna (HCI) is a rare bone disorder characterized by progressive intracranial bone overgrowth at the skull. Here we identified by whole-exome sequencing a dominant mutation (L441R) in SLC39A14 (ZIP14). We show that L441R ZIP14 is no longer trafficked towards the plasma membrane and excessively accumulates intracellular zinc, resulting in hyper-activation of cAMP-CREB and NFAT signaling. Conditional knock-in mice overexpressing L438R Zip14 in osteoblasts have a severe skeletal phenotype marked by a drastic increase in cortical thickness due to an enhanced endosteal bone formation, resembling the underlying pathology in HCI patients. Remarkably, L438R Zip14 also generates an osteoporotic trabecular bone phenotype. The effects of osteoblastic overexpression of L438R Zip14 therefore mimic the disparate actions of estrogen on cortical and trabecular bone through osteoblasts. Collectively, we reveal ZIP14 as a novel regulator of bone homeostasis, and that manipulating ZIP14 might be a therapeutic strategy for bone diseases.


Subject(s)
Cation Transport Proteins/genetics , Homeostasis/genetics , Hyperostosis/genetics , Mutation , Osteosclerosis/genetics , Skull Base/abnormalities , Animals , Cell Line , Cells, Cultured , Disease Models, Animal , HEK293 Cells , Humans , Hyperostosis/metabolism , Mice, Inbred C57BL , Mice, Knockout , Osteoblasts/cytology , Osteoblasts/metabolism , Osteosclerosis/metabolism , Signal Transduction/genetics , Skull Base/metabolism , Zinc/metabolism
4.
Am J Physiol Gastrointest Liver Physiol ; 318(4): G673-G681, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32003605

ABSTRACT

Impaired manganese (Mn) homeostasis can result in excess Mn accumulation in specific brain regions and neuropathology. Maintaining Mn homeostasis and detoxification is dependent on effective Mn elimination. Specific metal transporters control Mn homeostasis. Human carriers of mutations in the metal transporter ZIP14 and whole body Zip14-knockout (WB-KO) mice display similar phenotypes, including spontaneous systemic and brain Mn overload and motor dysfunction. Initially, it was believed that Mn accumulation due to ZIP14 mutations was caused by impaired hepatobiliary Mn elimination. However, liver-specific Zip14-KO mice did not show systemic Mn accumulation or motor deficits. ZIP14 is highly expressed in the small intestine and is localized to the basolateral surface of enterocytes. Thus, we hypothesized that basolaterally localized ZIP14 in enterocytes provides another route for the elimination of Mn. Using wild-type and intestine-specific Zip14-KO (I-KO) mice, we have shown that ablation of intestinal Zip14 is sufficient to cause systemic and brain Mn accumulation. The lack of intestinal ZIP14-mediated Mn excretion was compensated for by the hepatobiliary system; however, it was not sufficient to maintain Mn homeostasis. When supplemented with extra dietary Mn, I-KO mice displayed some motor dysfunctions and brain Mn accumulation based on both MRI imaging and chemical analysis, thus demonstrating the importance of intestinal ZIP14 as a route of Mn excretion. A defect in intestinal Zip14 expresssion likely could contribute to the Parkinson-like Mn accumulation of manganism.NEW & NOTEWORTHY Mn-induced parkinsonism is recognized as rising in frequency because of both environmental factors and genetic vulnerability; yet currently, there is no cure. We provide evidence in an integrative animal model that basolaterally localized ZIP14 regulates Mn excretion and detoxification and that deletion of intestinal ZIP14 leads to systemic and brain Mn accumulation, providing robust evidence for the indispensable role of intestinal ZIP14 in Mn excretion.


Subject(s)
Cation Transport Proteins/metabolism , Gait Disorders, Neurologic/chemically induced , Intestinal Mucosa/metabolism , Manganese/toxicity , Animals , Biological Transport , Brain/metabolism , Brain/pathology , Cation Transport Proteins/genetics , Dose-Response Relationship, Drug , Genotype , Inflammation/chemically induced , Manganese/administration & dosage , Mice , Mice, Knockout , Serous Membrane/metabolism
5.
Proc Natl Acad Sci U S A ; 114(29): E5805-E5814, 2017 07 18.
Article in English | MEDLINE | ID: mdl-28673968

ABSTRACT

Extensive endoplasmic reticulum (ER) stress damages the liver, causing apoptosis and steatosis despite the activation of the unfolded protein response (UPR). Restriction of zinc from cells can induce ER stress, indicating that zinc is essential to maintain normal ER function. However, a role for zinc during hepatic ER stress is largely unknown despite important roles in metabolic disorders, including obesity and nonalcoholic liver disease. We have explored a role for the metal transporter ZIP14 during pharmacologically and high-fat diet-induced ER stress using Zip14-/- (KO) mice, which exhibit impaired hepatic zinc uptake. Here, we report that ZIP14-mediated hepatic zinc uptake is critical for adaptation to ER stress, preventing sustained apoptosis and steatosis. Impaired hepatic zinc uptake in Zip14 KO mice during ER stress coincides with greater expression of proapoptotic proteins. ER stress-induced Zip14 KO mice show greater levels of hepatic steatosis due to higher expression of genes involved in de novo fatty acid synthesis, which are suppressed in ER stress-induced WT mice. During ER stress, the UPR-activated transcription factors ATF4 and ATF6α transcriptionally up-regulate Zip14 expression. We propose ZIP14 mediates zinc transport into hepatocytes to inhibit protein-tyrosine phosphatase 1B (PTP1B) activity, which acts to suppress apoptosis and steatosis associated with hepatic ER stress. Zip14 KO mice showed greater hepatic PTP1B activity during ER stress. These results show the importance of zinc trafficking and functional ZIP14 transporter activity for adaptation to ER stress associated with chronic metabolic disorders.


Subject(s)
Cation Transport Proteins/metabolism , Endoplasmic Reticulum Stress/physiology , Liver/metabolism , Zinc/metabolism , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Activating Transcription Factor 6/genetics , Activating Transcription Factor 6/metabolism , Animals , Apoptosis/genetics , Biological Transport/physiology , Cation Transport Proteins/genetics , Endoplasmic Reticulum Stress/genetics , Hep G2 Cells , Humans , Liver/drug effects , Mice, Inbred C57BL , Mice, Knockout , Non-alcoholic Fatty Liver Disease/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Tunicamycin/pharmacology , Unfolded Protein Response
6.
J Neurosci ; 37(25): 5996-6006, 2017 06 21.
Article in English | MEDLINE | ID: mdl-28536273

ABSTRACT

Mutations in human ZIP14 have been linked to symptoms of the early onset of Parkinsonism and Dystonia. This phenotype is likely related to excess manganese accumulation in the CNS. The metal transporter ZIP14 (SLC39A14) is viewed primarily as a zinc transporter that is inducible via proinflammatory stimuli. In vitro evidence shows that ZIP14 can also transport manganese. To examine a role for ZIP14 in manganese homeostasis, we used Zip14 knock-out (KO) male and female mice to conduct comparative metabolic, imaging, and functional studies. Manganese accumulation was fourfold to fivefold higher in brains of Zip14 KO mice compared with young adult wild-type mice. There was less accumulation of subcutaneously administered 54Mn in the liver, gallbladder, and gastrointestinal tract of the KO mice, suggesting that manganese elimination is impaired with Zip14 ablation. Impaired elimination creates the opportunity for atypical manganese accumulation in tissues, including the brain. The intensity of MR images from brains of the Zip14 KO mice is indicative of major manganese accumulation. In agreement with excessive manganese accumulation was the impaired motor function observed in the Zip14 KO mice. These results also demonstrate that ZIP14 is not essential for manganese uptake by the brain. Nevertheless, the upregulation of signatures of brain injury observed in the Zip14 KO mice demonstrates that normal ZIP14 function is an essential factor required to prevent manganese-linked neurodegeneration.SIGNIFICANCE STATEMENT Manganese is an essential micronutrient. When acquired in excess, manganese accumulates in tissues of the CNS and is associated with neurodegenerative disease, particularly Parkinson-like syndrome and dystonia. Some members of the ZIP metal transporter family transport manganese. Using mutant mice deficient in the ZIP14 metal transporter, we have discovered that ZIP14 is essential for manganese elimination via the gastrointestinal tract, and a lack of ZIP14 results in manganese accumulation in critical tissues such as the brain, as measured by MRI, and produces signatures of brain injury and impaired motor function. Humans with altered ZIP14 function would lack this gatekeeper function of ZIP14 and therefore would be prone to manganese-related neurological diseases.


Subject(s)
Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Manganese Poisoning/genetics , Manganese Poisoning/metabolism , Manganese/metabolism , Motor Activity/genetics , Animals , Brain Chemistry/genetics , Female , Gastrointestinal Motility/genetics , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Tissue Distribution , Zinc/metabolism , Zinc/pharmacology
7.
J Nutr ; 148(2): 174-184, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29490098

ABSTRACT

The SLC39A family of metal transporters was identified through homologies with the Zrt- and Irt-like (ZIP) proteins from yeast and plants. Of all the ZIP transporters, ZIP14 is arguably the most robustly characterized in terms of function at the integrative level. Mice with a global knockout of Zip14 are viable, thus providing the opportunity to conduct physiologic experiments. In mice, Zip14 expression is highly tissue specific, with the greatest abundance in the jejunum > liver > heart > kidney > white adipose tissue > skeletal muscle > spleen > pancreas. A unique feature of Zip14 is its upregulation by proinflammatory conditions, particularly increased interleukin 6 (IL-6) and nitric oxide. The transcription factors AP-1, ATF4, and ATF6α are involved in Zip14 regulation. ZIP14 does not appear to be zinc-regulated. The Zip14 knockout phenotype shows multiple sites of ZIP14 function, including the liver, adipose tissue, brain, pancreas, and bone. A prominent feature of the Zip14 ablation is a reduction in intestinal barrier function and onset of metabolic endotoxemia. Many aspects of the phenotype are accentuated with age and accompany increased circulating IL-6. Studies with 65Zn, 59Fe [nontransferrin-bound iron (NTBI)] and 54Mn show that ZIP14 transports these metals. At a steady state, the plasma concentrations of zinc, NTBI, and manganese are such that zinc ions are the major substrate available for ZIP14 at the cell surface. Upregulation of ZIP14 accounts for the hypozincemia and hepatic zinc accumulation associated with acute inflammation and sepsis and is required for liver regeneration and resistance to endoplasmic reticulum (ER) stress. Zip14 ablation in mice produces a defect in manganese excretion that leads to excess manganese accumulation in the brain that produces characteristics of Parkinsonism.


Subject(s)
Cation Transport Proteins/physiology , Adipose Tissue/chemistry , Adipose Tissue/physiology , Animals , Biological Transport/physiology , Bone and Bones/chemistry , Bone and Bones/physiology , Brain/physiology , Cation Transport Proteins/analysis , Cation Transport Proteins/genetics , Endotoxemia/etiology , Endotoxemia/metabolism , Interleukin-6/blood , Intestines/physiology , Iron/blood , Liver/metabolism , Liver/physiology , Manganese/analysis , Manganese/blood , Mice , Mice, Knockout , Neoplasms/metabolism , Nitric Oxide/physiology , Pancreas/chemistry , Pancreas/physiology , Tissue Distribution , Zinc/blood , Zinc/metabolism
8.
J Biol Chem ; 291(46): 23939-23951, 2016 Nov 11.
Article in English | MEDLINE | ID: mdl-27703010

ABSTRACT

Zinc influences signaling pathways through controlled targeted zinc transport. Zinc transporter Zip14 KO mice display a phenotype that includes impaired intestinal barrier function with low grade chronic inflammation, hyperinsulinemia, and increased body fat, which are signatures of diet-induced diabetes (type 2 diabetes) and obesity in humans. Hyperglycemia in type 2 diabetes and obesity is caused by insulin resistance. Insulin resistance results in inhibition of glucose uptake by liver and other peripheral tissues, principally adipose and muscle and with concurrently higher hepatic glucose production. Therefore, modulation of hepatic glucose metabolism is an important target for antidiabetic treatment approaches. We demonstrate that during glucose uptake, cell surface abundance of zinc transporter ZIP14 and mediated zinc transport increases. Zinc is distributed to multiple sites in hepatocytes through sequential translocation of ZIP14 from plasma membrane to early and late endosomes. Endosomes from Zip14 KO mice were zinc-deficient because activities of the zinc-dependent insulin-degrading proteases insulin-degrading enzyme and cathepsin D were impaired; hence insulin receptor activity increased. Transient increases in cytosolic zinc levels are concurrent with glucose uptake and suppression of glycogen synthesis. In contrast, Zip14 KO mice exhibited greater hepatic glycogen synthesis and impaired gluconeogenesis and glycolysis related to low cytosolic zinc levels. We can conclude that ZIP14-mediated zinc transport contributes to regulation of endosomal insulin receptor activity and glucose homeostasis in hepatocytes. Therefore, modulation of ZIP14 transport activity presents a new target for management of diabetes and other glucose-related disorders.


Subject(s)
Cation Transport Proteins/metabolism , Endosomes/metabolism , Glucose/metabolism , Hepatocytes/metabolism , Liver/metabolism , Receptor, Insulin/metabolism , Zinc/metabolism , Animals , Cation Transport Proteins/genetics , Endosomes/genetics , Glucose/genetics , Glycogen/biosynthesis , Glycogen/genetics , Mice , Mice, Knockout , Protein Transport/physiology , Receptor, Insulin/genetics
9.
Annu Rev Nutr ; 36: 1-15, 2016 07 17.
Article in English | MEDLINE | ID: mdl-27090747

ABSTRACT

After having written hundreds of research articles, reviews, and book chapters, I find it awkward to pen an autobiography. I still do use a pen. As stated by others in the nutrition field who have written of their own experiences in a perspective article for the Annual Review of Nutrition, my course through this field of science has been serendipitous. My interest in nutrition developed during my experiences with horses and then Angus cattle and entry into an animal science degree program. As the age of molecular biology was unfolding, I pursued a PhD in nutritional biochemistry with Hamilton Eaton at the University of Connecticut followed by postdoctoral work with Hector DeLuca at the University of Wisconsin, working on vitamins A and D, respectively. At Rutgers University, one of the two institutions where I have served on the faculty, I started my research program on trace elements with a focus on cadmium toxicity but soon thereafter began my research on zinc metabolism and function. I moved to the University of Florida in 1982 for an endowed position and have been a Florida Gator ever since. At the University of Florida, research expanded to include identification of zinc-responsive genes and physiological outcomes of zinc transport influencing health and disease, particularly as related to inflammation. I had the opportunity to contribute national science policy as president of both the Federation of American Societies for Experimental Biology and the American Society for Nutrition. As the time of this writing, I maintain an active laboratory.


Subject(s)
Biochemistry/history , Nutrition Policy , Nutritional Sciences/history , Zinc/metabolism , Animal Nutrition Sciences/history , Animal Nutrition Sciences/methods , Animal Nutrition Sciences/trends , Animals , Awards and Prizes , Biochemistry/methods , Biochemistry/trends , Biological Transport , Biomedical Research/economics , Biomedical Research/legislation & jurisprudence , Cadmium/toxicity , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , History, 20th Century , History, 21st Century , Humans , Leadership , National Academy of Sciences, U.S. , Nutrition Policy/history , Nutritional Sciences/methods , Nutritional Sciences/trends , Periodicals as Topic , Public Policy/history , Research Support as Topic/legislation & jurisprudence , Societies, Scientific/history , United States
10.
Am J Physiol Endocrinol Metab ; 310(4): E258-68, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26646099

ABSTRACT

Zinc is a signaling molecule in numerous metabolic pathways, the coordination of which occurs through activity of zinc transporters. The expression of zinc transporter Zip14 (Slc39a14), a zinc importer of the solute carrier 39 family, is stimulated under proinflammatory conditions. Adipose tissue upregulates Zip14 during lipopolysaccharide-induced endotoxemia. A null mutation of Zip14 (KO) revealed that phenotypic changes in adipose include increased cytokine production, increased plasma leptin, hypertrophied adipocytes, and dampened insulin signaling. Adipose tissue from KO mice had increased levels of preadipocyte markers, lower expression of the differentiation marker (PPARγ), and activation of NF-κB and STAT3 pathways. Our overall hypothesis was that ZIP14 would play a role in adipocyte differentiation and inflammatory obesity. Global Zip14 KO causes systemic endotoxemia. The observed metabolic changes in adipose metabolism were reversed when oral antibiotics were administrated, indicating that circulating levels of endotoxin were in part responsible for the adipose phenotype. To evaluate a mechanism, 3T3-L1 cells were differentiated into adipocytes and treated with siRNA to knock down Zip14. These cells had an impaired ability to mobilize zinc, which caused dysregulation of inflammatory pathways (JAK2/STAT3 and NF-κB). The Zip14 deletion may limit the availability of intracellular zinc, yielding the unique phenotype of inflammation coupled with hypertrophy. Taken together, these results suggest that aberrant zinc distribution observed with Zip14 ablation impacts adipose cytokine production and metabolism, ultimately increasing fat deposition when exposed to endotoxin. To our knowledge, this is the first investigation into the mechanistic role of ZIP14 in adipose tissue regulation and metabolism.


Subject(s)
Adipocytes/metabolism , Adipose Tissue, White/metabolism , Cation Transport Proteins/genetics , Cytokines/metabolism , Endotoxemia/metabolism , 3T3-L1 Cells , Adipocytes/pathology , Adipose Tissue , Adipose Tissue, White/pathology , Adiposity , Animals , Blotting, Western , Cation Transport Proteins/metabolism , Cell Differentiation , Gene Knockdown Techniques , Hypertrophy , Inflammation , Janus Kinase 2/metabolism , Leptin/metabolism , Lipopolysaccharides , Mice , Mice, Knockout , Microscopy, Confocal , NF-kappa B/metabolism , PPAR gamma/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction
11.
J Nutr ; 146(11): 2180-2186, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27605406

ABSTRACT

BACKGROUND: Several in vitro studies have shown that zinc deficiency could induce endoplasmic reticulum (ER) stress, resulting in activation of the unfolded protein response. OBJECTIVE: We aimed to determine whether consumption of a zinc-deficient diet (ZnD) triggers ER stress and to understand the impact of dietary zinc intake on ER stress-induced apoptosis using a mouse model. METHODS: Young adult (8-16 wk of age) male mice of strain C57BL/6 were fed either a ZnD (<1 mg/kg diet), or a zinc-adequate diet (ZnA; 30 mg/kg diet). After 2 wk, liver, pancreas, and serum samples were collected and analyzed for indexes of ER stress. In another experiment, mice were fed either a ZnD, a ZnA, or a zinc-supplementation diet (ZnS; 180 mg/kg diet). After 2 wk, vehicle or tunicamycin (TM; 2 mg/kg body weight) was administered to mice to model ER stress. Liver and serum were analyzed for indexes of ER stress to evaluate the effects of zinc status. RESULTS: Mice fed a ZnD did not activate the apoptotic and ER stress markers in the liver or pancreas. During the TM challenge, mice fed a ZnD showed greater C/EBP-homologous protein expression in the liver (3.8-fold, P < 0.01) than did ZnA-fed mice. TM-treated mice fed a ZnD also had greater terminal deoxynucleotidyl transferase deoxyuridine triphosphate nick-end labeling-positive cells in the liver (2.2-fold, P < 0.05), greater hepatic triglyceride accumulation (1.5-fold, P < 0.05), greater serum alanine aminotransferase activity (1.6-fold, P < 0.05), and greater protein-tyrosine phosphatase 1B activity (1.5-fold, P < 0.05), respectively, than did those fed a ZnA. No significant differences were observed in these parameters between mice fed ZnAs and ZnSs. CONCLUSIONS: Consumption of a ZnD per se is not a critical factor for induction of ER stress in mice; however, once ER stress is triggered, adequate dietary zinc intake is required for suppressing apoptotic cell death and further insults in the liver of mice.


Subject(s)
Activating Transcription Factor 4/metabolism , Apoptosis/physiology , Eukaryotic Initiation Factor-2/metabolism , Transcription Factor CHOP/metabolism , Zinc/pharmacology , Activating Transcription Factor 4/genetics , Animal Feed , Animals , Diet , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/physiology , Eukaryotic Initiation Factor-2/genetics , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Male , Mice , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Transcription Factor CHOP/genetics , Zinc/administration & dosage
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.
Am J Physiol Gastrointest Liver Physiol ; 308(3): G171-8, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25428902

ABSTRACT

ZIP14 is a zinc transport protein with high expression in the small intestine and liver. Zip14 is upregulated during endotoxemia and leads to increased liver zinc content and transient hypozinemia. Since body zinc status and inflammation are associated with changes in intestinal permeability, we hypothesized that ZIP14 may influence intestinal permeability. Wild-type (WT) and Zip14 knockout (KO) mice were used to determine ZIP14-associated intestinal zinc metabolism and effects on permeability. Fractionation of plasma membranes revealed that ZIP14 is localized to the basolateral membrane of enterocytes. Studies utilizing (65)Zn administered by subcutaneous injection revealed greater zinc accumulation in the SI of Zip14 KO mice compared with WT mice. Isolation of endosomes confirmed the presence of ZIP14. Quantification of endosomal zinc concentration by FluoZin-3AM fluorescence demonstrated that zinc is trapped in endosomes of Zip14 KO mice. Intestinal permeability assessed both by plasma FITC-dextran following gavage and by serum endotoxin content was greater in Zip14 KO mice. Threonine phosphorylation of the tight junction protein occludin, which is necessary for tight junction assembly, was reduced in KO mice. Claudin 1 and 2, known to have an inverse relationship in regards to tight junction integrity, reflected impaired barrier function in KO jejunum. These data suggest involvement of ZIP14 in providing zinc for a regulatory role needed for maintenance of the intestinal barrier. In conclusion, ZIP14 is a basolaterally localized protein in enterocytes and is involved in endosomal trafficking of zinc and is necessary for proper maintenance of intestinal tight junctions.


Subject(s)
Cation Transport Proteins/metabolism , Intestinal Mucosa/metabolism , Tight Junctions/metabolism , Zinc/metabolism , Animals , Cell Membrane/metabolism , Cells, Cultured , Hepatocytes/metabolism , Humans , Liver/metabolism , Mice, Inbred C57BL , Mice, Knockout , Permeability
14.
Proc Natl Acad Sci U S A ; 108(52): 20970-5, 2011 Dec 27.
Article in English | MEDLINE | ID: mdl-22171008

ABSTRACT

Implementation of zinc interventions for subjects suspected of being zinc-deficient is a global need, but is limited due to the absence of reliable biomarkers. To discover molecular signatures of human zinc deficiency, a combination of transcriptome, cytokine, and microRNA analyses was applied to a dietary zinc depletion/repletion protocol with young male human subjects. Concomitant with a decrease in serum zinc concentration, changes in buccal and blood gene transcripts related to zinc homeostasis occurred with zinc depletion. Microarray analyses of whole blood RNA revealed zinc-responsive genes, particularly, those associated with cell cycle regulation and immunity. Responses of potential signature genes of dietary zinc depletion were further assessed by quantitative real-time PCR. The diagnostic properties of specific serum microRNAs for dietary zinc deficiency were identified by acute responses to zinc depletion, which were reversible by subsequent zinc repletion. Depression of immune-stimulated TNFα secretion by blood cells was observed after low zinc consumption and may serve as a functional biomarker. Our findings introduce numerous novel candidate biomarkers for dietary zinc status assessment using a variety of contemporary technologies and which identify changes that occur prior to or with greater sensitivity than the serum zinc concentration which represents the current zinc status assessment marker. In addition, the results of gene network analysis reveal potential clinical outcomes attributable to suboptimal zinc intake including immune function defects and predisposition to cancer. These demonstrate through a controlled depletion/repletion dietary protocol that the illusive zinc biomarker(s) can be identified and applied to assessment and intervention strategies.


Subject(s)
Biomarkers/blood , Cytokines/blood , Diet , Homeostasis/physiology , MicroRNAs/blood , Zinc/deficiency , Adult , Analysis of Variance , DNA Primers/genetics , Gene Regulatory Networks , Genomics/methods , Humans , Male , Microarray Analysis , Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/blood , Zinc/blood
15.
Gastroenterology ; 142(7): 1536-46.e5, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22374166

ABSTRACT

BACKGROUND & AIMS: Zinc homeostasis in cells is maintained through tight regulation of zinc influx, efflux, and distribution to intracellular organelles by zinc transporters. The Zrt-Irt-like protein (ZIP) transporters facilitate zinc influx to the cytosol. Expression of the ZIP family member Zip14 can be induced by inflammatory cytokines, which also initiate liver regeneration. Hepatocyte proliferation is required for liver regeneration. Zinc regulates cell proliferation, tissue growth, and many mitogenic signaling pathways; we investigated its role in hepatocytes. METHODS: Wild-type and Zip14(-/-) mice that underwent partial hepatectomy (70% of liver removed) were used as models of liver regeneration. We also analyzed AML12 hepatocytes that overexpressed Zip14. Proliferation was assessed with proliferating cell nuclear antigen, CD1, and Ki67 markers and along with assays of zinc content was related to protein tyrosine phosphatase 1B (PTP1B) and extracellular signal-regulated kinase 1/2 signaling. RESULTS: Zip14 was up-regulated and hepatic zinc content increased during liver regeneration. Increased hepatic zinc inhibited activity of the phosphatase PTP1B and increased phosphorylation of c-Met, which promoted hepatocyte proliferation. AML12 cells that overexpressed Zip14 increased in zinc content and proliferation; PTP1B was inhibited and phosphorylation of c-Met increased. The increases in hepatic levels of zinc and hepatocyte proliferation that occurred following partial hepatectomy were not observed in Zip14(-/-) mice. CONCLUSIONS: The transporter Zip14 mediates hepatic uptake of zinc during liver regeneration and for hepatocyte proliferation. These findings indicate that zinc transporter activity regulates liver tissue growth by sequestering zinc. Reagents that regulate ZIP14 activity might be developed as therapeutics to promote liver regeneration in patients with chronic liver disease.


Subject(s)
Cation Transport Proteins/pharmacology , Cell Proliferation/drug effects , Hepatocytes/metabolism , Liver Regeneration/physiology , Proto-Oncogene Proteins c-met/metabolism , Animals , Cell Line , Hepatocyte Growth Factor/pharmacology , Hepatocytes/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphorylation , Proliferating Cell Nuclear Antigen/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Up-Regulation , Zinc/metabolism , Zinc/pharmacology
16.
J Nutr ; 143(12): 1882-8, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24089422

ABSTRACT

Zinc transporters have been characterized to further understand the absorption and metabolism of dietary zinc. Our goal was to characterize zinc transporter Slc39a11 (ZIP11) expression and its subcellular localization within cells of the murine gastrointestinal tract of mice and to determine if dietary zinc regulates ZIP11. The greatest ZIP11 expression was in the stomach, cecum, and colon. Both Zip11 mRNA and ZIP11 protein were shown to be downregulated during dietary zinc restriction (<1 mg Zn/kg) in the murine stomach tissue but were unaffected in the colon. Acute repletion with zinc did not restore Zip11 mRNA levels in the stomach. Immunohistochemistry (IHC) revealed high ZIP11 levels in the lower regions of gastric glands and parietal cells of the stomach. IHC analysis of the colon showed a marked ZIP11 abundance within the cytoplasm of the colonic epithelial cells. IHC also showed an increase in ZIP11 expression in the colon during zinc restriction. There is a robust abundance of ZIP11 in the nuclei of cells of both stomach and colon. Our experiments suggest that when dietary zinc intake is compromised, the colon may increase zinc transporter expression to improve the efficiency for absorption via increased expression of specific zinc transporters, including ZIP11 and also zinc transporter Slc39a4. In conclusion, ZIP11 is highly expressed within the murine stomach and colon and appears to be partially regulated by dietary zinc intake within these tissues. ZIP11 may play a specialized role in zinc homeostasis within these tissues, helping to maintain mucosal integrity and function.


Subject(s)
Cation Transport Proteins/metabolism , Cell Nucleus/drug effects , Colon/metabolism , Gastric Mucosa/metabolism , Zinc/pharmacology , Animals , Base Sequence , Cation Transport Proteins/genetics , DNA Primers , Mice , Mice, Inbred C57BL , Polymerase Chain Reaction , RNA, Messenger/genetics , Zinc/administration & dosage
17.
Proc Natl Acad Sci U S A ; 107(7): 2818-23, 2010 Feb 16.
Article in English | MEDLINE | ID: mdl-20133611

ABSTRACT

The exocrine pancreas plays an important role in endogenous zinc loss by regulating excretion into the intestinal tract and hence influences the dietary zinc requirement. The present experiments show that the zinc transporter ZnT2 (Slc30a2) is localized to the zymogen granules and that dietary zinc restriction in mice decreased the zinc concentration of zymogen granules and ZnT2 expression. Excess zinc given orally increased ZnT2 expression and was associated with increased pancreatic zinc accumulation. Rat AR42J acinar cells when induced into a secretory phenotype, using the glucocorticoid analog dexamethasone (DEX), exhibited increased ZnT2 expression and labile zinc as measured with a fluorophore. DEX administrated to mice also induced ZnT2 expression that accompanied a reduction of the pancreatic zinc content. ZnT2 promoter analyses identified elements required for responsiveness to zinc and DEX. Zinc regulation was traced to a MRE located downstream from the ZnT2 transcription start site. Responsiveness to DEX is produced by two upstream STAT5 binding sites that require the glucocorticoid receptor for activation. ZnT2 knockdown in the AR42J cells using siRNA resulted in increased cytoplasmic zinc and decreased zymogen granule zinc that further demonstrated that ZnT2 may mediate the sequestration of zinc into zymogen granules. We conclude, based upon experiments with intact mice and pancreatic acinar cells in culture, that ZnT2 participates in zinc transport into pancreatic zymogen granules through a glucocorticoid pathway requiring glucocorticoid receptor and STAT5, and zinc-regulated signaling pathways requiring MTF-1. The ZnT2 transporter appears to function in a physiologically responsive manner involving entero-pancreatic zinc trafficking.


Subject(s)
Cation Transport Proteins/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation/physiology , Pancreas/metabolism , Receptors, Glucocorticoid/metabolism , STAT5 Transcription Factor/metabolism , Transcription Factors/metabolism , Analysis of Variance , Animals , Cation Transport Proteins/genetics , Cell Line, Tumor , Chromatin Immunoprecipitation , Dexamethasone/pharmacology , Fluorescent Antibody Technique , Gene Expression Regulation/drug effects , Immunoblotting , Kinetics , Luciferases , Mice , Pancreas/cytology , RNA Interference , Rats , Reverse Transcriptase Polymerase Chain Reaction , Zinc/metabolism , Zinc/pharmacology , Transcription Factor MTF-1
18.
Nutrients ; 14(23)2022 Dec 01.
Article in English | MEDLINE | ID: mdl-36501144

ABSTRACT

Integration of non-coding RNAs and miRNAs with physiological processes in animals, including nutrient metabolism, is an important new focus. Twenty-three transporter proteins control cellular zinc homeostasis. The transporter Zip14 (Slc39a14) responds to proinflammatory stimuli. Using enterocyte-specific Zip14 knockout mice and RNA-sequencing and quantitative polymerase chain reaction (qPCR), we conducted transcriptome profiling of proximal small intestine, where Zip14 is highly expressed, using RNA from whole intestine tissue, isolated intestinal epithelial cells (IECs) and intestinal organoids. H19, U90926, Meg3, Bvht, Pvt1, Neat1 and miR-7027 were among the most highly expressed genes. Enterocyte-specific deletion of Zip14 demonstrated tissue specific expression, as such these changes were not observed with skeletal muscle. Chromatin immunoprecipitation (ChIP) assays of chromatin from isolated intestinal epithelial cells showed that enterocyte-specific Zip14 deletion enhanced binding of proinflammatory transcription factors (TFs) signal transducer and activator of transcription 3 (STAT3) and nuclear factor kappa beta (NF-ĸß) to promoters of H19, Meg3 and U90926. We conclude enterocyte-specific ablation of Zip14 restricts changes in those RNAs to the intestine. Binding of proinflammatory TFs, NF-Ä¸ß and STAT3 to the H19, Meg3 and U90926 promoters is consistent with a model where Zip14 ablation, leads to increased TF occupancy, allowing epigenetic regulation of specific lncRNA genes.


Subject(s)
Cation Transport Proteins , MicroRNAs , RNA, Long Noncoding , Mice , Animals , RNA, Long Noncoding/genetics , MicroRNAs/genetics , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Epigenesis, Genetic , Inflammation/genetics , Mice, Knockout , Zinc/metabolism
19.
Am J Physiol Cell Physiol ; 301(4): C862-71, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21653899

ABSTRACT

Recent studies have shown that overexpression of the transmembrane protein Zrt- and Irt-like protein 14 (Zip14) stimulates the cellular uptake of zinc and nontransferrin-bound iron (NTBI). Here, we directly tested the hypothesis that Zip14 transports free zinc, iron, and other metal ions by using the Xenopus laevis oocyte heterologous expression system, and use of this approach also allowed us to characterize the functional properties of Zip14. Expression of mouse Zip14 in RNA-injected oocytes stimulated the uptake of (55)Fe in the presence of l-ascorbate but not nitrilotriacetic acid, indicating that Zip14 is an iron transporter specific for ferrous ion (Fe(2+)) over ferric ion (Fe(3+)). Zip14-mediated (55)Fe(2+) uptake was saturable (K(0.5) ≈ 2 µM), temperature-dependent (apparent activation energy, E(a) = 15 kcal/mol), pH-sensitive, Ca(2+)-dependent, and inhibited by Co(2+), Mn(2+), and Zn(2+). HCO(3)(-) stimulated (55)Fe(2+) transport. These properties are in close agreement with those of NTBI uptake in the perfused rat liver and in isolated hepatocytes reported in the literature. Zip14 also mediated the uptake of (109)Cd(2+), (54)Mn(2+), and (65)Zn(2+) but not (64)Cu (I or II). (65)Zn(2+) uptake also was saturable (K(0.5) ≈ 2 µM) but, notably, the metal-ion inhibition profile and Ca(2+) dependence of Zn(2+) transport differed from those of Fe(2+) transport, and we propose a model to account for these observations. Our data reveal that Zip14 is a complex, broad-scope metal-ion transporter. Whereas zinc appears to be a preferred substrate under normal conditions, we found that Zip14 is capable of mediating cellular uptake of NTBI characteristic of iron-overload conditions.


Subject(s)
Cation Transport Proteins/metabolism , Iron/metabolism , Zinc/metabolism , Animals , Cation Transport Proteins/genetics , Gene Expression Regulation , Humans , Mice , Oocytes , Protein Isoforms , Rats , Xenopus
20.
Annu Rev Nutr ; 29: 153-76, 2009.
Article in English | MEDLINE | ID: mdl-19400752

ABSTRACT

Research advances defining how zinc is transported into and out of cells and organelles have increased exponentially within the past five years. Research has progressed through application of molecular techniques including genomic analysis, cell transfection, RNA interference, kinetic analysis of ion transport, and application of cell and animal models including knockout mice. The knowledge base has increased for most of 10 members of the ZnT family and 14 members of the Zrt-, Irt-like protein (ZIP) family. Relative to the handling of dietary zinc is the involvement of ZnT1, ZIP4, and ZIP5 in intestinal zinc transport, involvement of ZIP10 and ZnT1 in renal zinc reabsorption, and the roles of ZIP5, ZnT2, and ZnT1 in pancreatic release of endogenous zinc. These events are major factors in regulation of zinc homeostasis. Other salient findings are the involvement of ZnT2 in lactation, ZIP14 in the hypozincemia of inflammation, ZIP6, ZIP7, and ZIP10 in metastatic breast cancer, and ZnT8 in insulin processing and as an autoantigen in diabetes.


Subject(s)
Carrier Proteins/physiology , Zinc/administration & dosage , Zinc/metabolism , Animals , Biological Transport , Homeostasis , Humans , Kidney/metabolism , Pancreas/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL