Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 17 de 17
Filtrer
Plus de filtres











Gamme d'année
1.
Pediatr Investig ; 7(1): 6-12, 2023 Mar.
Article de Anglais | MEDLINE | ID: mdl-36967740

RÉSUMÉ

Importance: Transient neonatal zinc deficiency (TNZD) occurs in breastfed infants due to abnormally low breast milk zinc levels. Mutations in the solute carrier family 30 member 2 (SLC30A2) gene, which encodes the zinc transporter ZNT2, cause low zinc concentration in breast milk. Objective: This study aimed to provide further insights into TNZD pathophysiology. Methods: SLC30A2 sequencing was performed in three unrelated Japanese mothers, whose infants developed TNZD due to low-zinc milk consumption. The effects of the identified mutations were examined using cell-based assays and luciferase reporter analysis. Results: Novel SLC30A2 mutations were identified in each mother. One harbored a heterozygous missense mutation in the ZNT2 zinc-binding site, which resulted in defective zinc transport. The other two mothers exhibited multiple heterozygous mutations in the SLC30A2 promoter, the first mutations in the SLC30A2 regulatory region reported to date. Interpretation: This report provides new genetic insights into TNZD pathogenesis in breastfed infants.

2.
Int J Mol Sci ; 23(19)2022 Sep 28.
Article de Anglais | MEDLINE | ID: mdl-36232769

RÉSUMÉ

A wide range of microbial pathogens can enter the gastrointestinal tract, causing mucosal inflammation and infectious colitis and accounting for most cases of acute diarrhea. Severe cases of infectious colitis can persist for weeks, and if untreated, may lead to major complications and death. While the molecular pathogenesis of microbial infections is often well-characterized, host-associated epithelial factors that affect risk and severity of infectious colitis are less well-understood. The current study characterized functions of the zinc (Zn) transporter ZnT2 (SLC30A2) in cultured HT29 colonocytes and determined consequences of ZnT2 deletion in mice on the colonic response to enteric infection with Citrobacter rodentium. ZnT2 in colonocytes transported Zn into vesicles buffering cytoplasmic Zn pools, which was important for Toll-like receptor 4 (TLR4) expression, activation of pathogen-stimulated NF-κß translocation and cytokine expression. Additionally, ZnT2 was critical for lysosome biogenesis and bacterial-induced autophagy, both promoting robust host defense and resolution mechanisms in response to enteric pathogens. These findings reveal that ZnT2 is a novel regulator of mucosal inflammation in colonocytes and is critical to the response to infectious colitis, suggesting that manipulating the function of ZnT2 may offer new therapeutic strategies to treat specific intestinal infections.


Sujet(s)
Transporteurs de cations , Colite , Inflammation , Muqueuse intestinale , Récepteur de type Toll-4 , Animaux , Transporteurs de cations/génétique , Transporteurs de cations/métabolisme , Colite/étiologie , Colite/génétique , Colite/métabolisme , Cytokines/génétique , Cytokines/métabolisme , Cellules HT29 , Humains , Inflammation/génétique , Inflammation/métabolisme , Muqueuse intestinale/métabolisme , Souris , Souris de lignée C57BL , Récepteur de type Toll-4/génétique , Récepteur de type Toll-4/métabolisme , Zinc/métabolisme
3.
Biochem Biophys Rep ; 32: 101362, 2022 Dec.
Article de Anglais | MEDLINE | ID: mdl-36204728

RÉSUMÉ

Recently, we reported that TMEM163 is a zinc efflux transporter that likely belongs to the mammalian solute carrier 30 (Slc30/ZnT) subfamily of the cation diffusion facilitator (CDF) protein superfamily. We hypothesized that human TMEM163 forms functional heterodimers with certain ZNT proteins based on their overlapping subcellular localization with TMEM163 and previous reports that some ZNT monomers interact with each other. In this study, we heterologously expressed individual constructs with a unique peptide tag containing TMEM163, ZNT1, ZNT2, ZNT3, and ZNT4 (negative control) or co-expressed TMEM163 with each ZNT in cultured cells for co-immunoprecipitation (co-IP) experiments. We also co-expressed TMEM163 with two different peptide tags as a positive co-IP control. Western blot analyses revealed that TMEM163 dimerizes with itself but that it also heterodimerizes with ZNT1, ZNT2, ZNT3, and ZNT4 proteins. Confocal microscopy revealed that TMEM163 and ZNT proteins partially co-localize in cells, suggesting that they exist as homodimers and heterodimers in their respective subcellular sites. Functional zinc flux assays using Fluozin-3 and Newport Green dyes show that TMEM163/ZNT heterodimers exhibit similar efflux function as TMEM163 homodimers. Cell surface biotinylation revealed that the plasma membrane localization of TMEM163 is not markedly influenced by ZNT co-expression. Overall, our results show that the interaction between TMEM163 and distinct ZNT proteins is physiologically relevant and that their heterodimerization may serve to increase the functional diversity of zinc effluxers within specific tissues or cell types.

4.
5.
Gut Microbes ; 14(1): 2014739, 2022.
Article de Anglais | MEDLINE | ID: mdl-34965180

RÉSUMÉ

Loss of Paneth cell (PC) function is implicated in intestinal dysbiosis, mucosal inflammation, and numerous intestinal disorders, including necrotizing enterocolitis (NEC). Studies in mouse models show that zinc transporter ZnT2 (SLC30A2) is critical for PC function, playing a role in granule formation, secretion, and antimicrobial activity; however, no studies have investigated whether loss of ZnT2 function is associated with dysbiosis, mucosal inflammation, or intestinal dysfunction in humans. SLC30A2 was sequenced in healthy preterm infants (26-37 wks; n = 75), and structural analysis and functional assays determined the impact of mutations. In human stool samples, 16S rRNA sequencing and RNAseq of bacterial and human transcripts were performed. Three ZnT2 variants were common (>5%) in this population: H346Q, f = 19%; L293R, f = 7%; and a previously identified compound substitution in Exon7, f = 16%). H346Q had no effect on ZnT2 function or beta-diversity. Exon7 impaired zinc transport and was associated with a fractured gut microbiome. Analysis of microbial pathways suggested diverse effects on nutrient metabolism, glycan biosynthesis and metabolism, and drug resistance, which were associated with increased expression of host genes involved in tissue remodeling. L293R caused profound ZnT2 dysfunction and was associated with overt gut dysbiosis. Microbial pathway analysis suggested effects on nucleotide, amino acid and vitamin metabolism, which were associated with the increased expression of host genes involved in inflammation and immune response. In addition, L293R was associated with reduced weight gain in the early postnatal period. This implicates ZnT2 as a novel modulator of mucosal homeostasis in humans and suggests that genetic variants in ZnT2 may affect the risk of mucosal inflammation and intestinal disease.


Sujet(s)
Transporteurs de cations/génétique , Dysbiose/génétique , Maladies néonatales/génétique , Prématuré/métabolisme , Intestins/métabolisme , Mutation perte de fonction , Animaux , Bactéries/classification , Bactéries/génétique , Bactéries/isolement et purification , Transporteurs de cations/déficit , Dysbiose/métabolisme , Dysbiose/microbiologie , Exons , Femelle , Microbiome gastro-intestinal , Humains , Nouveau-né , Maladies néonatales/métabolisme , Maladies néonatales/microbiologie , Intestins/microbiologie , Mâle , Souris knockout , Mutation , Mutation faux-sens , Polyosides/métabolisme
6.
Biotech Histochem ; 96(8): 623-635, 2021 Nov.
Article de Anglais | MEDLINE | ID: mdl-33615931

RÉSUMÉ

We investigated the relations among levels of metallothionein (MT); zinc (Zn) transport proteins, ZnT2, ZIP2 (ZRT and IRT-like proteins); and ZIP4, which enable Zn absorption in the small intestine of rats. We also investigated tissue Zn levels in the small intestine. We used four groups of adult male rats: group 1, control; group 2, pinealectomy (Px); group 3, Px + melatonin (MEL); group 4, MEL only. Animals in groups 3 and 4 were administered 5 mg/kg/day MEL for four weeks. At the end of the study, all animals were sacrificed and samples of duodenum, jejunum and ileum were harvested to analyze ZnT2, ZIP2, ZIP4 and MT levels using immunohistochemistry, and tissue Zn levels were measured by atomic absorption spectrophotometry. The lowest ZnT2 levels in the duodenum, jejunum and ileum, and the lowest ZIP2 levels in the duodenum and ileum were found in group 2. The lowest ZIP4 levels were found in the duodenum and jejunum, and the lowest MT levels in the duodenum and ileum were found in group 2. The highest MT values in the ileum were found in group 4. We found that ZnT2, ZIP2, ZIP4 and MT levels were reduced in the ileum compared to controls following Px, but levels approached control values after MEL administration. By its effects on ZnT2, ZIP2, ZIP4 and MT levels, MEL participates in the absorption of Zn in the rat small intestine.


Sujet(s)
Mélatonine , Métallothionéine , Animaux , Compléments alimentaires , Intestin grêle , Mâle , Mélatonine/pharmacologie , Pinéalectomie , Rats , Zinc
7.
Am J Physiol Cell Physiol ; 318(6): C1166-C1177, 2020 06 01.
Article de Anglais | MEDLINE | ID: mdl-32320289

RÉSUMÉ

Suboptimal lactation is a common, yet underappreciated cause for early cessation of breastfeeding. Molecular regulation of mammary gland function is critical to the process lactation; however, physiological factors underlying insufficient milk production are poorly understood. The zinc (Zn) transporter ZnT2 is critical for regulation of mammary gland development and maturation during puberty, lactation, and postlactation gland remodeling. Numerous genetic variants in the gene encoding ZnT2 (SLC30A2) are associated with low milk Zn concentration and result in severe Zn deficiency in exclusively breastfed infants. However, the functional impacts of genetic variation in ZnT2 on key mammary epithelial cell functions have not yet been systematically explored at the cellular level. Here we determined a common mutation in SLC30A2/ZnT2 substituting serine for threonine at amino acid 288 (Thr288Ser) was found in 20% of women producing low milk volume (n = 2/10) but was not identified in women producing normal volume. Exploration of cellular consequences in vitro using phosphomimetics showed the serine substitution promoted preferential phosphorylation of ZnT2, driving localization to the lysosome and increasing lysosome biogenesis and acidification. While the substitution did not initiate lysosome-mediated cell death, cellular ATP levels were significantly reduced. Our findings demonstrate the Thr288Ser mutation in SLC30A2/ZnT2 impairs critical functions of mammary epithelial cells and suggest a role for genetic variation in the regulation of milk production and lactation performance.


Sujet(s)
Transporteurs de cations/métabolisme , Métabolisme énergétique , Cellules épithéliales/métabolisme , Lactation/métabolisme , Lysosomes/métabolisme , Glandes mammaires humaines/métabolisme , Lait humain/métabolisme , Mutation , Adénosine triphosphate/métabolisme , Adulte , Études cas-témoins , Transporteurs de cations/génétique , Lignée cellulaire , Métabolisme énergétique/génétique , Femelle , Humains , Concentration en ions d'hydrogène , Lactation/génétique , Lysosomes/génétique , Biogenèse des organelles , Phosphorylation , Jeune adulte
8.
Exp Dermatol ; 29(6): 556-561, 2020 06.
Article de Anglais | MEDLINE | ID: mdl-32278324

RÉSUMÉ

The SLC30A2 gene encodes zinc transporter ZnT2, which is indispensable for the transport of zinc into the breast milk in the mammary gland. Transient neonatal zinc deficiency (TNZD) is caused by a mutation in the maternal SLC30A2 gene and has a clinical presentation similar to that of acrodermatitis enteropathica (AE). We described the case of a Chinese infant who presented with AE-like lesions 10 days after birth. Sanger sequencing of the AE-causing gene SLC39A4 revealed no mutations in genomic DNA from the infant, excluding the possibility of AE. Detection of the mother's breast milk showed a significantly lower zinc level. Thus, SLC30A2 sequencing was performed on her genomic DNA and a previously unreported homozygous c.262G > A (p.E88K) mutation was disclosed. Functional analysis suggested the novel mutation could lead to a strong disruption of zinc secretion, which indicated a complete loss of function in the ZnT2 protein. We finally diagnosed the infant with TNZD. To the best of our knowledge, this is the first case of TNZD caused by a homozygous mutation in the maternal SLC30A2 gene. Compared to the heterozygous condition, a homozygous mutation seems to result in a more significant decrease in zinc secretion and a more rapid onset of TNZD.


Sujet(s)
Transporteurs de cations/génétique , Troubles de la croissance/génétique , Lait humain/composition chimique , Maladies de la peau/étiologie , Zinc/déficit , Transport biologique/génétique , Analyse de mutations d'ADN , Femelle , Troubles de la croissance/complications , Cellules HEK293/métabolisme , Homozygote , Humains , Nouveau-né , Mutation , Maladies de la peau/diagnostic , Zinc/métabolisme
9.
Am J Physiol Regul Integr Comp Physiol ; 315(2): R323-R335, 2018 08 01.
Article de Anglais | MEDLINE | ID: mdl-29718697

RÉSUMÉ

Mammary gland involution, a tightly regulated process of tissue remodeling by which a lactating mammary gland reverts to the prepregnant state, is characterized by the most profound example of regulated epithelial cell death in normal tissue. Defects in the execution of involution are associated with lactation failure and breast cancer. Initiation of mammary gland involution requires upregulation of lysosome biogenesis and acidification to activate lysosome-mediated cell death; however, specific mediators of this initial phase of involution are not well described. Zinc transporter 2 [ZnT2 ( SLC30A2)] has been implicated in lysosome biogenesis and lysosome-mediated cell death during involution; however, the direct role of ZnT2 in this process has not been elucidated. Here we showed that ZnT2-null mice had impaired alveolar regression and reduced activation of the involution marker phosphorylated Stat3, indicating insufficient initiation of mammary gland remodeling during involution. Moreover, we found that the loss of ZnT2 inhibited assembly of the proton transporter vacuolar ATPase on lysosomes, thereby decreasing lysosome abundance and size. Studies in cultured mammary epithelial cells revealed that while the involution signal TNFα promoted lysosome biogenesis and acidification, attenuation of ZnT2 impaired the lysosome response to this involution signal, which was not a consequence of cytoplasmic Zn accumulation. Our findings establish ZnT2 as a novel regulator of vacuolar ATPase assembly, driving lysosome biogenesis, acidification, and tissue remodeling during the initiation of mammary gland involution.


Sujet(s)
Transporteurs de cations/métabolisme , Cellules épithéliales/métabolisme , Lactation , Lysosomes/métabolisme , Glandes mammaires animales/métabolisme , Biogenèse des organelles , Animaux , Transporteurs de cations/déficit , Transporteurs de cations/génétique , Cellules cultivées , Cellules épithéliales/effets des médicaments et des substances chimiques , Femelle , Concentration en ions d'hydrogène , Lysosomes/effets des médicaments et des substances chimiques , Glandes mammaires animales/effets des médicaments et des substances chimiques , Souris , Souris knockout , Taille de l'organelle , Phosphorylation , Facteur de transcription STAT-3/métabolisme , Transduction du signal , Facteur de nécrose tumorale alpha/pharmacologie , Vacuolar Proton-Translocating ATPases/métabolisme
10.
Traffic ; 17(3): 267-88, 2016 Mar.
Article de Anglais | MEDLINE | ID: mdl-26728129

RÉSUMÉ

The solute carrier 30A (SLC30A) family of zinc exporters transports zinc into the lumen of intracellular organelles in order to prevent zinc toxicity. We reported that formation of tyrosine dimers is required for ZnT3 (zinc transporter 3) zinc transport activity and targeting to synaptic-like microvesicles (SLMVs) in PC12 cells and the formation of ZnT3/ZnT10 heterodimers. Here, we focused on ZnT10 to determine the role of heterodimerization in the sorting of ZnTs in the endolysosomal pathway. Using cell fractionation, immunoprecipitation and immunofluorescence approaches, we found that ZnT10 resides in transferrin receptor and Rab5-positive endosomes and forms covalent heterodimers and oligomers with ZnT2, ZnT3 and ZnT4. The interaction of ZnT10 with ZnT3, mediated by dityrosine bonds, was unable to target ZnT10 into SLMVs in vitro or into synaptic vesicles isolated from mouse brain in vivo. However, ZnT3/ZnT10 heterodimers regulate epidermal growth factor receptor (EGF-R) signaling by increasing the phosphorylation of mitogen-activated protein kinase kinase (MEK) and extracellular signal-regulated kinase (ERK1/2), but not EGF-R, C-Raf or Akt phosphorylation in response to EGF. Further, mutation of tyrosine 4 in ZnT10 reduced ZnT3/ZnT10 dityrosine-mediated heterodimerization and zinc transport, as well as MEK and ERK1/2 phosphorylation, which were also reduced by the zinc chelator TPEN. Phosphorylation of these kinases is likely to occur in the cytosol as no differences in phosphorylation were observed in membrane fractions of control and ZnT3/ZnT10-expressing cells. We propose that ZnT10 plays a role in signal transduction, which is mediated by homo and heterodimerization with other ZnTs.


Sujet(s)
Transporteurs de cations/métabolisme , Endosomes/métabolisme , Système de signalisation des MAP kinases , Animaux , Facteur de croissance épidermique/métabolisme , Récepteurs ErbB/métabolisme , Cellules HEK293 , Humains , MAP Kinase Kinase Kinases/métabolisme , Mitogen-Activated Protein Kinase 1/métabolisme , Mitogen-Activated Protein Kinase 3/métabolisme , Cellules PC12 , Multimérisation de protéines , Transport des protéines , Protéines proto-oncogènes c-akt/métabolisme , Rats , Vésicules synaptiques/métabolisme
11.
J Mammary Gland Biol Neoplasia ; 20(3-4): 159-72, 2015 Dec.
Article de Anglais | MEDLINE | ID: mdl-26293594

RÉSUMÉ

The zinc (Zn) transporter ZnT2 (SLC30A2) is expressed in specialized secretory cells including breast, pancreas and prostate, and imports Zn into mitochondria and vesicles. Mutations in SLC30A2 substantially reduce milk Zn concentration ([Zn]) and cause severe Zn deficiency in exclusively breastfed infants. Recent studies show that ZnT2-null mice have low milk [Zn], in addition to profound defects in mammary gland function during lactation. Here, we used breast milk [Zn] to identify novel non-synonymous ZnT2 variants in a population of lactating women. We also asked whether specific variants induce disturbances in intracellular Zn management or cause cellular dysfunction in mammary epithelial cells. Healthy, breastfeeding women were stratified into quartiles by milk [Zn] and exonic sequencing of SLC30A2 was performed. We found that 36% of women tested carried non-synonymous ZnT2 variants, all of whom had milk Zn levels that were distinctly above or below those in women without variants. We identified 12 novel heterozygous variants. Two variants (D(103)E and T(288)S) were identified with high frequency (9 and 16%, respectively) and expression of T(288)S was associated with a known hallmark of breast dysfunction (elevated milk sodium/potassium ratio). Select variants (A(28)D, K(66)N, Q(71)H, D(103)E, A(105)P, Q(137)H, T(288)S and T(312)K) were characterized in vitro. Compared with wild-type ZnT2, these variants were inappropriately localized, and most resulted in either 'loss-of-function' or 'gain-of-function', and altered sub-cellular Zn pools, Zn secretion, and cell cycle check-points. Our study indicates that SLC30A2 variants are common in this population, dysregulate Zn management and can lead to breast cell dysfunction. This suggests that genetic variation in ZnT2 could be an important modifier of infant growth/development and reproductive health/disease. Importantly, milk [Zn] level may serve as a bio-reporter of breast function during lactation.


Sujet(s)
Transporteurs de cations/génétique , Cellules épithéliales/physiologie , Lactation/génétique , Glandes mammaires humaines/physiopathologie , Lait humain/composition chimique , Zinc/métabolisme , Animaux , Allaitement naturel , Points de contrôle du cycle cellulaire/génétique , Lignée cellulaire , Analyse de mutations d'ADN , Exome , Femelle , Humains , Souris , Mutation , Analyse de séquence d'ADN , Zinc/analyse
12.
J Nutr ; 145(9): 1999-2005, 2015 Sep.
Article de Anglais | MEDLINE | ID: mdl-26203096

RÉSUMÉ

BACKGROUND: Lactation failure is common in overweight and obese women; however, the precise mechanism remains unknown. OBJECTIVE: We tested the hypothesis that obesity-induced inflammation in the mammary gland (MG) redistributes subcellular zinc pools to promote cell death of mammary epithelial cells (MECs) and premature involution. METHODS: Female DBA/2J mice were fed a high-fat (obese; 45% kcal from fat, n = 60) or control diet (lean; 10% kcal from fat, n = 50) for 5 wk and bred. MG cytokines and macrophage infiltration were determined by reverse transcriptase-polymerase chain reaction and F4/80 staining, respectively. Zinc concentration was analyzed by atomic absorption spectroscopy, and zinc transporters and markers of endoplasmic reticulum (ER) stress, autophagy, and involution were measured by immunoblot. To confirm effects of inflammation, tumor necrosis factor-α (TNF) or vehicle was injected into adjacent MGs of lean lactating C57BL/6 mice (n = 5) and cultured MECs (HC11 cells) were treated with TNF in vitro. RESULTS: Seventy-seven percent of obese mice failed to lactate (lean: 39%; P < 0.001). Obese mice capable of lactating had greater macrophage infiltration (obese: 135 ± 40.4 macrophages/mm(2); lean: 63.8 ± 8.9 macrophages/mm(2); P < 0.001) and elevated TNF expression (P < 0.05), concurrent with lower zrt- irt-like protein 7 abundance (P < 0.05) and higher ER zinc concentration (obese: 0.36 ± 0.004 µg Zn/mg protein; lean: 0.30 ± 0.02 µg Zn/mg protein; P < 0.05) compared with lean mice. Heat shock protein 5 (HSPA5) expression (P < 0.05) was suppressed in the MG of obese mice, which was consistent with HSPA5 suppression in TNF-injected MGs (P < 0.01) and MECs treated with TNF in vitro (P < 0.01). Moreover, obesity increased lysosomal activity (P < 0.05) and autophagy in the MG, which corresponded to increased zinc transporter 2 abundance and lysosomal zinc concentration compared with lean mice (obese: 0.20 ± 0.02 µg Zn/mg protein; lean: 0.14 ± 0.01 µg Zn/mg protein; P < 0.05). Importantly, MGs of obese mice exhibited markers of apoptosis (P = 0.05) and involution (P < 0.01), which were not observed in lean mice. CONCLUSIONS: Diet-induced obesity created a proinflammatory MG microenvironment in mice, which was associated with zinc-mediated ER stress and autophagy and the activation of premature involution.


Sujet(s)
Inflammation/anatomopathologie , Glandes mammaires animales/physiopathologie , Obésité/anatomopathologie , Zinc/composition chimique , Animaux , Autophagie , Cellules cultivées , Chaperonne BiP du réticulum endoplasmique , Cellules épithéliales/composition chimique , Cellules épithéliales/cytologie , Femelle , Inflammation/étiologie , Lactation , Macrophages/cytologie , Mâle , Glandes mammaires animales/cytologie , Souris , Souris de lignée C57BL , Souris de lignée DBA , Lait/composition chimique , Lait/métabolisme , Protéines de lait/composition chimique , Obésité/complications , Facteur de nécrose tumorale alpha/pharmacologie
13.
J Biol Chem ; 290(21): 13064-78, 2015 May 22.
Article de Anglais | MEDLINE | ID: mdl-25851903

RÉSUMÉ

The zinc transporter ZnT2 (SLC30A2) imports zinc into vesicles in secreting mammary epithelial cells (MECs) and is critical for zinc efflux into milk during lactation. Recent studies show that ZnT2 also imports zinc into mitochondria and is expressed in the non-lactating mammary gland and non-secreting MECs, highlighting the importance of ZnT2 in general mammary gland biology. In this study we used nulliparous and lactating ZnT2-null mice and characterized the consequences on mammary gland development, function during lactation, and milk composition. We found that ZnT2 was primarily expressed in MECs and to a limited extent in macrophages in the nulliparous mammary gland and loss of ZnT2 impaired mammary expansion during development. Secondly, we found that lactating ZnT2-null mice had substantial defects in mammary gland architecture and MEC function during secretion, including fewer, condensed and disorganized alveoli, impaired Stat5 activation, and unpolarized MECs. Loss of ZnT2 led to reduced milk volume and milk containing less protein, fat, and lactose compared with wild-type littermates, implicating ZnT2 in the regulation of mammary differentiation and optimal milk production during lactation. Together, these results demonstrate that ZnT2-mediated zinc transport is critical for mammary gland function, suggesting that defects in ZnT2 not only reduce milk zinc concentration but may compromise breast health and increase the risk for lactation insufficiency in lactating women.


Sujet(s)
Transporteurs de cations/physiologie , Lactation/physiologie , Glandes mammaires animales/croissance et développement , Zinc/métabolisme , Animaux , Transport biologique , Technique de Western , Prolifération cellulaire , Cellules cultivées , Femelle , Techniques immunoenzymatiques , Mâle , Glandes mammaires animales/métabolisme , Matrix metalloproteinase 2/métabolisme , Matrix metalloproteinase 9/métabolisme , Souris , Souris transgéniques , Lait/métabolisme
14.
J Biol Chem ; 289(34): 23653-61, 2014 Aug 22.
Article de Anglais | MEDLINE | ID: mdl-25016022

RÉSUMÉ

The zinc transporter ZnT2 imports zinc into secretory vesicles and regulates zinc export from the mammary epithelial cell. Mutations in ZnT2 substantially impair zinc secretion into milk. The lactogenic hormone prolactin (PRL) transcriptionally increases ZnT2 expression through the Jak2/STAT5 signaling pathway, increasing zinc accumulation in secretory vesicles and zinc secretion. Herein, we report that PRL post-translationally stimulated ZnT2 ubiquitination, which altered ZnT2 trafficking and augmented vesicular zinc accumulation and secretion from mammary epithelial cells in a transient manner. Ubiquitination then down-regulated zinc secretion by stimulating degradation of ZnT2. Mutagenesis of two N-terminal lysine residues (K4R and K6R) inhibited ZnT2 ubiquitination, vesicular zinc accumulation and secretion, and protein degradation. These findings establish that PRL post-translationally regulates ZnT2-mediated zinc secretion in a multifactorial manner, first by enhancing zinc accumulation in vesicles to transiently enhance zinc secretion and then by activating ubiquitin-dependent ZnT2 degradation. This provides insight into novel mechanisms through which ZnT2 and zinc transport is tightly regulated in mammary epithelial cells.


Sujet(s)
Transporteurs de cations/physiologie , Glandes mammaires animales/métabolisme , Prolactine/physiologie , Ubiquitination/physiologie , Animaux , Séquence nucléotidique , Transporteurs de cations/génétique , Transporteurs de cations/métabolisme , Lignée cellulaire , Cellules épithéliales/métabolisme , Femelle , Immunoprécipitation , Lysine/métabolisme , Glandes mammaires animales/cytologie , Souris , Maturation post-traductionnelle des protéines , Petit ARN interférent
15.
Exp Cell Res ; 321(2): 190-200, 2014 Feb 15.
Article de Anglais | MEDLINE | ID: mdl-24333596

RÉSUMÉ

Prolactin receptor (PRL-R) activation regulates cell differentiation, proliferation, cell survival and motility of breast cells. Prolactin (PRL) and PRL-R over-expression are strongly implicated in breast cancer, particularly contributing to tumor growth and invasion in the more aggressive estrogen-receptor negative (ER-) disease. PRL-R antagonists have been suggested as potential therapeutic agents; however, mechanisms through which PRL-R antagonists exert their actions are not well-understood. Zinc (Zn) is a regulatory factor for over 10% of the proteome, regulating critical cell processes such as proliferation, cell signaling, transcription, apoptosis and autophagy. PRL-R signaling regulates Zn metabolism in breast cells. Herein we determined effects of PRL-R attenuation on cellular Zn metabolism and cell function in a model of ER-, PRL-R over-expressing breast cancer cells (MDA-MB-453). PRL-R attenuation post-transcriptionally increased ZnT2 abundance and redistributed intracellular Zn pools into lysosomes and mitochondria. ZnT2-mediated lysosomal Zn sequestration was associated with reduced matrix metalloproteinase 2 (MMP-2) activity and decreased invasion. ZnT2-mediated Zn accumulation in mitochondria was associated with increased mitochondrial oxidation. Our results suggest that PRL-R antagonism in PRL-R over-expressing breast cancer cells may reduce invasion through the redistribution of intracellular Zn pools critical for cellular function.


Sujet(s)
Tumeurs du sein/anatomopathologie , Transporteurs de cations/physiologie , Antihormones/pharmacologie , Récepteur prolactine/antagonistes et inhibiteurs , Zinc/métabolisme , Transport biologique/effets des médicaments et des substances chimiques , Tumeurs du sein/génétique , Tumeurs du sein/métabolisme , Femelle , Humains , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Invasion tumorale , Oxydoréduction/effets des médicaments et des substances chimiques , Petit ARN interférent/génétique , Récepteur prolactine/génétique , Distribution tissulaire/effets des médicaments et des substances chimiques , Cellules cancéreuses en culture
16.
Exp Biol Med (Maywood) ; 239(2): 202-12, 2014 Feb.
Article de Anglais | MEDLINE | ID: mdl-24311739

RÉSUMÉ

Zinc (Zn) is an essential element for normal brain function; an abnormal Zn homeostasis in brain and the cerebrospinal fluid (CSF) has been implied in the etiology of Alzheimer's disease (AD). However, the mechanisms that regulate Zn transport in the blood-brain interface remain unknown. This study was designed to investigate Zn transport by the blood-CSF barrier (BCB) in the choroid plexus, with a particular focus on Zn transporter-2 (ZnT2), and to understand if lead (Pb) accumulation in the choroid plexus disturbed the Zn regulatory function in the BCB. Confocal microscopy, quantitative PCR and western blot demonstrated the presence of ZnT2 in the choroidal epithelia; ZnT2 was primarily in cytosol in freshly isolated plexus tissues but more toward the peripheral membrane in established choroidal Z310 cells. Exposure of rats to Pb (single ip injection of 50 mg Pb acetate/kg) for 24 h increased ZnT2 fluorescent signals in plexus tissues by confocal imaging and protein expression by western blot. Similar results were obtained by in vitro experiments using Z310 cells. Further studies using cultured cells and a two-chamber Transwell device showed that Pb treatment significantly reduced the cellular Zn concentration and led to an increased transport of Zn across the BCB, the effect that may be due to the increased ZnT2 by Pb exposure. Taken together, these results indicate that ZnT2 is present in the BCB; Pb exposure increases the ZnT2 expression in choroidal epithelial cells by a yet unknown mechanism and as a result, more Zn ions may be deposited into the intracellular Zn pool, leading to a relative Zn deficiency state in the cytoplasm at the BCB.


Sujet(s)
Transporteurs de cations/génétique , Plexus choroïde/effets des médicaments et des substances chimiques , Plomb/pharmacologie , Régulation positive/effets des médicaments et des substances chimiques , Zinc/métabolisme , Animaux , Transport biologique/effets des médicaments et des substances chimiques , Transporteurs de cations/métabolisme , Transporteurs de cations/physiologie , Lignée cellulaire , Plexus choroïde/métabolisme , Homéostasie , Mâle , Souris , Rats , Rat Sprague-Dawley , Zinc/sang , Zinc/liquide cérébrospinal
17.
Article de Chinois | WPRIM (Pacifique Occidental) | ID: wpr-559803

RÉSUMÉ

Objective:To investigate the effect of zinc supplementation on the expression profile of zinc transporters mRNA in placenta and small intestine of pregnant rats.Method:Eighteen pregnant SD rats were randomly divided into normal control(ZC)and zinc supplement group(ZS).Rats in ZC group drank deionized distilled water while those in ZS group drank water supplemented with zinc of 1.26 mmol/L.Placenta and small intestine were taken at gestational day 9.5 and 17.5,respectively.The expression of ZnT1,2,5 and 6 mRNA was examined by RT-PCR.Results:At gestational D9.5,the expression of placental ZnT1,2 and small intestinal ZnT1,2,6 mRNA was up-regulated,and placental ZnT5 mRNA down-regulated by dietary zinc supplementation.Dietary zinc intake had no effect on the expression of placental ZnT6 and small intestinal ZT5 mRNA.At gestational D17.5,the expression of placental ZnT5 and 6 mRNA was up-regulated by dietary zinc supplementation,and dietary zinc had no effect on the expression of placental and small intestinal ZnT1,2 mRNA.The expression of ZnT5 mRNA at gestational D17.5 in both groups was not detectable.Conclusion:Dietary zinc supplementation during pregnancy has significant effect on the expression profile of ZnT 1,2,5 and 6mRNA in placenta and small intestine of pregnant rats.

SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE