1
|
Wang X, Zhang M, Ma J, Tie Y, Wang S. Biochemical Markers of Zinc Nutrition. Biol Trace Elem Res 2024; 202:5328-5338. [PMID: 38319550 DOI: 10.1007/s12011-024-04091-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 01/29/2024] [Indexed: 02/07/2024]
Abstract
Zinc is an important trace element involved in the biochemical and physiological functions of the organism and is essential in the human body. It has been reported that 17.3% of people around the world are at risk of many diseases due to zinc deficiency, which has already affected people's healthy lives. Currently, mild zinc deficiency is difficult to diagnose early due to the lack of typical clinical manifestations, so finding zinc biomarkers is crucial for people's health. The present article reviews the main representative zinc biomarkers, such as body fluid zinc levels, zinc-dependent proteins, tissue zinc, and zinc-containing enzymes, to provide a reference for actively promoting the study of zinc nutritional status and early clinical diagnosis.
Collapse
Affiliation(s)
- Xinying Wang
- North China University of Science and Technology, Tangshan, Hebei Province, 063210, China
| | - Menghui Zhang
- North China University of Science and Technology, Tangshan, Hebei Province, 063210, China
| | - Jing Ma
- Hebei Key Laboratory of Reproductive Medicine, Hebei Reproductive Health Hospital, Shijiazhuang, Hebei Province, 050071, China
| | - Yanqing Tie
- Hebei General Hospital, Shijiazhuang, Hebei Province, 050051, China.
| | - Shusong Wang
- Hebei Key Laboratory of Reproductive Medicine, Hebei Reproductive Health Hospital, Shijiazhuang, Hebei Province, 050071, China.
| |
Collapse
|
2
|
Dorward AM, Stewart AJ, Pitt SJ. The role of Zn2+ in shaping intracellular Ca2+ dynamics in the heart. J Gen Physiol 2023; 155:e202213206. [PMID: 37326614 PMCID: PMC10276528 DOI: 10.1085/jgp.202213206] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 04/18/2023] [Accepted: 05/22/2023] [Indexed: 06/17/2023] Open
Abstract
Increasing evidence suggests that Zn2+ acts as a second messenger capable of transducing extracellular stimuli into intracellular signaling events. The importance of Zn2+ as a signaling molecule in cardiovascular functioning is gaining traction. In the heart, Zn2+ plays important roles in excitation-contraction (EC) coupling, excitation-transcription coupling, and cardiac ventricular morphogenesis. Zn2+ homeostasis in cardiac tissue is tightly regulated through the action of a combination of transporters, buffers, and sensors. Zn2+ mishandling is a common feature of various cardiovascular diseases. However, the precise mechanisms controlling the intracellular distribution of Zn2+ and its variations during normal cardiac function and during pathological conditions are not fully understood. In this review, we consider the major pathways by which the concentration of intracellular Zn2+ is regulated in the heart, the role of Zn2+ in EC coupling, and discuss how Zn2+ dyshomeostasis resulting from altered expression levels and efficacy of Zn2+ regulatory proteins are key drivers in the progression of cardiac dysfunction.
Collapse
Affiliation(s)
- Amy M. Dorward
- School of Medicine, University of St Andrews, St Andrews, UK
| | - Alan J. Stewart
- School of Medicine, University of St Andrews, St Andrews, UK
| | | |
Collapse
|
3
|
Ma C, Gong C. Considerations in production of the prokaryotic ZIP family transporters for structural and functional studies. Methods Enzymol 2023; 687:1-30. [PMID: 37666628 DOI: 10.1016/bs.mie.2023.04.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2023]
Abstract
Zinc ions play essential roles as components of enzymes and many other important biomolecules, and are associated with numerous diseases. The uptake of Zn2+ and other metal ions require a widely distributed transporter protein family called Zrt/Irt-like Proteins (ZIP family), the majority members of which tend to have eight transmembrane helices with both N- and C- termini located on the extracellular or periplasmic side. Their small sizes and dynamic conformations bring many difficulties in their production for structural studies either by crystallography or Cryo-EM. Here, we summarize the problems that may encounter at the various steps of processing the ZIP proteins from gene to structural and functional studies, and provide some solutions and examples from our and other labs for the cloning, expression, purification, stability screening, metal ion transport assays and structural studies of prokaryotic ZIP family transporters using Escherichia coli as a heterologous host.
Collapse
Affiliation(s)
- Cheng Ma
- Protein Facility, Zhejiang University School of Medicine, Hangzhou, P.R. China; The First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, P.R. China.
| | - Caixia Gong
- The First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, P.R. China; Zhejiang Provincial Key Laboratory for Diagnosis and Treatment of Aging and Physic-chemical Injury Diseases, Hangzhou, P.R. China.
| |
Collapse
|
4
|
Kelleher SL. The ins and outs of mammary gland calcium and zinc transport: A brief review. JDS COMMUNICATIONS 2023; 4:240-244. [PMID: 37360130 PMCID: PMC10285217 DOI: 10.3168/jdsc.2022-0291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 08/31/2022] [Indexed: 06/28/2023]
Abstract
Milk is an excellent source of all macrominerals and trace elements, which are essential for proper function of a wide variety of vital processes. The concentrations of minerals in milk are influenced by numerous factors, including stage of lactation, time of day, nutritional and health status of the mother, as well as maternal genotype and environmental exposures. Additionally, tight regulation of mineral transport within the secretory mammary epithelial cell itself is critical for the production and secretion of milk. In this brief review, we focus on the current understanding of how the essential divalent cations calcium (Ca) and zinc (Zn) are transported in the mammary gland (MG) with a focus on molecular regulation and the consequence of genotype. A deeper grasp of mechanisms and factors affecting Ca and Zn transport in the MG is important to understanding milk production, mineral output, and MG health to inform intervention design and novel diagnostic and therapeutic strategies in production animals and humans.
Collapse
|
5
|
Tan HW, Xu YM, Liang ZL, Cai NL, Wu YY, Lau ATY. Single-gene knockout-coupled omics analysis identifies C9orf85 and CXorf38 as two uncharacterized human proteins associated with ZIP8 malfunction. Front Mol Biosci 2022; 9:991308. [PMID: 36330220 PMCID: PMC9623088 DOI: 10.3389/fmolb.2022.991308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 09/13/2022] [Indexed: 02/05/2023] Open
Abstract
Human transmembrane protein metal cation symporter ZIP8 (SLC39A8) is a member of the solute carrier gene family responsible for intracellular transportation of essential micronutrients, including manganese, selenium, and zinc. Previously, we established a ZIP8-knockout (KO) human cell model using the CRISPR/Cas9 system and explored how the expression of ZIP8 could possibly contribute to a wide range of human diseases. To further assess the biophysiological role of ZIP8, in the current study, we employed isobaric tags for relative and absolute quantitation (iTRAQ) and detected the changes of the proteome in ZIP8-KO cells (proteomic data are available via ProteomeXchange with identifier PXD036680). A total of 286 differentially expressed proteins (206 downregulated and 80 upregulated proteins) were detected in the ZIP8-KO cell model, and subsequent bioinformatics analyses (GO, KEGG, KOG, and PPI) were performed on these proteins. Interestingly, four "uncharacterized" proteins (proteins with unknown biological function) were identified in the differentially expressed proteins: C1orf198, C9orf85, C17orf75, and CXorf38-all of which were under-expressed in the ZIP8-KO cells. Notably, C9orf85 and CXorf38 were amongst the top-10 most downregulated proteins, and their expressions could be selectively induced by essential micronutrients. Furthermore, clinical-based bioinformatic analysis indicated that positive correlations between the gene expressions of ZIP8 and C9orf85 or CXorf38 were observed in multiple cancer types. Overall, this study reveals the proteomic landscape of cells with impaired ZIP8 and uncovers the potential relationships between essential micronutrients and uncharacterized proteins C9orf85 and CXorf38. The differentially expressed proteins identified in ZIP8-KO cells could be the potential targets for diagnosing and/or treating human ZIP8-associated diseases, including but not limited to malnutrition, viral infection, and cancers.
Collapse
Affiliation(s)
- Heng Wee Tan
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong, China
| | | | | | | | | | - Andy T. Y. Lau
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong, China
| |
Collapse
|
6
|
Olea-Flores M, Kan J, Carlson A, Syed SA, McCann C, Mondal V, Szady C, Ricker HM, McQueen A, Navea JG, Caromile LA, Padilla-Benavides T. ZIP11 Regulates Nuclear Zinc Homeostasis in HeLa Cells and Is Required for Proliferation and Establishment of the Carcinogenic Phenotype. Front Cell Dev Biol 2022; 10:895433. [PMID: 35898402 PMCID: PMC9309433 DOI: 10.3389/fcell.2022.895433] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 06/13/2022] [Indexed: 12/12/2022] Open
Abstract
Zinc (Zn) is an essential trace element that plays a key role in several biological processes, including transcription, signaling, and catalysis. A subcellular network of transporters ensures adequate distribution of Zn to facilitate homeostasis. Among these are a family of importers, the Zrt/Irt-like proteins (ZIP), which consists of 14 members (ZIP1-ZIP14) that mobilize Zn from the extracellular domain and organelles into the cytosol. Expression of these transporters varies among tissues and during developmental stages, and their distribution at various cellular locations is essential for defining the net cellular Zn transport. Normally, the ion is bound to proteins or sequestered in organelles and vesicles. However, though research has focused on Zn internalization in mammalian cells, little is known about Zn mobilization within organelles, including within the nuclei under both normal and pathological conditions. Analyses from stomach and colon tissues isolated from mouse suggested that ZIP11 is the only ZIP transporter localized to the nucleus of mammalian cells, yet no clear cellular role has been attributed to this protein. We hypothesized that ZIP11 is essential to maintaining nuclear Zn homeostasis in mammalian cells. To test this, we utilized HeLa cells, as research in humans correlated elevated expression of ZIP11 with poor prognosis in cervical cancer patients. We stably knocked down ZIP11 in HeLa cancer cells and investigated the effect of Zn dysregulation in vitro. Our data show that ZIP11 knockdown (KD) reduced HeLa cells proliferation due to nuclear accumulation of Zn. RNA-seq analyses revealed that genes related to angiogenesis, apoptosis, mRNA metabolism, and signaling pathways are dysregulated. Although the KD cells undergoing nuclear Zn stress can activate the homeostasis response by MTF1 and MT1, the RNA-seq analyses showed that only ZIP14 (an importer expressed on the plasma membrane and endocytic vesicles) is mildly induced, which may explain the sensitivity to elevated levels of extracellular Zn. Consequently, ZIP11 KD HeLa cells have impaired migration, invasive properties and decreased mitochondrial potential. Furthermore, KD of ZIP11 delayed cell cycle progression and rendered an enhanced senescent state in HeLa cells, pointing to a novel mechanism whereby maintenance of nuclear Zn homeostasis is essential for cancer progression.
Collapse
Affiliation(s)
- Monserrat Olea-Flores
- Department of Molecular Biology and Biochemistry, Wesleyan University, Middletown, CT, United States
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA, United States
| | - Julia Kan
- Department of Molecular Biology and Biochemistry, Wesleyan University, Middletown, CT, United States
| | - Alyssa Carlson
- Department of Molecular Biology and Biochemistry, Wesleyan University, Middletown, CT, United States
| | - Sabriya A. Syed
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA, United States
| | - Cat McCann
- Department of Molecular Biology and Biochemistry, Wesleyan University, Middletown, CT, United States
| | - Varsha Mondal
- Department of Molecular Biology and Biochemistry, Wesleyan University, Middletown, CT, United States
| | - Cecily Szady
- Department of Chemistry, Skidmore College, Saratoga Springs, NY, United States
| | - Heather M. Ricker
- Department of Chemistry, Skidmore College, Saratoga Springs, NY, United States
| | - Amy McQueen
- Department of Molecular Biology and Biochemistry, Wesleyan University, Middletown, CT, United States
| | - Juan G. Navea
- Department of Chemistry, Skidmore College, Saratoga Springs, NY, United States
| | - Leslie A. Caromile
- Department of Cell Biology, Center for Vascular Biology, UCONN Health-Center, Farmington, CT, United States
| | - Teresita Padilla-Benavides
- Department of Molecular Biology and Biochemistry, Wesleyan University, Middletown, CT, United States
- *Correspondence: Teresita Padilla-Benavides,
| |
Collapse
|
7
|
Samuelson DR, Haq S, Knoell DL. Divalent Metal Uptake and the Role of ZIP8 in Host Defense Against Pathogens. Front Cell Dev Biol 2022; 10:924820. [PMID: 35832795 PMCID: PMC9273032 DOI: 10.3389/fcell.2022.924820] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 05/26/2022] [Indexed: 01/13/2023] Open
Abstract
Manganese (Mn) and Zinc (Zn) are essential micronutrients whose concentration and location within cells are tightly regulated at the onset of infection. Two families of Zn transporters (ZIPs and ZnTs) are largely responsible for regulation of cytosolic Zn levels and to a certain extent, Mn levels, although much less is known regarding Mn. The capacity of pathogens to persevere also depends on access to micronutrients, yet a fundamental gap in knowledge remains regarding the importance of metal exchange at the host interface, often referred to as nutritional immunity. ZIP8, one of 14 ZIPs, is a pivotal importer of both Zn and Mn, yet much remains to be known. Dietary Zn deficiency is common and commonly occurring polymorphic variants of ZIP8 that decrease cellular metal uptake (Zn and Mn), are associated with increased susceptibility to infection. Strikingly, ZIP8 is the only Zn transporter that is highly induced following bacterial exposure in key immune cells involved with host defense against leading pathogens. We postulate that mobilization of Zn and Mn into key cells orchestrates the innate immune response through regulation of fundamental defense mechanisms that include phagocytosis, signal transduction, and production of soluble host defense factors including cytokines and chemokines. New evidence also suggests that host metal uptake may have long-term consequences by influencing the adaptive immune response. Given that activation of ZIP8 expression by pathogens has been shown to influence parenchymal, myeloid, and lymphoid cells, the impact applies to all mucosal surfaces and tissue compartments that are vulnerable to infection. We also predict that perturbations in metal homeostasis, either genetic- or dietary-induced, has the potential to impact bacterial communities in the host thereby adversely impacting microbiome composition. This review will focus on Zn and Mn transport via ZIP8, and how this vital metal transporter serves as a "go to" conductor of metal uptake that bolsters host defense against pathogens. We will also leverage past studies to underscore areas for future research to better understand the Zn-, Mn- and ZIP8-dependent host response to infection to foster new micronutrient-based intervention strategies to improve our ability to prevent or treat commonly occurring infectious disease.
Collapse
Affiliation(s)
- Derrick R. Samuelson
- Division of Pulmonary, Critical Care, and Sleep, Department of Internal Medicine, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Sabah Haq
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, United States
| | - Daren L. Knoell
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, United States,*Correspondence: Daren L. Knoell,
| |
Collapse
|
8
|
Willekens J, Runnels LW. Impact of Zinc Transport Mechanisms on Embryonic and Brain Development. Nutrients 2022; 14:2526. [PMID: 35745255 PMCID: PMC9231024 DOI: 10.3390/nu14122526] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/09/2022] [Accepted: 06/09/2022] [Indexed: 12/04/2022] Open
Abstract
The trace element zinc (Zn) binds to over ten percent of proteins in eukaryotic cells. Zn flexible chemistry allows it to regulate the activity of hundreds of enzymes and influence scores of metabolic processes in cells throughout the body. Deficiency of Zn in humans has a profound effect on development and in adults later in life, particularly in the brain, where Zn deficiency is linked to several neurological disorders. In this review, we will summarize the importance of Zn during development through a description of the outcomes of both genetic and early dietary Zn deficiency, focusing on the pathological consequences on the whole body and brain. The epidemiology and the symptomology of Zn deficiency in humans will be described, including the most studied inherited Zn deficiency disease, Acrodermatitis enteropathica. In addition, we will give an overview of the different forms and animal models of Zn deficiency, as well as the 24 Zn transporters, distributed into two families: the ZIPs and the ZnTs, which control the balance of Zn throughout the body. Lastly, we will describe the TRPM7 ion channel, which was recently shown to contribute to intestinal Zn absorption and has its own significant impact on early embryonic development.
Collapse
Affiliation(s)
| | - Loren W. Runnels
- Department of Pharmacology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA;
| |
Collapse
|
9
|
Golgi Metal Ion Homeostasis in Human Health and Diseases. Cells 2022; 11:cells11020289. [PMID: 35053405 PMCID: PMC8773785 DOI: 10.3390/cells11020289] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/31/2021] [Accepted: 01/11/2022] [Indexed: 12/24/2022] Open
Abstract
The Golgi apparatus is a membrane organelle located in the center of the protein processing and trafficking pathway. It consists of sub-compartments with distinct biochemical compositions and functions. Main functions of the Golgi, including membrane trafficking, protein glycosylation, and sorting, require a well-maintained stable microenvironment in the sub-compartments of the Golgi, along with metal ion homeostasis. Metal ions, such as Ca2+, Mn2+, Zn2+, and Cu2+, are important cofactors of many Golgi resident glycosylation enzymes. The homeostasis of metal ions in the secretory pathway, which is required for proper function and stress response of the Golgi, is tightly regulated and maintained by transporters. Mutations in the transporters cause human diseases. Here we provide a review specifically focusing on the transporters that maintain Golgi metal ion homeostasis under physiological conditions and their alterations in diseases.
Collapse
|
10
|
Zinc Signaling in the Mammary Gland: For Better and for Worse. Biomedicines 2021; 9:biomedicines9091204. [PMID: 34572390 PMCID: PMC8469023 DOI: 10.3390/biomedicines9091204] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/06/2021] [Accepted: 09/06/2021] [Indexed: 02/07/2023] Open
Abstract
Zinc (Zn2+) plays an essential role in epithelial physiology. Among its many effects, most prominent is its action to accelerate cell proliferation, thereby modulating wound healing. It also mediates affects in the gastrointestinal system, in the testes, and in secretory organs, including the pancreas, salivary, and prostate glands. On the cellular level, Zn2+ is involved in protein folding, DNA, and RNA synthesis, and in the function of numerous enzymes. In the mammary gland, Zn2+ accumulation in maternal milk is essential for supporting infant growth during the neonatal period. Importantly, Zn2+ signaling also has direct roles in controlling mammary gland development or, alternatively, involution. During breast cancer progression, accumulation or redistribution of Zn2+ occurs in the mammary gland, with aberrant Zn2+ signaling observed in the malignant cells. Here, we review the current understanding of the role of in Zn2+ the mammary gland, and the proteins controlling cellular Zn2+ homeostasis and signaling, including Zn2+ transporters and the Gq-coupled Zn2+ sensing receptor, ZnR/GPR39. Significant advances in our understanding of Zn2+ signaling in the normal mammary gland as well as in the context of breast cancer provides new avenues for identification of specific targets for breast cancer therapy.
Collapse
|
11
|
Zhu B, Huo R, Zhi Q, Zhan M, Chen X, Hua ZC. Increased expression of zinc transporter ZIP4, ZIP11, ZnT1, and ZnT6 predicts poor prognosis in pancreatic cancer. J Trace Elem Med Biol 2021; 65:126734. [PMID: 33631610 DOI: 10.1016/j.jtemb.2021.126734] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 02/11/2021] [Accepted: 02/15/2021] [Indexed: 01/19/2023]
Abstract
INTRODUCTION Zinc homeostasis is regulated by SLC39A/ZIP, SLC30A/ZnT, and metallothionein (MT) families in human cells. Zinc dyshomeostasis may affect or be affected by the abnormal behavior of cancer cells. Although decreased serum zinc levels are observed in patients with pancreatic adenocarcinoma (PAAD), limited information is available regarding the expression pattern and prognostic roles of zinc homeostasis-related genes in PAAD. OBJECTIVES The primary objective of this study was to explore the expression pattern and prognostic roles of zinc homeostasis-related genes in PAAD. METHODS The expression pattern of 35 known zinc homeostasis-related genes in PAAD was systemically explored based on RNA-sequencing data from the Cancer Genome Atlas (TCGA) and the Genotype-Tissue Expression (GTEx) projects. The association between the expression levels of zinc homeostasis-related genes and survival of PAAD patients was evaluated using the Kaplan-Meier method and the log-rank test. Expressional correlation between zinc homeostasis-related genes with potential prognostic value in PAAD and normal pancreatic controls was evaluated using Pearson's correlation analysis. Functional enrichment analyses were performed to elucidate possible mechanisms for the potential prognostic and therapeutic roles of these zinc homeostasis-related genes in PAAD. Effects of ZIP11, ZnT1, or ZnT6 knockdown on the proliferation and the migration of Capan-1 pancreatic cancer cells were assessed by the CCK-8 assay and the wound healing assay respectively. RESULTS We demonstrated that the expression levels of ZIP1, ZIP3, ZIP4, ZIP6, ZIP7, ZIP9, ZIP10, ZIP11, ZIP13, ZnT1, ZnT5, ZnT6, ZnT7, and ZnT9 were increased, whereas the expression levels of ZIP5, ZIP14, ZnT2, MT1 G, MT1H, and MT1X were decreased in PAAD tumors compared with normal pancreatic controls. Among these differentially-expressed genes related to zinc homeostasis, higher expression of ZIP4, ZIP11, ZnT1 or ZnT6 predicted poorer prognosis with the possible involvement of several cancer-related processes and pathways in PAAD patients. We further demonstrated that knockdown of ZIP11 attenuated Capan-1 cell proliferation with decreased activation of ERK1/2 pathway; knockdown of ZnT1 attenuated Capan-1 cell proliferation with decreased activation of ERK1/2, p38 MAPK, NF-kB, and mTOR pathways; knockdown of ZnT6 attenuated Capan-1 cell proliferation with decreased activation of ERK1/2, p38 MAPK, and NF-kB pathways. CONCLUSIONS Higher expression of the zinc transporter ZIP4, ZIP11, ZnT1 or ZnT6 predicted poorer prognosis in patients with PAAD. These findings provide new clues for understanding the complex relationship between zinc homeostasis and pancreatic cancer.
Collapse
Affiliation(s)
- Bo Zhu
- School of Biopharmacy, China Pharmaceutical University, Nanjing 211198, PR China; School of Medicine and Holistic Integrative Medicine and Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210023, PR China; School of Life Sciences, Nanjing University, Nanjing 210023, PR China.
| | - Ruwei Huo
- School of Medicine and Holistic Integrative Medicine and Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Qi Zhi
- School of Medicine and Holistic Integrative Medicine and Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Mingjie Zhan
- School of Biopharmacy, China Pharmaceutical University, Nanjing 211198, PR China
| | - Xiao Chen
- School of Medicine and Holistic Integrative Medicine and Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210023, PR China; School of Life Sciences, Nanjing University, Nanjing 210023, PR China
| | - Zi-Chun Hua
- School of Biopharmacy, China Pharmaceutical University, Nanjing 211198, PR China; School of Life Sciences, Nanjing University, Nanjing 210023, PR China; Changzhou High-Tech Research Institute of Nanjing University and Jiangsu Target Pharma Laboratories Inc., Changzhou 213164, PR China.
| |
Collapse
|
12
|
Han Y, Sanford L, Simpson DM, Dowell RD, Palmer AE. Remodeling of Zn 2+ homeostasis upon differentiation of mammary epithelial cells. Metallomics 2021; 12:346-362. [PMID: 31950952 DOI: 10.1039/c9mt00301k] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Zinc is the second most abundant transition metal in humans and an essential nutrient required for growth and development of newborns. During lactation, mammary epithelial cells differentiate into a secretory phenotype, uptake zinc from blood circulation, and export it into mother's milk. At the cellular level, many zinc-dependent cellular processes, such as transcription, metabolism of nutrients, and proliferation are involved in the differentiation of mammary epithelial cells. Using mouse mammary epithelial cells as a model system, we investigated the remodeling of zinc homeostasis during differentiation induced by treatment with the lactogenic hormones cortisol and prolactin. RNA-Seq at different stages of differentiation revealed changes in global gene expression, including genes encoding zinc-dependent proteins and regulators of zinc homeostasis. Increases in mRNA levels of three zinc homeostasis genes, Slc39a14 (ZIP14) and metallothioneins (MTs) I and II were induced by cortisol but not by prolactin. The cortisol-induced increase was partially mediated by the nuclear glucocorticoid receptor signaling pathway. An increase in the cytosolic labile Zn2+ pool was also detected in lactating mammary cells, consistent with upregulation of MTs. We found that the zinc transporter ZIP14 was important for the expression of a major milk protein, whey acid protein (WAP), as knockdown of ZIP14 dramatically decreased WAP mRNA levels. In summary, our study demonstrated remodeling of zinc homeostasis upon differentiation of mammary epithelial cells resulting in changes in cytosolic Zn2+ and differential expression of zinc homeostasis genes, and these changes are important for establishing the lactation phenotype.
Collapse
Affiliation(s)
- Yu Han
- Department of Biochemistry, University of Colorado Boulder, 3415 Colorado Ave., Boulder, CO 80303, USA. and BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, USA
| | - Lynn Sanford
- Department of Biochemistry, University of Colorado Boulder, 3415 Colorado Ave., Boulder, CO 80303, USA. and BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, USA
| | - David M Simpson
- Department of Biochemistry, University of Colorado Boulder, 3415 Colorado Ave., Boulder, CO 80303, USA. and BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, USA
| | - Robin D Dowell
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, USA and Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Amy E Palmer
- Department of Biochemistry, University of Colorado Boulder, 3415 Colorado Ave., Boulder, CO 80303, USA. and BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, USA
| |
Collapse
|
13
|
The role of labile Zn 2+ and Zn 2+-transporters in the pathophysiology of mitochondria dysfunction in cardiomyocytes. Mol Cell Biochem 2020; 476:971-989. [PMID: 33225416 DOI: 10.1007/s11010-020-03964-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 10/23/2020] [Indexed: 02/06/2023]
Abstract
An important energy supplier of cardiomyocytes is mitochondria, similar to other mammalian cells. Studies have demonstrated that any defect in the normal processes controlled by mitochondria can lead to abnormal ROS production, thereby high oxidative stress as well as lack of ATP. Taken into consideration, the relationship between mitochondrial dysfunction and overproduction of ROS as well as the relation between increased ROS and high-level release of intracellular labile Zn2+, those bring into consideration the importance of the events related with those stimuli in cardiomyocytes responsible from cellular Zn2+-homeostasis and responsible Zn2+-transporters associated with the Zn2+-homeostasis and Zn2+-signaling. Zn2+-signaling, controlled by cellular Zn2+-homeostatic mechanisms, is regulated with intracellular labile Zn2+ levels, which are controlled, especially, with the two Zn2+-transporter families; ZIPs and ZnTs. Our experimental studies in mammalian cardiomyocytes and human heart tissue showed that Zn2+-transporters localizes to mitochondria besides sarco(endo)plasmic reticulum and Golgi under physiological condition. The protein levels as well as functions of those transporters can re-distribute under pathological conditions, therefore, they can interplay among organelles in cardiomyocytes to adjust a proper intracellular labile Zn2+ level. In the present review, we aimed to summarize the already known Zn2+-transporters localize to mitochondria and function to stabilize not only the cellular Zn2+ level but also cellular oxidative stress status. In conclusion, one can propose that a detailed understanding of cellular Zn2+-homeostasis and Zn2+-signaling through mitochondria may emphasize the importance of new mitochondria-targeting agents for prevention and/or therapy of cardiovascular dysfunction in humans.
Collapse
|
14
|
Growth Modulatory Role of Zinc in Prostate Cancer and Application to Cancer Therapeutics. Int J Mol Sci 2020; 21:ijms21082991. [PMID: 32340289 PMCID: PMC7216164 DOI: 10.3390/ijms21082991] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 04/16/2020] [Accepted: 04/18/2020] [Indexed: 02/06/2023] Open
Abstract
Zinc is a group IIB heavy metal. It is an important regulator of major cell signaling pathways in most mammalian cells, functions as an antioxidant and plays a role in maintaining genomic stability. Zinc deficiency leads to severe diseases in the brain, pancreas, liver, kidneys and reproductive organs. Zinc loss occurs during tumor development in a variety of cancers. The prostate normally contains abundant intracellular zinc and zinc loss is a hallmark of the development of prostate cancer development. The underlying mechanism of this loss is not clearly understood. The knowledge that excess zinc prevents the growth of prostate cancers suggests that zinc-mediated therapeutics could be an effective approach for cancer prevention and treatment, although challenges remain. This review summarizes the specific roles of zinc in several cancer types focusing on prostate cancer. The relationship between prostate cancer and the dysregulation of zinc homeostasis is examined in detail in an effort to understand the role of zinc in prostate cancer.
Collapse
|
15
|
Thokala S, Bodiga VL, Kudle MR, Bodiga S. Comparative Response of Cardiomyocyte ZIPs and ZnTs to Extracellular Zinc and TPEN. Biol Trace Elem Res 2019; 192:297-307. [PMID: 30778755 DOI: 10.1007/s12011-019-01671-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Accepted: 02/07/2019] [Indexed: 12/19/2022]
Abstract
Intracellular zinc concentrations are tightly regulated by the coordinated regulation of ZIPs and ZnTs. Very little is known about the regulation of these transporters in cardiomyocytes, in response to extracellular zinc. Adult rat cardiomyocytes express ZnTs 1, 2, 5, and 9, in addition to ZIPs 1, 2, 3, 6, 7, 9, 10, 11, 13, and 14. We have determined the intracellular free zinc levels using Zinpyr-1 fluorescence and studied response of ZIP and ZnT mRNA by real-time PCR to the changes in extracellular zinc and TPEN in adult rat ventricular myocytes. TPEN downregulated ZnT1, ZnT2, and ZIP11 mRNAs but upregulated ZnT5, ZIP2, ZIP7, ZIP10, ZIP13, and ZIP14 mRNAs. Zinc supplementation upregulated ZnT1, ZnT2 mRNA but downregulated ZnT5, ZIP1, ZIP2, ZIP3, ZIP7, ZIP9, and ZIP10 mRNA. The negative regulation of ZIPs by zinc excess can be explained in terms of zinc homeostasis as these transporters may act to protect cells from zinc over accumulation by reducing zinc influx when the extracellular concentration of zinc is high. Similarly, the ZnT expression appears to be regulated to avoid loss of zinc from the intracellular milieu, under zinc-deficient conditions.
Collapse
Affiliation(s)
- Sandhya Thokala
- Department of Biochemistry, Kakatiya University, Vidyaranyapuri, Warangal Urban, Telangana, India
| | - Vijaya Lakshmi Bodiga
- Department of Biochemistry and Molecular Biology, Institute of Genetics and Hospital for Genetic Diseases, Osmania University, Begumpet, Hyderabad, Telangana, India
| | - Madhukar Rao Kudle
- Department of Biochemistry, Kakatiya University, Vidyaranyapuri, Warangal Urban, Telangana, India
| | - Sreedhar Bodiga
- Department of Biochemistry, Kakatiya University, Vidyaranyapuri, Warangal Urban, Telangana, India.
| |
Collapse
|
16
|
Nebert DW, Liu Z. SLC39A8 gene encoding a metal ion transporter: discovery and bench to bedside. Hum Genomics 2019; 13:51. [PMID: 31521203 PMCID: PMC6744627 DOI: 10.1186/s40246-019-0233-3] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 08/14/2019] [Indexed: 01/08/2023] Open
Abstract
SLC39A8 is an evolutionarily highly conserved gene that encodes the ZIP8 metal cation transporter in all vertebrates. SLC39A8 is ubiquitously expressed, including pluripotent embryonic stem cells; SLC39A8 expression occurs in every cell type examined. Uptake of ZIP8-mediated Mn2+, Zn2+, Fe2+, Se4+, and Co2+ represents endogenous functions-moving these cations into the cell. By way of mouse genetic differences, the phenotype of "subcutaneous cadmium-induced testicular necrosis" was assigned to the Cdm locus in the 1970s. This led to identification of the mouse Slc39a8 gene, its most closely related Slc39a14 gene, and creation of Slc39a8-overexpressing, Slc39a8(neo/neo) knockdown, and cell type-specific conditional knockout mouse lines; the Slc39a8(-/-) global knockout mouse is early-embryolethal. Slc39a8(neo/neo) hypomorphs die between gestational day 16.5 and postnatal day 1-exhibiting severe anemia, dysregulated hematopoiesis, hypoplastic spleen, dysorganogenesis, stunted growth, and hypomorphic limbs. Not surprisingly, genome-wide association studies subsequently revealed human SLC39A8-deficiency variants exhibiting striking pleiotropy-defects correlated with clinical disorders in virtually every organ, tissue, and cell-type: numerous developmental and congenital disorders, the immune system, cardiovascular system, kidney, lung, liver, coagulation system, central nervous system, musculoskeletal system, eye, and gastrointestinal tract. Traits with which SLC39A8-deficiency variants are currently associated include Mn2+-deficient hypoglycosylation; numerous birth defects; Leigh syndrome-like mitochondrial redox deficiency; decreased serum high-density lipoprotein-cholesterol levels; increased body mass index; greater risk of coronary artery disease, hypotension, cardiovascular death, allergy, ischemic stroke, schizophrenia, Parkinson disease, inflammatory bowel disease, Crohn disease, myopia, and adolescent idiopathic scoliosis; systemic lupus erythematosus with primary Sjögren syndrome; decreased height; and inadvertent participation in the inflammatory progression of osteoarthritis.
Collapse
Affiliation(s)
- Daniel W Nebert
- Department of Environmental Health and Center for Environmental Genetics, University of Cincinnati Medical Center, Cincinnati, OH, 45267-0056, USA.
- Division of Human Genetics, Department of Pediatrics & Molecular Developmental Biology, Cincinnati Children's Hospital, Cincinnati, OH, 45229-2899, USA.
| | - Zijuan Liu
- Department of Biological Sciences, Oakland University, Rochester, MI, 48309, USA
| |
Collapse
|
17
|
Milk-derived miRNA profiles elucidate molecular pathways that underlie breast dysfunction in women with common genetic variants in SLC30A2. Sci Rep 2019; 9:12686. [PMID: 31481661 PMCID: PMC6722070 DOI: 10.1038/s41598-019-48987-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 08/15/2019] [Indexed: 12/19/2022] Open
Abstract
Studies in humans and pre-clinical animal models show milk-derived miRNAs reflect mammary gland function during lactation. The zinc transporter SLC30A2/ZnT2 plays a critical role in mammary gland function; ZnT2-null mice have profound defects in mammary epithelial cell (MEC) polarity and secretion, resulting in sub-optimal lactation. Non-synonymous genetic variation in SLC30A2 is common in humans, and several common ZnT2 variants are associated with changes in milk components that suggest breast dysfunction in women. To identify novel mechanisms through which dysfunction might occur, milk-derived miRNA profiles were characterized in women harboring three common genetic variants in SLC30A2 (D103E, T288S, and Exon 7). Expression of ten miRNAs differed between genotypes, and contributed to distinct spatial separation. Studies in breast milk and cultured MECs confirmed expression of ZnT2 variants alters abundance of protein levels of several predicted mRNA targets critical for breast function (PRLR, VAMP7, and SOX4). Moreover, bioinformatic analysis identified two novel gene networks that may underlie normal MEC function. Thus, we propose that genetic variation in genes critical for normal breast function such as SLC30A2 has important implications for lactation performance in women, and that milk-derived miRNAs can be used to identify novel mechanisms and for diagnostic potential.
Collapse
|
18
|
Clancey E, Kiser JN, Moraes JGN, Dalton JC, Spencer TE, Neibergs HL. Genome-wide association analysis and gene set enrichment analysis with SNP data identify genes associated with 305-day milk yield in Holstein dairy cows. Anim Genet 2019; 50:254-258. [PMID: 30994194 DOI: 10.1111/age.12792] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/23/2018] [Indexed: 11/29/2022]
Abstract
Milk production traits, such as 305-day milk yield (305MY), have been under direct selection to improve production in dairy cows. Over the past 50 years, the average milk yield has nearly doubled, and over 56% of the increase is attributable to genetic improvement. As such, additional improvements in milk yield are still possible as new loci are identified. The objectives of this study were to detect SNPs and gene sets associated with 305MY in order to identify new candidate genes contributing to variation in milk production. A population of 781 primiparous Holstein cows from six central Washington dairies with records of 305MY and energy corrected milk were used to perform a genome-wide association analysis (GWAA) using the Illumina BovineHD BeadChip (777 962 SNPs) to identify QTL associated with 305MY (P < 1.0 × 10-5 ). A gene set enrichment analysis with SNP data (GSEA-SNP) was performed to identify gene sets (normalized enrichment score > 3.0) and leading edge genes (LEGs) influencing 305MY. The GWAA identified three QTL comprising 34 SNPs and 30 positional candidate genes. In the GSEA-SNP, five gene sets with 58 unique and 24 shared LEGs contributed to 305MY. Identification of QTL and LEGs associated with 305MY can provide additional targets for genomic selection to continue to improve 305MY in dairy cattle.
Collapse
Affiliation(s)
- E Clancey
- Department of Animal Sciences, Center for Reproductive Biology, Washington State University, PO Box 646310, Pullman, WA, 99164-6310, USA
| | - J N Kiser
- Department of Animal Sciences, Center for Reproductive Biology, Washington State University, PO Box 646310, Pullman, WA, 99164-6310, USA
| | - J G N Moraes
- Division of Animal Sciences, S158A Animal Sciences Research Center, University of Missouri, Columbia, MO, 65211, USA
| | - J C Dalton
- Department of Animal and Veterinary Sciences, Caldwell Research and Extension Center, University of Idaho, 1904 E Chicago St, Suite A, B, Caldwell, ID, 83605, USA
| | - T E Spencer
- Division of Animal Sciences, S158A Animal Sciences Research Center, University of Missouri, Columbia, MO, 65211, USA
| | - H L Neibergs
- Department of Animal Sciences, Center for Reproductive Biology, Washington State University, PO Box 646310, Pullman, WA, 99164-6310, USA
| |
Collapse
|
19
|
He B, Bortoluzzi C, King WD, Graugnard D, Dawson KA, Applegate TJ. Zinc source influences the gene expression of zinc transporters in jejunum and cecal tonsils during broiler challenge with Eimeria maxima and Clostridium perfringens. Poult Sci 2019; 98:1146-1152. [PMID: 30285259 DOI: 10.3382/ps/pey484] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 09/19/2018] [Indexed: 11/20/2022] Open
Abstract
The objective of this experiment was to study the effects of dietary zinc (Zn) source on gene expression of Zn transporters (metallothionein [MT], ZIP 3, 5, 8, 9, 10, 11, 13, and 14, and ZnT 1, 4, 5, 6, 7, 9, and 10) in the jejunum and cecal tonsils of broilers challenged with coccidia or coccidia plus Clostridium perfringens. A 2 × 2 factorial design was used with 2 Zn sources (90 mg Zn/kg from either ZnSO4 or an organic Zn, Bioplex® Zn) and challenged with approximately 5,000 oocysts of Eimeria maxima at 14 d of age with or without C. perfringens (108 CFU/bird) at 18, 19, and 20 d of age (8 pens per treatment and 8 birds per pen) after which 1 bird/pen was sampled at 21 d of age. In the jejunum, co-infection resulted in higher ZnT 5 and 6 gene expression, while organic Zn fed birds had lower ZIP 5 and 11, and higher ZnT1. Additionally, an interaction of challenge by Zn source was noted wherein ZnT10 was unaffected by the C. perfringens in the organic Zn treatment but was 2.7-fold lower in the co-infected ZnSO4 fed birds. S100A9 gene expression, a biomarker of inflammatory response in necrotic enteritis, increased 2 and 2.8-fold in the cecal tonsils and jejunum with the co-infection, respectively. Supplementation with organic Zn lowered S100A9 by 1.9 and 4.4-fold in the cecal tonsils and jejunum, respectively, when birds were supplemented with ZnSO4. Notably, MT, ZIP 3, 8, 9, 10, 13, or 14, and ZnT 4, 7, and 9 were unaffected by Zn source and/or method of challenge. An interaction of challenge by Zn source was also noted for serum Zn concentration, which was reduced when birds were challenged with C. perfringens and fed ZnSO4 but no difference between challenge method when birds were fed organic Zn. Based on the expression of ZnT and ZIP genes, more Zn trafficking due to treatment occured in the jejunum than cecal tonsils, but further studies are needed to ascertain how Zn source regulates intracellular free Zn concentrations and whole-body Zn status during an enteric challenge.
Collapse
Affiliation(s)
- B He
- Department of Poultry Science, The University of Georgia, Athens, GA 30602, USA.,Department of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - C Bortoluzzi
- Department of Poultry Science, The University of Georgia, Athens, GA 30602, USA
| | - W D King
- Alltech-University of Kentucky Nutrition Research Alliance, Lexington, KY 40356, USA
| | - D Graugnard
- Alltech-University of Kentucky Nutrition Research Alliance, Lexington, KY 40356, USA
| | - K A Dawson
- Alltech-University of Kentucky Nutrition Research Alliance, Lexington, KY 40356, USA
| | - T J Applegate
- Department of Poultry Science, The University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
20
|
Paskavitz AL, Quintana J, Cangussu D, Tavera-Montañez C, Xiao Y, Ortiz-Miranda S, Navea JG, Padilla-Benavides T. Differential expression of zinc transporters accompanies the differentiation of C2C12 myoblasts. J Trace Elem Med Biol 2018; 49:27-34. [PMID: 29895369 PMCID: PMC6082398 DOI: 10.1016/j.jtemb.2018.04.024] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 04/16/2018] [Accepted: 04/20/2018] [Indexed: 12/11/2022]
Abstract
Zinc transporters facilitate metal mobilization and compartmentalization, playing a key role in cellular development. Little is known about the mechanisms and pathways of Zn movement between Zn transporters and metalloproteins during myoblast differentiation. We analyzed the differential expression of ZIP and ZnT transporters during C2C12 myoblast differentiation. Zn transporters account for a transient decrease of intracellular Zn upon myogenesis induction followed by a gradual increase of Zn in myotubes. Considering the subcellular localization and function of each of the Zn transporters, our findings indicate that a fine regulation is necessary to maintain correct metal concentrations in the cytosol and subcellular compartments to avoid toxicity, maintain homeostasis, and for loading metalloproteins needed during myogenesis. This study advances our basic understanding of the complex Zn transport network during muscle differentiation.
Collapse
Affiliation(s)
- Amanda L Paskavitz
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA, 01605, USA; Department of Chemistry, Skidmore College, 815 North Broadway, Saratoga Springs, NY, 12866, USA
| | - Julia Quintana
- Department of Chemistry and Biochemistry, Worcester Polytechnic Institute, 100 Institute Road, Worcester, MA, 01609, USA
| | - Daniella Cangussu
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA, 01605, USA
| | - Cristina Tavera-Montañez
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA, 01605, USA; Department of Chemistry and Biochemistry, Worcester Polytechnic Institute, 100 Institute Road, Worcester, MA, 01609, USA
| | - Yao Xiao
- Department of Chemistry, Skidmore College, 815 North Broadway, Saratoga Springs, NY, 12866, USA
| | - Sonia Ortiz-Miranda
- Department of Neurobiology, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA, 01605, USA
| | - Juan G Navea
- Department of Chemistry, Skidmore College, 815 North Broadway, Saratoga Springs, NY, 12866, USA
| | - Teresita Padilla-Benavides
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA, 01605, USA.
| |
Collapse
|
21
|
Gartmann L, Wex T, Grüngreiff K, Reinhold D, Kalinski T, Malfertheiner P, Schütte K. Expression of zinc transporters ZIP4, ZIP14 and ZnT9 in hepatic carcinogenesis-An immunohistochemical study. J Trace Elem Med Biol 2018; 49:35-42. [PMID: 29895370 DOI: 10.1016/j.jtemb.2018.04.034] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 04/25/2018] [Accepted: 04/26/2018] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Dysregulation of both, systemic zinc levels and tissue-specific zinc transporters, is reported in chronic inflammatory and malignant liver disease (hepatocellular carcinoma, HCC). Aim of this study is to assess the expression level of three zinc transporters in liver tissue and HCC: ZIP4, ZIP14 and ZnT9. METHODS The study is based on tissue samples obtained from 138 patients with histologically proven HCC. Tissue specimens from tumor (n = 138) and extra-lesional specimens (n = 72) were assessed immunohistochemically for the expression of the three zinc transporters. Expression levels were semi-quantitatively scored and statistically analyzed with respect to the etiology of HCC (alcohol, AFLD; non-alcoholic fatty liver disease, NAFLD; virus-hepatitis, VH) and survival. RESULTS Overall, expression levels of ZIP4, ZIP14 and ZnT9 were significantly higher in HCC tissue than in adjacent extra-lesional liver tissue. Expression levels in tumor tissue and survival time revealed a negative correlation for ZIP4 and ZIP14, and in part for ZnT9 (nuclear staining) (p < 0.05), whereas cytoplasmic staining of ZnT9 did not correlate with survival. Furthermore, the expression level of ZIP4 in extra-lesional tissue showed inverse correlation with survival time. CONCLUSION The upregulation of zinc transporters in hepatic carcinogenesis and its negative correlation with survival time implies a regulatory/functional link between zinc-homeostasis and development/progression of HCC that deserves to be further explored.
Collapse
Affiliation(s)
- Laura Gartmann
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University, Leipziger Str. 44, D-39120 Magdeburg, Germany; Medical Laboratory for Clinical Chemistry, Microbiology and Infectious Diseases "Prof. Schenk/Dr. Ansorge & Colleagues", Department Molecular Genetics, Schwiesaustr. 11, D-39124, Magdeburg, Germany
| | - Thomas Wex
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University, Leipziger Str. 44, D-39120 Magdeburg, Germany; Medical Laboratory for Clinical Chemistry, Microbiology and Infectious Diseases "Prof. Schenk/Dr. Ansorge & Colleagues", Department Molecular Genetics, Schwiesaustr. 11, D-39124, Magdeburg, Germany
| | - Kurt Grüngreiff
- Clinic of Gastroenterology, City Hospital Magdeburg, Klinikum Magdeburg GmbH, Birkenallee 34, D-39130, Magdeburg, Germany
| | - Dirk Reinhold
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke University, Leipziger Str. 44, D-39120 Magdeburg, Germany
| | - Thomas Kalinski
- Institute of Pathology, Otto-von-Guericke University, Leipziger Str. 44, D-39120, Magdeburg, Germany
| | - Peter Malfertheiner
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University, Leipziger Str. 44, D-39120 Magdeburg, Germany
| | - Kerstin Schütte
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University, Leipziger Str. 44, D-39120 Magdeburg, Germany; Department of Internal Medicine and Gastroenterology, Niels-Stensen-Kliniken, Marienhospital Osnabrück GmbH, Bischofsstraße 1, D-49074, Osnabrück, Germany.
| |
Collapse
|
22
|
Ha E, Bae JH. Zinc transporter SLC39A11 polymorphisms are associated with chronic gastritis in the Korean population: the possible effect on spicy food intake. Nutr Res 2018; 57:78-85. [PMID: 30122198 DOI: 10.1016/j.nutres.2018.04.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 01/23/2018] [Accepted: 04/17/2018] [Indexed: 12/13/2022]
Abstract
Herein, we hypothesized that ZIP11 variants would be important risk factors for chronic gastritis and that there would be an interaction effect of the relationship between their variants and spicy food intake on the development of chronic gastritis. Participants in this cross-sectional study (n = 3882 + 252) were recruited from a cohort of the Korean Genome and Epidemiology Study in 2001. Age, sex, education, smoking and drinking status, exercise, stress, and income level of all participants were determined by a questionnaire. Demographic and anthropometric data were collected. Fasting blood samples were collected to determine the serum levels of glucose, insulin, total bilirubin, total cholesterol, high-density lipoprotein cholesterol, and triglycerides. The presence of chronic gastritis was defined as a confirmed diagnosis by a physician. Food consumption was determined using a semiquantitative food frequency questionnaire. We found 8 different single nucleotide polymorphisms (SNPs) that are significantly different between subjects without gastritis and those with gastritis. Of these 8 SNPs, 3 SNP (rs17183225 [C/T], rs17780814 [A/C], and rs17780820 [A/G]) are closely located in the intronic region of zinc transporter SLC39A11, commonly known as ZIP11, and show linkage disequilibrium (D' = 1.0). We also found that participants with (TCA + TCG) haplotype of ZIP11 at high levels of dietary intake of spicy foods show a significantly increasing tendency in the odds of having chronic gastritis when compared with those with CAA haplotype (odds ratio, 2.620; 95% confidence interval, 1.207-5.689). The data indicate positive associations between higher meal frequency and lower spicy food preference and gastritis. In conclusion, we found that zinc transporter gene ZIP11 is associated with chronic gastritis in the Korean population and it may interact with spicy food, which suggests ZIP11 as a therapeutic target for precision nutrition.
Collapse
Affiliation(s)
- Eunyoung Ha
- Department of Biochemistry, School of Medicine, Keimyung University, Daegu, Republic of Korea
| | - Ji-Hyun Bae
- Department of Food Science and Nutrition, College of Natural Sciences, Keimyung University, Daegu, Republic of Korea.
| |
Collapse
|
23
|
Bowers K, Srai SKS. The trafficking of metal ion transporters of the Zrt- and Irt-like protein family. Traffic 2018; 19:813-822. [PMID: 29952128 DOI: 10.1111/tra.12602] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 06/25/2018] [Accepted: 06/25/2018] [Indexed: 12/28/2022]
Abstract
Metal ion transporters of the Zrt- and Irt-like protein (ZIP, or SLC39A) family transport zinc, iron, manganese and/or cadmium across cellular membranes and into the cytosol. The 14 human ZIP family proteins are expressed in a wide variety of tissues and function in many different cellular processes. Many of these proteins (including ZIP1, 2, 3, 4, 5, 6/10, 8, 9, 11, 12, 14) are situated, at least some of the time, on the plasma membrane, where they mediate metal ion uptake into cells. Their level on the cell surface can be controlled rapidly via protein trafficking in response to the ions they transport. For example, the cell surface level of many ZIPs (including ZIP1, 3, 4, 8 and 12) is mediated by the available concentration of zinc. Zinc depletion causes a decrease in endocytosis and degradation, resulting in more ZIP on the surface to take up the essential ion. ZIP levels on the cell surface are a balance between endocytosis, recycling and degradation. We review the trafficking mechanisms of human ZIP proteins, highlighting possible targeting motifs and suggesting a model of zinc-mediated endocytic trafficking. We also provide two possible models for ZIP14 trafficking and degradation.
Collapse
Affiliation(s)
- Katherine Bowers
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London, UK
| | - Surjit K S Srai
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London, UK
| |
Collapse
|
24
|
Zinc Transporter Proteins. Neurochem Res 2017; 43:517-530. [PMID: 29243032 DOI: 10.1007/s11064-017-2454-y] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 12/07/2017] [Accepted: 12/11/2017] [Indexed: 01/06/2023]
Abstract
Zinc, which is involved in the structure of all enzyme classes, is a micro nutrient element and necessary for growth and development. The ability of zinc to function without causing toxic effects is depends on the protection of its homeostasis. Zinc transporter proteins are responsible for keeping zinc at certain concentrations. Based on their predicted membrane topology, Zn transporters are divided into two major families, SLC39s/ZIPs and SLC30s/ZnTs, which transport Zn in opposite directions through cellular and intracellular membranes. ZIPs increases the zinc concentration in the cytosol. For this, the ZIPs carries the zinc from extracellular and intracellular compartments to the cytosol. ZnTs, reduces the concentration of zinc in the cytosol. For this, ZnTs carries the zinc from the cytosol to extracellular and intracellular compartments. After being transported to the cell, 50% of the zinc is found in the cytoplasm, 30-40% in the nucleus, and 10% in the plasma and organelle membranes. The expression of many zinc transporter proteins in the cell is depending on the concentration of zinc and the physiological problems. The aim of this study is to give information about association of zinc transporter proteins with physiological events and health problems.
Collapse
|
25
|
Recent Advances in the Role of SLC39A/ZIP Zinc Transporters In Vivo. Int J Mol Sci 2017; 18:ijms18122708. [PMID: 29236063 PMCID: PMC5751309 DOI: 10.3390/ijms18122708] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 11/27/2017] [Accepted: 12/08/2017] [Indexed: 02/07/2023] Open
Abstract
Zinc (Zn), which is an essential trace element, is involved in numerous mammalian physiological events; therefore, either a deficiency or excess of Zn impairs cellular machineries and influences physiological events, such as systemic growth, bone homeostasis, skin formation, immune responses, endocrine function, and neuronal function. Zn transporters are thought to mainly contribute to Zn homeostasis within cells and in the whole body. Recent genetic, cellular, and molecular studies of Zn transporters highlight the dynamic role of Zn as a signaling mediator linking several cellular events and signaling pathways. Dysfunction in Zn transporters causes various diseases. This review aims to provide an update of Zn transporters and Zn signaling studies and discusses the remaining questions and future directions by focusing on recent progress in determining the roles of SLC39A/ZIP family members in vivo.
Collapse
|
26
|
Lehtovirta-Morley LE, Alsarraf M, Wilson D. Pan-Domain Analysis of ZIP Zinc Transporters. Int J Mol Sci 2017; 18:E2631. [PMID: 29211002 PMCID: PMC5751234 DOI: 10.3390/ijms18122631] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 12/01/2017] [Accepted: 12/01/2017] [Indexed: 12/13/2022] Open
Abstract
The ZIP (Zrt/Irt-like protein) family of zinc transporters is found in all three domains of life. However, little is known about the phylogenetic relationship amongst ZIP transporters, their distribution, or their origin. Here we employed phylogenetic analysis to explore the evolution of ZIP transporters, with a focus on the major human fungal pathogen, Candida albicans. Pan-domain analysis of bacterial, archaeal, fungal, and human proteins revealed a complex relationship amongst the ZIP family members. Here we report (i) a eukaryote-wide group of cellular zinc importers, (ii) a fungal-specific group of zinc importers having genetic association with the fungal zincophore, and, (iii) a pan-kingdom supercluster made up of two distinct subgroups with orthologues in bacterial, archaeal, and eukaryotic phyla.
Collapse
Affiliation(s)
- Laura E Lehtovirta-Morley
- Aberdeen Fungal Group, Medical Research Council Centre for Medical Mycology, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK.
- School of Biological Sciences, University of East Anglia, Norwich NR4 7TJ, UK.
| | - Mohammad Alsarraf
- Aberdeen Fungal Group, Medical Research Council Centre for Medical Mycology, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK.
| | - Duncan Wilson
- Aberdeen Fungal Group, Medical Research Council Centre for Medical Mycology, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK.
| |
Collapse
|
27
|
Kambe T, Matsunaga M, Takeda TA. Understanding the Contribution of Zinc Transporters in the Function of the Early Secretory Pathway. Int J Mol Sci 2017; 18:ijms18102179. [PMID: 29048339 PMCID: PMC5666860 DOI: 10.3390/ijms18102179] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 10/12/2017] [Accepted: 10/15/2017] [Indexed: 01/07/2023] Open
Abstract
More than one-third of newly synthesized proteins are targeted to the early secretory pathway, which is comprised of the endoplasmic reticulum (ER), Golgi apparatus, and other intermediate compartments. The early secretory pathway plays a key role in controlling the folding, assembly, maturation, modification, trafficking, and degradation of such proteins. A considerable proportion of the secretome requires zinc as an essential factor for its structural and catalytic functions, and recent findings reveal that zinc plays a pivotal role in the function of the early secretory pathway. Hence, a disruption of zinc homeostasis and metabolism involving the early secretory pathway will lead to pathway dysregulation, resulting in various defects, including an exacerbation of homeostatic ER stress. The accumulated evidence indicates that specific members of the family of Zn transporters (ZNTs) and Zrt- and Irt-like proteins (ZIPs), which operate in the early secretory pathway, play indispensable roles in maintaining zinc homeostasis by regulating the influx and efflux of zinc. In this review, the biological functions of these transporters are discussed, focusing on recent aspects of their roles. In particular, we discuss in depth how specific ZNT transporters are employed in the activation of zinc-requiring ectoenzymes. The means by which early secretory pathway functions are controlled by zinc, mediated by specific ZNT and ZIP transporters, are also subjects of this review.
Collapse
Affiliation(s)
- Taiho Kambe
- Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kyoto 606-8502, Japan.
| | - Mayu Matsunaga
- Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kyoto 606-8502, Japan.
| | - Taka-Aki Takeda
- Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kyoto 606-8502, Japan.
| |
Collapse
|
28
|
Bodiga VL, Thokala S, Kovur SM, Bodiga S. Zinc Dyshomeostasis in Cardiomyocytes after Acute Hypoxia/Reoxygenation. Biol Trace Elem Res 2017; 179:117-129. [PMID: 28181174 DOI: 10.1007/s12011-017-0957-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 01/30/2017] [Indexed: 02/06/2023]
Abstract
Zinc dyshomeostasis may play a role in the pathogenesis of myocardial ischemia/reperfusion injury. The objective of this study was to investigate the expression profile of zinc regulated transporter like- and iron-regulated transporter-like proteins (ZIPs) and zinc transporter proteins (ZnTs) in cardiomyocytes and their modulation in response to hypoxia and reoxygenation. Adult rat ventricular myocytes (ARVMs) were subjected to 6 h of hypoxia, followed by 18 h of reoxygenation. Intracellular and extracellular zinc concentrations were determined using Fluozin-3 and Newport Green fluorescence, respectively. Expression of ZnTs 1, 2, 5, and 9 along with ZIPs 1, 2, 3, 6, 7, 9, 10, 11, 13, and 14 was detectable in the cardiomyocytes by real-time reverse transcriptase polymerase chain reaction. Hypoxia elicited accumulation of intracellular free zinc, but subsequent reoxygenation resulted in striking loss of intracellular free zinc and decreased the cardiomyocyte viability. Concomitantly, extracellular zinc levels dropped rapidly during hypoxia, but increased after reoxygenation. Immunoblotting analysis revealed that hypoxia increased the expression of ZnT1, but reoxygenation significantly increased the expression of ZnTs 2 and 5. Neither hypoxia nor reoxygenation altered the levels of ZnT9. Increased intracellular zinc at the end of hypoxia is related to enhanced expression of ZIPs, whereas decreased intracellular zinc during reoxygenation appears to be due to lowered expression of all ZIPs, in addition to elevated levels of ZnTs 2 and 5. These results thus suggest that there is impaired accumulation of intracellular zinc during reoxygenation, due to overexpression of specific ZnTs and downregulation of ZIP expression.
Collapse
Affiliation(s)
- Vijaya Lakshmi Bodiga
- Institute of Genetics & Hospital for Genetic Diseases, Begumpet, Osmania University, Hyderabad, Telangana, 500016, India
| | - Sandhya Thokala
- Department of Biochemistry, Kakatiya University, Warangal, Telangana, 506009, India
| | - Sita Mahalaxmi Kovur
- Institute of Genetics & Hospital for Genetic Diseases, Begumpet, Osmania University, Hyderabad, Telangana, 500016, India
| | - Sreedhar Bodiga
- Department of Biochemistry, Kakatiya University, Warangal, Telangana, 506009, India.
| |
Collapse
|
29
|
Bafaro E, Liu Y, Xu Y, Dempski RE. The emerging role of zinc transporters in cellular homeostasis and cancer. Signal Transduct Target Ther 2017; 2:17029. [PMID: 29218234 PMCID: PMC5661630 DOI: 10.1038/sigtrans.2017.29] [Citation(s) in RCA: 163] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 04/21/2017] [Accepted: 04/25/2017] [Indexed: 12/21/2022] Open
Abstract
Zinc is an essential micronutrient that plays a role in the structural or enzymatic functions of many cellular proteins. Cellular zinc homeostasis involves the opposing action of two families of metal transporters: the ZnT (SLC30) family that functions to reduce cytoplasmic zinc concentrations and the ZIP (SLC39) family that functions to increase cytoplasmic zinc concentrations. Fluctuations in intracellular zinc levels mediated by these transporter families affect signaling pathways involved in normal cell development, growth, differentiation and death. Consequently, changes in zinc transporter localization and function resulting in zinc dyshomeostasis have pathophysiological effects. Zinc dyshomeostasis has been implicated in the progression of cancer. Here we review recent progress toward understanding the structural basis for zinc transport by ZnT and ZIP family proteins, as well as highlight the roles of zinc as a signaling molecule in physiological conditions and in various cancers. As zinc is emerging as an important signaling molecule in the development and progression of cancer, the ZnT and ZIP transporters that regulate cellular zinc homeostasis are promising candidates for targeted cancer therapy.
Collapse
Affiliation(s)
- Elizabeth Bafaro
- Department of Chemistry and Biochemistry, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
| | - Yuting Liu
- Department of Chemistry and Biochemistry, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
| | - Yan Xu
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Robert E Dempski
- Department of Chemistry and Biochemistry, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
| |
Collapse
|
30
|
Zong N, Ma SX, Wang ZY. Localization of zinc transporters in the spinal cord of cynomolgus monkey. J Chem Neuroanat 2017; 82:56-59. [PMID: 28455213 DOI: 10.1016/j.jchemneu.2017.04.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 04/09/2017] [Accepted: 04/09/2017] [Indexed: 11/17/2022]
Abstract
Zinc is abundant in the spinal cord, where it participates in several physiological and pathophysiological processes, including neurotransmission, spinal cord injury, and amyotrophic lateral sclerosis. However, the mechanisms underlying zinc homeostasis in the spinal cord are largely unknown. Zinc transporters (ZnTs) are responsible for transporting zinc from the cytoplasm to the extracellular space or to intracellular compartments. In the present study, we examined the distribution of ZnT1-10 proteins in the spinal cord of cynomolgus monkey. Immunohistochemical studies demonstrate that all detected ZnT family members are expressed in the gray matter. ZnT1-10 immunoreactivity can be seen in both motor and sensory neurons in the dorsal and ventral horn from the cervical to sacral segments. No obvious immunostaining was found in the glia cells. The present study demonstrates that ZnT proteins are functionally important for regulating zinc metabolism in both motor and sensory functions in monkey spinal cord.
Collapse
Affiliation(s)
- Ni Zong
- Institute of Neuroscience, College of Life Health Sciences, Northeastern University, Shenyang 110169, China
| | - Shi-Xin Ma
- Institute of Neuroscience, College of Life Health Sciences, Northeastern University, Shenyang 110169, China
| | - Zhan-You Wang
- Institute of Neuroscience, College of Life Health Sciences, Northeastern University, Shenyang 110169, China.
| |
Collapse
|
31
|
Lee S, Kelleher SL. Molecular regulation of lactation: The complex and requisite roles for zinc. Arch Biochem Biophys 2016; 611:86-92. [DOI: 10.1016/j.abb.2016.04.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 03/10/2016] [Accepted: 04/04/2016] [Indexed: 12/22/2022]
|
32
|
Napoli E, Ross-Inta C, Song G, Wong S, Hagerman R, Gane LW, Smilowitz JT, Tassone F, Giulivi C. Premutation in the Fragile X Mental Retardation 1 (FMR1) Gene Affects Maternal Zn-milk and Perinatal Brain Bioenergetics and Scaffolding. Front Neurosci 2016; 10:159. [PMID: 27147951 PMCID: PMC4835505 DOI: 10.3389/fnins.2016.00159] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 03/29/2016] [Indexed: 12/12/2022] Open
Abstract
Fragile X premutation alleles have 55–200 CGG repeats in the 5′ UTR of the FMR1 gene. Altered zinc (Zn) homeostasis has been reported in fibroblasts from >60 years old premutation carriers, in which Zn supplementation significantly restored Zn-dependent mitochondrial protein import/processing and function. Given that mitochondria play a critical role in synaptic transmission, brain function, and cognition, we tested FMRP protein expression, brain bioenergetics, and expression of the Zn-dependent synaptic scaffolding protein SH3 and multiple ankyrin repeat domains 3 (Shank3) in a knock-in (KI) premutation mouse model with 180 CGG repeats. Mitochondrial outcomes correlated with FMRP protein expression (but not FMR1 gene expression) in KI mice and human fibroblasts from carriers of the pre- and full-mutation. Significant deficits in brain bioenergetics, Zn levels, and Shank3 protein expression were observed in the Zn-rich regions KI hippocampus and cerebellum at PND21, with some of these effects lasting into adulthood (PND210). A strong genotype × age interaction was observed for most of the outcomes tested in hippocampus and cerebellum, whereas in cortex, age played a major role. Given that the most significant effects were observed at the end of the lactation period, we hypothesized that KI milk might have a role at compounding the deleterious effects on the FMR1 genetic background. A higher gene expression of ZnT4 and ZnT6, Zn transporters abundant in brain and lactating mammary glands, was observed in the latter tissue of KI dams. A cross-fostering experiment allowed improving cortex bioenergetics in KI pups nursing on WT milk. Conversely, WT pups nursing on KI milk showed deficits in hippocampus and cerebellum bioenergetics. A highly significant milk type × genotype interaction was observed for all three-brain regions, being cortex the most influenced. Finally, lower milk-Zn levels were recorded in milk from lactating women carrying the premutation as well as other Zn-related outcomes (Zn-dependent alkaline phosphatase activity and lactose biosynthesis—whose limiting step is the Zn-dependent β-1,4-galactosyltransferase). In premutation carriers, altered Zn homeostasis, brain bioenergetics and Shank3 levels could be compounded by Zn-deficient milk, increasing the risk of developing emotional and neurological/cognitive problems and/or FXTAS later in life.
Collapse
Affiliation(s)
- Eleonora Napoli
- Department of Molecular Biosciences, School of Veterinary Medicine Davis, CA, USA
| | - Catherine Ross-Inta
- Department of Molecular Biosciences, School of Veterinary Medicine Davis, CA, USA
| | - Gyu Song
- Department of Molecular Biosciences, School of Veterinary Medicine Davis, CA, USA
| | - Sarah Wong
- Department of Molecular Biosciences, School of Veterinary Medicine Davis, CA, USA
| | - Randi Hagerman
- Medical Investigations of Neurodevelopmental Disorders Institute, University of California, DavisDavis, CA, USA; Department of Pediatrics, University of California Davis Medical CenterSacramento, CA, USA
| | - Louise W Gane
- Medical Investigations of Neurodevelopmental Disorders Institute, University of California, Davis Davis, CA, USA
| | - Jennifer T Smilowitz
- Department of Food Science and Technology and Foods for Health Institute, University of California, Davis Davis, CA, USA
| | - Flora Tassone
- Medical Investigations of Neurodevelopmental Disorders Institute, University of California, DavisDavis, CA, USA; Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, DavisDavis, CA, USA
| | - Cecilia Giulivi
- Department of Molecular Biosciences, School of Veterinary MedicineDavis, CA, USA; Medical Investigations of Neurodevelopmental Disorders Institute, University of California, DavisDavis, CA, USA
| |
Collapse
|
33
|
Podany AB, Wright J, Lamendella R, Soybel DI, Kelleher SL. ZnT2-Mediated Zinc Import Into Paneth Cell Granules Is Necessary for Coordinated Secretion and Paneth Cell Function in Mice. Cell Mol Gastroenterol Hepatol 2016; 2:369-383. [PMID: 28174721 PMCID: PMC5042355 DOI: 10.1016/j.jcmgh.2015.12.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Accepted: 12/22/2015] [Indexed: 12/30/2022]
Abstract
BACKGROUND & AIMS Defects in Paneth cell (PC) function are associated with microbial dysbiosis and intestinal inflammation. PC granules contain antimicrobial peptides, cytokines, and substantial stores of zinc (Zn). We hypothesized that Zn, transported into the granule through the Zn transporter (ZnT)2, is critical for signature PC functions. METHODS ZnT2 was localized to PC granules using immunofluorescence and sucrose gradient fractionation in wild-type (wt) mice, and consequences of ZnT2 loss were characterized in ZnT2 knockout (ZnT2ko) mice. Terminal ilea were harvested for immunofluorescence, electron microscopy, and fluorescent imaging with the Zn reporter Zinpyr-1. Alterations in fecal microbiota were characterized using 16s ribosomal RNA sequencing. PC degranulation, bacterial translocation, cytokine response to Escherichia coli endotoxin lipopolysaccharide, crypt viability after exposure to the oxidant monochloramine (NH2Cl), and bactericidal activity of luminal contents of terminal ilea against enteropathogenic E coli were assessed. RESULTS ZnT2 was localized to the membrane of PC granules. In ZnT2ko mice, spontaneous degranulation was observed more frequently than among wt mice. Secretory granules were hypodense with less active lysozyme, and there was evidence of autophagosome accumulation and granule degradation in PCs from ZnT2ko mice. Gut microbiota of ZnT2ko mice were enriched in Bacteroidales S24-7 and relatively depleted of species commonly found in wt mice. Evidence of PC dysfunction in ZnT2ko mice included impaired granule secretion and increased inflammatory response to lipopolysaccharide, less bactericidal activity, and greater susceptibility to cell death from NH2Cl. CONCLUSIONS ZnT2 is critical for Zn import into PC granules, and the inability to import Zn leads to profound defects in PC function and uncoordinated granule secretion.
Collapse
Key Words
- CFU, colony forming unit
- EPEC, enteropathogenic Escherichia coli
- ER, endoplasmic reticulum
- IF, immunofluorescent
- IL, interleukin
- IP, intraperitoneal
- LPS, lipopolysaccharide
- Microbiota
- NEC, necrotizing enterocolitis
- OTU, organizational taxonomic unit
- PBS, phosphate-buffered saline
- PC, Paneth cell
- PCR, polymerase chain reaction
- Small Intestine
- TNF, tumor necrosis factor
- ZIP, ZRT, IRT-like protein
- Zinc Transporter
- Zn, zinc
- ZnT, zinc transporter
- ko, knockout
- wt, wild-type
Collapse
Affiliation(s)
- Abigail B. Podany
- Department of Surgery, Penn State Hershey College of Medicine, Hershey, Pennsylvania,Department of Cellular and Molecular Physiology, Penn State Hershey College of Medicine, Hershey, Pennsylvania
| | - Justin Wright
- Department of Biology, Juniata College, Huntingdon, Pennsylvania
| | | | - David I. Soybel
- Department of Surgery, Penn State Hershey College of Medicine, Hershey, Pennsylvania,Department of Cellular and Molecular Physiology, Penn State Hershey College of Medicine, Hershey, Pennsylvania
| | - Shannon L. Kelleher
- Department of Surgery, Penn State Hershey College of Medicine, Hershey, Pennsylvania,Department of Cellular and Molecular Physiology, Penn State Hershey College of Medicine, Hershey, Pennsylvania,Department of Pharmacology, Penn State Hershey College of Medicine, Hershey, Pennsylvania,Correspondence Address correspondence to: Shannon L. Kelleher, PhD, Penn State Hershey College of Medicine, 500 University Drive, Hershey, Pennsylvania 17033. fax: (717) 531-5393.Penn State Hershey College of Medicine500 University DriveHersheyPennsylvania 17033
| |
Collapse
|
34
|
Chandler P, Kochupurakkal BS, Alam S, Richardson AL, Soybel DI, Kelleher SL. Subtype-specific accumulation of intracellular zinc pools is associated with the malignant phenotype in breast cancer. Mol Cancer 2016; 15:2. [PMID: 26728511 PMCID: PMC4700748 DOI: 10.1186/s12943-015-0486-y] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 12/11/2015] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Zinc (Zn) hyper-accumulates in breast tumors and malignant cell lines compared to normal mammary epithelium. The mechanisms responsible for Zn accumulation and the consequence of Zn dysregulation are poorly understood. METHODS Microarrays were performed to assess differences in the expression of Zn transporters and metallothioneins (MTs) in human breast tumors and breast cancer cell lines. Real-time PCR and immunoblotting were employed to profile Zn transporter expression in representative luminal (T47D), basal (MDA-MB-231), and non-malignant (MCF10A) cell lines. Zn distribution in human tumors was assessed by X-ray fluorescence imaging. Zn distribution and content in cell lines was measured using FluoZin-3 imaging, and quantification and atomic absorption spectroscopy. Functional consequences of ZnT2 over-expression in MDA-MB-231 cells including invasion, proliferation, and cell cycle were measured using Boyden chambers, MTT assays, and flow cytometry, respectively. RESULTS Gene expression profiling of human breast tumors and breast cancer cell lines identified subtype-specific dysregulation in the Zn transporting network. X-ray fluorescence imaging of breast tumor tissues revealed Zn hyper-accumulation at the margins of Luminal breast tumors while Zn was more evenly distributed within Basal tumors. While both T47D and MDA-MB-231 cells hyper-accumulated Zn relative to MCF10A cells, T47D cells accumulated 2.5-fold more Zn compared to MDA-MB-231 cells. FluoZin-3 imaging indicated that Zn was sequestered into numerous large vesicles in T47D cells, but was retained in the cytoplasm and found in fewer and larger, amorphous sub-cellular compartments in MDA-MB-231 cells. The differences in Zn localization mirrored the relative abundance of the Zn transporter ZnT2; T47D cells over-expressed ZnT2, whereas MDA-MB-231 cells did not express ZnT2 protein due to proteasomal degradation. To determine the functional relevance of the lack of ZnT2 in MDA-MB-231cells, cells were transfected to express ZnT2. ZnT2 over-expression led to Zn vesicularization, shifts in cell cycle, enhanced apoptosis, and reduced proliferation and invasion. CONCLUSIONS This comprehensive analysis of the Zn transporting network in malignant breast tumors and cell lines illustrates that distinct subtype-specific dysregulation of Zn management may underlie phenotypic characteristics of breast cancers such as grade, invasiveness, metastatic potential, and response to therapy.
Collapse
Affiliation(s)
- Paige Chandler
- The Interdisciplinary Graduate Program in Physiology, Penn State Hershey College of Medicine, Hershey, PA, 17033, USA
- The Department of Cellular and Molecular Physiology, Penn State Hershey College of Medicine, Hershey, PA, 17033, USA
| | - Bose S Kochupurakkal
- Dana Farber Cancer Institute, Boston, MA, 02115, USA
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Samina Alam
- The Department of Cellular and Molecular Physiology, Penn State Hershey College of Medicine, Hershey, PA, 17033, USA
- The Department of Surgery, Penn State Hershey College of Medicine, Hershey, PA, 17033, USA
| | - Andrea L Richardson
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - David I Soybel
- The Department of Cellular and Molecular Physiology, Penn State Hershey College of Medicine, Hershey, PA, 17033, USA
- The Department of Surgery, Penn State Hershey College of Medicine, Hershey, PA, 17033, USA
| | - Shannon L Kelleher
- The Interdisciplinary Graduate Program in Physiology, Penn State Hershey College of Medicine, Hershey, PA, 17033, USA.
- The Department of Cellular and Molecular Physiology, Penn State Hershey College of Medicine, Hershey, PA, 17033, USA.
- The Department of Pharmacology, Penn State Hershey College of Medicine, Hershey, PA, 17033, USA.
- The Department of Surgery, Penn State Hershey College of Medicine, Hershey, PA, 17033, USA.
| |
Collapse
|
35
|
Alam S, Hennigar SR, Gallagher C, Soybel DI, Kelleher SL. Exome Sequencing of SLC30A2 Identifies Novel Loss- and Gain-of-Function Variants Associated with Breast Cell Dysfunction. J Mammary Gland Biol Neoplasia 2015; 20:159-72. [PMID: 26293594 DOI: 10.1007/s10911-015-9338-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 07/22/2015] [Indexed: 11/29/2022] Open
Abstract
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.
Collapse
Affiliation(s)
- Samina Alam
- Department of Cellular and Molecular Physiology, The Pennsylvania State University Hershey College of Medicine, Hershey, PA, USA
- Department of Surgery, The Pennsylvania State University Hershey College of Medicine, Hershey, PA, USA
| | - Stephen R Hennigar
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Carla Gallagher
- Department of Public Health Sciences, The Pennsylvania State University Hershey College of Medicine, Hershey, PA, USA
| | - David I Soybel
- Department of Cellular and Molecular Physiology, The Pennsylvania State University Hershey College of Medicine, Hershey, PA, USA
- Department of Surgery, The Pennsylvania State University Hershey College of Medicine, Hershey, PA, USA
| | - Shannon L Kelleher
- Department of Cellular and Molecular Physiology, The Pennsylvania State University Hershey College of Medicine, Hershey, PA, USA.
- Department of Pharmacology, The Pennsylvania State University Hershey College of Medicine, Hershey, PA, USA.
- Department of Surgery, The Pennsylvania State University Hershey College of Medicine, Hershey, PA, USA.
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, USA.
| |
Collapse
|
36
|
McCormick NH, Lee S, Hennigar SR, Kelleher SL. ZnT4 (SLC30A4)-null ("lethal milk") mice have defects in mammary gland secretion and hallmarks of precocious involution during lactation. Am J Physiol Regul Integr Comp Physiol 2015; 310:R33-40. [PMID: 26538236 DOI: 10.1152/ajpregu.00315.2014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 11/02/2015] [Indexed: 02/01/2023]
Abstract
During lactation, highly specialized secretory mammary epithelial cells (MECs) produce and secrete huge quantities of nutrients and nonnutritive factors into breast milk. The zinc (Zn) transporter ZnT4 (SLC30A4) transports Zn into the trans-Golgi apparatus for lactose synthesis, and across the apical cell membrane for efflux from MECs into milk. This is consistent with observations in "lethal milk" (lm/lm) mice, which have a truncation mutation in SLC30A4, and present with not only low milk Zn concentration, but also smaller mammary glands, decreased milk volume, and lactation failure by lactation day 2. However, the molecular underpinnings of these defects are not understood. Here, we used lactating C57BL/6J(lm/lm) (ZnT4-null) mice to explore the consequences of a ZnT4-null phenotype on mammary gland function during early lactation. Lactating C57BL/6J(lm/lm) mice had significantly fewer, smaller, and collapsed alveoli comprising swollen, lipid-filled MECs during early lactation. These defects were associated with decreased Akt expression and STAT5 activation, indicative of defects in MEC secretion. In addition, increased expression of ZnT2, TNF-α, and cleaved e-cadherin concomitant with increased activation of STAT3 implicated the loss of ZnT4 in precocious activation of involution. Collectively, our study indicates that the loss of ZnT4 has profound consequences on MEC secretion and may promote tissue remodeling in the mammary gland during early lactation.
Collapse
Affiliation(s)
- Nicholas H McCormick
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, Pennsylvania
| | - Sooyeon Lee
- Department of Cellular and Molecular Physiology, Penn State Hershey College of Medicine, Hershey, Pennsylvania
| | - Stephen R Hennigar
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, Pennsylvania
| | - Shannon L Kelleher
- Department of Cellular and Molecular Physiology, Penn State Hershey College of Medicine, Hershey, Pennsylvania; Department of Pharmacology, Penn State Hershey College of Medicine, Hershey, Pennsylvania; Department of Surgery, Penn State Hershey College of Medicine, Hershey, Pennsylvania; and Department of Nutritional Sciences, The Pennsylvania State University, University Park, Pennsylvania
| |
Collapse
|
37
|
Hennigar SR, Velasquez V, Kelleher SL. Obesity-Induced Inflammation Is Associated with Alterations in Subcellular Zinc Pools and Premature Mammary Gland Involution in Lactating Mice. J Nutr 2015; 145:1999-2005. [PMID: 26203096 PMCID: PMC4548167 DOI: 10.3945/jn.115.214122] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 06/24/2015] [Indexed: 11/14/2022] Open
Abstract
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.
Collapse
Affiliation(s)
- Stephen R Hennigar
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA; and
| | - Vanessa Velasquez
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA; and
| | - Shannon L Kelleher
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA; and Departments of Cell and Molecular Physiology, Pharmacology, and Surgery, Penn State Hershey College of Medicine, Hershey, PA
| |
Collapse
|
38
|
Kambe T, Tsuji T, Hashimoto A, Itsumura N. The Physiological, Biochemical, and Molecular Roles of Zinc Transporters in Zinc Homeostasis and Metabolism. Physiol Rev 2015; 95:749-84. [DOI: 10.1152/physrev.00035.2014] [Citation(s) in RCA: 556] [Impact Index Per Article: 61.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Zinc is involved in a variety of biological processes, as a structural, catalytic, and intracellular and intercellular signaling component. Thus zinc homeostasis is tightly controlled at the whole body, tissue, cellular, and subcellular levels by a number of proteins, with zinc transporters being particularly important. In metazoan, two zinc transporter families, Zn transporters (ZnT) and Zrt-, Irt-related proteins (ZIP) function in zinc mobilization of influx, efflux, and compartmentalization/sequestration across biological membranes. During the last two decades, significant progress has been made in understanding the molecular properties, expression, regulation, and cellular and physiological roles of ZnT and ZIP transporters, which underpin the multifarious functions of zinc. Moreover, growing evidence indicates that malfunctioning zinc homeostasis due to zinc transporter dysfunction results in the onset and progression of a variety of diseases. This review summarizes current progress in our understanding of each ZnT and ZIP transporter from the perspective of zinc physiology and pathogenesis, discussing challenging issues in their structure and zinc transport mechanisms.
Collapse
Affiliation(s)
- Taiho Kambe
- Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Tokuji Tsuji
- Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Ayako Hashimoto
- Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Naoya Itsumura
- Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| |
Collapse
|
39
|
Chemek M, Boughammoura S, Mimouna SB, Chouchene L, Banni M, Messaoudi I. Changes of the mRNA expression pattern of Zn transporters: a probable mechanism for cadmium retention and zinc redistribution in the suckling rat tissues. Biol Trace Elem Res 2015; 165:173-82. [PMID: 25653005 DOI: 10.1007/s12011-015-0255-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 01/25/2015] [Indexed: 12/27/2022]
Abstract
The present study was conducted to provide potential mechanism that may be responsible for Cd retention and Cd-induced Zn redistribution in tissues of suckling rat. For this purpose, suckling rats from mother receiving either tap water, Cd, or Cd + Zn during lactation period were sacrificed on postnatal day (PND) 14 and PND 21 for performing chemical and molecular analysis. Our results show that Cd exposure, although it does not affect the milk consumption, it clearly alters the lactational transfer, absorption, and distribution of Zn in the suckling rat organism. At the molecular level, Cd caused upregulation of ZIP 3, ZIP 4, and ZIP 8 gene expressions in the mammary gland of mothers rats and in the intestine of their pups but decreased the expression of ZnT 2 and ZnT 4 only in the mammary tissue at PND 14 and PND 21. Zn supply reversed the Cd-induced decrease in the neonatal Zn apparent absorption and restores the gastrointestinal, brain, and plasma levels of this essential element in the suckling rat organism at PND 14 and PND 21. Also, with this treatment, the gene expressions of ZnT 1 in the mammary gland and ZnT 4 in the neonatal intestine were found to be upregulated at PND 21. Furthermore, our results show that Cd or Cd + Zn treatment increase the neonatal hepatic MTs accumulation at PND 14 only. These results imply that the downregulation of ZnT as well as the overexpression of ZIP transporters, in responses to the Zn depletion induced by Cd in the tissues of lactating rat and their offspring, play a major role in Cd accumulation and Zn redistribution in tissues of suckling rat.
Collapse
Affiliation(s)
- Marouane Chemek
- LR11ES41: Génétique, Biodiversité et Valorisation des Bioressources, Institut de Biotechnologie, 5000, Monastir, Tunisia
| | | | | | | | | | | |
Collapse
|
40
|
Lee S, Hennigar SR, Alam S, Nishida K, Kelleher SL. Essential Role for Zinc Transporter 2 (ZnT2)-mediated Zinc Transport in Mammary Gland Development and Function during Lactation. J Biol Chem 2015; 290:13064-78. [PMID: 25851903 DOI: 10.1074/jbc.m115.637439] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Indexed: 01/28/2023] Open
Abstract
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.
Collapse
Affiliation(s)
- Sooyeon Lee
- From the Interdisciplinary Graduate Physiology Program and Department of Nutritional Sciences, The Pennsylvania State University, University Park, Pennsylvania 16802, Departments of Cellular and Molecular Physiology
| | - Stephen R Hennigar
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, Pennsylvania 16802
| | - Samina Alam
- Departments of Cellular and Molecular Physiology, Surgery, Penn State Hershey College of Medicine, Hershey, Pennsylvania 17033
| | - Keigo Nishida
- Laboratory for Homeostatic Network, RCAI, RIKEN Center for Integrative Medical Sciences (IMS-RCAI), Yokohama 230-0045, Japan, and Laboratory of Immune Regulation, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka 513-8670, Japan
| | - Shannon L Kelleher
- From the Interdisciplinary Graduate Physiology Program and Department of Nutritional Sciences, The Pennsylvania State University, University Park, Pennsylvania 16802, Departments of Cellular and Molecular Physiology, Surgery, Penn State Hershey College of Medicine, Hershey, Pennsylvania 17033, Pharmacology, and
| |
Collapse
|
41
|
Manservisi F, Gopalakrishnan K, Tibaldi E, Hysi A, Iezzi M, Lambertini L, Teitelbaum S, Chen J, Belpoggi F. Effect of maternal exposure to endocrine disrupting chemicals on reproduction and mammary gland development in female Sprague-Dawley rats. Reprod Toxicol 2014; 54:110-9. [PMID: 25554385 DOI: 10.1016/j.reprotox.2014.12.013] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 12/09/2014] [Accepted: 12/19/2014] [Indexed: 01/27/2023]
Abstract
The aim of the study is to determine whether low doses of "endocrine disrupting chemicals" (EDCs) affect the development and proliferative activity of the mammary glands (MGs). Adult parous/nulliparous female Sprague-Dawley (SD) rats were treated from post-natal day (PND) 1 until PND 180 with diethylphthalate (DEP), methylparaben (MPB), triclosan (TCS) and a mixture at doses comparable to human exposure. The doses (mg/kg b.w./day) were: DEP=0.173; MPB=0.105; TCS=0.05. EDC treatment resulted in mortality rates >20% in pups as early as lactation day 7. Significant morphological/histological changes were observed at the end of lactation in the MGs of EDC-treated dams. The total transcriptome profile as well as lactation-related genes in MGs also corroborate the morphological findings as more profound gene expression changes are present only at the weaning period. The study highlights the heightened sensitivity of the MGs during critical windows of exposure, particularly pregnancy and lactation, with an impact on pups' survival.
Collapse
Affiliation(s)
- Fabiana Manservisi
- Cesare Maltoni Cancer Research Center, Ramazzini Institute, Bologna, Italy(1).
| | - Kalpana Gopalakrishnan
- Department of Preventive Medicine, Icahn School of Medicine at Mount Sinai, NY, United States
| | - Eva Tibaldi
- Cesare Maltoni Cancer Research Center, Ramazzini Institute, Bologna, Italy(1)
| | - Albana Hysi
- Immuno-Oncology Laboratory Aging Research Center (CeSI), G. D'Annunzio University Foundation, Department of Medicine and Aging Sciences, G. D'Annunzio University of Chieti-Pescara, Italy
| | - Manuela Iezzi
- Immuno-Oncology Laboratory Aging Research Center (CeSI), G. D'Annunzio University Foundation, Department of Medicine and Aging Sciences, G. D'Annunzio University of Chieti-Pescara, Italy
| | - Luca Lambertini
- Department of Preventive Medicine, Icahn School of Medicine at Mount Sinai, NY, United States
| | - Susan Teitelbaum
- Department of Preventive Medicine, Icahn School of Medicine at Mount Sinai, NY, United States
| | - Jia Chen
- Department of Preventive Medicine, Icahn School of Medicine at Mount Sinai, NY, United States
| | - Fiorella Belpoggi
- Cesare Maltoni Cancer Research Center, Ramazzini Institute, Bologna, Italy(1).
| |
Collapse
|
42
|
Bostanci Z, Mack RP, Lee S, Soybel DI, Kelleher SL. Paradoxical zinc toxicity and oxidative stress in the mammary gland during marginal dietary zinc deficiency. Reprod Toxicol 2014; 54:84-92. [PMID: 25088245 DOI: 10.1016/j.reprotox.2014.07.076] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 07/07/2014] [Accepted: 07/22/2014] [Indexed: 01/06/2023]
Abstract
Zinc (Zn) regulates numerous cellular functions. Zn deficiency is common in females; ∼80% of women and 40% of adolescent girls consume inadequate Zn. Zn deficiency enhances oxidative stress, inflammation and DNA damage. Oxidative stress and inflammation is associated with breast disease. We hypothesized that Zn deficiency increases oxidative stress in the mammary gland, altering the microenvironment and architecture. Zn accumulated in the mammary glands of Zn deficient mice and this was associated with macrophage infiltration, enhanced oxidative stress and over-expression of estrogen receptor α. Ductal and stromal hypercellularity was associated with aberrant collagen deposition and disorganized e-cadherin. Importantly, these microenvironmental alterations were associated with substantial impairments in ductal expansion and mammary gland development. This is the first study to show that marginal Zn deficiency creates a toxic microenvironment in the mammary gland impairing breast development. These changes are consistent with hallmarks of potential increased risk for breast disease and cancer.
Collapse
Affiliation(s)
- Zeynep Bostanci
- Department of Nutritional Sciences, The Pennsylvania State University, United States; Department of Surgery, Penn State Hershey College of Medicine, United States
| | - Ronald P Mack
- Department of Nutritional Sciences, The Pennsylvania State University, United States; Department of Kinesiology, The Pennsylvania State University, United States
| | - Sooyeon Lee
- Department of Nutritional Sciences, The Pennsylvania State University, United States; Interdisciplinary Graduate Program in Physiology, The Pennsylvania State University, United States
| | - David I Soybel
- Department of Nutritional Sciences, The Pennsylvania State University, United States; Department of Surgery, Penn State Hershey College of Medicine, United States; Department of Cell and Molecular Physiology, Penn State Hershey College of Medicine, United States
| | - Shannon L Kelleher
- Department of Nutritional Sciences, The Pennsylvania State University, United States; Interdisciplinary Graduate Program in Physiology, The Pennsylvania State University, United States; Department of Surgery, Penn State Hershey College of Medicine, United States; Department of Cell and Molecular Physiology, Penn State Hershey College of Medicine, United States.
| |
Collapse
|
43
|
Seo YA, Lee S, Hennigar SR, Kelleher SL. Prolactin (PRL)-stimulated ubiquitination of ZnT2 mediates a transient increase in zinc secretion followed by ZnT2 degradation in mammary epithelial cells. J Biol Chem 2014; 289:23653-61. [PMID: 25016022 DOI: 10.1074/jbc.m113.531145] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
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.
Collapse
Affiliation(s)
- Young Ah Seo
- the Department of Nutritional Sciences, Pennsylvania State University, University Park, Pennsylvania 16802, and the Departments of Genetics and Complex Diseases and Nutrition, Harvard School of Public Health, Boston, Massachusetts 02115
| | - Sooyeon Lee
- the Department of Nutritional Sciences, Pennsylvania State University, University Park, Pennsylvania 16802, and From the Departments of Cell and Molecular Physiology
| | - Stephen R Hennigar
- the Department of Nutritional Sciences, Pennsylvania State University, University Park, Pennsylvania 16802, and
| | - Shannon L Kelleher
- the Department of Nutritional Sciences, Pennsylvania State University, University Park, Pennsylvania 16802, and From the Departments of Cell and Molecular Physiology, Pharmacology, and Surgery, Penn State Hershey College of Medicine, Hershey, Pennsylvania 17033,
| |
Collapse
|
44
|
McCormick NH, Hennigar SR, Kiselyov K, Kelleher SL. The biology of zinc transport in mammary epithelial cells: implications for mammary gland development, lactation, and involution. J Mammary Gland Biol Neoplasia 2014; 19:59-71. [PMID: 24338187 DOI: 10.1007/s10911-013-9314-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Accepted: 12/04/2013] [Indexed: 02/07/2023] Open
Abstract
Zinc plays a critical role in a vast array of cellular functions including gene transcription, protein translation, cell proliferation, differentiation, bioenergetics, and programmed cell death. The mammary gland depends upon tight coordination of these processes during development and reproduction for optimal expansion, differentiation, and involution. For example, zinc is required for activation of matrix metalloproteinases, intracellular signaling cascades such as MAPK and PKC, and the activation of both mitochondrial-mediated apoptosis and lysosomal-mediated cell death. In addition to functional needs, during lactation the mammary gland must balance providing optimal zinc for cellular requirements with the need to secrete a substantial amount of zinc into milk to meet the requirements of the developing neonate. Finally, the mammary gland exhibits the most profound example of programmed cell death, which is driven by both apoptotic and lysosomal-mediated cell death. Two families of zinc-specific transporters regulate zinc delivery for these diverse functions. Members of the ZIP family of zinc transporters (ZIP1-14) import zinc into the cytoplasm from outside the cell or from subcellular organelles, while members of the ZnT family (ZnT1-10) export zinc from the cytoplasm. Recently, the ion channel transient receptor potential mucolipin 1 (TRPML1) has also been implicated in zinc transport. Herein, we review our current understanding of the molecular mechanisms through which mammary epithelial cells utilize zinc with a focus on the transport of zinc into discrete subcellular organelles for specific cellular functions during mammary gland development, lactation, and involution.
Collapse
Affiliation(s)
- Nicholas H McCormick
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, USA
| | | | | | | |
Collapse
|
45
|
Lasry I, Golan Y, Berman B, Amram N, Glaser F, Assaraf YG. In situ dimerization of multiple wild type and mutant zinc transporters in live cells using bimolecular fluorescence complementation. J Biol Chem 2014; 289:7275-92. [PMID: 24451381 DOI: 10.1074/jbc.m113.533786] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Zinc transporters (ZnTs) facilitate zinc efflux and zinc compartmentalization, thereby playing a key role in multiple physiological processes and pathological disorders, presumed to be modulated by transporter dimerization. We recently proposed that ZnT2 homodimerization is the underlying basis for the dominant negative effect of a novel heterozygous G87R mutation identified in women producing zinc-deficient milk. To provide direct visual evidence for the in situ dimerization and function of multiple normal and mutant ZnTs, we applied here the bimolecular fluorescence complementation (BiFC) technique, which enables direct visualization of specific protein-protein interactions. BiFC is based upon reconstitution of an intact fluorescent protein including YFP when its two complementary, non-fluorescent N- and C-terminal fragments (termed YN and YC) are brought together by a pair of specifically interacting proteins. Homodimerization of ZnT1, -2, -3, -4, and -7 was revealed by high subcellular fluorescence observed upon co-transfection of non-fluorescent ZnT-YC and ZnT-YN; this homodimer fluorescence localized in the characteristic compartments of each ZnT. The validity of the BiFC assay in ZnT dimerization was further corroborated when high fluorescence was obtained upon co-transfection of ZnT5-YC and ZnT6-YN, which are known to form heterodimers. We further show that BiFC recapitulated the pathogenic role that ZnT mutations play in transient neonatal zinc deficiency. Zinquin, a fluorescent zinc probe applied along with BiFC, revealed the in situ functionality of ZnT dimers. Hence, the current BiFC-Zinquin technique provides the first in situ evidence for the dimerization and function of wild type and mutant ZnTs in live cells.
Collapse
Affiliation(s)
- Inbal Lasry
- From the Fred Wyszkowski Cancer Research Laboratory, Department of Biology, and
| | | | | | | | | | | |
Collapse
|
46
|
Liuzzi JP, Yoo C. Role of zinc in the regulation of autophagy during ethanol exposure in human hepatoma cells. Biol Trace Elem Res 2013; 156:350-6. [PMID: 24061963 DOI: 10.1007/s12011-013-9816-3] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Accepted: 09/02/2013] [Indexed: 02/07/2023]
Abstract
Faulty autophagy has been linked to various diseases including neurodegenerative disorders, diabetes, and cancer. Increasing evidence support the notion that activation of autophagy protects against ethanol-induced steatosis and liver injury. Herein, we investigated the role of zinc in autophagy in human hepatoma cells VL-17A exposed or not to ethanol. LC3II/LC3I ratio, p62, and Beclin-1 expression and autophagosomes number were determined in cells incubated in medium containing various concentrations of zinc with or without ethanol. In addition, labile zinc and mRNA expression of metallothionein and the zinc transporters SLC39A8, SLC39A14, and SLC30A10 were evaluated in cells exposed to ethanol and the autophagy inhibitor 3-methyladenine. Zinc depletion caused a significant suppression of autophagy in cells. Conversely, zinc addition to medium stimulated autophagy in cells. Moreover, cotreatment with ethanol and excess zinc (40 μM) had an additive effect on the induction of autophagy. 3-methyadenine treatment decreased labile zinc, but this effect was more pronounced in cells exposed to ethanol. Lastly, ethanol and 3-methyladenine caused significant changes in the expression of metallothionein and zinc transporters. The results from this study support the hypothesis that zinc is critical for autophagy under basal conditions and during ethanol exposure.
Collapse
Affiliation(s)
- J P Liuzzi
- Department of Dietetics and Nutrition, Florida International University, Miami, USA,
| | | |
Collapse
|
47
|
Yu Y, Wu A, Zhang Z, Yan G, Zhang F, Zhang L, Shen X, Hu R, Zhang Y, Zhang K, Wang F. Characterization of the GufA subfamily member SLC39A11/Zip11 as a zinc transporter. J Nutr Biochem 2013; 24:1697-708. [PMID: 23643525 DOI: 10.1016/j.jnutbio.2013.02.010] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Revised: 01/29/2013] [Accepted: 02/14/2013] [Indexed: 11/18/2022]
Abstract
Cellular zinc influx and efflux are maintained by two major transporter families, the ZIP (SLC39A) and ZnT (SLC30A or CDF) molecules. The functions of one molecule in this class, ZIP11/SLC39A11, remain unclear. Bioinformatics analysis of the distribution and evolutionary relationships of different ZIP members in eukaryotes and prokaryotes indicated that Zip11, the sole member of gufA subfamily, is an ancient ZIP family member that might have originated in early eukaryotic ancestors. Murine Zip11 mRNA is abundantly expressed in testes and the digestive system including stomach, ileum and cecum. Analysis of cellular zinc content, metallothionein levels, and cell viability under high or low zinc conditions in cells transfected with a murine Zip11 expression plasmid, suggest that Zip11 is a zinc importer. Further, cellular zinc concentrations and metallothionein levels decreased when Zip11 was knocked down. In mice supplemented with zinc, both mRNA and protein levels of Zip11 were slightly up-regulated in several tissues. The metal response element sequences (MREs) upstream of the first exon of Zip11 responded to elevated extracellular zinc concentrations, as assessed by luciferase reporter assays. Mutagenic analysis showed that several of the MREs could regulate Zip11 promoter activity, and metal-responsive transcription factor-1 (MTF-1) was shown to be involved in this process. Collectively, these data suggest that Zip11 has unique protein sequence and structure features, it functions as a cellular zinc transporter, and its expression is at least partially regulated by zinc via hMTF-1 binding to MREs of the Zip11 promoter.
Collapse
Affiliation(s)
- Yu Yu
- Department of Nutrition, Institute of Nutrition and Food Safety, School of Public Health, Zhejiang University, Hangzhou 310058, P.R. China; Key Laboratory of Nutrition and Metabolism, Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, P.R. China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Lisle RS, Anthony K, Randall MA, Diaz FJ. Oocyte-cumulus cell interactions regulate free intracellular zinc in mouse oocytes. Reproduction 2013; 145:381-90. [PMID: 23404848 DOI: 10.1530/rep-12-0338] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Zinc increases in the oocyte during maturation and is required for progression and completion of meiosis. The objective of this study was to determine whether cumulus cells regulate the levels of free intracellular zinc in the oocyte during maturation. In the cumulus-oocyte complex (COC) the relative level of free intracellular zinc was almost fourfold higher in cumulus cells compared with the resident germinal vesicle-stage oocyte. Removal of cumulus cells caused a fourfold increase in intracellular zinc in the oocyte by 1 h after cumulus cell removal, but subsequent coculture of denuded oocytes with COC decreased free intracellular zinc in the oocyte by 65%. Thus, cumulus cells suppress free intracellular zinc in the oocyte. The mRNA transcripts for the zinc transporter proteins Slc39a6, Slc39a8, Slc39a9, Slc39a10, Slc39a12, Slc30a2, Slc30a4, Slc30a5 and Slc30a8 mRNAs were higher in oocytes, while Slc39a1, Slc39a7, Slc39a13, Slc39a14, Slc30a6, Slc30a7 and Slc30a9 mRNAs were higher in cumulus cells. Thus a complex zinc transport network is present in the COC. Pretreatment with epidermal growth factor for 4 h abolished the ability of COCs to restrict free intracellular zinc in denuded oocytes. Coculture of denuded metaphase II oocytes with COC lowers free intracellular zinc in mature oocytes. Oocytes matured in vivo or oocytes from older mice had lower levels of free intracellular zinc than oocytes matured in vitro or from younger mice. Thus, a precise mechanism for regulating oocyte zinc homeostasis has been uncovered in the COC that is disrupted with increasing age or by removal of cumulus cells.
Collapse
Affiliation(s)
- R S Lisle
- Department of Animal Science, , Center for Reproductive Biology and Health, The Pennsylvania State University, University Park, PA 16802, USA
| | | | | | | |
Collapse
|
49
|
Myers SA, Nield A, Myers M. Zinc transporters, mechanisms of action and therapeutic utility: implications for type 2 diabetes mellitus. J Nutr Metab 2012; 2012:173712. [PMID: 23304467 PMCID: PMC3530793 DOI: 10.1155/2012/173712] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Revised: 11/07/2012] [Accepted: 11/07/2012] [Indexed: 12/29/2022] Open
Abstract
Zinc is an essential trace element that plays a vital role in maintaining many biological processes and cellular homeostasis. Dysfunctional zinc signaling is associated with a number of chronic disease states including cancer, cardiovascular disease, Alzheimer's disease, and diabetes. Cellular homeostasis requires mechanisms that tightly control the uptake, storage, and distribution of zinc. This is achieved through the coordinated actions of zinc transporters and metallothioneins. Evidence on the role of these proteins in type 2 diabetes mellitus (T2DM) is now emerging. Zinc plays a key role in the synthesis, secretion and action of insulin in both physiological and pathophysiological states. Moreover, recent studies highlight zinc's dynamic role as a "cellular second messenger" in the control of insulin signaling and glucose homeostasis. This suggests that zinc plays an unidentified role as a novel second messenger that augments insulin activity. This previously unexplored concept would raise a whole new area of research into the pathophysiology of insulin resistance and introduce a new class of drug target with utility for diabetes pharmacotherapy.
Collapse
Affiliation(s)
- Stephen A. Myers
- School of Health Sciences, University of Ballarat, University Drive, Mount Helen, VIC 3350, Australia
- Collaborative Research Network, University of Ballarat, Mount Helen, VIC 3350, Australia
| | - Alex Nield
- School of Health Sciences, University of Ballarat, University Drive, Mount Helen, VIC 3350, Australia
| | - Mark Myers
- School of Health Sciences, University of Ballarat, University Drive, Mount Helen, VIC 3350, Australia
| |
Collapse
|
50
|
Park JG, Qin Y, Galati DF, Palmer AE. New sensors for quantitative measurement of mitochondrial Zn(2+). ACS Chem Biol 2012; 7:1636-40. [PMID: 22850482 DOI: 10.1021/cb300171p] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Zinc (Zn(2+)) homeostasis plays a vital role in cell function, and the dysregulation of intracellular Zn(2+) is associated with mitochondrial dysfunction. Few tools exist to quantitatively monitor the buffered, free Zn(2+) concentration in mitochondria of living cells ([Zn(2+)](mito)). We have validated three high dynamic range, ratiometric, genetically encoded, fluorescent Zn(2+) sensors that we have successfully used to precisely measure and monitor [Zn(2+)](mito) in several cell types. Using one of these sensors, called mito-ZapCY1, we report observations that free Zn(2+) is buffered at concentrations about 3 orders of magnitude lower in mitochondria than in the cytosol and that HeLa cells expressing mito-ZapCY1 have an average [Zn(2+)](mito) of 0.14 pM, which differs significantly from other cell types. These optimized mitochondrial Zn(2+) sensors could improve our understanding of the relationship between Zn(2+) homeostasis and mitochondrial function.
Collapse
Affiliation(s)
- J. Genevieve Park
- Department of Chemistry
and Biochemistry and BioFrontiers Institute and ‡Department of Molecular, Cellular,
and Developmental Biology, University of Colorado Boulder, Boulder, Colorado 80309, United States
| | - Yan Qin
- Department of Chemistry
and Biochemistry and BioFrontiers Institute and ‡Department of Molecular, Cellular,
and Developmental Biology, University of Colorado Boulder, Boulder, Colorado 80309, United States
| | - Domenico F. Galati
- Department of Chemistry
and Biochemistry and BioFrontiers Institute and ‡Department of Molecular, Cellular,
and Developmental Biology, University of Colorado Boulder, Boulder, Colorado 80309, United States
| | - Amy E. Palmer
- Department of Chemistry
and Biochemistry and BioFrontiers Institute and ‡Department of Molecular, Cellular,
and Developmental Biology, University of Colorado Boulder, Boulder, Colorado 80309, United States
| |
Collapse
|