1
|
Xia Z, Tang B, Li X, Li X, Jia Y, Jiang J, Chen J, Song J, Liu S, Min J, Wang F. A Novel Role for the Longevity-Associated Protein SLC39A11 as a Manganese Transporter. RESEARCH (WASHINGTON, D.C.) 2024; 7:0440. [PMID: 39114488 PMCID: PMC11304475 DOI: 10.34133/research.0440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 07/11/2024] [Indexed: 08/10/2024]
Abstract
The identification of aging- and longevity-associated genes is important for promoting healthy aging. By analyzing a large cohort of Chinese centenarians, we previously found that single-nucleotide polymorphisms (SNPs) in the SLC39A11 gene (also known as ZIP11) are associated with longevity in males. However, the function of the SLC39A11 protein remains unclear. Here, we found that SLC39A11 expression is significantly reduced in patients with Hutchinson-Gilford progeria syndrome (HGPS). In addition, we found that zebrafish with a mutation in slc39a11 that significantly reduces its expression have an accelerated aging phenotype, including a shortened average lifespan, muscle atrophy and reduced swimming, impaired muscle regeneration, gut damage, and abnormal morphology in the reproductive system. Interestingly, these signs of premature aging were more pronounced in male zebrafish than in females. RNA-sequencing analysis revealed that cellular senescence may serve as a potential mechanism for driving this slc39a11 deficiency-induced phenotype in mutant zebrafish. Moreover, immunofluorescence showed significantly increased DNA damage and reactive oxygen species signaling in slc39a11 mutant zebrafish. Using inductively coupled plasma mass spectrometry (ICP-MS), we found that manganese significantly accumulates in slc39a11 mutant zebrafish, as well as in the serum of both global Slc39a11 knockout and hepatocyte-specific Slc39a11 knockout mice, suggesting that this metal transporter regulates systemic manganese levels. Finally, using cultured human fibroblasts, we found that both knocking down SLC39A11 and exposure to high extracellular manganese increased cellular senescence. These findings provide compelling evidence that SLC39A11 serves to protect against the aging process, at least in part by regulating cellular manganese homeostasis.
Collapse
Affiliation(s)
- Zhidan Xia
- The First Affiliated Hospital, The Second Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Zhejiang Provincial Key Laboratory of Bioelectromagnetics, State Key Laboratory of Experimental Hematology,
Zhejiang University School of Medicine, Hangzhou, China
| | - Biyao Tang
- The First Affiliated Hospital, The Second Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Zhejiang Provincial Key Laboratory of Bioelectromagnetics, State Key Laboratory of Experimental Hematology,
Zhejiang University School of Medicine, Hangzhou, China
- School of Public Health, Basic Medical Sciences, The First Affiliated Hospital, Hengyang Medical School,
University of South China, Hengyang, China
- School of Public Health, School of Basic Medical Sciences, The First Affiliated Hospital,
Xinxiang Medical University, Xinxiang, China
| | - Xiaopeng Li
- The First Affiliated Hospital, The Second Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Zhejiang Provincial Key Laboratory of Bioelectromagnetics, State Key Laboratory of Experimental Hematology,
Zhejiang University School of Medicine, Hangzhou, China
| | - Xinran Li
- The First Affiliated Hospital, The Second Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Zhejiang Provincial Key Laboratory of Bioelectromagnetics, State Key Laboratory of Experimental Hematology,
Zhejiang University School of Medicine, Hangzhou, China
| | - Yangfan Jia
- The First Affiliated Hospital, The Second Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Zhejiang Provincial Key Laboratory of Bioelectromagnetics, State Key Laboratory of Experimental Hematology,
Zhejiang University School of Medicine, Hangzhou, China
| | - Jianwei Jiang
- The First Affiliated Hospital, The Second Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Zhejiang Provincial Key Laboratory of Bioelectromagnetics, State Key Laboratory of Experimental Hematology,
Zhejiang University School of Medicine, Hangzhou, China
| | - Jingyao Chen
- The Core Facilities,
Zhejiang University School of Medicine, Hangzhou, China
| | - Jingshu Song
- The First Affiliated Hospital, The Second Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Zhejiang Provincial Key Laboratory of Bioelectromagnetics, State Key Laboratory of Experimental Hematology,
Zhejiang University School of Medicine, Hangzhou, China
| | - Siyi Liu
- The First Affiliated Hospital, The Second Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Zhejiang Provincial Key Laboratory of Bioelectromagnetics, State Key Laboratory of Experimental Hematology,
Zhejiang University School of Medicine, Hangzhou, China
| | - Junxia Min
- The First Affiliated Hospital, The Second Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Zhejiang Provincial Key Laboratory of Bioelectromagnetics, State Key Laboratory of Experimental Hematology,
Zhejiang University School of Medicine, Hangzhou, China
| | - Fudi Wang
- The First Affiliated Hospital, The Second Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Zhejiang Provincial Key Laboratory of Bioelectromagnetics, State Key Laboratory of Experimental Hematology,
Zhejiang University School of Medicine, Hangzhou, China
- School of Public Health, Basic Medical Sciences, The First Affiliated Hospital, Hengyang Medical School,
University of South China, Hengyang, China
- School of Public Health, School of Basic Medical Sciences, The First Affiliated Hospital,
Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
2
|
Liu H, Li L, Lu R. ZIP transporters-regulated Zn 2+ homeostasis: A novel determinant of human diseases. J Cell Physiol 2024; 239:e31223. [PMID: 38530191 DOI: 10.1002/jcp.31223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 01/16/2024] [Accepted: 02/06/2024] [Indexed: 03/27/2024]
Abstract
As an essential trace element for organisms, zinc participates in various physiological processes, such as RNA transcription, DNA replication, cell proliferation, and cell differentiation. The destruction of zinc homeostasis is associated with various diseases. Zinc homeostasis is controlled by the cooperative action of zinc transporter proteins that are responsible for the influx and efflux of zinc. Zinc transporter proteins are mainly categorized into two families: Zrt/Irt-like protein (SLC39A/ZIP) family and zinc transporter (SLC30A/ZNT) family. ZIP transporters contain 14 members, namely ZIP1-14, which can be further divided into four subfamilies. Currently, ZIP transporters-regulated zinc homeostasis is one of the research hotspots. Cumulative evidence suggests that ZIP transporters-regulated zinc homeostasis may cause physiological dysfunction and contribute to the onset and progression of diverse diseases, such as cancers, neurological diseases, and cardiovascular diseases. In this review, we initially discuss the structure and distribution of ZIP transporters. Furthermore, we comprehensively review the latest research progress of ZIP transporters-regulated zinc homeostasis in diseases, providing a new perspective into new therapeutic targets for treating related diseases.
Collapse
Affiliation(s)
- Huimei Liu
- Department of Pharmacology, Hengyang Medical School, University of South China, Hengyang, China
| | - Lanfang Li
- Department of Pharmacology, Hengyang Medical School, University of South China, Hengyang, China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, China
| | - Ruirui Lu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
3
|
Kim EY, Verdejo-Torres O, Diaz-Rodriguez K, Hasanain F, Caromile L, Padilla-Benavides T. Single nucleotide polymorphisms and Zn transport by ZIP11 shape functional phenotypes of HeLa cells. Metallomics 2024; 16:mfae006. [PMID: 38285610 DOI: 10.1093/mtomcs/mfae006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 01/27/2024] [Indexed: 01/31/2024]
Abstract
Zinc (Zn) is a vital micronutrient with essential roles in biological processes like enzyme function, gene expression, and cell signaling. Disruptions in the cellular regulation of Zn2+ ions often lead to pathological states. Mammalian Zn transporters, such as ZIP11, play a key role in homeostasis of this ion. ZIP11 resides predominately in the nucleus and Golgi apparatus. Our laboratory reported a function of ZIP11 in maintaining nuclear Zn levels in HeLa cervical cancer cells. Analyses of cervical and ovarian cancer patients' datasets identified four coding, single nucleotide polymorphisms (SNPs) in SLC39A11, the gene that encodes ZIP11, correlating with disease severity. We hypothesized that these SNPs might translate to functional changes in the ZIP11 protein by modifying access to substrate availability. We also proposed that a metal-binding site (MBS) in ZIP11 is crucial for transmembrane Zn2+ transport and required for maintenance of various pathogenic phenotypes observed in HeLa cells. Here, we investigated these claims by re-introducing single the SLC39A11 gene encoding for mutant residues associated with the SNPs, as well as MBS mutations into HeLa cells knocked down for the transporter. Some SNPs-encoding ZIP11 variants rescued Zn levels, proliferation, migration, and invasiveness of knockdown (KD) cells. Conversely, single MBS mutations mimicked the traits of KD cells, confirming the transporter's role in establishing and maintaining proliferative, migratory, and invasive traits. Overall, the intricate role of Zn in cellular dynamics and cancer progression underscores the significance of Zn transporters like ZIP11 in potential therapeutic interventions.
Collapse
Affiliation(s)
- Elizabeth Y Kim
- Department of Molecular Biology & Biochemistry, Wesleyan University, 52 Lawn Ave., Middletown, CT 06459, USA
| | - Odette Verdejo-Torres
- Department of Molecular Biology & Biochemistry, Wesleyan University, 52 Lawn Ave., Middletown, CT 06459, USA
| | - Karla Diaz-Rodriguez
- Department of Chemistry and Biochemistry, Worcester Polytechnic Institute, 60 Prescott St., Worcester, MA 01605, USA
| | - Farah Hasanain
- Department of Molecular Biology & Biochemistry, Wesleyan University, 52 Lawn Ave., Middletown, CT 06459, USA
| | - Leslie Caromile
- Departmentof Cell Biology, Center for Vascular Biology, UCONN Health-Center, Farmington, CT 06030, USA
| | - Teresita Padilla-Benavides
- Department of Molecular Biology & Biochemistry, Wesleyan University, 52 Lawn Ave., Middletown, CT 06459, USA
| |
Collapse
|
4
|
Bendellaa M, Lelièvre P, Coll JL, Sancey L, Deniaud A, Busser B. Roles of zinc in cancers: From altered metabolism to therapeutic applications. Int J Cancer 2024; 154:7-20. [PMID: 37610131 DOI: 10.1002/ijc.34679] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 07/10/2023] [Accepted: 07/24/2023] [Indexed: 08/24/2023]
Abstract
Zinc (Zn) is a crucial trace element involved in various cellular processes, including oxidative stress, apoptosis and immune response, contributing to cellular homeostasis. Dysregulation of Zn homeostasis occurs in certain cancers. This review discusses the role of Zn in cancer and its associated components, such as Zn-related proteins, their potential as biomarkers and the use of Zn-based strategies for tumor treatment. ZIP and ZnT proteins regulate Zn metabolism under normal conditions, but their expression is aberrant in cancer. These Zn proteins can serve as prognostic or diagnostic biomarkers, aiding in early cancer detection and disease monitoring. Moreover, targeting Zn and its pathways offers potential therapeutic approaches for cancer treatment. Modulating Zn biodistribution within cells using metal-binding agents allows for the control of downstream signaling pathways. Direct utilization of zinc as a therapeutic agent, including Zn supplementation or Zn oxide nanoparticle administration, holds promise for improving the prognosis of cancer patients.
Collapse
Affiliation(s)
- Mohamed Bendellaa
- Grenoble Alpes University, Institute for Advanced Biosciences, INSERM U1209, CNRS UMR5309, Grenoble, France
| | - Pierre Lelièvre
- Grenoble Alpes University, Institute for Advanced Biosciences, INSERM U1209, CNRS UMR5309, Grenoble, France
| | - Jean-Luc Coll
- Grenoble Alpes University, Institute for Advanced Biosciences, INSERM U1209, CNRS UMR5309, Grenoble, France
| | - Lucie Sancey
- Grenoble Alpes University, Institute for Advanced Biosciences, INSERM U1209, CNRS UMR5309, Grenoble, France
| | - Aurélien Deniaud
- Grenoble Alpes University, CNRS, CEA, IRIG, Laboratoire de Chimie et Biologie des Métaux, Grenoble, France
| | - Benoit Busser
- Grenoble Alpes University, Institute for Advanced Biosciences, INSERM U1209, CNRS UMR5309, Grenoble, France
- Department of Laboratory Medicine, Grenoble Alpes University Hospital, Grenoble, France
- Institut Universitaire de France (IUF), Paris, France
| |
Collapse
|
5
|
Fan YG, Wu TY, Zhao LX, Jia RJ, Ren H, Hou WJ, Wang ZY. From zinc homeostasis to disease progression: Unveiling the neurodegenerative puzzle. Pharmacol Res 2024; 199:107039. [PMID: 38123108 DOI: 10.1016/j.phrs.2023.107039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 11/16/2023] [Accepted: 12/10/2023] [Indexed: 12/23/2023]
Abstract
Zinc is a crucial trace element in the human body, playing a role in various physiological processes such as oxidative stress, neurotransmission, protein synthesis, and DNA repair. The zinc transporters (ZnTs) family members are responsible for exporting intracellular zinc, while Zrt- and Irt-like proteins (ZIPs) are involved in importing extracellular zinc. These processes are essential for maintaining cellular zinc homeostasis. Imbalances in zinc metabolism have been linked to the development of neurodegenerative diseases. Disruptions in zinc levels can impact the survival and activity of neurons, thereby contributing to the progression of neurodegenerative diseases through mechanisms like cell apoptosis regulation, protein phase separation, ferroptosis, oxidative stress, and neuroinflammation. Therefore, conducting a systematic review of the regulatory network of zinc and investigating the relationship between zinc dysmetabolism and neurodegenerative diseases can enhance our understanding of the pathogenesis of these diseases. Additionally, it may offer new insights and approaches for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Yong-Gang Fan
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, China.
| | - Ting-Yao Wu
- First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121000, China
| | - Ling-Xiao Zhao
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, China
| | - Rong-Jun Jia
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, China
| | - Hang Ren
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, China
| | - Wen-Jia Hou
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, China
| | - Zhan-You Wang
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, 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: 13] [Impact Index Per Article: 4.3] [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
|
Barman SK, Zaman MS, Veljanoski F, Malladi CS, Mahns DA, Wu MJ. Expression profiles of the genes associated with zinc homeostasis in normal and cancerous breast and prostate cells. Metallomics 2022; 14:6601457. [PMID: 35657662 DOI: 10.1093/mtomcs/mfac038] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 05/12/2022] [Indexed: 11/14/2022]
Abstract
Zn2+ dyshomeostasis is an intriguing phenomenon in breast and prostate cancers, with breast cancer cells exhibiting higher intracellular Zn2+ level compared to their corresponding normal epithelial cells, in contrast to the low Zn2+ level in prostate cancer cells. In order to gain molecular insights into the zinc homeostasis of breast and prostate cancer cells, this study profiled the expression of 28 genes, including 14 zinc importer genes (SLC39A1-14) which encode ZIP1-14 to transport Zn2+ into the cytoplasm, 10 zinc exporter genes (SLC30A1-10) which encode ZnT1-10 to transport Zn2+ out of the cytoplasm and 4 metallothionein genes (MT1B, MT1F, MT1X, MT2A) in breast (MCF10A, MCF-7, MDA-MB-231) and prostate (RWPE-1, PC3, DU145) cell lines in response to extracellular zinc exposures at a mild cytotoxic dosage and a benign dosage. The RNA samples were prepared at 0 min (T0), 30 min (T30) and 120 min (T120) in a time course with or without zinc exposure, which were used for profiling the baseline and dynamic gene expression. The up-regulation of MT genes was observed across the breast and prostate cancer cell lines. The expression landscape of SLC39A and SLC30A was revealed by the qRT-PCR data of this study, which sheds light on the divergence of intracellular Zn2+ levels for breast and prostate cancer cells. Taken together, the findings are valuable in unravelling the molecular intricacy of zinc homeostasis in breast and prostate cancer cells.
Collapse
Affiliation(s)
- Shital K Barman
- School of Science, Western Sydney University, Locked Bag 1797, Penrith, NSW 2751
| | - Mohammad S Zaman
- School of Science, Western Sydney University, Locked Bag 1797, Penrith, NSW 2751
| | - Filip Veljanoski
- School of Science, Western Sydney University, Locked Bag 1797, Penrith, NSW 2751
| | - Chandra S Malladi
- Proteomics and Lipidomics Lab, School of Medicine, Western Sydney University, Locked
| | - David A Mahns
- School of Medicine, Western Sydney University, Locked Bag 1797, Penrith, NSW 2751
| | - Ming J Wu
- School of Science, Western Sydney University, Locked Bag 1797, Penrith, NSW 2751
| |
Collapse
|
8
|
Csikós A, Kozma B, Pór Á, Kovács I, Lampé R, Miklós I, Takacs P. Zinc Transporter 9 (SLC30A9) Expression Is Decreased in the Vaginal Tissues of Menopausal Women. Biol Trace Elem Res 2021; 199:4011-4019. [PMID: 33409913 DOI: 10.1007/s12011-020-02525-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 11/29/2020] [Indexed: 12/11/2022]
Abstract
Our aim was to compare zinc transporter (ZnT/SLC30A, and ZIP/SLC39A) expression between pre- and postmenopausal women in human vaginal tissues. Zinc transporter families are responsible for the maintenance of intracellular zinc concentrations. Zinc has significant effects on the extracellular matrix composition. Vaginal wall biopsies were obtained from seven premenopausal and seven postmenopausal women. mRNA expression of twenty-four zinc transporters was determined by quantitative real-time PCR. Zinc transporter expression at the protein level was assessed by immunohistochemistry. Student's t test and Mann-Whitney U test were used to compare data. ZnT4 and ZnT9 mRNA expression were significantly lower in postmenopausal women compared with premenopausal women (mean ± SD mRNA expression in relative units, 96.43 ± 140.61 vs. 410.59 ± 304.34, p = 0.03 and 0.62 ± 0.39 vs. 1.13 ± 0.31, p = 0.02). In addition, ZIP2, ZIP3, and ZIP6 mRNA expressions were significantly lower in postmenopausal women compared with premenopausal women (mean ± SD mRNA expression in relative units, 1.11 ± 0.61 vs. 2.29 ± 1.20, p = 0.04; 2.32 ± 1.90 vs. 15.82 ± 12.97, p = 0.02 and 1.10 ± 0.80 vs. 5.73 ± 4.72, p = 0.03). ZnT9 protein expression in the stratum spinosum was significantly lower in postmenopausal women (p = 0.012). Zinc transporters were expressed differentially in the vaginal tissues. ZnT9 expression was significantly lower in postmenopausal women compared with premenopausal women.
Collapse
Affiliation(s)
- Anett Csikós
- Molecular Biology Group, FemPharma, LLC, Vígkedvű Mihály utca 21. II/5, Debrecen, 4024, Hungary.
| | - Bence Kozma
- Department of Obstetrics and Gynecology, University of Debrecen, Debrecen, Hungary
| | - Ágnes Pór
- Department of Pathology, Gyula Kenézy County Hospital, University of Debrecen, Debrecen, Hungary
| | - Ilona Kovács
- Department of Pathology, Gyula Kenézy County Hospital, University of Debrecen, Debrecen, Hungary
| | - Rudolf Lampé
- Department of Obstetrics and Gynecology, University of Debrecen, Debrecen, Hungary
| | - Ida Miklós
- Department of Genetics and Applied Microbiology, University of Debrecen, Debrecen, Hungary
| | - Peter Takacs
- Department of Obstetrics and Gynecology, Eastern Virginia Medical School, Norfolk, VA, USA
| |
Collapse
|
9
|
Jiang Y, Zhan H, Zhang Y, Yang J, Liu M, Xu C, Fan X, Zhang J, Zhou Z, Shi X, Ramesh R, Li M. ZIP4 promotes non-small cell lung cancer metastasis by activating snail-N-cadherin signaling axis. Cancer Lett 2021; 521:71-81. [PMID: 34450198 DOI: 10.1016/j.canlet.2021.08.025] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/16/2021] [Accepted: 08/22/2021] [Indexed: 02/07/2023]
Abstract
Non-small cell lung cancer (NSCLC) is one of the most critical health problems worldwide, with high incidence and poor survival rate. A zinc importer ZIP4 has been implicated in the process of tumor growth and metastasis of many cancers. However, its exact role and the underlying mechanism in NSCLC remains to be elucidated. In the present study, we found that human ZIP4 was substantially overexpressed in NSCLC tissues and was correlated with poor overall survival (OS) and progression-free survival (PFS). Overexpression of ZIP4 promoted cell migration, invasion and metastasis both in vitro and in a mouse lung metastasis model. Silencing of ZIP4 attenuated migration, invasion and metastasis. Mechanistically, overexpression of ZIP4 increased the expression of Snail, Slug and N-cadherin while genetic inactivation of ZIP4 downregulated the expression of above-mentioned genes. Further analysis showed that transcriptional factor Snail which modulates N-cadherin was involved in the process of ZIP4-mediated NSCLC migration and invasion. We also demonstrated that ZIP4 positively correlates with the levels of Snail, Slug and N-cadherin in mice lung metastasis tumors. Together, these results suggest that ZIP4 acts as an important regulator of Snail-N-cadherin signaling axis in promoting NSCLC progression and may serve as a novel predictive marker and therapeutic target in NSCLC.
Collapse
Affiliation(s)
- Yuanyuan Jiang
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital, Shandong University, Jinan, Shandong, China; Department of Medicine, the University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Surgery, the University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Hanxiang Zhan
- Department of Medicine, the University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Surgery, the University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Yuqing Zhang
- Department of Medicine, the University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Surgery, the University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Jingxuan Yang
- Department of Medicine, the University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Surgery, the University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Mingyang Liu
- Department of Medicine, the University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Surgery, the University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Chao Xu
- Department of Biostatistics and Epidemiology, Hudson College of Public Health, the University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Xiao Fan
- Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Junxia Zhang
- Department of Medicine, the University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Surgery, the University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhijun Zhou
- Department of Medicine, the University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Surgery, the University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Xiuhui Shi
- Department of Medicine, the University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Surgery, the University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Rajagopal Ramesh
- Department of Pathology, the University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Min Li
- Department of Medicine, the University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Surgery, the University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
10
|
Expression Analysis of Zinc Transporters in Nervous Tissue Cells Reveals Neuronal and Synaptic Localization of ZIP4. Int J Mol Sci 2021; 22:ijms22094511. [PMID: 33925953 PMCID: PMC8123391 DOI: 10.3390/ijms22094511] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 04/19/2021] [Accepted: 04/23/2021] [Indexed: 12/14/2022] Open
Abstract
In the last years, research has shown that zinc ions play an essential role in the physiology of brain function. Zinc acts as a potent neuromodulatory agent and signaling ions, regulating healthy brain development and the function of both neurons and glial cells. Therefore, the concentration of zinc within the brain and its cells is tightly controlled. Zinc transporters are key regulators of (extra-) cellular zinc levels, and deregulation of zinc homeostasis and zinc transporters has been associated with neurodegenerative and neuropsychiatric disorders. However, to date, the presence of specific family members and their subcellular localization within brain cells have not been investigated in detail. Here, we analyzed the expression of all zinc transporters (ZnTs) and Irt-like proteins (ZIPs) in the rat brain. We further used primary rat neurons and rat astrocyte cell lines to differentiate between the expression found in neurons or astrocytes or both. We identified ZIP4 expressed in astrocytes but significantly more so in neurons, a finding that has not been reported previously. In neurons, ZIP4 is localized to synapses and found in a complex with major postsynaptic scaffold proteins of excitatory synapses. Synaptic ZIP4 reacts to short-term fluctuations in local zinc levels. We conclude that ZIP4 may have a so-far undescribed functional role at excitatory postsynapses.
Collapse
|
11
|
Michalczyk K, Cymbaluk-Płoska A. The Role of Zinc and Copper in Gynecological Malignancies. Nutrients 2020; 12:E3732. [PMID: 33287452 PMCID: PMC7761859 DOI: 10.3390/nu12123732] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 11/29/2020] [Accepted: 12/02/2020] [Indexed: 12/14/2022] Open
Abstract
Zinc (Zn) and copper (Cu) are essential microelements, which take part in cellular metabolism, feature in enzymatic systems, and regulate enzyme activity. Homeostasis of these micronutrients is tightly regulated by multiple compensatory mechanisms that balance their concentrations including transporters, importers, and metallothioneins. An altered intake of only one of these trace elements may cause an imbalance in their levels and result in their competition for absorption. Relatively low levels of zinc and increased levels of copper may result in an increased level of oxidative stress and impair the antioxidant properties of multiple enzymes. Altered levels of trace elements were discovered in various pathologies including immunological, degenerative, and inflammatory diseases. Moreover, due to the role of Zn and Cu in oxidative stress and chronic inflammation, they were found to influence cancerogenesis. We review the roles of zinc and copper and their mechanisms in tumor growth, metastasis potential, microenvironment remodeling, and drug resistance. We highlight their role as potential biomarkers for cancer diagnosis, treatment, and prognosis, concentrating on their impact on gynecological malignancies.
Collapse
Affiliation(s)
- Kaja Michalczyk
- Department of Gynecological Surgery and Oncology of Adults and Adolescents, Pomeranian Medical University, al. Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland;
| | | |
Collapse
|
12
|
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: 24] [Impact Index Per Article: 3.4] [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
|
13
|
Baltaci AK, Yuce K. Zinc Transporter Proteins. Neurochem Res 2018; 43:517-530. [PMID: 29243032 DOI: 10.1007/s11064-017-2454-y] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [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
Affiliation(s)
| | - Kemal Yuce
- Department of Physiology, Medical Faculty, Selcuk University, Konya, Turkey
| |
Collapse
|
14
|
Fan Q, Cai Q, Li P, Wang W, Wang J, Gerry E, Wang TL, Shih IM, Nephew KP, Xu Y. The novel ZIP4 regulation and its role in ovarian cancer. Oncotarget 2017; 8:90090-90107. [PMID: 29163813 PMCID: PMC5685734 DOI: 10.18632/oncotarget.21435] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 08/26/2017] [Indexed: 12/20/2022] Open
Abstract
Our RNAseq analyses revealed that ZIP4 is a top gene up-regulated in more aggressive ovarian cancer cells. ZIP4's role in cancer stem cells has not been reported in any type of cancer. In addition, the role and regulation of ZIP4, a zinc transporter, have been studied in the context of extracellular zinc transporting. Factors other than zinc with ZIP4 regulatory effects are essentially unknown. ZIP4 expression and its regulation in epithelial ovarian cancer cells was assessed by immunoblotting, quantitative PCR, or immunohistochemistry staining in human ovarian tissues. Cancer stem cell-related activities were examined to evaluate the role of ZIP4 in human high-grade serous ovarian cancer cells in vitro and in vivo. RNAi and CRISPR techniques were used to knockdown or knockout ZIP4 and related genes. Ovarian cancer tissues overexpressed ZIP4 when compared with normal and benign tissues. ZIP4 knockout significantly reduced several cancer stem cell-related activities in EOC cells, including proliferation, anoikis-resistance, colony-formation, spheroid-formation, drug-resistance, and side-population in vitro. ZIP4-expressing side-population highly expressed known CSC markers ALDH1 and OCT4. ZIP4 knockout dramatically reduced tumorigenesis and ZIP4 overexpression increased tumorigenesis in vivo. In addition, the ZIP4-expressing side-population had the tumor initiating activity. Moreover, the oncolipid lysophosphatic acid effectively up-regulated ZIP4 expression via the nuclear receptor peroxisome proliferator-activated receptor gamma and lysophosphatic acid 's promoting effects in cancer stem cell-related activities in HGSOC cells was at least partially mediated by ZIP4 in an extracellular zinc-independent manner. Our critical data imply that ZIP4 is a new and important cancer stem cell regulator in ovarian cancer. Our data also provide an innovative interpretation for the apparent disconnection between low levels of zinc and up-regulation of ZIP4 in ovarian cancer tissues.
Collapse
Affiliation(s)
- Qipeng Fan
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Qingchun Cai
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Pengfei Li
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Pharmaceutical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, P.R. China
| | - Wenyan Wang
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Obstetrics and Gynecology, The Second Hospital of Anhui Medical University, Hefei City, 230601, P.R. China
| | - Jing Wang
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- MASDINO (Beijing) Medical Research Co. Ltd., Beijing, 100123, P.R. China
| | - Emily Gerry
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD 21231, USA
| | - Tian-Li Wang
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD 21231, USA
| | - Ie-Ming Shih
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD 21231, USA
| | - Kenneth P. Nephew
- Medical Sciences, Indiana University School of Medicine, Jordan Hall 302, Bloomington, IN 47405, USA
| | - Yan Xu
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
15
|
SLC39A4 expression is associated with enhanced cell migration, cisplatin resistance, and poor survival in non-small cell lung cancer. Sci Rep 2017; 7:7211. [PMID: 28775359 PMCID: PMC5543149 DOI: 10.1038/s41598-017-07830-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 07/04/2017] [Indexed: 12/13/2022] Open
Abstract
The zinc transporter SLC39A4 influences epithelial cell morphology and migration in various cancers; however, its role in regulating cell invasion and chemotherapeutic resistance in human lung cancer is not yet clear. Here, integrated analysis of gene expression in non-small cell lung cancer revealed that SLC39A4 expression is significantly correlated with increased tumour size and regional lymph node spread, as well as shorter overall survival (OS) and disease-free survival (DFS). SLC39A4 silencing by lentivirus-mediated shRNA blocked human lung cancer cell epithelial-mesenchymal transition and metastasis in vitro and in vivo, respectively. Moreover, SLC39A4 knockdown enhanced cancer cell sensitivity to cisplatin-induced death by inhibiting stemness in lung cancer cells. Collectively, these data suggest that SLC39A4 may be a novel therapeutic target and predictive marker of tumour metastasis in non-small cell lung cancer.
Collapse
|
16
|
Li Q, Jin J, Liu J, Wang L, He Y. Knockdown of Zinc Transporter ZIP5 by RNA Interference Inhibits Esophageal Cancer Growth In Vivo. Oncol Res 2017; 24:205-14. [PMID: 27458102 PMCID: PMC7838672 DOI: 10.3727/096504016x14648701447896] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
We recently found that SLC39A5 (ZIP5), a zinc transporter, is overexpressed in esophageal cancer. Downregulation of ZIP5 inhibited the proliferation, migration, and invasion of the esophageal cancer cell line KYSE170 in vitro. In this study, we found that downregulation of SLC39A5 (ZIP5) by interference resulted in a significant reduction in esophageal cancer tumor volume and weight in vivo. COX2 (cyclooxygenase 2) expression was decreased and E-cadherin expression was increased in the KYSE170K xenografts, which was caused by the downregulation of ZIP5. However, we did not find that the downregulation of ZIP5 caused a change in the relative expressions of cyclin D1, VEGF (vascular endothelial growth factor), MMP9 (matrix metalloprotein 9), and Bcl-2 (B-cell lymphoma/leukmia-2) mRNA or an alteration in the average level of zinc in the peripheral blood and xenografts in vivo. Collectively, these findings indicate that knocking down ZIP5 by small interfering RNA (siRNA) might be a novel treatment strategy for esophageal cancer with ZIP5 overexpression.
Collapse
Affiliation(s)
- Qian Li
- Cancer Institute, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | | | | | | | | |
Collapse
|
17
|
Melo MCC, de Souza JS, Kizys MML, Vidi AC, Dorta HS, Kunii IS, Giannocco G, Carvalheira G, Dias-da-Silva MR. Novel lincRNA Susceptibility Gene and Its Role in Etiopathogenesis of Thyrotoxic Periodic Paralysis. J Endocr Soc 2017; 1:809-815. [PMID: 29264532 PMCID: PMC5686567 DOI: 10.1210/js.2017-00015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 02/23/2017] [Indexed: 11/19/2022] Open
Abstract
Thyrotoxic periodic paralysis (TPP) is a life-threatening neuromuscular complication of thyrotoxicosis characterized by muscle weakness and hypokalemia and with an unclear etiopathogenesis. However, the 17q24.3 locus had been genetically linked to TPP, in which the genetic variant rs312691 (TC genotype) in long intergenic noncoding RNA (lincRNA) CTD-2378E21.1 is located downstream of inward-rectifier potassium (Kir) channel genes [KCNJ2 and its antisense KCNJ2 (AS-KCNJ2)]. A TPP patient with a suppressed thyroid-stimulating hormone level, a high free thyroxine level of (5.8 ng/dL), and low serum potassium level of (2 mEq/L) was evaluated for Kir channel expression during and after recovery from thyrotoxicosis. We observed that circulating lincRNA and Kir expression varied in accordance with thyroid status and TC genotype. To endorse this association of a lincRNA-rs312691 variant with a genetic risk of TPP, an additional series of 37 patients with TPP and 32 patients with thyrotoxic without paralysis (TWP) were assessed. We verified that the risk of minor allele C was greater in TPP than in TWP (odds ratio, 5.289; P = 0.0062), and protective major allele T was more frequent than observed in the 1000 genome controls (odds ratio, 11.90; P < 0.0001). AS-KCNJ2 was downregulated during thyrotoxicosis in the TWP controls carrying allele T and were upregulated in those with TPP with risk allele C. Moreover, KCNJ2 (Kir2.1) expression was reduced during thyrotoxicosis and restored in euthyroid status. We further excluded any other coding variant by performing targeted exome sequencing mutational screening in 17q24.3. Our data suggest that high lincRNA AS-KCNJ2 and CDT-2378E21.1 expression, possibly driven by the triiodothyronine regulatory mechanism, reduces the Kir2.1 expression observed during thyrotoxicosis. This finding could contribute to the understanding of the reduced inward-rectifying current observed during muscle weakness in genetically susceptible TPP patients.
Collapse
Affiliation(s)
- Maria Clara C Melo
- Laboratory of Molecular and Translational Endocrinology, Department of Medicine, Universidade Federal de São Paulo, São Paulo, 04039-032, Brazil
| | - Janaína S de Souza
- Biological Sciences, Universidade Federal de São Paulo, Diadema, 09972-270, Brazil
| | - Marina M L Kizys
- Laboratory of Molecular and Translational Endocrinology, Department of Medicine, Universidade Federal de São Paulo, São Paulo, 04039-032, Brazil
| | - Angela C Vidi
- Molecular Biology Program, Universidade Federal de São Paulo, São Paulo, 04044-020, Brazil
| | - Haron S Dorta
- Laboratory of Molecular and Translational Endocrinology, Department of Medicine, Universidade Federal de São Paulo, São Paulo, 04039-032, Brazil
| | - Ilda S Kunii
- Laboratory of Molecular and Translational Endocrinology, Department of Medicine, Universidade Federal de São Paulo, São Paulo, 04039-032, Brazil
| | - Gisele Giannocco
- Molecular Biology Program, Universidade Federal de São Paulo, São Paulo, 04044-020, Brazil
| | - Gianna Carvalheira
- Morphology and Genetics, Universidade Federal de São Paulo, São Paulo, 04039-032, Brazil
| | - Magnus R Dias-da-Silva
- Laboratory of Molecular and Translational Endocrinology, Department of Medicine, Universidade Federal de São Paulo, São Paulo, 04039-032, Brazil.,Biological Sciences, Universidade Federal de São Paulo, Diadema, 09972-270, Brazil
| |
Collapse
|
18
|
Davanian F, Faeghi F, Shahzadi S, Farshifar Z. Diffusion Tensor Imaging for Glioma Grading: Analysis of Fiber Density Index. Basic Clin Neurosci 2017; 8:13-18. [PMID: 28446945 PMCID: PMC5396168 DOI: 10.15412/j.bcn.03080102] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Introduction: The most common primary tumors of brain are gliomas and tumor grading is essential for designing proper treatment strategies. The gold standard choice to determine grade of glial tumor is biopsy which is an invasive method. The purpose of this study was to investigate the role of fiber density index (FDi) by means of diffusion tensor imaging (DTI) (as a noninvasive method) in glial tumor grading. Methods: A group of 20 patients with histologically confirmed diagnosis of gliomas were evaluated in this study. We used a 1.5 Tesla MR system (AVANTO; Siemens, Germany) with a standard head coil for scanning. Multidirectional diffusion weighted imaging (measured in 12 noncollinear directions), and T1 weighted nonenhanced were performed for all patients. We defined two regions of interest (ROIs); 1) White matter fibers near the tumor and 2) Similar fibers in the contralateral hemisphere. Results: FDi of the low-grade gliomas was higher than those of high-grade gliomas, which was significant (P=0.017). FDi ratio (ratio of fiber density in vicinity of the tumor to homologous fiber tracts in the contralateral hemisphere) is higher in low-grade than high-grade tumors, (P=0.05). In addition, we performed ROC (receiver operating characteristic) curve and the area under curve (AUC) was 0.813(P=0.013). Conclusion: Our findings prove significant difference in FDi near by low-grade and high-grade gliomas. Therefore, FDi values and ratios are helpful in glial tumor grading.
Collapse
Affiliation(s)
- Fariba Davanian
- Department of Radiology Technology, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fariborz Faeghi
- Department of Radiology Technology, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sohrab Shahzadi
- Department of Neurosurgery, Shohada Tajrish Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Farshifar
- Department of Radiology Technology, School of Paramedical, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
19
|
Pan Z, Choi S, Ouadid-Ahidouch H, Yang JM, Beattie JH, Korichneva I. Zinc transporters and dysregulated channels in cancers. Front Biosci (Landmark Ed) 2017; 22:623-643. [PMID: 27814637 DOI: 10.2741/4507] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
As a nutritionally essential metal ion, zinc (Zn) not only constitutes a structural element for more than 3000 proteins but also plays important regulatory functions in cellular signal transduction. Zn homeostasis is tightly controlled by regulating the flux of Zn across cell membranes through specific transporters, i.e. ZnT and ZIP family proteins. Zn deficiency and malfunction of Zn transporters have been associated with many chronic diseases including cancer. However, the mechanisms underlying Zn regulatory functions in cellular signaling and their impact on the pathogenesis and progression of cancers remain largely unknown. In addition to these acknowledged multifunctions, Zn modulates a wide range of ion channels that in turn may also play an important role in cancer biology. The goal of this review is to propose how zinc deficiency, through modified Zn homeostasis, transporter activity and the putative regulatory function of Zn can influence ion channel activity, and thereby contribute to carcinogenesis and tumorigenesis. This review intends to stimulate interest in, and support for research into the understanding of Zn-modulated channels in cancers, and to search for novel biomarkers facilitating effective clinical stratification of high risk cancer patients as well as improved prevention and therapy in this emerging field.
Collapse
Affiliation(s)
- Zui Pan
- The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA,
| | - Sangyong Choi
- Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Halima Ouadid-Ahidouch
- University of Picardie Jules Verne, UFR Sciences, EA 4667, Laboratory of Cell and Molecular Physiology, SFR CAP-SANTE (FED 4231), Amiens, France
| | - Jin-Ming Yang
- Department of Pharmacology, College of Medicine, Penn State University, 500 University Drive Hershey, PA 17033, USA
| | - John H Beattie
- Rowett Institute of Nutrition and Health, University of Aberdeen, Foresterhill, Bucksburn, Aberdeen AB25 2ZD, Scotland, UK
| | - Irina Korichneva
- University of Picardie Jules Verne, UFR Sciences, EA 4667, Laboratory of Cell and Molecular Physiology, SFR CAP-SANTE (FED 4231), Amiens, France
| |
Collapse
|
20
|
Münnich N, Wernhart S, Hogstrand C, Schlomann U, Nimsky C, Bartsch JW. Expression of the zinc importer protein ZIP9/SLC39A9 in glioblastoma cells affects phosphorylation states of p53 and GSK-3β and causes increased cell migration. Biometals 2016; 29:995-1004. [DOI: 10.1007/s10534-016-9971-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 09/08/2016] [Indexed: 12/14/2022]
|
21
|
Zinc and zinc-containing biomolecules in childhood brain tumors. J Mol Med (Berl) 2016; 94:1199-1215. [PMID: 27638340 DOI: 10.1007/s00109-016-1454-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 07/13/2016] [Accepted: 07/27/2016] [Indexed: 12/21/2022]
Abstract
Zinc ions are essential cofactors of a wide range of enzymes, transcription factors, and other regulatory proteins. Moreover, zinc is also involved in cellular signaling and enzymes inhibition. Zinc dysregulation, deficiency, over-supply, and imbalance in zinc ion transporters regulation are connected with various diseases including cancer. A zinc ion pool is maintained by two types of proteins: (i) zinc-binding proteins, which act as a buffer and intracellular donors of zinc and (ii) zinc transporters responsible for zinc fluxes into/from cells and organelles. The decreased serum zinc ion levels have been identified in patients suffering from various cancer diseases, including head and neck tumors and breast, prostate, liver, and lung cancer. On the contrary, increased zinc ion levels have been found in breast cancer and other malignant tissues. Zinc metalloproteomes of a majority of tumors including brain ones are still not yet fully understood. Current knowledge show that zinc ion levels and detection of certain zinc-containing proteins may be utilized for diagnostic and prognostic purposes. In addition, these proteins can also be promising therapeutic targets. The aim of the present work is an overview of the importance of zinc ions, zinc transporters, and zinc-containing proteins in brain tumors, which are, after leukemia, the second most common type of childhood cancer and the second leading cause of death in children after accidents.
Collapse
|