1
|
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: 6] [Impact Index Per Article: 6.0] [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
|
2
|
Giorgi G, Mascaró M, Gandini NA, Rabassa ME, Coló GP, Arévalo J, Curino AC, Facchinetti MM, Roque ME. Iron cycle disruption by heme oxygenase-1 activation leads to a reduced breast cancer cell survival. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166621. [PMID: 36539019 DOI: 10.1016/j.bbadis.2022.166621] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 12/01/2022] [Accepted: 12/01/2022] [Indexed: 12/23/2022]
Abstract
Heme oxygenase-1 (HO-1), which catalyzes heme degradation releasing iron, regulates several processes related to breast cancer. Iron metabolism deregulation is also connected with several tumor processes. However the regulatory relationship between HO-1 and iron proteins in breast cancer remains unclear. Using human breast cancer biopsies, we found that high HO-1 levels significantly correlated with low DMT1 levels. Contrariwise, high HO-1 levels significantly correlated with high ZIP14 and prohepcidin expression, as well as hemosiderin storage. At mRNA level, we found that high HO-1 expression significantly correlated with low DMT1 expression but high ZIP14, L-ferritin and hepcidin expression. In in vivo experiments in mice with genetic overexpression or pharmacological activation of HO-1, we detected the same expression pattern observed in human biopsies. In in vitro experiments, HO-1 activation induced changes in iron proteins expression leading to an increase of hemosiderin, ROS levels, lipid peroxidation and a decrease of the growth rate. Such low growth rate induced by HO-1 activation was reversed when iron levels or ROS levels were reduced. Our findings demonstrate an important role of HO-1 on iron homeostasis in breast cancer. The changes in iron proteins expression when HO-1 is modulated led to the iron accumulation deregulating the iron cell cycle, and consequently, generating oxidative stress and low viability, all contributing to impair breast cancer progression.
Collapse
Affiliation(s)
- G Giorgi
- Laboratorio de Fisiología Humana, Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS), 8000 Bahía Blanca, Argentina
| | - M Mascaró
- Laboratorio de Biología del Cáncer, Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Universidad Nacional del Sur (UNS)-CONICET, Departamento de Biología, Bioquímica y Farmacia (UNS), 8000 Bahía Blanca, Argentina
| | - N A Gandini
- Laboratorio de Biología del Cáncer, Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Universidad Nacional del Sur (UNS)-CONICET, Departamento de Biología, Bioquímica y Farmacia (UNS), 8000 Bahía Blanca, Argentina
| | - M E Rabassa
- Centro de Investigaciones Inmunológicas Básicas y Aplicadas (CINIBA), Facultad de Ciencias Médicas, Universidad Nacional de La Plata (UNLP), 1900 La Plata, Buenos Aires, Argentina
| | - G P Coló
- Laboratorio de Biología del Cáncer, Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Universidad Nacional del Sur (UNS)-CONICET, Departamento de Biología, Bioquímica y Farmacia (UNS), 8000 Bahía Blanca, Argentina
| | - J Arévalo
- Servicio de Patología, Hospital Interzonal de Agudos "Dr. José Penna", 8000 Bahía Blanca, Argentina
| | - A C Curino
- Laboratorio de Biología del Cáncer, Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Universidad Nacional del Sur (UNS)-CONICET, Departamento de Biología, Bioquímica y Farmacia (UNS), 8000 Bahía Blanca, Argentina.
| | - M M Facchinetti
- Laboratorio de Biología del Cáncer, Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Universidad Nacional del Sur (UNS)-CONICET, Departamento de Biología, Bioquímica y Farmacia (UNS), 8000 Bahía Blanca, Argentina
| | - M E Roque
- Laboratorio de Fisiología Humana, Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS), 8000 Bahía Blanca, Argentina
| |
Collapse
|
3
|
Renteria M, Belkin O, Aickareth J, Jang D, Hawwar M, Zhang J. Zinc's Association with the CmPn/CmP Signaling Network in Breast Cancer Tumorigenesis. Biomolecules 2022; 12:1672. [PMID: 36421686 PMCID: PMC9687477 DOI: 10.3390/biom12111672] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/05/2022] [Accepted: 11/09/2022] [Indexed: 08/24/2023] Open
Abstract
It is well-known that serum and cellular concentrations of zinc are altered in breast cancer patients. Specifically, there are notable zinc hyper-aggregates in breast tumor cells when compared to normal mammary epithelial cells. However, the mechanisms responsible for zinc accumulation and the consequences of zinc dysregulation are poorly understood. In this review, we detailed cellular zinc regulation/dysregulation under the influence of varying levels of sex steroids and breast cancer tumorigenesis to try to better understand the intricate relationship between these factors based on our current understanding of the CmPn/CmP signaling network. We also made some efforts to propose a relationship between zinc signaling and the CmPn/CmP signaling network.
Collapse
Affiliation(s)
| | | | | | | | | | - Jun Zhang
- Department of Molecular and Translational Medicine (MTM), Texas Tech University Health Science Center El Paso, El Paso, TX 79905, USA
| |
Collapse
|
4
|
Saravanan R, Balasubramanian V, Swaroop Balamurugan SS, Ezhil I, Afnaan Z, John J, Sundaram S, Gouthaman S, Pakala SB, Rayala SK, Venkatraman G. Zinc transporter LIV1: A promising cell surface target for triple negative breast cancer. J Cell Physiol 2022; 237:4132-4156. [PMID: 36181695 DOI: 10.1002/jcp.30880] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 08/22/2022] [Accepted: 08/30/2022] [Indexed: 11/05/2022]
Abstract
Breast cancer is one of the leading causes contributing to the global cancer burden. The triple negative breast cancer (TNBC) molecular subtype accounts for the most aggressive type. Despite progression in therapeutic options and prognosis in breast cancer treatment options, there remains a high rate of distant relapse. With advancements in understanding the role of zinc and zinc carriers in the prognosis and treatment of the disease, the scope of precision treatment/targeted therapy has been expanded. Zinc levels and zinc transporters play a vital role in maintaining cellular homeostasis, tumor surveillance, apoptosis, and immune function. This review focuses on the zinc transporter, LIV1, as an essential target for breast cancer prognosis and emerging treatment options. Previous studies give an insight into the role of LIV1 in fulfilling the most important hallmarks of cancer such as apoptosis, metastasis, invasion, and evading the immune system. Normal tissue expression of LIV1 is limited. Higher expression of LIV1 has been linked to Epithelial-Mesenchymal Transition, histological grade of cancer, and early node metastasis. LIV1 was found to be one of the attractive targets in the therapeutic hunt for TNBCs. TNBCs are an immunogenic breast cancer subtype. As zinc transporters are known to serve as the metabolic gatekeepers of immune cells, this review bridges tumor infiltrating lymphocytes, TNBC and LIV1. In addition, the suitability of LIV1 as an antibody-drug conjugate (Seattle genetics [SGN]-LIV1A) target in TNBC, represents a promising strategy for patients. Early clinical trial results reveal that this novel agent reduces tumor burden by inducing mitotic arrest, immunomodulation, and immunogenic cell death, warranting further investigation of SGN-LIV1A in combination with immuno-oncology agents. Priming the patient's immune response in combination with SGN-LIV1A could eventually change the landscape for the TNBC patient population.
Collapse
Affiliation(s)
- Roshni Saravanan
- Department of Human Genetics, Sri Ramachandra Faculty of Biomedical Sciences and Technology, Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu, India
| | - Vaishnavi Balasubramanian
- Department of Human Genetics, Sri Ramachandra Faculty of Biomedical Sciences and Technology, Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu, India
| | - Srikanth Swamy Swaroop Balamurugan
- Department of Human Genetics, Sri Ramachandra Faculty of Biomedical Sciences and Technology, Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu, India
| | - Inemai Ezhil
- Department of Biotechnology, Indian Institute of Technology-Madras, Chennai, Tamil Nadu, India
| | - Zeba Afnaan
- Department of Human Genetics, Sri Ramachandra Faculty of Biomedical Sciences and Technology, Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu, India
| | - Jisha John
- Department of Human Genetics, Sri Ramachandra Faculty of Biomedical Sciences and Technology, Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu, India
| | - Sandhya Sundaram
- Department of Pathology, Sri Ramachandra Medical College and Research Institute, Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu, India
| | - Shanmugasundaram Gouthaman
- Department of Surgical Oncology, Sri Ramachandra Medical College and Research Institute, Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu, India
| | - Suresh B Pakala
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, India
| | - Suresh Kumar Rayala
- Department of Biotechnology, Indian Institute of Technology-Madras, Chennai, Tamil Nadu, India
| | - Ganesh Venkatraman
- Department of Human Genetics, Sri Ramachandra Faculty of Biomedical Sciences and Technology, Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu, India
| |
Collapse
|
5
|
Althobiti M, El-sharawy KA, Joseph C, Aleskandarany M, Toss MS, Green AR, Rakha EA. Oestrogen-regulated protein SLC39A6: a biomarker of good prognosis in luminal breast cancer. Breast Cancer Res Treat 2021; 189:621-630. [PMID: 34453638 PMCID: PMC8505289 DOI: 10.1007/s10549-021-06336-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 07/15/2021] [Indexed: 10/27/2022]
Abstract
PURPOSE The outcome of the luminal oestrogen receptor-positive (ER +) subtype of breast cancer (BC) is highly variable and patient stratification needs to be refined. We assessed the prognostic significance of oestrogen-regulated solute carrier family 39 member 6 (SLC39A6) in BC, with emphasis on ER + tumours. MATERIALS AND METHODS SLC39A6 mRNA expression and copy number alterations were assessed using the METABRIC cohort (n = 1980). SLC39A6 protein expression was evaluated in a large (n = 670) and annotated series of early-stage (I-III) operable BC using tissue microarrays and immunohistochemistry. The associations between SLC39A6 expression and clinicopathological parameters, patient outcomes and other ER-related markers were evaluated using Chi-square tests and Kaplan-Meier curves. RESULTS High SLC39A6 mRNA and protein expression was associated with features characteristic of less aggressive tumours in the entire BC cohort and ER + subgroup. SLC39A6 protein expression was detected in the cytoplasm and nuclei of the tumour cells. High SLC39A6 nuclear expression and mRNA levels were positively associated with ER + tumours and expression of ER-related markers, including the progesterone receptor, forkhead box protein A1 and GATA binding protein 3. In the ER + luminal BC, high SLC39A6 expression was independently associated with longer BC-specific survival (BCSS) (P = 0.015, HR 0.678, 95% CI 0.472‒0.972) even in those who did not receive endocrine therapy (P = 0.001, HR 0.701, 95% CI 0.463‒1.062). CONCLUSION SLC39A6 may be prognostic for a better outcome in ER + luminal BC. Further functional studies to investigate the role of SLC39A6 in ER + luminal BC are warranted.
Collapse
Affiliation(s)
- Maryam Althobiti
- grid.4563.40000 0004 1936 8868Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham, University Park, Nottingham, NG7 2RD England ,grid.449644.f0000 0004 0441 5692Department of Clinical Laboratory Science, College of Applied Medical Science, Shaqra University 33, Shaqra, 11961 Saudi Arabia
| | - Khloud A. El-sharawy
- grid.4563.40000 0004 1936 8868Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham, University Park, Nottingham, NG7 2RD England ,grid.462079.e0000 0004 4699 2981Faculty of Science, Damietta University, Damietta, Egypt
| | - Chitra Joseph
- grid.4563.40000 0004 1936 8868Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham, University Park, Nottingham, NG7 2RD England
| | - Mohammed Aleskandarany
- grid.4563.40000 0004 1936 8868Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham, University Park, Nottingham, NG7 2RD England
| | - Michael S. Toss
- grid.4563.40000 0004 1936 8868Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham, University Park, Nottingham, NG7 2RD England
| | - Andrew R. Green
- grid.4563.40000 0004 1936 8868Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham, University Park, Nottingham, NG7 2RD England ,grid.4563.40000 0004 1936 8868Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, University of Nottingham Biodiscovery Institute, University Park, Nottingham, NG7 2RD England
| | - Emad A. Rakha
- grid.4563.40000 0004 1936 8868Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham, University Park, Nottingham, NG7 2RD England ,grid.4563.40000 0004 1936 8868Present Address: Department of Histopathology, School of Medicine, The University of Nottingham, City Hospital Campus, Hucknall Road, Nottingham, NG5 1PB, UK
| |
Collapse
|
6
|
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: 62] [Impact Index Per Article: 12.4] [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
|
7
|
Liu L, Yang J, Wang C. Analysis of the prognostic significance of solute carrier (SLC) family 39 genes in breast cancer. Biosci Rep 2020; 40:BSR20200764. [PMID: 32744318 PMCID: PMC7426635 DOI: 10.1042/bsr20200764] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 07/15/2020] [Accepted: 07/31/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Breast cancer (BC) is the most common malignancy in females and remains a main cause of cancer-associated death worldwide. The solute carrier (SLC) groups of membrane transport proteins, which control the influx of zinc, participate in ranging of physiological processes and may provide novel therapeutic targets of cancers. However, the prognostic values of individual SLC family 39 (SLC39A) genes in patients with BC are not clarified. MATERIALS AND METHODS The mRNA expression of SLC family 39 genes in BC was evaluated by using the UALCAN database. The prognostic values of overall survival (OS) of SLC family 39 genes in patients with BC were investigated by Kaplan-Meier plotter. The survival analysis of cells was determined by Project Achilles. RESULTS The analytic results suggested that SLC39A1, SLC39A3, SLC39A4, SLC39A5, SLC39A6, SLC39A7, SLC39A9, SLC39A10, SLC39A11 and SLC39A13 were significantly up-regulated in BC tissues compared with normal breast tissues. However, SLC39A8 and SLC39A14 were expressed higher in normal tissues than in BC tissues. High expression of SLC39A2, SLC39A3, SLC39A4, SLC39A5, SLC39A7, SLC39A12 and SLC39A13 was significantly associated with worse OS in patients with BC. In contrast, high mRNA levels of SLC39A6 and SLC39A14 indicated favorable OS. Through subgroup analysis, all abnormal expressed SLC family members were correlated with prognoses of patients with specific BC. Moreover, SLC39A7 was associated with proliferation and cloning of BC. CONCLUSIONS Our results suggested that SLC family 39 members were promising prognostic biomarkers of BC. The SLC39A7 played a key role in growth and survival of BC cells.
Collapse
Affiliation(s)
- Limei Liu
- Department of Keratonosus, Weifang Eye Hospital, Weifang 261053, Shandong, China
| | - Jiaomin Yang
- Department of Laboratory Medicine, Weifang Yidu Central Hospital, Qingzhou 262500, Shandong, China
| | - Chao Wang
- Department of Pathogen Biology, Weifang Medical University, Weifang 261053, Shandong, China
| |
Collapse
|
8
|
Tafreshi NK, Morse DL, Lee MC. Narrowing the focus: Therapeutic cell surface targets for refractory triple-negative breast cancer. World J Clin Oncol 2020; 11:169-179. [PMID: 32355639 PMCID: PMC7186233 DOI: 10.5306/wjco.v11.i4.169] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 03/25/2020] [Accepted: 03/30/2020] [Indexed: 02/06/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is defined as a type of breast cancer with lack of expression of estrogen receptor, progesterone receptor and human epidermal growth factor 2 protein. In comparison to other types of breast cancer, TNBC characterizes for its aggressive behavior, more prone to early recurrence and a disease with poor response to molecular target therapy. Although TNBC is identified in only 25%-30% of American breast cancer cases annually, these tumors continue to be a therapeutic challenge for clinicians for several reasons: Tumor heterogeneity, limited and toxic systemic therapy options, and often resistance to current standard therapy, characterized by progressive disease on treatment, residual tumor after cytotoxic chemotherapy, and early recurrence after complete surgical excision. Cell-surface targeted therapies have been successful for breast cancer in general, however there are currently no approved cell-surface targeted therapies specifically indicated for TNBC. Recently, several cell-surface targets have been identified as candidates for treatment of TNBC and associated targeted therapies are in development. The purpose of this work is to review the current clinical challenges posed by TNBC, the therapeutic approaches currently in use, and provide an overview of developing cell surface targeting approaches to improve outcomes for treatment resistant TNBC.
Collapse
Affiliation(s)
- Narges K Tafreshi
- Department of Cancer Physiology, Moffitt Cancer Center, Tampa, FL 33612, United States
- Department of Physics, University of South Florida, Tampa, FL 33612, United States
- Division of Oncologic Sciences, University of South Florida, Tampa, FL 33612 FL, United States
| | - David L Morse
- Department of Cancer Physiology, Moffitt Cancer Center, Tampa, FL 33612, United States
- Department of Physics, University of South Florida, Tampa, FL 33612, United States
- Division of Oncologic Sciences, University of South Florida, Tampa, FL 33612 FL, United States
| | - Marie Catherine Lee
- Division of Oncologic Sciences, University of South Florida, Tampa, FL 33612 FL, United States
- Comprehensive Breast Program, Moffitt Cancer Center, Tampa, FL 33612, United States
| |
Collapse
|
9
|
How cellular Zn 2+ signaling drives physiological functions. Cell Calcium 2018; 75:53-63. [PMID: 30145429 DOI: 10.1016/j.ceca.2018.08.004] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 08/16/2018] [Accepted: 08/17/2018] [Indexed: 01/10/2023]
Abstract
Zinc is an essential micronutrient affecting many aspects of human health. Cellular Zn2+ homeostasis is critical for cell function and survival. Zn2+, acting as a first or second messenger, triggers signaling pathways that mediate the physiological roles of Zn2+. Transient changes in Zn2+ concentrations within the cell or in the extracellular region occur following its release from Zn2+ binding metallothioneins, its transport across membranes by the ZnT or ZIP transporters, or release of vesicular Zn2+. These transients activate a distinct Zn2+ sensing receptor, ZnR/GPR39, or modulate numerous proteins and signaling pathways. Importantly, Zn2+ signaling regulates cellular physiological functions such as: proliferation, differentiation, ion transport and secretion. Indeed, novel therapeutic approaches aimed to maintain Zn2+ homeostasis and signaling are evolving. This review focuses on recent findings describing roles of Zn2+ and its transporters in regulating physiological or pathological processes.
Collapse
|
10
|
Ventura-Bixenshpaner H, Asraf H, Chakraborty M, Elkabets M, Sekler I, Taylor KM, Hershfinkel M. Enhanced ZnR/GPR39 Activity in Breast Cancer, an Alternative Trigger of Signaling Leading to Cell Growth. Sci Rep 2018; 8:8119. [PMID: 29802348 PMCID: PMC5970167 DOI: 10.1038/s41598-018-26459-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 04/20/2018] [Indexed: 12/14/2022] Open
Abstract
Acquired resistance to the estrogen receptor (ER) antagonist tamoxifen, is a major obstacle in treatment of breast cancer. Changes in Zn2+ accumulation and distribution are associated with tamoxifen-resistance and breast cancer progression. The Zn2+-sensing G-protein coupled receptor, ZnR/GPR39, triggers signaling leading to cell growth, but a role for this receptor in breast cancer in unknown. Using fluorescence imaging, we found Zn2+-dependent Ca2+ release, mediated by ZnR/GPR39 activity, in TAMR tamoxifen-resistant cells derived from MCF-7 cells, but not in ER-expressing MCF-7 or T47D cells. Furthermore, ZnR/GPR39 signaling was monitored in ER negative BT20, MDA-MB-453 and JIMT-1 cells. Expression of ZnR/GPR39 was increased in grade 3 human breast cancer biopsies compared to grade 2. Consistently, analysis of two breast cancer patient cohorts, GDS4057 and TCGA, indicated that in ER-negative tumors higher ZnR/GPR39 mRNA levels are associated with more aggressive tumors. Activation of ZnR/GPR39 in TAMR cells triggered MAPK, mTOR and PI3K signaling. Importantly, enhanced cell growth and invasiveness was observed in the ER negative breast cancer cells, TAMR, MDA-MB-453 and BT20 cells but not in the ER expressing MCF-7 cells. Thus, we suggest ZnR/GPR39 as a potential therapeutic target for combination treatment in breast cancer, particularly relevant in ER negative tumors.
Collapse
Affiliation(s)
- Hila Ventura-Bixenshpaner
- Department of Physiology and Cell Biology and The Zlotowski Center for Neuroscience, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Hila Asraf
- Department of Physiology and Cell Biology and The Zlotowski Center for Neuroscience, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Moumita Chakraborty
- Department of Physiology and Cell Biology and The Zlotowski Center for Neuroscience, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Moshe Elkabets
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Israel Sekler
- Department of Physiology and Cell Biology and The Zlotowski Center for Neuroscience, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Kathryn M Taylor
- Breast Cancer Molecular Pharmacology Group, School of Pharmacy and Pharmaceutical Sciences, Redwood Building, Cardiff University, King Edward VII Avenue, Cardiff, CF10 3NB, UK
| | - Michal Hershfinkel
- Department of Physiology and Cell Biology and The Zlotowski Center for Neuroscience, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
| |
Collapse
|
11
|
Hershfinkel M. The Zinc Sensing Receptor, ZnR/GPR39, in Health and Disease. Int J Mol Sci 2018; 19:ijms19020439. [PMID: 29389900 PMCID: PMC5855661 DOI: 10.3390/ijms19020439] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Revised: 01/24/2018] [Accepted: 01/29/2018] [Indexed: 02/07/2023] Open
Abstract
A distinct G-protein coupled receptor that senses changes in extracellular Zn2+, ZnR/GPR39, was found in cells from tissues in which Zn2+ plays a physiological role. Most prominently, ZnR/GPR39 activity was described in prostate cancer, skin keratinocytes, and colon epithelial cells, where zinc is essential for cell growth, wound closure, and barrier formation. ZnR/GPR39 activity was also described in neurons that are postsynaptic to vesicular Zn2+ release. Activation of ZnR/GPR39 triggers Gαq-dependent signaling and subsequent cellular pathways associated with cell growth and survival. Furthermore, ZnR/GPR39 was shown to regulate the activity of ion transport mechanisms that are essential for the physiological function of epithelial and neuronal cells. Thus, ZnR/GPR39 provides a unique target for therapeutically modifying the actions of zinc in a specific and selective manner.
Collapse
Affiliation(s)
- Michal Hershfinkel
- Department of Physiology and Cell Biology and The Zlotowski Center for Neuroscience, Faculty of Health Sciences, POB 653, Ben-Gurion Ave. Ben-Gurion University of the Negev, Beer Sheva 84105, Israel.
| |
Collapse
|
12
|
Wessels I, Maywald M, Rink L. Zinc as a Gatekeeper of Immune Function. Nutrients 2017; 9:E1286. [PMID: 29186856 PMCID: PMC5748737 DOI: 10.3390/nu9121286] [Citation(s) in RCA: 376] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 11/20/2017] [Accepted: 11/22/2017] [Indexed: 12/27/2022] Open
Abstract
After the discovery of zinc deficiency in the 1960s, it soon became clear that zinc is essential for the function of the immune system. Zinc ions are involved in regulating intracellular signaling pathways in innate and adaptive immune cells. Zinc homeostasis is largely controlled via the expression and action of zinc "importers" (ZIP 1-14), zinc "exporters" (ZnT 1-10), and zinc-binding proteins. Anti-inflammatory and anti-oxidant properties of zinc have long been documented, however, underlying mechanisms are still not entirely clear. Here, we report molecular mechanisms underlying the development of a pro-inflammatory phenotype during zinc deficiency. Furthermore, we describe links between altered zinc homeostasis and disease development. Consequently, the benefits of zinc supplementation for a malfunctioning immune system become clear. This article will focus on underlying mechanisms responsible for the regulation of cellular signaling by alterations in zinc homeostasis. Effects of fast zinc flux, intermediate "zinc waves", and late homeostatic zinc signals will be discriminated. Description of zinc homeostasis-related effects on the activation of key signaling molecules, as well as on epigenetic modifications, are included to emphasize the role of zinc as a gatekeeper of immune function.
Collapse
Affiliation(s)
- Inga Wessels
- Institute of Immunology, Faculty of Medicine, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074 Aachen, Germany.
| | - Martina Maywald
- Institute of Immunology, Faculty of Medicine, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074 Aachen, Germany.
| | - Lothar Rink
- Institute of Immunology, Faculty of Medicine, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074 Aachen, Germany.
| |
Collapse
|
13
|
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: 73] [Impact Index Per Article: 9.1] [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
|
14
|
Taylor KA, Pugh N. The contribution of zinc to platelet behaviour during haemostasis and thrombosis. Metallomics 2016; 8:144-55. [PMID: 26727074 DOI: 10.1039/c5mt00251f] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Platelets are the primary cellular determinants of haemostasis and pathological thrombus formation leading to myocardial infarction and stroke. Following vascular injury or atherosclerotic plaque rupture, platelets are recruited to sites of damage and undergo activation induced by a variety of soluble and/or insoluble agonists. Platelet activation is a multi-step process culminating in the formation of thrombi, which contribute to the haemostatic process. Zinc (Zn(2+)) is acknowledged as an important signalling molecule in a diverse range of cellular systems, however there is limited understanding of the influence of Zn(2+) on platelet behaviour during thrombus formation. This review evaluates the contributions of exogenous and intracellular Zn(2+) to platelet function and assesses the potential pathophysiological implications of Zn(2+) signalling. We also provide a speculative assessment of the mechanisms by which platelets could respond to changes in extracellular and intracellular Zn(2+) concentration.
Collapse
Affiliation(s)
- K A Taylor
- Department of Biomedical and Forensic Sciences, Faculty of Science and Technology, Anglia Ruskin University, Cambridge, CB1 1PT, UK.
| | - N Pugh
- Department of Biomedical and Forensic Sciences, Faculty of Science and Technology, Anglia Ruskin University, Cambridge, CB1 1PT, UK.
| |
Collapse
|
15
|
Pisano A, Santolla MF, De Francesco EM, De Marco P, Rigiracciolo DC, Perri MG, Vivacqua A, Abonante S, Cappello AR, Dolce V, Belfiore A, Maggiolini M, Lappano R. GPER, IGF-IR, and EGFR transduction signaling are involved in stimulatory effects of zinc in breast cancer cells and cancer-associated fibroblasts. Mol Carcinog 2016; 56:580-593. [PMID: 27341075 DOI: 10.1002/mc.22518] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 06/08/2016] [Accepted: 06/22/2016] [Indexed: 01/23/2023]
Abstract
Zinc (Zn) is an essential trace mineral that contributes to the regulation of several cellular functions; however, it may be also implicated in the progression of breast cancer through different mechanisms. It has been largely reported that the classical estrogen receptor (ER), as well as the G protein estrogen receptor (GPER, previously known as GPR30) can exert a main role in the development of breast tumors. In the present study, we demonstrate that zinc chloride (ZnCl2 ) involves GPER in the activation of insulin-like growth factor receptor I (IGF-IR)/epidermal growth factor receptor (EGFR)-mediated signaling, which in turn triggers downstream pathways like ERK and AKT in breast cancer cells, and main components of the tumor microenvironment namely cancer-associated fibroblasts (CAFs). Further corroborating these findings, ZnCl2 stimulates a functional crosstalk of GPER with IGF-IR and EGFR toward the transcription of diverse GPER target genes. Then, we show that GPER contributes to the stimulatory effects induced by ZnCl2 on cell-cycle progression, proliferation, and migration of breast cancer cells as well as migration of CAFs. Together, our data provide novel insights into the molecular mechanisms through which zinc may exert stimulatory effects in breast cancer cells and CAFs toward tumor progression. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Assunta Pisano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | | | | | - Paola De Marco
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | | | - Maria Grazia Perri
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Adele Vivacqua
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | | | - Anna Rita Cappello
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Vincenza Dolce
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Antonino Belfiore
- Division of Endocrinology, Department of Health, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Marcello Maggiolini
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Rosamaria Lappano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| |
Collapse
|
16
|
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: 66] [Impact Index Per Article: 7.3] [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
|
17
|
Myers SA. Zinc transporters and zinc signaling: new insights into their role in type 2 diabetes. Int J Endocrinol 2015; 2015:167503. [PMID: 25983752 PMCID: PMC4423030 DOI: 10.1155/2015/167503] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 04/07/2015] [Accepted: 04/12/2015] [Indexed: 12/13/2022] Open
Abstract
Zinc is an essential trace element that plays a vital role in many biological processes including growth and development, immunity, and metabolism. Recent studies have highlighted zinc's dynamic role as a "cellular second messenger" in the control of insulin signaling and glucose homeostasis. Accordingly, mechanisms that contribute to dysfunctional zinc signaling are suggested to be associated with metabolic disease states including cancer, cardiovascular disease, Alzheimer's disease, and diabetes. The actions of the proteins that control the uptake, storage, and distribution of zinc, the zinc transporters, are under intense investigation due to their emerging role in type 2 diabetes. The synthesis, secretion, and action of insulin are dependent on zinc and the transporters that make this ion available to cellular processes. This suggests that zinc plays a previously unidentified role where changes in zinc status over time may affect 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
- University of Tasmania (UTAS), School of Health Sciences, Newnham Campus, Launceston, TAS 7250, Australia
- *Stephen A. Myers:
| |
Collapse
|
18
|
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: 2.9] [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
|
19
|
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: 4.7] [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
|
20
|
Alam S, Kelleher SL. Cellular mechanisms of zinc dysregulation: a perspective on zinc homeostasis as an etiological factor in the development and progression of breast cancer. Nutrients 2012; 4:875-903. [PMID: 23016122 PMCID: PMC3448077 DOI: 10.3390/nu4080875] [Citation(s) in RCA: 121] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Revised: 07/11/2012] [Accepted: 07/17/2012] [Indexed: 12/29/2022] Open
Abstract
Worldwide, breast cancer is the most commonly diagnosed cancer among women and is the leading cause of female cancer deaths. Zinc (Zn) functions as an antioxidant and plays a role in maintaining genomic stability. Zn deficiency results in oxidative DNA damage and increased cancer risk. Studies suggest an inverse association between dietary and plasma Zn levels and the risk for developing breast cancer. In contrast, breast tumor biopsies display significantly higher Zn levels compared with normal tissue. Zn accumulation in tumor tissue also correlates with increased levels of Zn importing proteins. Further, aberrant expression of Zn transporters in tumors correlates with malignancy, suggesting that altered metal homeostasis in the breast could contribute to malignant transformation and the severity of cancer. However, studies have yet to link dysregulated Zn transport and abnormal Zn-dependent functions in breast cancer development. Herein, we summarize studies that address the multi-modal role of Zn dyshomeostasis in breast cancer with respect to the role of Zn in modulating oxidative stress, DNA damage response/repair pathways and cell proliferation/apoptosis, and the relationship to aberrant regulation of Zn transporters. We also compare Zn dysregulation in breast tissue to that of prostate, pancreatic and ovarian cancer where possible.
Collapse
Affiliation(s)
- Samina Alam
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA 16802, USA;
| | - Shannon L. Kelleher
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA 16802, USA;
- Department of Surgery, the Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
- Cell and Molecular Physiology, the Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
- Author to whom correspondence should be addressed; ; Tel.: +1-814-863-9680; Fax: +1-814-863-6103
| |
Collapse
|
21
|
Yang ZM, Chen WW, Wang YF. Gene expression profiling in gastric mucosa from Helicobacter pylori-infected and uninfected patients undergoing chronic superficial gastritis. PLoS One 2012; 7:e33030. [PMID: 22438889 PMCID: PMC3306372 DOI: 10.1371/journal.pone.0033030] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Accepted: 02/09/2012] [Indexed: 12/13/2022] Open
Abstract
Helicobacter pylori infection reprograms host gene expression and influences various cellular processes, which have been investigated by cDNA microarray using in vitro culture cells and in vivo gastric biopsies from patients of the Chronic Abdominal Complaint. To further explore the effects of H. pylori infection on host gene expression, we have collected the gastric antral mucosa samples from 6 untreated patients with gastroscopic and pathologic confirmation of chronic superficial gastritis. Among them three patients were infected by H. pylori and the other three patients were not. These samples were analyzed by a microarray chip which contains 14,112 cloned cDNAs, and microarray data were analyzed via BRB ArrayTools software and Ingenuity Pathways Analysis (IPA) website. The results showed 34 genes of 38 differentially expressed genes regulated by H. pylori infection had been annotated. The annotated genes were involved in protein metabolism, inflammatory and immunological reaction, signal transduction, gene transcription, trace element metabolism, and so on. The 82% of these genes (28/34) were categorized in three molecular interaction networks involved in gene expression, cancer progress, antigen presentation and inflammatory response. The expression data of the array hybridization was confirmed by quantitative real-time PCR assays. Taken together, these data indicated that H. pylori infection could alter cellular gene expression processes, escape host defense mechanism, increase inflammatory and immune responses, activate NF-κB and Wnt/β-catenin signaling pathway, disturb metal ion homeostasis, and induce carcinogenesis. All of these might help to explain H. pylori pathogenic mechanism and the gastroduodenal pathogenesis induced by H. pylori infection.
Collapse
Affiliation(s)
- Ze-Min Yang
- Pi-Wei Institute, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- School of Basic Courses, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Wei-Wen Chen
- Pi-Wei Institute, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- E-Institute of Traditional Chinese Medicine Internal Medicine, Shanghai Municipal Education Committee, Shanghai, China
- * E-mail:
| | - Ying-Fang Wang
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| |
Collapse
|
22
|
Kelleher SL, Velasquez V, Croxford TP, McCormick NH, Lopez V, MacDavid J. Mapping the zinc-transporting system in mammary cells: molecular analysis reveals a phenotype-dependent zinc-transporting network during lactation. J Cell Physiol 2012; 227:1761-70. [PMID: 21702047 DOI: 10.1002/jcp.22900] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The mammary epithelial cell transitions from a non-secreting to a terminally differentiated, secreting cell during lactation. Zinc (Zn) is a key modulator of phenotypic transition as it regulates over 300 biological functions including transcription, translation, energy transformation, intracellular signaling, and apoptosis. In addition, Zn must be redirected from normal cellular functions into the secretory compartment, as many components of the secretory system are Zn-dependent and an extraordinary amount of Zn is secreted (1-3 mg Zn/day) into milk. Herein, we utilized a "systems biology" approach of genomic and proteomic profiling to explore mechanisms through which Zn is reallocated during phenotype transition in the lactating mammary gland from mice and cultured mammary cells. Nine Zn transporters play key roles in Zn redistribution within the network during lactation. Protein abundance of six Zip (Zip3, Zip5, Zip7, Zip8, Zip10, Zip11) and three ZnT (ZnT2, ZnT4, ZnT9) proteins was expanded >2-fold during lactation, which was not necessarily reflected by changes in mRNA expression. Our data suggest that Zip5, Zip8, and Zip10 may be key to Zn acquisition from maternal circulation, while multiple Zip proteins reuptake Zn from milk. Confocal microscopy of cultured mammary cells identified the Golgi apparatus (modulated in part by ZnT5, Zip7, and Zip11) and the late endosomal compartment (modulated in part by ZnT2 and Zip3) as key intracellular compartments through which Zn is reallocated during lactation. These results provide an important framework for understanding the "Zn-transporting network" through which mammary gland Zn pools are redistributed and secreted into milk.
Collapse
Affiliation(s)
- Shannon L Kelleher
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, Pennsylvania 16802, USA.
| | | | | | | | | | | |
Collapse
|
23
|
Taylor KM, Hiscox S, Nicholson RI, Hogstrand C, Kille P. Protein kinase CK2 triggers cytosolic zinc signaling pathways by phosphorylation of zinc channel ZIP7. Sci Signal 2012; 5:ra11. [PMID: 22317921 PMCID: PMC3428905 DOI: 10.1126/scisignal.2002585] [Citation(s) in RCA: 199] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The transition element zinc, which has recently been identified as an intracellular second messenger, has been implicated in various signaling pathways, including those leading to cell proliferation. Zinc channels of the ZIP (ZRT1- and IRT1-like protein) family [also known as solute carrier family 39A (SLC39A)] transiently increase the cytosolic free zinc (Zn(2+)) concentration in response to extracellular signals. We show that phosphorylation of evolutionarily conserved residues in endoplasmic reticulum zinc channel ZIP7 is associated with the gated release of Zn(2+) from intracellular stores, leading to activation of tyrosine kinases and the phosphorylation of AKT and extracellular signal-regulated kinases 1 and 2. Through pharmacological manipulation, proximity ligation assay, and mutagenesis, we identified protein kinase CK2 as the kinase responsible for ZIP7 activation. Together, the present results show that transition element channels in eukaryotes can be activated posttranslationally by phosphorylation, as part of a cell signaling cascade. Our study links the regulated release of zinc from intracellular stores to phosphorylation of kinases involved in proliferative responses and cell migration, suggesting a functional role for ZIP7 and zinc signals in these events. The connection with proliferation and migration, as well as the activation of ZIP7 by CK2, a kinase that is antiapoptotic and promotes cell division, suggests that ZIP7 may provide a target for anticancer drug development.
Collapse
Affiliation(s)
- Kathryn M Taylor
- Breast Cancer Molecular Pharmacology Group, School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Redwood Building, King Edward VIIth Avenue, Cardiff CF10 3NB, UK.
| | | | | | | | | |
Collapse
|
24
|
Chasapis CT, Loutsidou AC, Spiliopoulou CA, Stefanidou ME. Zinc and human health: an update. Arch Toxicol 2011; 86:521-34. [PMID: 22071549 DOI: 10.1007/s00204-011-0775-1] [Citation(s) in RCA: 555] [Impact Index Per Article: 39.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2011] [Accepted: 10/26/2011] [Indexed: 02/07/2023]
Abstract
The importance of micronutrients in health and nutrition is undisputable, and among them, zinc is an essential element whose significance to health is increasingly appreciated and whose deficiency may play an important role in the appearance of diseases. Zinc is one of the most important trace elements in the organism, with three major biological roles, as catalyst, structural, and regulatory ion. Zinc-binding motifs are found in many proteins encoded by the human genome physiologically, and free zinc is mainly regulated at the single-cell level. Zinc has critical effect in homeostasis, in immune function, in oxidative stress, in apoptosis, and in aging, and significant disorders of great public health interest are associated with zinc deficiency. In many chronic diseases, including atherosclerosis, several malignancies, neurological disorders, autoimmune diseases, aging, age-related degenerative diseases, and Wilson's disease, the concurrent zinc deficiency may complicate the clinical features, affect adversely immunological status, increase oxidative stress, and lead to the generation of inflammatory cytokines. In these diseases, oxidative stress and chronic inflammation may play important causative roles. It is therefore important that status of zinc is assessed in any case and zinc deficiency is corrected, since the unique properties of zinc may have significant therapeutic benefits in these diseases. In the present paper, we review the zinc as a multipurpose trace element, its biological role in homeostasis, proliferation and apoptosis and its role in immunity and in chronic diseases, such as cancer, diabetes, depression, Wilson's disease, Alzheimer's disease, and other age-related diseases.
Collapse
Affiliation(s)
- Christos T Chasapis
- Department of Pharmacy, School of Health Sciences, University of Patras, Patras, Greece
| | | | | | | |
Collapse
|
25
|
Capturing changes in gene expression dynamics by gene set differential coordination analysis. Genomics 2011; 98:469-77. [PMID: 21971296 DOI: 10.1016/j.ygeno.2011.09.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2011] [Revised: 09/01/2011] [Accepted: 09/16/2011] [Indexed: 12/31/2022]
Abstract
Analyzing gene expression data at the gene set level greatly improves feature extraction and data interpretation. Currently most efforts in gene set analysis are focused on differential expression analysis--finding gene sets whose genes show first-order relationship with the clinical outcome. However the regulation of the biological system is complex, and much of the change in gene expression dynamics do not manifest in the form of differential expression. At the gene set level, capturing the change in expression dynamics is difficult due to the complexity and heterogeneity of the gene sets. Here we report a systematic approach to detect gene sets that show differential coordination patterns with the rest of the transcriptome, as well as pairs of gene sets that are differentially coordinated with each other. We demonstrate that the method can identify biologically relevant gene sets, many of which do not show first-order relationship with the clinical outcome.
Collapse
|
26
|
Nam H, Knutson MD. Effect of dietary iron deficiency and overload on the expression of ZIP metal-ion transporters in rat liver. Biometals 2011; 25:115-24. [PMID: 21826460 DOI: 10.1007/s10534-011-9487-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2011] [Accepted: 07/31/2011] [Indexed: 01/22/2023]
Abstract
The mammalian ZIP (Zrt-, Irt-like Protein) family of transmembrane transport proteins consists of 14 members that share considerable homology. ZIP proteins have been shown to mediate the cellular uptake of the essential trace elements zinc, iron, and manganese. The aim of the present study was to determine the effect of dietary iron deficiency and overload on the expression of all 14 ZIP transporters in the liver, the main site of iron storage. Weanling male rats (n = 6/group) were fed iron-deficient (FeD), iron-adequate (FeA), or iron-overloaded (FeO) diets in two independent feeding studies. In study 1, diets were based on the TestDiet 5755 formulation and contained iron at 9 ppm (FeD), 215 ppm (FeA), and 27,974 ppm (3% FeO). In study 2, diets were based on the AIN-93G formulation and contained iron at 9 ppm Fe (FeD), 50 ppm Fe (FeA), or 18916 ppm (2% FeO). After 3 weeks, the FeD diets depleted liver non-heme iron stores and induced anemia, whereas FeO diets resulted in hepatic iron overload. Quantitative RT-PCR revealed that ZIP5 mRNA levels were 3- and 8-fold higher in 2% FeO and 3% FeO livers, respectively, compared with FeA controls. In both studies, a consistent downregulation of ZIP6, ZIP7, and ZIP10 was also observed in FeO liver relative to FeA controls. Studies in H4IIE hepatoma cells further documented that iron loading affects the expression of these ZIP transporters. Overall, our data suggest that ZIP5, ZIP6, ZIP7, and ZIP10 are regulated by iron, indicating that they may play a role in hepatic iron/metal homeostasis during iron deficiency and overload.
Collapse
Affiliation(s)
- Hyeyoung Nam
- Food Science and Human Nutrition Department, University of Florida, Gainesville, FL 32611, USA
| | | |
Collapse
|
27
|
Croxford TP, McCormick NH, Kelleher SL. Moderate zinc deficiency reduces testicular Zip6 and Zip10 abundance and impairs spermatogenesis in mice. J Nutr 2011; 141:359-65. [PMID: 21248196 PMCID: PMC3040901 DOI: 10.3945/jn.110.131318] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Male infertility accounts for ~40% of cases of failure to conceive. Testes have a strict zinc (Zn) requirement and severe Zn deficiency compromises spermatogenesis, sperm viability, and motility, compromising fertility in men. Despite the high prevalence of marginal Zn deficiency in humans, less emphasis has been placed on understanding the consequences on male reproduction. Swiss Webster mice were used to visualize Zip protein expression during spermatogenesis using immunohistochemistry. Data suggest Zip5 imports Zn into Sertoli cells and spermatocytes, augmented by Zip10 (primary spermatocytes) and Zip8 (secondary spermatocytes). Zip6, 8, and 10 expression was retained in round spermatids, although Zip8 and Zip10 expression disappears during spermatid maturation. Zip1 and Zip6 expression was detected in mature, elongated spermatids. Zip14 was detected in undifferentiated spermatogonia and Leydig cells. Mice fed diets (n = 10/group) reduced in Zn concentration [marginal-Zn diet (MZD), 10 mg Zn/kg; low-Zn diet (ZD), 7 mg Zn/kg] for 30 d had >35% lower liver Zn concentrations than mice fed the control diet (C; 30 mg Zn/kg) (P < 0.05). Plasma Zn and testosterone concentrations and the testes Zn concentration and weight were not significantly lower than in controls. Plasma Zn was greater in the ZD group than in the C and MZD groups. Mice fed ZD had a reduced number of terminal deoxynucleotidyl transferase dUTP nick end labeling-positive cells (~50%; P < 0.05), compromised seminiferous tubule structure, and reduced Zip10 and Zip6 abundance (>50%; P < 0.5) compared with mice fed C. Our data provide compelling evidence that reduced Zn intake may be associated with infertility in men, perhaps independent of decreased levels of circulating Zn or testosterone, which warrants further investigation in human populations.
Collapse
|
28
|
Schulte L, Glanfield A, Nawaratna S, Gobert GN, McManus DP, Jones MK. Molecular analysis of zinc transporters in Schistosoma japonicum. Exp Parasitol 2011; 127:768-76. [PMID: 21255570 DOI: 10.1016/j.exppara.2011.01.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2010] [Revised: 11/19/2010] [Accepted: 01/11/2011] [Indexed: 11/30/2022]
Abstract
Members of the zinc-regulated transporter/iron-regulated transporter-like protein (ZIP) family of proteins transport metal ions across cell membranes. Genes encoding ZIPs are present in the genomes of schistosomes. Here, we describe molecular characterisation of six ZIPs (SjZIPA-F) from Schistosoma japonicum. Quantitative PCR analyses of these ZIPs through the lifecycle showed that each is expressed predominantly during the intramammalian stage and are particularly enriched in adult females. Using laser microdissected tissue as template, SjZIPA-D were transcriptionally enriched in female reproductive tissues, SjZIPE was not expressed in specific tissues and SjZIPF was expressed similarly in each tissue. Whole mount in situ hybridization revealed that SjZIPA and SjZIPB were localised to the oesophageal gland of adults and the vitellaria. We have shown that multiple ZIPs are expressed by schistosomes during the intramammalian parasitic phases and propose that the encoded products perform diverse cellular functions related to metal transport in different cells of S. japonicum.
Collapse
Affiliation(s)
- Leigh Schulte
- Queensland Institute of Medical Research, 300 Herston Road, Herston, Qld 4006, Australia
| | | | | | | | | | | |
Collapse
|
29
|
John E, Laskow TC, Buchser WJ, Pitt BR, Basse PH, Butterfield LH, Kalinski P, Lotze MT. Zinc in innate and adaptive tumor immunity. J Transl Med 2010; 8:118. [PMID: 21087493 PMCID: PMC3002329 DOI: 10.1186/1479-5876-8-118] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2010] [Accepted: 11/18/2010] [Indexed: 12/19/2022] Open
Abstract
Zinc is important. It is the second most abundant trace metal with 2-4 grams in humans. It is an essential trace element, critical for cell growth, development and differentiation, DNA synthesis, RNA transcription, cell division, and cell activation. Zinc deficiency has adverse consequences during embryogenesis and early childhood development, particularly on immune functioning. It is essential in members of all enzyme classes, including over 300 signaling molecules and transcription factors. Free zinc in immune and tumor cells is regulated by 14 distinct zinc importers (ZIP) and transporters (ZNT1-8). Zinc depletion induces cell death via apoptosis (or necrosis if apoptotic pathways are blocked) while sufficient zinc levels allows maintenance of autophagy. Cancer cells have upregulated zinc importers, and frequently increased zinc levels, which allow them to survive. Based on this novel synthesis, approaches which locally regulate zinc levels to promote survival of immune cells and/or induce tumor apoptosis are in order.
Collapse
Affiliation(s)
- Erica John
- Department of Surgery, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Thomas C Laskow
- Department of Surgery, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - William J Buchser
- Department of Surgery, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Bruce R Pitt
- Department of Occupational Health, University of Pittsburgh, 100 Technology Drive, Pittsburgh, PA 15219, USA
| | - Per H Basse
- Department of Immunology, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Lisa H Butterfield
- Department of Medicine, University of Pittsburgh, 3550 Terrace Street, Pittsburgh, PA 15261, USA
| | - Pawel Kalinski
- Department of Surgery, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Michael T Lotze
- Department of Surgery, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| |
Collapse
|
30
|
Sheridan P, Kamimura T, Shimodaira H. A scale-free structure prior for graphical models with applications in functional genomics. PLoS One 2010; 5:e13580. [PMID: 21079769 PMCID: PMC2974640 DOI: 10.1371/journal.pone.0013580] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2010] [Accepted: 09/28/2010] [Indexed: 11/18/2022] Open
Abstract
The problem of reconstructing large-scale, gene regulatory networks from gene expression data has garnered considerable attention in bioinformatics over the past decade with the graphical modeling paradigm having emerged as a popular framework for inference. Analysis in a full Bayesian setting is contingent upon the assignment of a so-called structure prior—a probability distribution on networks, encoding a priori biological knowledge either in the form of supplemental data or high-level topological features. A key topological consideration is that a wide range of cellular networks are approximately scale-free, meaning that the fraction, , of nodes in a network with degree is roughly described by a power-law with exponent between and . The standard practice, however, is to utilize a random structure prior, which favors networks with binomially distributed degree distributions. In this paper, we introduce a scale-free structure prior for graphical models based on the formula for the probability of a network under a simple scale-free network model. Unlike the random structure prior, its scale-free counterpart requires a node labeling as a parameter. In order to use this prior for large-scale network inference, we design a novel Metropolis-Hastings sampler for graphical models that includes a node labeling as a state space variable. In a simulation study, we demonstrate that the scale-free structure prior outperforms the random structure prior at recovering scale-free networks while at the same time retains the ability to recover random networks. We then estimate a gene association network from gene expression data taken from a breast cancer tumor study, showing that scale-free structure prior recovers hubs, including the previously unknown hub SLC39A6, which is a zinc transporter that has been implicated with the spread of breast cancer to the lymph nodes. Our analysis of the breast cancer expression data underscores the value of the scale-free structure prior as an instrument to aid in the identification of candidate hub genes with the potential to direct the hypotheses of molecular biologists, and thus drive future experiments.
Collapse
Affiliation(s)
- Paul Sheridan
- Department of Mathematical and Computing Sciences, Tokyo Institute of Technology, Tokyo, Japan.
| | | | | |
Collapse
|
31
|
Weaver BP, Zhang Y, Hiscox S, Guo GL, Apte U, Taylor KM, Sheline CT, Wang L, Andrews GK. Zip4 (Slc39a4) expression is activated in hepatocellular carcinomas and functions to repress apoptosis, enhance cell cycle and increase migration. PLoS One 2010; 5. [PMID: 20957146 PMCID: PMC2950147 DOI: 10.1371/journal.pone.0013158] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2010] [Accepted: 09/10/2010] [Indexed: 12/27/2022] Open
Abstract
Background The zinc transporter ZIP4 (Slc39a4) is important for proper mammalian development and is an essential gene in mice. Recent studies suggest that this gene may also play a role in pancreatic cancer. Methods/Principal Findings Herein, we present evidence that this essential zinc transporter is expressed in hepatocellular carcinomas. Zip4 mRNA and protein were dramatically elevated in hepatocytes in the majority of human hepatocellular carcinomas relative to noncancerous surrounding tissues, as well as in hepatocytes in hepatocellular carcinomas occurring in farnesoid X receptor-knockout mice. Interestingly, meta-analysis of microarray data in the Geo and Oncomine databases suggests that Zip4 mRNA may also be elevated in many types of cancer. Potential mechanisms of action of ZIP4 were examined in cultured cell lines. RNAi knockdown of Zip4 in mouse Hepa cells significantly increased apoptosis and modestly slowed progression from G0/G1 to S phase when cells were released from hydroxyurea block into zinc-deficient medium. Cell migration assays revealed that RNAi knockdown of Zip4 in Hepa cells depressed in vitro migration whereas forced over-expression in Hepa cells and MCF-7 cells enhanced in vitro migration. Conclusions ZIP4 may play a role in the acquisition of zinc by hepatocellular carcinomas, and potentially many different cancerous cell-types, leading to repressed apoptosis, enhanced growth rate and enhanced invasive behavior.
Collapse
Affiliation(s)
- Benjamin P. Weaver
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Yuxia Zhang
- Departments of Medicine and Oncological Sciences, Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
| | - Stephen Hiscox
- Tenovus Centre for Cancer Research, Welsh School of Pharmacy, Cardiff University, Cardiff, United Kingdom
| | - Grace L. Guo
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Udayan Apte
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Kathryn M. Taylor
- Tenovus Centre for Cancer Research, Welsh School of Pharmacy, Cardiff University, Cardiff, United Kingdom
| | - Christian T. Sheline
- Neuroscience Center of Excellence, Louisiana State University Health Science Center, New Orleans, Louisiana, United States of America
| | - Li Wang
- Departments of Medicine and Oncological Sciences, Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
| | - Glen K. Andrews
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
- * E-mail:
| |
Collapse
|
32
|
Abstract
"Oh, Jerusalem of gold, and of light, and of bronze..." goes the popular song. But it was another metal that towered above the Jerusalem landscape during the meeting of the International Society for Zinc Biology (ISZB; http://www.iszb.org/), held at Mishkenot Sha'ananim, a whisper away from the Old City walls. More than 100 scientists gathered on 1 to 5 December 2009 to discuss their research on the biology of this metal. Zinc is a double-edged sword. Zinc supplementation accelerates wound healing and growth and promotes an effective immune response. On the other hand, zinc deficiency leads to growth retardation and impaired learning and memory function, and has been linked to mood disorders. At the cellular level, however, uncontrolled increases in zinc concentrations can lead to neuronal cell death and may be involved in neurodegenerative disorders. Through regulation of various intracellular signaling pathways, zinc can accelerate cell growth and possibly contribute to cancer. However, despite the physiological and clinical importance of this metal, research on the molecular basis of these effects is still in its infancy. The 2009 ISZB meeting provided a venue for investigators working on various zinc-related issues to share their thoughts and ideas and to promote the growth of this field.
Collapse
Affiliation(s)
- Michal Hershfinkel
- Department of Morphology, Ben Gurion University, Beer Sheva, 84105, Israel
| | | | | | | |
Collapse
|
33
|
McCormick N, Velasquez V, Finney L, Vogt S, Kelleher SL. X-ray fluorescence microscopy reveals accumulation and secretion of discrete intracellular zinc pools in the lactating mouse mammary gland. PLoS One 2010; 5:e11078. [PMID: 20552032 PMCID: PMC2884036 DOI: 10.1371/journal.pone.0011078] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2010] [Accepted: 05/20/2010] [Indexed: 11/18/2022] Open
Abstract
Background The mammary gland is responsible for the transfer of a tremendous amount of zinc (∼1–3 mg zinc/day) from maternal circulation into milk during lactation to support the growth and development of the offspring. When this process is compromised, severe zinc deficiency compromises neuronal development and immune function and increases infant morbidity and/or mortality. It remains unclear as to how the lactating mammary gland dynamically integrates zinc import from maternal circulation with the enormous amount of zinc that is secreted into milk. Methodology/Principal Findings Herein we utilized X-ray fluorescence microscopy (XFM) which allowed for the visualization and quantification of the process of zinc transfer through the mammary gland of the lactating mouse. Our data illustrate that a large amount of zinc first accumulates in the mammary gland during lactation. Interestingly, this zinc is not cytosolic, but accumulated in large, discrete sub-cellular compartments. These zinc pools were then redistributed to small intracellular vesicles destined for secretion in a prolactin-responsive manner. Confocal microscopy identified mitochondria and the Golgi apparatus as the sub-cellular compartments which accumulate zinc; however, zinc pools in the Golgi apparatus, but not mitochondria are redistributed to vesicles destined for secretion during lactation. Conclusions/Significance Our data directly implicate the Golgi apparatus in providing a large, mobilizable zinc storage pool to assist in providing for the tremendous amount of zinc that is secreted into milk. Interestingly, our study also provides compelling evidence that mitochondrial zinc pools expand in the mammary gland during lactation which we speculate may play a role in regulating mammary gland function.
Collapse
Affiliation(s)
- Nicholas McCormick
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Vanessa Velasquez
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Lydia Finney
- Argonne National Laboratory, Argonne, Illinois, United States of America
| | - Stefan Vogt
- Argonne National Laboratory, Argonne, Illinois, United States of America
| | - Shannon L. Kelleher
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
34
|
Sharir H, Zinger A, Nevo A, Sekler I, Hershfinkel M. Zinc released from injured cells is acting via the Zn2+-sensing receptor, ZnR, to trigger signaling leading to epithelial repair. J Biol Chem 2010; 285:26097-106. [PMID: 20522546 DOI: 10.1074/jbc.m110.107490] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
A role for Zn(2+) in accelerating wound healing is established, yet, the signaling pathways linking Zn(2+) to tissue repair are not well known. We show that in the human HaCaT keratinocytes extracellular Zn(2+) induces a metabotropic Ca(2+) response that is abolished by silencing the expression of the G-protein-coupled receptor GPR39, suggesting that this Zn(2+)-sensing receptor, ZnR, is mediating the response. Keratinocytic-ZnR signaling is highly selective for Zn(2+) and can be triggered by nanomolar concentrations of this ion. Interestingly, Zn(2+) was also released following cellular injury, as monitored by a specific non-permeable fluorescent Zn(2+) probe, ZnAF-2. Chelation of Zn(2+) and scavenging of ATP from conditioned medium, collected from injured epithelial cultures, was sufficient to eliminate the metabotropic Ca(2+) signaling. The signaling triggered by Zn(2+), via ZnR, or by ATP further activated MAP kinase and induced up-regulation of the sodium/proton exchanger NHE1 activity. Finally, activation of ZnR/GPR39 signaling or application of ATP enhanced keratinocytes scratch closure in an in vitro model. Thus our results indicate that extracellular Zn(2+), which is either applied or released following injury, activates ZnR/GPR39 to promote signaling leading to epithelial repair.
Collapse
Affiliation(s)
- Haleli Sharir
- Departments of Morphology, Ben Gurion University, Beer-Sheva 84105, Israel
| | | | | | | | | |
Collapse
|
35
|
Lichten LA, Cousins RJ. Mammalian zinc transporters: nutritional and physiologic regulation. Annu Rev Nutr 2009; 29:153-76. [PMID: 19400752 DOI: 10.1146/annurev-nutr-033009-083312] [Citation(s) in RCA: 538] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Research advances defining how zinc is transported into and out of cells and organelles have increased exponentially within the past five years. Research has progressed through application of molecular techniques including genomic analysis, cell transfection, RNA interference, kinetic analysis of ion transport, and application of cell and animal models including knockout mice. The knowledge base has increased for most of 10 members of the ZnT family and 14 members of the Zrt-, Irt-like protein (ZIP) family. Relative to the handling of dietary zinc is the involvement of ZnT1, ZIP4, and ZIP5 in intestinal zinc transport, involvement of ZIP10 and ZnT1 in renal zinc reabsorption, and the roles of ZIP5, ZnT2, and ZnT1 in pancreatic release of endogenous zinc. These events are major factors in regulation of zinc homeostasis. Other salient findings are the involvement of ZnT2 in lactation, ZIP14 in the hypozincemia of inflammation, ZIP6, ZIP7, and ZIP10 in metastatic breast cancer, and ZnT8 in insulin processing and as an autoantigen in diabetes.
Collapse
Affiliation(s)
- Louis A Lichten
- Nutritional Genomics Laboratory, Food Science and Human Nutrition Department and Center for Nutritional Sciences, University of Florida, Gainesville, FL 32611-2710, USA
| | | |
Collapse
|
36
|
Kelleher SL, Lopez V, Lönnerdal B, Dufner-Beattie J, Andrews GK. Zip3 (Slc39a3) functions in zinc reuptake from the alveolar lumen in lactating mammary gland. Am J Physiol Regul Integr Comp Physiol 2009; 297:R194-201. [PMID: 19458277 DOI: 10.1152/ajpregu.00162.2009] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The lactating mammary gland is composed of multiple cell types that tightly coordinate the accumulation, production, and secretion of milk components, including essential metals such as zinc (Zn). Our previous studies in animal and cell models implicated the Zn transporter Zip3 (Slc39a3) in mammary gland Zn acquisition. Herein, we investigated this hypothesis directly by utilizing Zip3-null mice. Our data verify that Zip3 is expressed in secretory mammary cells; however, Zip3 does not play a major role in Zn import from the maternal circulation. Importantly, the primary localization of Zip3 was associated with the luminal membrane of the secretory mammary cells. Consistent with this localization, Zn transfer studies using (65)Zn revealed that Zn retention in the secreted milk pool and milk Zn concentration was higher in Zip3-null compared with wild-type mice. Although total mammary gland Zn concentration was not altered, Zip3-null mice also had altered mammary tissue architecture, increased number of apoptotic cells, and reduced mammary gland weight implicating subtle changes in Zip3-mediated intracellular Zn pools in apoptosis regulation. Taken together, our data indicate that Zip3 does not participate in the acquisition of Zn from maternal circulation for secretion into milk but, in contrast, primarily plays a role in the reuptake and cellular retention of Zn in the mammary gland from the previously secreted milk pool, thus regulating cellular function.
Collapse
Affiliation(s)
- Shannon L Kelleher
- Dept. of Nutritional Sciences, 222 Chandlee Laboratory, University Park, PA 16802-6110, USA.
| | | | | | | | | |
Collapse
|