1
|
Schulz MT, Rink L. Zinc deficiency as possible link between immunosenescence and age-related diseases. Immun Ageing 2025; 22:19. [PMID: 40390089 PMCID: PMC12087153 DOI: 10.1186/s12979-025-00511-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 04/23/2025] [Indexed: 05/21/2025]
Abstract
As global life expectancy increases, research reveals a critical challenge in aging: the progressive deterioration of immune function, termed immunosenescence. This age-related immune decline is characterized by a complex dysregulation of immune responses, which leaves older adults increasingly vulnerable to infections, chronic inflammatory states, and various degenerative diseases. Without intervention, immunosenescence significantly contributes to morbidity and mortality among the elderly, intensifying healthcare burdens and diminishing quality of life on both individual and societal levels. This review explores the essential role of zinc, a trace element critical for immune health, in mitigating the impact of immunosenescence and slowing the cascade of immunological dysfunctions associated with aging. By modulating the activity of key immune cells and pathways, zinc supplementation emerges as a promising approach to strengthen immunity, reduce oxidative stress, and counteract "inflammaging," a state of chronic, low-grade inflammation that accelerates tissue damage and drives disease progression. Zinc's involvement in cellular defense and repair mechanisms across the immune system highlights its ability to enhance immune cell functionality, resilience, and adaptability, strengthening the body's resistance to infection and its ability to manage stressors that contribute to diseases of aging. Indeed, zinc has demonstrated potential to improve immune responses, decrease inflammation, and mitigate the risk of age-related conditions including diabetes, depression, cardiovascular disease, and vision loss. Given the prevalent barriers to adequate zinc intake among older adults, including dietary limitations, decreased absorption, and interactions with medications, this review underscores the urgent need to address zinc deficiency in aging populations. Recent findings on zinc's cellular and molecular effects on immune health present zinc supplementation as a practical, accessible intervention for supporting healthier aging and improving quality of life. By integrating zinc into targeted strategies, public health efforts may not only sustain immunity in the elderly but also extend healthy longevity, reduce healthcare costs, and potentially mitigate the incidence and impact of chronic diseases that strain healthcare systems worldwide.
Collapse
Affiliation(s)
- Michael Tobias Schulz
- Institute of Immunology, Faculty of Medicine, RWTH Aachen University Hospital, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Lothar Rink
- Institute of Immunology, Faculty of Medicine, RWTH Aachen University Hospital, Pauwelsstraße 30, 52074, Aachen, Germany.
| |
Collapse
|
2
|
Huang L, Nguyen ST, Yang Z, Kirschke CP, Prouteau C, Copin MC, Bonneau D, Blanchet O, Mallebranche C, Pellier I, Coutant R, Miot C, Ziegler A. Reduced AKT activation accompanied with high TP53 expression is implicated in the impaired hematogenesis in Ziegler-Huang syndrome and the Znt7 null mice partially recapitulates the human disease linked to pancytopenia. J Trace Elem Med Biol 2025; 89:127658. [PMID: 40286389 DOI: 10.1016/j.jtemb.2025.127658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 04/10/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Inherited bone marrow failure (IBMF) is a life-threatening condition. Excessive expression of TP53 induces cell cycle arrest and apoptosis of hematopoietic cells in individuals with IBMF. We recently discovered two pathogenic variants, NM_001144884:c.21dup;p.(Asp8ArgfsTer3) and NM_001144884:c.842 + 15 T > C, in ZNT7 associated with IBMF (Ziegler-Huang Syndrome; BMF8). However, the pathophysiologic mechanism of IBMF caused by ZNT7 mutations remained unknown. METHOD We investigated TP53 expression and the activation of its upstream regulator, AKT, in cell lines from affected individuals. We rescued the wild-type phenotype of AKT activation via transduction of wild-type ZNT7 into patient's fibroblasts. We performed fluorescence microscopy to assess co-expression patterns of ZNT7 with hematopoietic cell markers in different human and mouse bone marrow cell types. Finally, we evaluated the hematological features of Znt7 deficient mice. RESULTS The growth of patient's EBV-transformed B (B-EBV) lymphoblasts was impaired. We observed excessive expression of TP53 in the patient's B-EBV lymphoblasts accompanied by a significant decrease in AKT activation. Importantly, overexpression of wild-type ZNT7 in patient's fibroblasts rescued the activation of the AKT pathway by insulin. Additionally, human ZNT7 was expressed in myeloid and lymphoid lineage cells, whereas mouse ZnT7 was mainly expressed in the nucleated hematopoietic cells in the respective bone marrow. Despite these differences, we observed progressive cytopenia in Znt7KO mice, partially recapitulating BMF8 in humans. CONCLUSION Excessive expression of TP53 and down-regulation of AKT activation induced by ZNT7 deficiency might impair cell survival, which may contribute to the pathophysiology of bone marrow failure in affected individuals with BMF8.
Collapse
Affiliation(s)
- Liping Huang
- USDA/ARS/Western Human Nutrition Research Center, 430 West Health Sciences Drive, Davis, CA 95616, USA; Graduate Group of Nutritional Biology, Department of Nutrition, University of California at Davis, One Shields Ave, Davis, CA 95616, USA; Integrative Genetics and Genomics, University of California at Davis, One Shields Ave, Davis, CA 95616, USA.
| | - Steven T Nguyen
- USDA/ARS/Western Human Nutrition Research Center, 430 West Health Sciences Drive, Davis, CA 95616, USA
| | - Zhongyue Yang
- Graduate Group of Nutritional Biology, Department of Nutrition, University of California at Davis, One Shields Ave, Davis, CA 95616, USA
| | - Catherine P Kirschke
- USDA/ARS/Western Human Nutrition Research Center, 430 West Health Sciences Drive, Davis, CA 95616, USA
| | - Clément Prouteau
- Department of Genetics, University Hospital of Angers, 4 rue Larrey, Cedex 9, Angers 49333, France
| | | | - Dominique Bonneau
- Department of Genetics, University Hospital of Angers, 4 rue Larrey, Cedex 9, Angers 49333, France
| | - Odile Blanchet
- Biobank Resource Center, CHU Angers, University Hospital of Angers, 4 rue Larrey, Cedex 9, BB-0033-00038, 49333, France; University Angers Nantes Université CHU Angers INSERM CNRS CRCI2NA SFR ICAT, Cedex 9, Angers 49333, France
| | - Coralie Mallebranche
- University Angers Nantes Université CHU Angers INSERM CNRS CRCI2NA SFR ICAT, Cedex 9, Angers 49333, France
| | - Isabelle Pellier
- University Angers Nantes Université CHU Angers INSERM CNRS CRCI2NA SFR ICAT, Cedex 9, Angers 49333, France
| | - Régis Coutant
- Department of Pediatric Endocrinology, University Hospital of Angers, Cedex 9, Angers 49333, France; Reference Center for Rare Pituitary Diseases, University of Hospital of Angers, Cedex 9, Angers 49333, France
| | - Charline Miot
- University Angers Nantes Université CHU Angers INSERM CNRS CRCI2NA SFR ICAT, Cedex 9, Angers 49333, France; Immunology and Allergology Laboratory, University Hospital of Angers, 4 rue Larrey, Cedex 9, Angers 49333, France
| | - Alban Ziegler
- Department of Genetics, University Hospital of Angers, 4 rue Larrey, Cedex 9, Angers 49333, France; Deparment of Medical Genetics, University Hospital of Toulouse, Toulouse 31100, France.
| |
Collapse
|
3
|
Du J, Shen M, Chen J, Yan H, Xu Z, Yang X, Yang B, Luo P, Ding K, Hu Y, He Q. The impact of solute carrier proteins on disrupting substance regulation in metabolic disorders: insights and clinical applications. Front Pharmacol 2025; 15:1510080. [PMID: 39850557 PMCID: PMC11754210 DOI: 10.3389/fphar.2024.1510080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 12/20/2024] [Indexed: 01/25/2025] Open
Abstract
Carbohydrates, lipids, bile acids, various inorganic salt ions and organic acids are the main nutrients or indispensable components of the human body. Dysregulation in the processes of absorption, transport, metabolism, and excretion of these metabolites can lead to the onset of severe metabolic disorders, such as type 2 diabetes, non-alcoholic fatty liver disease, gout and hyperbilirubinemia. As the second largest membrane receptor supergroup, several major families in the solute carrier (SLC) supergroup have been found to play key roles in the transport of substances such as carbohydrates, lipids, urate, bile acids, monocarboxylates and zinc ions. Based on common metabolic dysregulation and related metabolic substances, we explored the relationship between several major families of SLC supergroup and metabolic diseases, providing examples of drugs targeting SLC proteins that have been approved or are currently in clinical/preclinical research as well as SLC-related diagnostic techniques that are in clinical use or under investigation. By highlighting these connections, we aim to provide insights that may contribute to the development of improved treatment strategies and targeted therapies for metabolic disorders.
Collapse
Affiliation(s)
- Jiangxia Du
- Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Minhui Shen
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jiajia Chen
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hao Yan
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zhifei Xu
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaochun Yang
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Bo Yang
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Peihua Luo
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Department of Pharmaceutical and Translational Toxicology, Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, Zhejiang, China
| | - Kefeng Ding
- Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yuhuai Hu
- Yuhong Pharmaceutical Technology Co., Ltd., Hangzhou, Zhejiang, China
| | - Qiaojun He
- Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
- Department of Pharmaceutical and Translational Toxicology, Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
4
|
Kiouri DP, Chasapis CT, Mavromoustakos T, Spiliopoulou CA, Stefanidou ME. Zinc and its binding proteins: essential roles and therapeutic potential. Arch Toxicol 2025; 99:23-41. [PMID: 39508885 DOI: 10.1007/s00204-024-03891-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 10/09/2024] [Indexed: 11/15/2024]
Abstract
Zinc is an essential micronutrient that participates in a multitude of cellular and biochemical processes. It is indispensable for normal growth and the maintenance of physiological functions. As one of the most significant trace elements in the body, zinc fulfills three primary biological roles: catalytic, structural, and regulatory. It serves as a cofactor in over 300 enzymes, and more than 3000 proteins require zinc, underscoring its crucial role in numerous physiological processes such as cell division and growth, immune function, tissue maintenance, as well as synthesis protein and collagen synthesis. Zinc deficiency has been linked to increased oxidative stress and inflammation, which may contribute to the pathogenesis of a multitude of diseases, like neurological disorders and cancer. In addition, zinc is a key constituent of zinc-binding proteins, which play a pivotal role in maintaining cellular zinc homeostasis. This review aims to update and expand upon the understanding of zinc biology, highlighting the fundamental roles of zinc in biological processes and the health implications of zinc deficiency. This work also explores the diverse functions of zinc in immune regulation, cellular growth, and neurological health, emphasizing the need for further research to fully elucidate the therapeutic potential of zinc supplementation in disease prevention and management.
Collapse
Affiliation(s)
- Despoina P Kiouri
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635, Athens, Greece
- Department of Chemistry, Laboratory of Organic Chemistry, National and Kapodistrian University of Athens, 15772, Athens, Greece
| | - Christos T Chasapis
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635, Athens, Greece.
| | - Thomas Mavromoustakos
- Department of Chemistry, Laboratory of Organic Chemistry, National and Kapodistrian University of Athens, 15772, Athens, Greece
| | - Chara A Spiliopoulou
- Department of Forensic Medicine and Toxicology, School of Medicine, National and Kapodistrian University of Athens, 11527, Athens, Greece
| | - Maria E Stefanidou
- Department of Forensic Medicine and Toxicology, School of Medicine, National and Kapodistrian University of Athens, 11527, Athens, Greece.
| |
Collapse
|
5
|
Chantarasakha K, Yangchum A, Isaka M, Tepaamorndech S. Fungal Depsidones Stimulate AKT-Dependent Glucose Uptake in 3T3-L1 Adipocytes. JOURNAL OF NATURAL PRODUCTS 2024; 87:1673-1681. [PMID: 38597733 PMCID: PMC11287747 DOI: 10.1021/acs.jnatprod.3c01134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 03/27/2024] [Accepted: 03/29/2024] [Indexed: 04/11/2024]
Abstract
Enhanced glucose uptake in insulin-sensitive tissues is one of the therapeutic strategies to ameliorate hyperglycemia and maintain glucose homeostasis in type 2 diabetes. This study disclosed the role of fungal depsidones in glucose uptake and the underlying mechanism in 3T3-L1 adipocytes. Depsidones, including nidulin, nornidulin, and unguinol, isolated from Aspergillus unguis, stimulate glucose uptake in adipocytes. Compared to the others, nidulin exhibited an upward trend in glucose uptake. The effect of nidulin was found to be dose- and time-dependent. Nidulin also enhanced insulin- and metformin-stimulated glucose uptake. Upregulation of GLUT4 expression and AKT and AMPK phosphorylation were observed with nidulin treatment. Blockage of AKT, but not AMPK, phosphorylation was largely accompanied by diminished glucose uptake. In agreement, nidulin triggered the translocation of GLUT4 to the plasma membrane. Importantly, nidulin elevated glucose uptake associated with increased AKT phosphorylation in insulin-resistant adipocytes. Taken together, nidulin could stimulate glucose uptake mainly through AKT-dependent GLUT4 translocation, serving as a seed compound in drug discovery for type 2 diabetes.
Collapse
Affiliation(s)
- Kanittha Chantarasakha
- National
Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency
(NSTDA), 111 Thailand
Science Park, Phahonyothin Road, Klong Luang, Pathumthani 12120, Thailand
| | - Arunrat Yangchum
- National
Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency
(NSTDA), 111 Thailand
Science Park, Phahonyothin Road, Klong Luang, Pathumthani 12120, Thailand
| | - Masahiko Isaka
- National
Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency
(NSTDA), 111 Thailand
Science Park, Phahonyothin Road, Klong Luang, Pathumthani 12120, Thailand
| | - Surapun Tepaamorndech
- Department
of Microbiology, Faculty of Medicine, Chulalongkorn
University and King Chulalongkorn Memorial Hospital, Bangkok 10330, Thailand
| |
Collapse
|
6
|
Franco C, Canzoniero LMT. Zinc homeostasis and redox alterations in obesity. Front Endocrinol (Lausanne) 2024; 14:1273177. [PMID: 38260166 PMCID: PMC10800374 DOI: 10.3389/fendo.2023.1273177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 12/18/2023] [Indexed: 01/24/2024] Open
Abstract
Impairment of both cellular zinc and redox homeostasis is a feature of several chronic diseases, including obesity. A significant two-way interaction exists between redox metabolism and the relatively redox-inert zinc ion. Redox metabolism critically influences zinc homeostasis and controls its cellular availability for various cellular functions by regulating zinc exchange from/to zinc-binding proteins. Zinc can regulate redox metabolism and exhibits multiple pro-antioxidant properties. On the other hand, even minor disturbances in zinc status and zinc homeostasis affect systemic and cellular redox homeostasis. At the cellular level, zinc homeostasis is regulated by a multi-layered machinery consisting of zinc-binding molecules, zinc sensors, and two selective families of zinc transporters, the Zinc Transporter (ZnT) and Zrt, Irt-like protein (ZIP). In the present review, we summarize the current state of knowledge on the role of the mutual interaction between zinc and redox homeostasis in physiology and pathophysiology, pointing to the role of zinc in the alterations responsible for redox stress in obesity. Since zinc transporters primarily control zinc homeostasis, we describe how changes in the expression and activity of these zinc-regulating proteins are associated with obesity.
Collapse
|
7
|
Smerchek DT, Rients EL, McLaughlin AM, Thornton KJ, Hansen SL. Influence of steroidal implants and zinc sulfate supplementation on growth performance, trace mineral status, circulating metabolites, and transcriptional changes in skeletal muscle of feedlot steers. J Anim Sci 2024; 102:skae154. [PMID: 38828800 PMCID: PMC11190378 DOI: 10.1093/jas/skae154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 05/31/2024] [Indexed: 06/05/2024] Open
Abstract
Angus-cross steers (n = 144; 362 kg ± 20.4) were used to determine the effect of Zn and steroidal implants on performance, trace mineral status, circulating metabolites, and transcriptional changes occurring in skeletal muscle. Steers (n = 6 per pen) were stratified by body weight (BW) in a 3 × 2 factorial. GrowSafe bunks recorded individual feed intake (steer as experimental unit; n = 24 per treatment). Dietary treatments (ZINC; eight pens per treatment) included supplemental Zn as ZnSO4 at 1) 0 (analyzed 54 mg Zn/kg DM; Zn0); 2) 30 mg/kg DM (Zn30); 3) 100 mg Zn/kg DM (Zn100). After 60 d of Zn treatment, steers received a steroidal implant treatment (IMP) on day 0: 1) no implant; NO; or 2) high-potency combination implant (TE-200, Elanco, Greenfield, IN; 200 mg TBA, 20 mg E2; TE200). BWs were taken at days -60, 0, and in 28 d increments thereafter. Liver biopsies for TM analysis and blood for TM, serum glucose, insulin, nonesterified fatty acids (NEFA), urea-N, and IGF-1 analysis were collected on days 0, 20, 40, and 84. Glucose, NEFA, and insulin were used to calculate the revised quantitative insulin sensitivity check index (RQUICKI). Linear and quadratic effects of ZINC were evaluated in SAS 9.4. Means for IMP were separated using the LSMEANS statement with the PDIFF option. Day -60 BW was a covariate for performance and carcass data. Growth performance, plasma, liver, and metabolite data were analyzed as repeated measures. TE200 tended to decrease plasma Zn by 8.4% from days 0 to 20 while NO decreased by 3.6% (IMP × day; P = 0.08). A tendency for a ZINC × day effect on G:F was noted (P = 0.06) driven by Zn30 and Zn100 decreasing significantly from period 0-28 to period 28-56 while Zn0 was similar in both periods. An IMP × day effect was noted for RQUICKI where (P = 0.02) TE200 was greater on day 40 compared to NO cattle, but by day 84 RQUICKI was not different between TE200 and NO. On day 20, increasing Zn supplementation linearly increased mRNA abundance (P ≤ 0.09) of protein kinase B (AKT1), mammalian target of rapamycin (mTOR), matrix metalloproteinase 2 (MMP2), and myogenic factor 5 (MYF5). In this study, Zn and implants differentially affected genes related to energy metabolism, satellite cell function, and TM homeostasis on days 20 and 84 postimplant. These results suggest steroidal implants increase demand for Zn immediately following implant administration to support growth and may influence insulin sensitivity in finishing cattle.
Collapse
Affiliation(s)
- Dathan T Smerchek
- Department of Animal Science, Iowa State University, Ames, IA 50011, USA
| | - Emma L Rients
- Department of Animal Science, Iowa State University, Ames, IA 50011, USA
| | - Amy M McLaughlin
- Department of Animal Science, Iowa State University, Ames, IA 50011, USA
| | - Kara J Thornton
- Department of Animal, Dairy and Veterinary Science, Utah State University, Logan, UT 84322, USA
| | - Stephanie L Hansen
- Department of Animal Science, Iowa State University, Ames, IA 50011, USA
| |
Collapse
|
8
|
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
|
9
|
Tuncay E, Aktay I, Turan B. Overexpression of Slc30a7/ZnT7 increases the mitochondrial matrix levels of labile Zn 2+ and modifies histone modification in hyperinsulinemic cardiomyoblasts. J Trace Elem Med Biol 2023; 78:127198. [PMID: 37196548 DOI: 10.1016/j.jtemb.2023.127198] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 04/26/2023] [Accepted: 05/11/2023] [Indexed: 05/19/2023]
Abstract
BACKGROUND Cellular free Zn2+ concentrations ([Zn2+]) are primarily coordinated by Zn2+-transporters, although their roles are not well established in cardiomyocytes. Since we previously showed the important contribution of a Zn2+-transporter ZnT7 to [Zn2+]i regulation in hyperglycemic cardiomyocytes, here, we aimed to examine a possible regulatory role of ZnT7 not only on [Zn2+]i but also both the mitochondrial-free Zn2+ and/or Ca2+ in cardiomyocytes, focusing on the contribution of its overexpression to the mitochondrial function. METHODS We mimicked either hyperinsulinemia (by 50-μM palmitic acid, PA-cells, for 24-h) or overexpressed ZnT7 (ZnT7OE-cells) in H9c2 cardiomyoblasts. RESULTS Opposite to PA-cells, the [Zn2+]i in ZnT7OE-cells was not different from untreated H9c2-cells. An investigation of immunofluorescence imaging by confocal microscopy demonstrated a ZnT7 localization on the mitochondrial matrix. We demonstrated the ZnT7 localization on the mitochondrial matrix by using immunofluorescence imaging. Later, we determined the mitochondrial levels of [Zn2+]Mit and [Ca2+]Mit by using the Zn2+ and Ca2+ sensitive FRET probe and a Ca2+-sensitive dye Fluo4, respectively. The [Zn2+]Mit was found to increase significantly in ZnT7OE-cells, similar to the PA-cells while no significant changes in the [Ca2+]Mit in these cells. To examine the contribution of ZnT7 overexpression on the mitochondria function, we determined the level of reactive oxygen species (ROS) and the mitochondrial membrane potential (MMP) in these cells in comparison to the PA-cells. There were significantly increased production of ROS and depolarization in MMP and increases in marker proteins of mitochondria-associated apoptosis and autophagy in ZnT7-OE cells, similar to the PA-cells, parallel to increases in K-acetylation. Moreover, we determined significant increases in trimethylation of histone H3 lysine27, H3K27me3, and the mono-methylation of histone H3 lysine36, H3K36 in the ZnT7OE-cells, demonstrating the role of [Zn2+]Mit in epigenetic regulation of cardiomyocytes under hyperinsulinemia through histone modification. CONCLUSIONS Overall, our data have shown an important contribution of high expression of ZnT7-OE, through its buffering and muffling capacity in cardiomyocytes, on the regulation of not only [Zn2+]i but also both [Zn2+]Mit and [Ca2+]Mit affecting mitochondria function, in part, via histone modification.
Collapse
Affiliation(s)
- Erkan Tuncay
- Department of Biophysics, Ankara University, Faculty of Medicine, Ankara, Turkiye
| | - Irem Aktay
- Department of Biophysics, Ankara University, Faculty of Medicine, Ankara, Turkiye
| | - Belma Turan
- Department of Biophysics, Lokman Hekim University, Faculty of Medicine, Ankara, Turkiye.
| |
Collapse
|
10
|
Ruz M, Andrews-Guzmán M, Arredondo-Olguín M. Modulation of Zinc Transporter Expressions by Additional Zinc in C2C12 Cells Cultured in a High Glucose Environment and in the Presence of Insulin or Interleukin-6. Biol Trace Elem Res 2022; 201:3428-3437. [PMID: 36227447 DOI: 10.1007/s12011-022-03443-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 10/03/2022] [Indexed: 11/02/2022]
Abstract
Zn status has been related to various chronic diseases presenting oxidative stress and inflammation, such as type 2 diabetes. Zn supplementation has been suggested to be a potential coadjuvant in the management of this condition. Zn transporters constitute a key component in the maintenance of Zn homeostasis. Our aim was to evaluate the modulatory effect of additional Zn (10 or 100 µM; as a ZnSO4*7H20) on the mRNA relative expression of selected Zn transporters (ZnT1, ZnT5, ZnT7, ZIP6, ZIP7, ZIP10, ZIP14), in myoblast (C2C12) cells cultured in normal (10 mM) and high glucose (30 mM), and in the absence or presence of insulin (1 nM), and interleukin-6 (IL-6; 5 nM) for 24 h. The main findings of our study were that in high glucose conditions in absence of insulin or IL-6, additional Zn increased ZnT1 and ZIP6, and decreased ZnT5 and ZIP7 expressions. However, this situation is modified by insulin, where incremental Zn induced increased expressions of ZnT1, ZnT5, and all the ZIP transporters studied. In high glucose conditions and in the presence of IL-6, additional Zn caused increased expressions of ZnT7, ZIP7, and ZIP14, compared with results in the absence of IL-6. This study provides preliminary evidence for the differential expression of selected Zn transporters in C2C12 cells subjected to high glucose and incremental Zn, suggesting that important changes in intracellular Zn distribution take place in response to inflammatory and high-insulin environments. Further study is necessary to understand the implications of these findings.
Collapse
Affiliation(s)
- Manuel Ruz
- Department of Nutrition, Faculty of Medicine, University of Chile, Avenida Independencia 1027, Independencia, Santiago, Chile
| | - Mónica Andrews-Guzmán
- Micronutrient Laboratory, Institute of Nutrition and Food Technology, University of Chile, Macul 5540, Macul, Santiago, Chile
| | - Miguel Arredondo-Olguín
- Micronutrient Laboratory, Institute of Nutrition and Food Technology, University of Chile, Macul 5540, Macul, Santiago, Chile.
| |
Collapse
|
11
|
Roles of ZnT86D in Neurodevelopment and Pathogenesis of Alzheimer Disease in a Drosophila melanogaster Model. Int J Mol Sci 2022; 23:ijms231911832. [PMID: 36233134 PMCID: PMC9569493 DOI: 10.3390/ijms231911832] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/06/2022] [Accepted: 09/07/2022] [Indexed: 11/18/2022] Open
Abstract
Zinc is a fundamental trace element essential for numerous biological processes, and zinc homeostasis is regulated by the Zrt-/Irt-like protein (ZIP) and zinc transporter (ZnT) families. ZnT7 is mainly localized in the Golgi apparatus and endoplasmic reticulum (ER) and transports zinc into these organelles. Although previous studies have reported the role of zinc in animal physiology, little is known about the importance of zinc in the Golgi apparatus and ER in animal development and neurodegenerative diseases. In this study, we demonstrated that ZnT86D, a Drosophila ortholog of ZnT7, plays a pivotal role in the neurodevelopment and pathogenesis of Alzheimer disease (AD). When ZnT86D was silenced in neurons, the embryo-to-adult survival rate, locomotor activity, and lifespan were dramatically reduced. The toxic phenotypes were accompanied by abnormal neurogenesis and neuronal cell death. Furthermore, knockdown of ZnT86D in the neurons of a Drosophila AD model increased apoptosis and exacerbated neurodegeneration without significant changes in the deposition of amyloid beta plaques and susceptibility to oxidative stress. Taken together, our results suggest that an appropriate distribution of zinc in the Golgi apparatus and ER is important for neuronal development and neuroprotection and that ZnT7 is a potential protective factor against AD.
Collapse
|
12
|
Heiderscheit KJ, Hansen SL. Effect of increasing zinc supplementation on post-transit performance, behavior, blood and muscle metabolites, and gene expression in growing beef feedlot steers. J Anim Sci 2022; 100:6653514. [PMID: 35917831 PMCID: PMC9512101 DOI: 10.1093/jas/skac246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 07/30/2022] [Indexed: 11/12/2022] Open
Abstract
Fifty-four Angus-cross steers (297 kg ± 12) were stratified by body weight (BW) to pens (six steers per pen) to determine the effects of supplemental Zn on posttransit growth performance and blood and muscle metabolites. Dietary treatments started 25 d before trucking: control (CON; analyzed 54 mg Zn/kg DM), industry (IND; CON + 70 mg supplemental Zn/kg DM), and supranutritional Zn (SUPZN; CON + 120 mg supplemental Zn/kg DM). Supplemental Zn was bis-glycinate bound Zn (Plexomin Zn; Phytobiotics North America, Cary, NC). On day 0, steers were loaded onto a commercial trailer and transported in 18 h (1,822 km). Individual BW was recorded on days –26, –25, –1, and 0 (pre-transit), 1 (posttransit), 6, 27, and 28. Blood was collected on days –1, 1, 6, and 27. Longissimus thoracis biopsies were collected on days –1, 1, and 28. Daily individual feed disappearance was recorded via GrowSafe bunks. Data were analyzed using Proc Mixed of SAS with fixed effect of diet and steer as the experimental unit (growth performance, blood: n = 18 steers per treatment; muscle: n = 12 steers per treatment). Individual initial BW was used as a covariate in BW analysis. Contrast statements to test linear, quadratic, and Zn effects were used to analyze performance and blood parameters. Repeated measures analysis was used for posttransit DMI recovery and weekly posttransit DMI and Zn intake with the repeated effect of time. MetaboAnalyst 5.0 was utilized for statistical analysis of day 1 (off truck) muscle metabolites. Plasma Zn linearly increased due to Zn on days 1, 6, and 27 (P = 0.01), and off-truck (day 1) serum lactate increased over day –1 by 20%, 0%, and 20% in CON, IND, and SUPZN, respectively (Quadratic: P = 0.01). Muscle lactate tended to increase posttransit in CON and IND (P ≤ 0.07) but not SUPZN. Muscle metabolites relating to amino acid and nitrogen metabolism were increased in all treatments posttransit (P ≤ 0.02), and alanine-glucose cycle metabolites tended to increase in CON and IND (P ≤ 0.07). Steers supplemented with Zn recovered pretransit DMI quicker than CON (by d 2: P = 0.01), while IND had greater overall posttransit DMI than CON with SUPZN intermediate (P = 0.04), and Zn-fed steers had greater ADG posttransit (P = 0.04). Zinc supplementation mitigated muscle or serum lactate increases due to transit and increased posttransit ADG.
Collapse
|
13
|
Pérez-García A, Torrecilla-Parra M, Fernández-de Frutos M, Martín-Martín Y, Pardo-Marqués V, Ramírez CM. Posttranscriptional Regulation of Insulin Resistance: Implications for Metabolic Diseases. Biomolecules 2022; 12:biom12020208. [PMID: 35204710 PMCID: PMC8961590 DOI: 10.3390/biom12020208] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 01/14/2022] [Accepted: 01/17/2022] [Indexed: 12/14/2022] Open
Abstract
Insulin resistance defines an impairment in the biologic response to insulin action in target tissues, primarily the liver, muscle, adipose tissue, and brain. Insulin resistance affects physiology in many ways, causing hyperglycemia, hypertension, dyslipidemia, visceral adiposity, hyperinsulinemia, elevated inflammatory markers, and endothelial dysfunction, and its persistence leads to the development metabolic disease, including diabetes, obesity, cardiovascular disease, or nonalcoholic fatty liver disease (NAFLD), as well as neurological disorders such as Alzheimer’s disease. In addition to classical transcriptional factors, posttranscriptional control of gene expression exerted by microRNAs and RNA-binding proteins constitutes a new level of regulation with important implications in metabolic homeostasis. In this review, we describe miRNAs and RBPs that control key genes involved in the insulin signaling pathway and related regulatory networks, and their impact on human metabolic diseases at the molecular level, as well as their potential use for diagnosis and future therapeutics.
Collapse
|
14
|
Li W, Yang S, Chen G, He S. MiR-200c-3p regulates pyroptosis by targeting SLC30A7 in diabetic retinopathy. Hum Exp Toxicol 2022; 41:9603271221099589. [PMID: 35607288 DOI: 10.1177/09603271221099589] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
CLINICAL RELEVANCE MicroRNAs (miRNAs) have been reported to be involved in the progression of various diseases. Studying the regulatory mechanisms of miRNAs can help clinical treatment. BACKGROUND Diabetic retinopathy (DR) is one of the complications of diabetes. The objective of this study was to elucidate the underlying molecular mechanisms by which miR-200c-3p regulates the pyroptosis of DR cell. METHODS Human retinal microvascular endothelial cells (HRMECs) and high glucose (HG) cultures established DR cell model in vitro. RT-qPCR is used to detect the expression level of miRNAs. CCK-8 assays and flow cytometry are used to detect apoptosis of HRMECs cell. Western blotting is used to detect cleaved caspase-3, cleaved caspase-1, and N-GSDMD proteins levels in HRMECs. The ELISA assay is used to detect the expression of IL-1β and IL-18. Predict and validate potential binding sites between miR-200c-3p and SLC30A7 by dual luciferase reporter gene analysis. RESULTS The results showed that HG caused damage to HRMECs through the pyroptosis pathway rather than the apoptosis pathway. MiR-200c-3p is highly expressed in HG induced-HRMECs, and knockdown of miR-200c-3p mitigates HG-induced HRMECs pyroptosis. MiR-200c-3p negatively targets SLC30A7 in HRMECs, and miR-200c-3p regulates pyroptosis of HG-induced HRMECs by targeting SLC30A7. CONCLUSION The results suggest that miR-200c-3p might be a promising interference target for DR prevention and treatment. The results of current study may provide new insights into development of therapeutic strategies for DR.
Collapse
Affiliation(s)
- Weina Li
- Department of Glaucoma and Cataract, Liuzhou Aier Eye Hospital, Affiliated Hospital of Aier Ophthalmology College of Central South University, Liuzhou, China
| | - Sheng Yang
- Department of Glaucoma and Cataract, Liuzhou Aier Eye Hospital, Affiliated Hospital of Aier Ophthalmology College of Central South University, Liuzhou, China
| | - Guangsheng Chen
- Department of Glaucoma and Cataract, Liuzhou Aier Eye Hospital, Affiliated Hospital of Aier Ophthalmology College of Central South University, Liuzhou, China
| | - Shiping He
- Department of Glaucoma and Cataract, Liuzhou Aier Eye Hospital, Affiliated Hospital of Aier Ophthalmology College of Central South University, Liuzhou, China
| |
Collapse
|
15
|
Tamura Y. The Role of Zinc Homeostasis in the Prevention of Diabetes Mellitus and Cardiovascular Diseases. J Atheroscler Thromb 2021; 28:1109-1122. [PMID: 34148917 PMCID: PMC8592709 DOI: 10.5551/jat.rv17057] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 05/25/2021] [Indexed: 11/30/2022] Open
Abstract
Zinc is an essential micronutrient for human health and is involved in various biological functions, such as growth, metabolism, and immune function. In recent years, research on intracellular zinc dynamics has progressed, and it has become clear that zinc transporters strictly control intracellular zinc localization, zinc regulates the functions of various proteins and signal transduction pathways as a second messenger similar to calcium ions, and intracellular zinc dyshomeostasis is associated with impaired insulin synthesis, secretion, sensitivity, lipid metabolism, and vascular function. Numerous animal and human studies have shown that zinc deficiency may be associated with the risk factors for diabetes and cardiovascular diseases (CVDs) and zinc administration might be beneficial for the prevention and treatment of these diseases. Therefore, an understanding of zinc biology may help the establishment of novel strategies for the prevention and treatment of diabetes and CVDs. This review will summarize the current knowledge on the role of zinc homeostasis in the pathogenesis of diabetes and atherosclerosis and will discuss the potential of zinc in the prevention of these diseases.
Collapse
Affiliation(s)
- Yukinori Tamura
- Division of Physiology and Biochemistry, Faculty of Nutrition, Kobe Gakuin University, Kobe, Japan
| |
Collapse
|
16
|
Abstract
Since the discovery of manifest Zn deficiency in 1961, the increasing number of studies demonstrated the association between altered Zn status and multiple diseases. In this chapter, we provide a review of the most recent advances on the role of Zn in health and disease (2010-20), with a special focus on the role of Zn in neurodegenerative and neurodevelopmental disorders, diabetes and obesity, male and female reproduction, as well as COVID-19. In parallel with the revealed tight association between ASD risk and severity and Zn status, the particular mechanisms linking Zn2+ and ASD pathogenesis like modulation of synaptic plasticity through ProSAP/Shank scaffold, neurotransmitter metabolism, and gut microbiota, have been elucidated. The increasing body of data indicate the potential involvement of Zn2+ metabolism in neurodegeneration. Systemic Zn levels in Alzheimer's and Parkinson's disease were found to be reduced, whereas its sequestration in brain may result in modulation of amyloid β and α-synuclein processing with subsequent toxic effects. Zn2+ was shown to possess adipotropic effects through the role of zinc transporters, zinc finger proteins, and Zn-α2-glycoprotein in adipose tissue physiology, underlying its particular role in pathogenesis of obesity and diabetes mellitus type 2. Recent findings also contribute to further understanding of the role of Zn2+ in spermatogenesis and sperm functioning, as well as oocyte development and fertilization. Finally, Zn2+ was shown to be the potential adjuvant therapy in management of novel coronavirus infection (COVID-19), underlining the perspectives of zinc in management of old and new threats.
Collapse
Affiliation(s)
- Anatoly V Skalny
- IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia; Yaroslavl State University, Yaroslavl, Russia
| | - Michael Aschner
- IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia; Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Alexey A Tinkov
- IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia; Yaroslavl State University, Yaroslavl, Russia.
| |
Collapse
|
17
|
Zhang X, Guan T, Yang B, Chi Z, Wan Q, Gu HF. SLC30A7 has anti-oxidant stress effects in high glucose-induced apoptosis via the NFE2L2/HMOX1 signal transduction pathway. Diabetes Res Clin Pract 2021; 172:108445. [PMID: 32949653 DOI: 10.1016/j.diabres.2020.108445] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 09/10/2020] [Indexed: 01/13/2023]
Abstract
AIMS Apoptosis and oxidant stress are known to be involved in the pathogenesis of diabetic kidney disease (DKD). We have previously reported that zinc transporter 7 in SLC30 family (SLC30A7) inhibits apoptosis in rat peritoneal mesothelial cells under high glucose (HG) conditions. In the current study, we aimed to investigate whether SLC30A7 had effect for anti-oxidant stress in renal tubular epithelial cells under HG. METHODS SLC30A7 in HG-induced apoptosis in a normal rat kidney tubular epithelial cell line (NRK-52E cells)/kidneys of STZ-induced diabetic mice was examined and the activity of nuclear factor erythroid 2-related factor 2 (NFE2L2) was further analyzed by using real time RT-PCR, siRNA and Western blot protocols. RESULTS SLC30A7 was found to be up-regulated, while NFE2L2 was activated in kidneys of STZ-induced diabetic mice and HG-induced apoptosis of NRK-52E cells. Knock-down of SLC30A7 with siRNA protocol resulted in lower intracellular free zinc levels in the cells and decreased zinc distribution in the Golgi apparatus. Furthermore, knock-down of NFE2L2 down-regulated its target HMOX1 gene expression, decreased SLC30A7 activity but increased HG-induced apoptosis. CONCLUSION The current study provides new evidence that SLC30A7 has anti-oxidant stress effects in HG-induced apoptosis via the NFE2L2/HMOX1 signal transduction pathway.
Collapse
Affiliation(s)
- Xiuli Zhang
- Department of Nephrology, Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong Province 518000, PR China; Department of Pathophysiology, China Medical University, Shenyang, Liaoning Province 110001, PR China.
| | - Tingwen Guan
- Department of Pathophysiology, China Medical University, Shenyang, Liaoning Province 110001, PR China
| | - Boxuan Yang
- Department of Pathophysiology, China Medical University, Shenyang, Liaoning Province 110001, PR China
| | - Zhihong Chi
- Department of Pathophysiology, China Medical University, Shenyang, Liaoning Province 110001, PR China
| | - Qijun Wan
- Department of Nephrology, Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong Province 518000, PR China
| | - Harvest F Gu
- Center for Pathophysiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province 210009, PR China.
| |
Collapse
|
18
|
The role of labile Zn 2+ and Zn 2+-transporters in the pathophysiology of mitochondria dysfunction in cardiomyocytes. Mol Cell Biochem 2020; 476:971-989. [PMID: 33225416 DOI: 10.1007/s11010-020-03964-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 10/23/2020] [Indexed: 02/06/2023]
Abstract
An important energy supplier of cardiomyocytes is mitochondria, similar to other mammalian cells. Studies have demonstrated that any defect in the normal processes controlled by mitochondria can lead to abnormal ROS production, thereby high oxidative stress as well as lack of ATP. Taken into consideration, the relationship between mitochondrial dysfunction and overproduction of ROS as well as the relation between increased ROS and high-level release of intracellular labile Zn2+, those bring into consideration the importance of the events related with those stimuli in cardiomyocytes responsible from cellular Zn2+-homeostasis and responsible Zn2+-transporters associated with the Zn2+-homeostasis and Zn2+-signaling. Zn2+-signaling, controlled by cellular Zn2+-homeostatic mechanisms, is regulated with intracellular labile Zn2+ levels, which are controlled, especially, with the two Zn2+-transporter families; ZIPs and ZnTs. Our experimental studies in mammalian cardiomyocytes and human heart tissue showed that Zn2+-transporters localizes to mitochondria besides sarco(endo)plasmic reticulum and Golgi under physiological condition. The protein levels as well as functions of those transporters can re-distribute under pathological conditions, therefore, they can interplay among organelles in cardiomyocytes to adjust a proper intracellular labile Zn2+ level. In the present review, we aimed to summarize the already known Zn2+-transporters localize to mitochondria and function to stabilize not only the cellular Zn2+ level but also cellular oxidative stress status. In conclusion, one can propose that a detailed understanding of cellular Zn2+-homeostasis and Zn2+-signaling through mitochondria may emphasize the importance of new mitochondria-targeting agents for prevention and/or therapy of cardiovascular dysfunction in humans.
Collapse
|
19
|
Kable ME, Riazati N, Kirschke CP, Zhao J, Tepaamorndech S, Huang L. The Znt7-null mutation has sex dependent effects on the gut microbiota and goblet cell population in the mouse colon. PLoS One 2020; 15:e0239681. [PMID: 32991615 PMCID: PMC7523961 DOI: 10.1371/journal.pone.0239681] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 09/11/2020] [Indexed: 01/01/2023] Open
Abstract
Cellular homeostasis of zinc, an essential element for living organisms, is tightly regulated by a family of zinc transporters. The zinc transporter 7, ZnT7, is highly expressed on the membrane of the Golgi complex of intestinal epithelial cells and goblet cells. It has previously been shown that Znt7 knockout leads to zinc deficiency and decreased weight gain in C57BL/6 mice on a defined diet. However, effects within the colon are unknown. Given the expression profile of Znt7, we set out to analyze the changes in mucin density and gut microbial composition in the mouse large intestine induced by Znt7 knockout. We fed a semi-purified diet containing 30 mg Zn/kg to Znt7-/- mice with their heterozygous and wild type littermates and found a sex specific effect on colonic mucin density, goblet cell number, and microbiome composition. In male mice Znt7 knockout led to increased goblet cell number and mucin density but had little effect on gut microbiome composition. However, in female mice Znt7 knockout was associated with decreased goblet cell number and mucin density, with increased proportions of the microbial taxa, Allobaculum, relative to wild type. The gut microbial composition was correlated with mucin density in both sexes. These findings suggest that a sex-specific relationship exists between zinc homeostasis, mucin production and the microbial community composition within the colon.
Collapse
Affiliation(s)
- Mary E. Kable
- Immunity and Disease Prevention Research Unit, USDA-ARS, Western Human Nutrition Research Center, Davis, California, United States of America
- Department of Nutrition, University of California Davis, Davis, California, United States of America
- * E-mail: (MEK); (LH)
| | - Niknaz Riazati
- Department of Nutrition, University of California Davis, Davis, California, United States of America
| | - Catherine P. Kirschke
- Obesity and Metabolism Research Unit, USDA-ARS, Western Human Nutrition Research Center, Davis, California, United States of America
| | - Junli Zhao
- School of Food Science, Nanjing Xiaozhuang University, Nanjing, Jiangsu, China
| | - Surapun Tepaamorndech
- Food Biotechnology Research Unit, National Center for Genetic Engineering and Biotechnology (BIOTEC), Khlong Luang, Pathum Thani, Thailand
| | - Liping Huang
- Department of Nutrition, University of California Davis, Davis, California, United States of America
- Obesity and Metabolism Research Unit, USDA-ARS, Western Human Nutrition Research Center, Davis, California, United States of America
- * E-mail: (MEK); (LH)
| |
Collapse
|
20
|
Hernández-Camacho JD, Vicente-García C, Parsons DS, Navas-Enamorado I. Zinc at the crossroads of exercise and proteostasis. Redox Biol 2020; 35:101529. [PMID: 32273258 PMCID: PMC7284914 DOI: 10.1016/j.redox.2020.101529] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 03/25/2020] [Accepted: 03/27/2020] [Indexed: 12/11/2022] Open
Abstract
Zinc is an essential element for all forms of life, and one in every ten human proteins is a zinc protein. Zinc has catalytic, structural and signalling functions and its correct homeostasis affects many cellular processes. Zinc deficiency leads to detrimental consequences, especially in tissues with high demand such as skeletal muscle. Zinc cellular homeostasis is tightly regulated by different transport and buffer protein systems. Specifically, in skeletal muscle, zinc has been found to affect myogenesis and muscle regeneration due to its effects on muscle cell activation, proliferation and differentiation. In relation to skeletal muscle, exercise has been shown to modulate zinc serum and urinary levels and could directly affect cellular zinc transport. The oxidative stress induced by exercise may provide the basis for the mild zinc deficiency observed in athletes and could have severe consequences on health and sport performance. Proteostasis is induced during exercise and zinc plays an essential role in several of the associated pathways. Zinc deficiency could be a crucial issue in sport performance for athletes. Exercise could modulate zinc serum and cellular homeostasis. Zinc is part of proteostatic systems critical during exercise.
Collapse
Affiliation(s)
- Juan Diego Hernández-Camacho
- Centro Andaluz de Biología del Desarrollo, CSIC-UPO-JA, Universidad Pablo de Olavide, Sevilla, 41013, Spain; CIBERER, Instituto de Salud Carlos III, Madrid, 28000, Spain
| | - Cristina Vicente-García
- Centro Andaluz de Biología del Desarrollo, CSIC-UPO-JA, Universidad Pablo de Olavide, Sevilla, 41013, Spain
| | | | | |
Collapse
|
21
|
Schumann T, König J, Henke C, Willmes DM, Bornstein SR, Jordan J, Fromm MF, Birkenfeld AL. Solute Carrier Transporters as Potential Targets for the Treatment of Metabolic Disease. Pharmacol Rev 2020; 72:343-379. [PMID: 31882442 DOI: 10.1124/pr.118.015735] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The solute carrier (SLC) superfamily comprises more than 400 transport proteins mediating the influx and efflux of substances such as ions, nucleotides, and sugars across biological membranes. Over 80 SLC transporters have been linked to human diseases, including obesity and type 2 diabetes (T2D). This observation highlights the importance of SLCs for human (patho)physiology. Yet, only a small number of SLC proteins are validated drug targets. The most recent drug class approved for the treatment of T2D targets sodium-glucose cotransporter 2, product of the SLC5A2 gene. There is great interest in identifying other SLC transporters as potential targets for the treatment of metabolic diseases. Finding better treatments will prove essential in future years, given the enormous personal and socioeconomic burden posed by more than 500 million patients with T2D by 2040 worldwide. In this review, we summarize the evidence for SLC transporters as target structures in metabolic disease. To this end, we identified SLC13A5/sodium-coupled citrate transporter, and recent proof-of-concept studies confirm its therapeutic potential in T2D and nonalcoholic fatty liver disease. Further SLC transporters were linked in multiple genome-wide association studies to T2D or related metabolic disorders. In addition to presenting better-characterized potential therapeutic targets, we discuss the likely unnoticed link between other SLC transporters and metabolic disease. Recognition of their potential may promote research on these proteins for future medical management of human metabolic diseases such as obesity, fatty liver disease, and T2D. SIGNIFICANCE STATEMENT: Given the fact that the prevalence of human metabolic diseases such as obesity and type 2 diabetes has dramatically risen, pharmacological intervention will be a key future approach to managing their burden and reducing mortality. In this review, we present the evidence for solute carrier (SLC) genes associated with human metabolic diseases and discuss the potential of SLC transporters as therapeutic target structures.
Collapse
Affiliation(s)
- Tina Schumann
- Section of Metabolic and Vascular Medicine, Medical Clinic III, Dresden University School of Medicine (T.S., C.H., D.M.W., S.R.B.), and Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine (T.S., C.H., D.M.W.), Technische Universität Dresden, Dresden, Germany; Deutsches Zentrum für Diabetesforschung e.V., Neuherberg, Germany (T.S., C.H., D.M.W., A.L.B.); Clinical Pharmacology and Clinical Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany (J.K., M.F.F.); Institute for Aerospace Medicine, German Aerospace Center and Chair for Aerospace Medicine, University of Cologne, Cologne, Germany (J.J.); Diabetes and Nutritional Sciences, King's College London, London, United Kingdom (S.R.B., A.L.B.); Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, Tübingen, Germany (A.L.B.); and Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany (A.L.B.)
| | - Jörg König
- Section of Metabolic and Vascular Medicine, Medical Clinic III, Dresden University School of Medicine (T.S., C.H., D.M.W., S.R.B.), and Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine (T.S., C.H., D.M.W.), Technische Universität Dresden, Dresden, Germany; Deutsches Zentrum für Diabetesforschung e.V., Neuherberg, Germany (T.S., C.H., D.M.W., A.L.B.); Clinical Pharmacology and Clinical Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany (J.K., M.F.F.); Institute for Aerospace Medicine, German Aerospace Center and Chair for Aerospace Medicine, University of Cologne, Cologne, Germany (J.J.); Diabetes and Nutritional Sciences, King's College London, London, United Kingdom (S.R.B., A.L.B.); Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, Tübingen, Germany (A.L.B.); and Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany (A.L.B.)
| | - Christine Henke
- Section of Metabolic and Vascular Medicine, Medical Clinic III, Dresden University School of Medicine (T.S., C.H., D.M.W., S.R.B.), and Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine (T.S., C.H., D.M.W.), Technische Universität Dresden, Dresden, Germany; Deutsches Zentrum für Diabetesforschung e.V., Neuherberg, Germany (T.S., C.H., D.M.W., A.L.B.); Clinical Pharmacology and Clinical Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany (J.K., M.F.F.); Institute for Aerospace Medicine, German Aerospace Center and Chair for Aerospace Medicine, University of Cologne, Cologne, Germany (J.J.); Diabetes and Nutritional Sciences, King's College London, London, United Kingdom (S.R.B., A.L.B.); Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, Tübingen, Germany (A.L.B.); and Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany (A.L.B.)
| | - Diana M Willmes
- Section of Metabolic and Vascular Medicine, Medical Clinic III, Dresden University School of Medicine (T.S., C.H., D.M.W., S.R.B.), and Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine (T.S., C.H., D.M.W.), Technische Universität Dresden, Dresden, Germany; Deutsches Zentrum für Diabetesforschung e.V., Neuherberg, Germany (T.S., C.H., D.M.W., A.L.B.); Clinical Pharmacology and Clinical Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany (J.K., M.F.F.); Institute for Aerospace Medicine, German Aerospace Center and Chair for Aerospace Medicine, University of Cologne, Cologne, Germany (J.J.); Diabetes and Nutritional Sciences, King's College London, London, United Kingdom (S.R.B., A.L.B.); Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, Tübingen, Germany (A.L.B.); and Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany (A.L.B.)
| | - Stefan R Bornstein
- Section of Metabolic and Vascular Medicine, Medical Clinic III, Dresden University School of Medicine (T.S., C.H., D.M.W., S.R.B.), and Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine (T.S., C.H., D.M.W.), Technische Universität Dresden, Dresden, Germany; Deutsches Zentrum für Diabetesforschung e.V., Neuherberg, Germany (T.S., C.H., D.M.W., A.L.B.); Clinical Pharmacology and Clinical Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany (J.K., M.F.F.); Institute for Aerospace Medicine, German Aerospace Center and Chair for Aerospace Medicine, University of Cologne, Cologne, Germany (J.J.); Diabetes and Nutritional Sciences, King's College London, London, United Kingdom (S.R.B., A.L.B.); Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, Tübingen, Germany (A.L.B.); and Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany (A.L.B.)
| | - Jens Jordan
- Section of Metabolic and Vascular Medicine, Medical Clinic III, Dresden University School of Medicine (T.S., C.H., D.M.W., S.R.B.), and Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine (T.S., C.H., D.M.W.), Technische Universität Dresden, Dresden, Germany; Deutsches Zentrum für Diabetesforschung e.V., Neuherberg, Germany (T.S., C.H., D.M.W., A.L.B.); Clinical Pharmacology and Clinical Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany (J.K., M.F.F.); Institute for Aerospace Medicine, German Aerospace Center and Chair for Aerospace Medicine, University of Cologne, Cologne, Germany (J.J.); Diabetes and Nutritional Sciences, King's College London, London, United Kingdom (S.R.B., A.L.B.); Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, Tübingen, Germany (A.L.B.); and Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany (A.L.B.)
| | - Martin F Fromm
- Section of Metabolic and Vascular Medicine, Medical Clinic III, Dresden University School of Medicine (T.S., C.H., D.M.W., S.R.B.), and Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine (T.S., C.H., D.M.W.), Technische Universität Dresden, Dresden, Germany; Deutsches Zentrum für Diabetesforschung e.V., Neuherberg, Germany (T.S., C.H., D.M.W., A.L.B.); Clinical Pharmacology and Clinical Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany (J.K., M.F.F.); Institute for Aerospace Medicine, German Aerospace Center and Chair for Aerospace Medicine, University of Cologne, Cologne, Germany (J.J.); Diabetes and Nutritional Sciences, King's College London, London, United Kingdom (S.R.B., A.L.B.); Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, Tübingen, Germany (A.L.B.); and Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany (A.L.B.)
| | - Andreas L Birkenfeld
- Section of Metabolic and Vascular Medicine, Medical Clinic III, Dresden University School of Medicine (T.S., C.H., D.M.W., S.R.B.), and Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine (T.S., C.H., D.M.W.), Technische Universität Dresden, Dresden, Germany; Deutsches Zentrum für Diabetesforschung e.V., Neuherberg, Germany (T.S., C.H., D.M.W., A.L.B.); Clinical Pharmacology and Clinical Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany (J.K., M.F.F.); Institute for Aerospace Medicine, German Aerospace Center and Chair for Aerospace Medicine, University of Cologne, Cologne, Germany (J.J.); Diabetes and Nutritional Sciences, King's College London, London, United Kingdom (S.R.B., A.L.B.); Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, Tübingen, Germany (A.L.B.); and Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany (A.L.B.)
| |
Collapse
|
22
|
Alluri K, Nair KPM, Ghosh S. Differential expression of zinc transporters in functionally contrasting tissues involved in zinc homeostasis. NUCLEOSIDES NUCLEOTIDES & NUCLEIC ACIDS 2019; 39:615-629. [PMID: 31852371 DOI: 10.1080/15257770.2019.1670838] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Zinc homeostasis is maintained by 24 tissue-specific zinc transporters which include ZnTs (ZnT1-10), ZIPs (ZIP1-14), in addition to metallothionein (MT). Current study aimed the role of zinc transporters in maintaining the basal levels of zinc in functionally contrasting tissue specific THP-1 (monocyte), RD (muscle), and Saos-2 (bone) cells. Zinc transporters expression was assessed by qRT-PCR. The mRNA levels of ZnTs (ZnT5-7 & ZnT9), ZIPs (ZIP6-10, ZIP13-14), and MT were significantly (p < 0.05) higher in Saos-2 compared to THP-1 and RD. The present study suggests that distinct expression pattern of zinc transporters and metallothionein might be responsible for the differential zinc assimilation.
Collapse
Affiliation(s)
- Kiran Alluri
- Molecular Biology Division, ICMR-National Institute of Nutrition, Indian Council of Medical Research, Hyderabad, India
| | - Krishna Pillay Madhavan Nair
- Micronutrient Research Group, ICMR- National Institute of Nutrition, Indian Council of Medical Research, Hyderabad, India
| | - Sudip Ghosh
- Molecular Biology Division, ICMR-National Institute of Nutrition, Indian Council of Medical Research, Hyderabad, India
| |
Collapse
|
23
|
Rodríguez-Maya M, Domínguez-Vara I, Trujillo-Gutiérrez D, Morales-Almaráz E, Sánchez-Torres J, Bórquez-Gastelum J, Acosta-Dibarrat J, Grageola-Nuñez F, Rodríguez-Carpena J. Growth performance parameters, carcass traits, and meat quality of lambs supplemented with zinc methionine and (or) zinc oxide in feedlot system. CANADIAN JOURNAL OF ANIMAL SCIENCE 2019. [DOI: 10.1139/cjas-2018-0153] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- M.A. Rodríguez-Maya
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma del Estado de México, Campus Universitario “El Cerrillo”, Toluca, Estado de México CP. 50090, México
| | - I.A. Domínguez-Vara
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma del Estado de México, Campus Universitario “El Cerrillo”, Toluca, Estado de México CP. 50090, México
| | - D. Trujillo-Gutiérrez
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma del Estado de México, Campus Universitario “El Cerrillo”, Toluca, Estado de México CP. 50090, México
| | - E. Morales-Almaráz
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma del Estado de México, Campus Universitario “El Cerrillo”, Toluca, Estado de México CP. 50090, México
| | - J.E. Sánchez-Torres
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma del Estado de México, Campus Universitario “El Cerrillo”, Toluca, Estado de México CP. 50090, México
| | - J.L. Bórquez-Gastelum
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma del Estado de México, Campus Universitario “El Cerrillo”, Toluca, Estado de México CP. 50090, México
| | - J. Acosta-Dibarrat
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma del Estado de México, Campus Universitario “El Cerrillo”, Toluca, Estado de México CP. 50090, México
| | - F. Grageola-Nuñez
- Unidad Académica de Medicina Veterinaria y Zootecnia, Universidad Autónoma de Nayarit, Ciudad de la Cultura “Amado Nervo”, Tepic, Nayarit CP. 63155, México
| | - J.G. Rodríguez-Carpena
- Unidad Académica de Medicina Veterinaria y Zootecnia, Universidad Autónoma de Nayarit, Ciudad de la Cultura “Amado Nervo”, Tepic, Nayarit CP. 63155, México
| |
Collapse
|
24
|
Turan B. A Brief Overview from the Physiological and Detrimental Roles of Zinc Homeostasis via Zinc Transporters in the Heart. Biol Trace Elem Res 2019; 188:160-176. [PMID: 30091070 DOI: 10.1007/s12011-018-1464-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 07/26/2018] [Indexed: 12/15/2022]
Abstract
Zinc (mostly as free/labile Zn2+) is an essential structural constituent of many proteins, including enzymes in cellular signaling pathways via functioning as an important signaling molecule in mammalian cells. In cardiomyocytes at resting condition, intracellular labile Zn2+ concentration ([Zn2+]i) is in the nanomolar range, whereas it can increase dramatically under pathological conditions, including hyperglycemia, but the mechanisms that affect its subcellular redistribution is not clear. Therefore, overall, very little is known about the precise mechanisms controlling the intracellular distribution of labile Zn2+, particularly via Zn2+ transporters during cardiac function under both physiological and pathophysiological conditions. Literature data demonstrated that [Zn2+]i homeostasis in mammalian cells is primarily coordinated by Zn2+ transporters classified as ZnTs (SLC30A) and ZIPs (SLC39A). To identify the molecular mechanisms of diverse functions of labile Zn2+ in the heart, the recent studies focused on the discovery of subcellular localization of these Zn2+ transporters in parallel to the discovery of novel physiological functions of [Zn2+]i in cardiomyocytes. The present review summarizes the current understanding of the role of [Zn2+]i changes in cardiomyocytes under pathological conditions, and under high [Zn2+]i and how Zn2+ transporters are important for its subcellular redistribution. The emerging importance and the promise of some Zn2+ transporters for targeted cardiac therapy against pathological stimuli are also provided. Taken together, the review clearly outlines cellular control of cytosolic Zn2+ signaling by Zn2+ transporters, the role of Zn2+ transporters in heart function under hyperglycemia, the role of Zn2+ under increased oxidative stress and ER stress, and their roles in cancer are discussed.
Collapse
Affiliation(s)
- Belma Turan
- Department of Biophysics, Faculty of Medicine, Ankara University, Ankara, Turkey.
| |
Collapse
|
25
|
Huang L, Tepaamorndech S, Kirschke CP, Cai Y, Zhao J, Cao X, Rao A. Subcongenic analysis of a quantitative trait locus affecting body weight and glucose metabolism in zinc transporter 7 (znt7)-knockout mice. BMC Genet 2019; 20:19. [PMID: 30777014 PMCID: PMC6378724 DOI: 10.1186/s12863-019-0715-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 01/11/2019] [Indexed: 11/24/2022] Open
Abstract
Background A genome-wide mapping study using male F2zinc transporter 7-knockout mice (znt7-KO) and their wild type littermates in a mixed 129P1/ReJ (129P1) and C57BL/6J (B6) background identified a quantitative trait locus (QTL) on chromosome 7, which had a synergistic effect on body weight gain and fat deposit with the znt7-null mutation. Results The genetic segment for body weight on mouse chromosome 7 was investigated by newly created subcongenic znt7-KO mouse strains carrying different lengths of genomic segments of chromosome 7 from the 129P1 donor strain in the B6 background. We mapped the sub-QTL for body weight in the proximal region of the previously mapped QTL, ranging from 47.4 to 64.4 megabases (Mb) on chromosome 7. The 129P1 donor allele conferred lower body weight gain and better glucose handling during intraperitoneal glucose challenge than the B6 allele control. We identified four candidate genes, including Htatip2, E030018B13Rik, Nipa1, and Atp10a, in this sub-QTL using quantitative RT-PCR and cSNP detection (single nucleotide polymorphisms in the protein coding region). Conclusions This study dissected the genetic determinates of body weight and glucose metabolism in znt7-KO mice. The study demonstrated that a 17-Mb long 129P1 genomic region on mouse chromosome 7 conferred weight reduction and improved glucose tolerance in znt7-KO male mice. Among the four candidate genes identified, Htatip2 is the most likely candidate gene involved in the control of body weight based on its function in regulation of lipid metabolism. The candidate genes discovered in this study lay a foundation for future studies of their roles in development of metabolic diseases, such as obesity and type 2 diabetes. Electronic supplementary material The online version of this article (10.1186/s12863-019-0715-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- L Huang
- Obesity and Metabolism Research Unit, USDA/ARS/Western Human Nutrition Research Center, 430 West Health Sciences Drive, Davis, CA, 95616, USA.
| | - S Tepaamorndech
- Integrative Genetics and Genomics Graduate Group, University of California Davis, One Shields Avenue, Davis, CA, 95616, USA.,Present Address: Food Biotechnology Research Unit, National Center for Genetic Engineering and Biotechnology (BIOTEC), 113 Thailand Science Park, Phahonyothin Road, Pathum Thani, 12120, Thailand
| | - C P Kirschke
- Obesity and Metabolism Research Unit, USDA/ARS/Western Human Nutrition Research Center, 430 West Health Sciences Drive, Davis, CA, 95616, USA
| | - Y Cai
- Graduate Group of Nutritional Biology, University of California Davis, One Shields Avenue, Davis, CA, 95616, USA
| | - J Zhao
- Department of Nutrition, University of California Davis, One Shields Avenue, Davis, CA, 95616, USA.,School of Food Science, Nanjing Xiaozhuang University, Nanjing, 211171, Jiangsu, China
| | - Xiaohan Cao
- Food Science and Technology, University of California Davis, One Shields Avenue, Davis, CA, 95616, USA
| | - Andrew Rao
- Department of Nutrition, University of California Davis, One Shields Avenue, Davis, CA, 95616, USA
| |
Collapse
|
26
|
Tuncay E, Bitirim CV, Olgar Y, Durak A, Rutter GA, Turan B. Zn2+-transporters ZIP7 and ZnT7 play important role in progression of cardiac dysfunction via affecting sarco(endo)plasmic reticulum-mitochondria coupling in hyperglycemic cardiomyocytes. Mitochondrion 2019; 44:41-52. [DOI: 10.1016/j.mito.2017.12.011] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 12/13/2017] [Accepted: 12/27/2017] [Indexed: 12/20/2022]
|
27
|
Cruz KJC, de Oliveira ARS, Morais JBS, Severo JS, Mendes PMV, de Sousa Melo SR, de Sousa GS, Marreiro DDN. Zinc and Insulin Resistance: Biochemical and Molecular Aspects. Biol Trace Elem Res 2018; 186:407-412. [PMID: 29564656 DOI: 10.1007/s12011-018-1308-z] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 03/14/2018] [Indexed: 01/31/2023]
Abstract
Studies have shown the participation of minerals in mechanisms involved in the pathogenesis of insulin resistance. Zinc, in particular, seems to play an important role in the secretion and action of this hormone. Therefore, the aim of this review is to understand the role of zinc in increasing insulin sensitivity. We conducted a search of articles published in the PubMed and ScienceDirect database selected from March 2016 to February 2018, using the keywords "zinc," "insulin," "insulin resistance," "insulin sensitivity," and "supplementation." Following the eligibility criteria were selected 53 articles. The scientific evidences presented in this review show the importance of zinc and their carrier proteins in the synthesis and secretion of insulin, as well as in the signaling pathway of action of this hormone. Zinc deficiency is associated with glucose intolerance and insulin resistance; however, the effectiveness of the intervention with the zinc supplementation is still inconclusive.
Collapse
Affiliation(s)
- Kyria Jayanne Clímaco Cruz
- Department of Nutrition, Federal University of Piauí, Ministro Petrônio Portella Campus, Ininga, Teresina, Piauí, Brazil
| | - Ana Raquel Soares de Oliveira
- Department of Nutrition, Federal University of Piauí, Ministro Petrônio Portella Campus, Ininga, Teresina, Piauí, Brazil
| | - Jennifer Beatriz Silva Morais
- Department of Nutrition, Federal University of Piauí, Ministro Petrônio Portella Campus, Ininga, Teresina, Piauí, Brazil
| | - Juliana Soares Severo
- Department of Nutrition, Federal University of Piauí, Ministro Petrônio Portella Campus, Ininga, Teresina, Piauí, Brazil
| | - Priscyla Maria Vieira Mendes
- Department of Nutrition, Federal University of Piauí, Ministro Petrônio Portella Campus, Ininga, Teresina, Piauí, Brazil
| | | | | | - Dilina do Nascimento Marreiro
- Department of Nutrition, Federal University of Piauí, Ministro Petrônio Portella Campus, Ininga, Teresina, Piauí, Brazil.
| |
Collapse
|
28
|
Paskavitz AL, Quintana J, Cangussu D, Tavera-Montañez C, Xiao Y, Ortiz-Miranda S, Navea JG, Padilla-Benavides T. Differential expression of zinc transporters accompanies the differentiation of C2C12 myoblasts. J Trace Elem Med Biol 2018; 49:27-34. [PMID: 29895369 PMCID: PMC6082398 DOI: 10.1016/j.jtemb.2018.04.024] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 04/16/2018] [Accepted: 04/20/2018] [Indexed: 12/11/2022]
Abstract
Zinc transporters facilitate metal mobilization and compartmentalization, playing a key role in cellular development. Little is known about the mechanisms and pathways of Zn movement between Zn transporters and metalloproteins during myoblast differentiation. We analyzed the differential expression of ZIP and ZnT transporters during C2C12 myoblast differentiation. Zn transporters account for a transient decrease of intracellular Zn upon myogenesis induction followed by a gradual increase of Zn in myotubes. Considering the subcellular localization and function of each of the Zn transporters, our findings indicate that a fine regulation is necessary to maintain correct metal concentrations in the cytosol and subcellular compartments to avoid toxicity, maintain homeostasis, and for loading metalloproteins needed during myogenesis. This study advances our basic understanding of the complex Zn transport network during muscle differentiation.
Collapse
Affiliation(s)
- Amanda L Paskavitz
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA, 01605, USA; Department of Chemistry, Skidmore College, 815 North Broadway, Saratoga Springs, NY, 12866, USA
| | - Julia Quintana
- Department of Chemistry and Biochemistry, Worcester Polytechnic Institute, 100 Institute Road, Worcester, MA, 01609, USA
| | - Daniella Cangussu
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA, 01605, USA
| | - Cristina Tavera-Montañez
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA, 01605, USA; Department of Chemistry and Biochemistry, Worcester Polytechnic Institute, 100 Institute Road, Worcester, MA, 01609, USA
| | - Yao Xiao
- Department of Chemistry, Skidmore College, 815 North Broadway, Saratoga Springs, NY, 12866, USA
| | - Sonia Ortiz-Miranda
- Department of Neurobiology, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA, 01605, USA
| | - Juan G Navea
- Department of Chemistry, Skidmore College, 815 North Broadway, Saratoga Springs, NY, 12866, USA
| | - Teresita Padilla-Benavides
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA, 01605, USA.
| |
Collapse
|
29
|
Zhang X, Lian X, Liang D, Zhang L, Liu S, Yang L, Chi ZH, Gu H. Protective Effect of Znt7 on High Glucose-Induced Epithelial-to-Mesenchymal Transition in Renal Tubular Epithelial Cells. Kidney Blood Press Res 2018; 43:500-512. [DOI: 10.1159/000488697] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 03/23/2018] [Indexed: 11/19/2022] Open
|
30
|
Huang L, Tepaamorndech S, Kirschke CP, Newman JW, Keyes WR, Pedersen TL, Dumnil J. Aberrant fatty acid metabolism in skeletal muscle contributes to insulin resistance in zinc transporter 7 ( znt7)-knockout mice. J Biol Chem 2018; 293:7549-7563. [PMID: 29555680 DOI: 10.1074/jbc.m117.817692] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 03/12/2018] [Indexed: 12/22/2022] Open
Abstract
ZnT7 (Slc30a7) is a widely expressed zinc transporter involved in sequestration of zinc into the Golgi apparatus and vesicular compartments. znt7-knockout (KO) mice are mildly zinc-deficient and lean. Despite their lean phenotype, adult male znt7-KO mice are prone to insulin resistance. We hypothesized that fat partitioning from adipose to nonadipose tissues causes insulin resistance in znt7-KO mice. Here, we used biological and biochemical methods, including fatty acid and oxylipin profiling, EM, immunohistochemistry, quantitative RT-PCR, and Western blot analysis, to identify the underlying mechanism of insulin resistance in znt7-KO mice. We found that insulin resistance in this model was primarily associated with increased intracellular fatty acid levels in the skeletal muscle, which promoted intracellular lipid accumulation and production of bioactive lipid mediators, such as 12,13-dihydroxyoctadecanoic acid (12,13-DiHOME) and 12-hydroxyeicosatetraenoic acid (12-HETE). The expression of fatty acid-binding protein 3 (Fabp3) was dramatically up-regulated in the znt7-KO muscle cells accompanied by increased expression of Cd36, Slc27a1, and Slc27a4, the three major fatty acid transporters in the skeletal muscle. We also demonstrated that znt7-KO muscle cells had increased fatty acid oxidative capacity, indicated by enlarged mitochondria and increased mRNA or protein expression of key enzymes involved in the fatty acid mitochondrial shuttle and β-oxidation. We conclude that increased fatty acid uptake in the znt7-KO skeletal muscle is a key factor that contributes to the excessive intracellular lipid deposit and elevated production of bioactive lipid mediators. These mediators may play pivotal roles in oxidative stress and inflammation, leading to insulin resistance.
Collapse
Affiliation(s)
- Liping Huang
- From the Obesity and Metabolism Research Unit, United States Department of Agriculture/Agricultural Research Service/Western Human Nutrition Research Center, Davis, California 95616, .,Department of Nutrition and.,Integrative Genetics and Genomics Graduate Group, University of California, Davis, California 95616, and
| | - Surapun Tepaamorndech
- Integrative Genetics and Genomics Graduate Group, University of California, Davis, California 95616, and.,Food Biotechnology Research Unit, National Center for Genetic Engineering and Biotechnology (BIOTEC), 113 Thailand Science Park, Phahonyothin Road, Pathum Thani 12120, Thailand
| | - Catherine P Kirschke
- From the Obesity and Metabolism Research Unit, United States Department of Agriculture/Agricultural Research Service/Western Human Nutrition Research Center, Davis, California 95616
| | - John W Newman
- From the Obesity and Metabolism Research Unit, United States Department of Agriculture/Agricultural Research Service/Western Human Nutrition Research Center, Davis, California 95616.,Department of Nutrition and
| | - William R Keyes
- From the Obesity and Metabolism Research Unit, United States Department of Agriculture/Agricultural Research Service/Western Human Nutrition Research Center, Davis, California 95616
| | - Theresa L Pedersen
- From the Obesity and Metabolism Research Unit, United States Department of Agriculture/Agricultural Research Service/Western Human Nutrition Research Center, Davis, California 95616
| | - Jureeporn Dumnil
- Food Biotechnology Research Unit, National Center for Genetic Engineering and Biotechnology (BIOTEC), 113 Thailand Science Park, Phahonyothin Road, Pathum Thani 12120, Thailand
| |
Collapse
|
31
|
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
|
32
|
Role of Zinc Homeostasis in the Pathogenesis of Diabetes and Obesity. Int J Mol Sci 2018; 19:ijms19020476. [PMID: 29415457 PMCID: PMC5855698 DOI: 10.3390/ijms19020476] [Citation(s) in RCA: 159] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 01/30/2018] [Accepted: 02/02/2018] [Indexed: 12/11/2022] Open
Abstract
Zinc deficiency is a risk factor for obesity and diabetes. However, until recently, the underlying molecular mechanisms remained unclear. The breakthrough discovery that the common polymorphism in zinc transporter SLC30A8/ZnT8 may increase susceptibility to type 2 diabetes provided novel insights into the role of zinc in diabetes. Our group and others showed that altered ZnT8 function may be involved in the pathogenesis of type 2 diabetes, indicating that the precise control of zinc homeostasis is crucial for maintaining health and preventing various diseases, including lifestyle-associated diseases. Recently, the role of the zinc transporter ZIP13 in the regulation of beige adipocyte biogenesis was clarified, which indicated zinc homeostasis regulation as a possible therapeutic target for obesity and metabolic syndrome. Here we review advances in the role of zinc homeostasis in the pathophysiology of diabetes, and propose that inadequate zinc distribution may affect the onset of diabetes and metabolic diseases by regulating various critical biological events.
Collapse
|
33
|
Cai Y, Kirschke CP, Huang L. SLC30A family expression in the pancreatic islets of humans and mice: cellular localization in the β-cells. J Mol Histol 2018; 49:133-145. [DOI: 10.1007/s10735-017-9753-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 12/30/2017] [Indexed: 10/18/2022]
|
34
|
Olechnowicz J, Tinkov A, Skalny A, Suliburska J. Zinc status is associated with inflammation, oxidative stress, lipid, and glucose metabolism. J Physiol Sci 2018; 68:19-31. [PMID: 28965330 PMCID: PMC5754376 DOI: 10.1007/s12576-017-0571-7] [Citation(s) in RCA: 340] [Impact Index Per Article: 48.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 09/18/2017] [Indexed: 12/14/2022]
Abstract
A number of studies have reported that zinc plays a substantial role in the development of metabolic syndrome, taking part in the regulation of cytokine expression, suppressing inflammation, and is also required to activate antioxidant enzymes that scavenge reactive oxygen species, reducing oxidative stress. Zinc also plays a role in the correct functioning of lipid and glucose metabolism, regulating and forming the expression of insulin. In numerous studies, zinc supplementation has been found to improve blood pressure, glucose, and LDL cholesterol serum level. Deeper knowledge of zinc's properties may help in treating metabolic syndrome, thus protecting against stroke and angina pectoris, and ultimately against death.
Collapse
Affiliation(s)
- J Olechnowicz
- Poznan University of Life Sciences, ul. Wojska Polskiego 31, 62-624, Poznan , Poland
| | - A Tinkov
- Orenburg State Medical University, Sovetskaya St., 6, Orenburg, 460000, Russia
- Orenburg State University, Pobedy Avenue, 13, Orenburg, 460018, Russia
- RUDN University, Miklukho-Maklay St., 10/2, Moscow, 117198, Russia
- Yaroslavl State University, Sovetskaya St., 14, Yaroslavl, 150000, Russia
| | - A Skalny
- Orenburg State University, Pobedy Avenue, 13, Orenburg, 460018, Russia
- RUDN University, Miklukho-Maklay St., 10/2, Moscow, 117198, Russia
- Yaroslavl State University, Sovetskaya St., 14, Yaroslavl, 150000, Russia
- All-Russian Research Institute of Medicinal and Aromatic Plants (VILAR), Grina St., 7, Moscow, 117216, Russia
| | - Joanna Suliburska
- Poznan University of Life Sciences, ul. Wojska Polskiego 31, 62-624, Poznan , Poland.
| |
Collapse
|
35
|
Giacconi R, Cai L, Costarelli L, Cardelli M, Malavolta M, Piacenza F, Provinciali M. Implications of impaired zinc homeostasis in diabetic cardiomyopathy and nephropathy. Biofactors 2017; 43:770-784. [PMID: 28845600 DOI: 10.1002/biof.1386] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 07/12/2017] [Accepted: 07/27/2017] [Indexed: 12/18/2022]
Abstract
Impaired zinc homeostasis is observed in diabetes mellitus (DM2) and its complications. Zinc has a specific role in pancreatic β-cells via insulin synthesis, storage, and secretion. Intracellular zinc homeostasis is tightly controlled by zinc transporters (ZnT and Zip families) and metallothioneins (MT) which modulate the uptake, storage, and distribution of zinc. Several investigations in animal models demonstrate the protective role of MT in DM2 and its cardiovascular or renal complications, while a copious literature shows that a common polymorphism (R325W) in ZnT8, which affects the protein's zinc transport activity, is associated with increased DM2 risk. Emerging studies highlight a role of other zinc transporters in β-cell function, suggesting that targeting them could make a possible contribution in managing the hyperglycemia in diabetic patients. This article summarizes the current findings concerning the role of zinc homeostasis in DM2 pathogenesis and development of diabetic cardiomyopathy and nephropathy and suggests novel therapeutic targets. © 2017 BioFactors, 43(6):770-784, 2017.
Collapse
Affiliation(s)
- Robertina Giacconi
- Translational Research Center of Nutrition and Ageing, Scientific and Technological Pole, Italian National Institute of Health and Science on Aging (INRCA), Ancona, Italy
| | - Lu Cai
- Pediatric Research Institute at the Department of Pediatrics, Wendy L. Novak Diabetes Care Center, University of Louisville, Louisville, KY, USA
| | - Laura Costarelli
- Translational Research Center of Nutrition and Ageing, Scientific and Technological Pole, Italian National Institute of Health and Science on Aging (INRCA), Ancona, Italy
| | - Maurizio Cardelli
- Advanced Technology Center for Aging Research, Scientific and Technological Pole, Italian National Institute of Health and Science on Aging (INRCA), Ancona, Italy
| | - Marco Malavolta
- Translational Research Center of Nutrition and Ageing, Scientific and Technological Pole, Italian National Institute of Health and Science on Aging (INRCA), Ancona, Italy
| | - Francesco Piacenza
- Translational Research Center of Nutrition and Ageing, Scientific and Technological Pole, Italian National Institute of Health and Science on Aging (INRCA), Ancona, Italy
| | - Mauro Provinciali
- Advanced Technology Center for Aging Research, Scientific and Technological Pole, Italian National Institute of Health and Science on Aging (INRCA), Ancona, Italy
| |
Collapse
|
36
|
Kambe T, Matsunaga M, Takeda TA. Understanding the Contribution of Zinc Transporters in the Function of the Early Secretory Pathway. Int J Mol Sci 2017; 18:ijms18102179. [PMID: 29048339 PMCID: PMC5666860 DOI: 10.3390/ijms18102179] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 10/12/2017] [Accepted: 10/15/2017] [Indexed: 01/07/2023] Open
Abstract
More than one-third of newly synthesized proteins are targeted to the early secretory pathway, which is comprised of the endoplasmic reticulum (ER), Golgi apparatus, and other intermediate compartments. The early secretory pathway plays a key role in controlling the folding, assembly, maturation, modification, trafficking, and degradation of such proteins. A considerable proportion of the secretome requires zinc as an essential factor for its structural and catalytic functions, and recent findings reveal that zinc plays a pivotal role in the function of the early secretory pathway. Hence, a disruption of zinc homeostasis and metabolism involving the early secretory pathway will lead to pathway dysregulation, resulting in various defects, including an exacerbation of homeostatic ER stress. The accumulated evidence indicates that specific members of the family of Zn transporters (ZNTs) and Zrt- and Irt-like proteins (ZIPs), which operate in the early secretory pathway, play indispensable roles in maintaining zinc homeostasis by regulating the influx and efflux of zinc. In this review, the biological functions of these transporters are discussed, focusing on recent aspects of their roles. In particular, we discuss in depth how specific ZNT transporters are employed in the activation of zinc-requiring ectoenzymes. The means by which early secretory pathway functions are controlled by zinc, mediated by specific ZNT and ZIP transporters, are also subjects of this review.
Collapse
Affiliation(s)
- Taiho Kambe
- Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kyoto 606-8502, Japan.
| | - Mayu Matsunaga
- Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kyoto 606-8502, Japan.
| | - Taka-Aki Takeda
- Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kyoto 606-8502, Japan.
| |
Collapse
|
37
|
Tuncay E, Bitirim VC, Durak A, Carrat GRJ, Taylor KM, Rutter GA, Turan B. Hyperglycemia-Induced Changes in ZIP7 and ZnT7 Expression Cause Zn 2+ Release From the Sarco(endo)plasmic Reticulum and Mediate ER Stress in the Heart. Diabetes 2017; 66:1346-1358. [PMID: 28232492 DOI: 10.2337/db16-1099] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Accepted: 02/10/2017] [Indexed: 11/13/2022]
Abstract
Changes in cellular free Zn2+ concentration, including those in the sarco(endo)plasmic reticulum [S(E)R], are primarily coordinated by Zn2+ transporters (ZnTs) whose identity and role in the heart are not well established. We hypothesized that ZIP7 and ZnT7 transport Zn2+ in opposing directions across the S(E)R membrane in cardiomyocytes and that changes in their activity play an important role in the development of ER stress during hyperglycemia. The subcellular S(E)R localization of ZIP7 and ZnT7 was determined in cardiomyocytes and in isolated S(E)R preparations. Markedly increased mRNA and protein levels of ZIP7 were observed in ventricular cardiomyocytes from diabetic rats or high-glucose-treated H9c2 cells while ZnT7 expression was low. In addition, we observed increased ZIP7 phosphorylation in response to high glucose in vivo and in vitro. By using recombinant-targeted Förster resonance energy transfer sensors, we show that hyperglycemia induces a marked redistribution of cellular free Zn2+, increasing cytosolic free Zn2+ and lowering free Zn2+ in the S(E)R. These changes involve alterations in ZIP7 phosphorylation and were suppressed by small interfering RNA-mediated silencing of CK2α. Opposing changes in the expression of ZIP7 and ZnT7 were also observed in hyperglycemia. We conclude that subcellular free Zn2+ redistribution in the hyperglycemic heart, resulting from altered ZIP7 and ZnT7 activity, contributes to cardiac dysfunction in diabetes.
Collapse
Affiliation(s)
- Erkan Tuncay
- Department of Biophysics, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Verda C Bitirim
- Department of Biophysics, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Aysegul Durak
- Department of Biophysics, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Gaelle R J Carrat
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College London, London, U.K
| | - Kathryn M Taylor
- School of Pharmacy and Pharmaceutical Sciences, College of Biomedical and Life Sciences, Cardiff University, Cardiff, U.K
| | - Guy A Rutter
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College London, London, U.K
| | - Belma Turan
- Department of Biophysics, Faculty of Medicine, Ankara University, Ankara, Turkey
| |
Collapse
|
38
|
Tepaamorndech S, Oort P, Kirschke CP, Cai Y, Huang L. ZNT7 binds to CD40 and influences CD154-triggered p38 MAPK activity in B lymphocytes-a possible regulatory mechanism for zinc in immune function. FEBS Open Bio 2017; 7:675-690. [PMID: 28469980 PMCID: PMC5407898 DOI: 10.1002/2211-5463.12211] [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] [Received: 11/09/2016] [Revised: 01/09/2017] [Accepted: 02/14/2017] [Indexed: 01/04/2023] Open
Abstract
Zinc deficiency impairs the immune system leading to frequent infections. Although zinc is known to play critical roles in maintaining healthy immune function, the underlying molecular targets are largely unknown. In this study, we demonstrate that zinc is important for the CD154–CD40‐mediated activation of downstream signaling pathways in human B lymphocytes. CD40 is a receptor localized on the cell surface of many immune cells, including B lymphocytes. It binds to CD154, a membrane protein expressed on antigen‐activated T helper (Th) lymphocytes. This CD154‐CD40 interaction leads to B‐cell activation. We showed that cellular zinc deficiency impaired the CD154‐CD40‐mediated p38 mitogen‐activated protein kinase (p38 MAPK) phosphorylation. We also showed that zinc supplemental treatment of B lymphocytes had limited effect on this CD40‐mediated p38 MAPK signaling. Most importantly, we demonstrated that the zinc transporter protein zinc transporter 7 (ZNT7) interacted with CD40 using immunoprecipitation analyses. ZNT7 knockdown in B lymphocytes had a negative effect on the cell surface expression of CD40. Consequently, the CD40‐mediated p38 MAPK signaling transduction was down‐regulated in ZNT7KD B lymphocytes. Conversely, this p38 MAPK signaling activity was up‐regulated by overexpression (OE) of ZNT7 in B lymphocytes. Moreover, we found that ZNT7 knockdown in B lymphocytes constitutively up‐ and down‐regulated the inhibitor of i kappa B kinase and AKT serine/threonine kinase phosphorylation, respectively, which implies the activation of survival signaling in ZNT7KD B cells. We conclude that CD40 is the target molecule for ZNT7 in regulation of immune function of B lymphocytes.
Collapse
Affiliation(s)
- Surapun Tepaamorndech
- Integrative Genetics and Genomics Graduate Group University of California Davis CA USA.,Food Biotechnology Research Unit National Center for Genetic Engineering and Biotechnology Pathum Thani Thailand
| | - Pieter Oort
- Obesity and Metabolism Research Unit USDA/ARS/Western Human Nutrition Research Center Davis CA USA.,Present address: Astrona Biotechnologies HM Clause Innovation Center 28605 County Road 104 Davis CA 95618 USA
| | - Catherine P Kirschke
- Obesity and Metabolism Research Unit USDA/ARS/Western Human Nutrition Research Center Davis CA USA
| | - Yimeng Cai
- Graduate Group of Nutritional Biology University of California Davis CA USA
| | - Liping Huang
- Integrative Genetics and Genomics Graduate Group University of California Davis CA USA.,Obesity and Metabolism Research Unit USDA/ARS/Western Human Nutrition Research Center Davis CA USA.,Graduate Group of Nutritional Biology University of California Davis CA USA
| |
Collapse
|
39
|
Hara T, Takeda TA, Takagishi T, Fukue K, Kambe T, Fukada T. Physiological roles of zinc transporters: molecular and genetic importance in zinc homeostasis. J Physiol Sci 2017; 67:283-301. [PMID: 28130681 PMCID: PMC10717645 DOI: 10.1007/s12576-017-0521-4] [Citation(s) in RCA: 287] [Impact Index Per Article: 35.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 01/04/2017] [Indexed: 02/07/2023]
Abstract
Zinc (Zn) is an essential trace mineral that regulates the expression and activation of biological molecules such as transcription factors, enzymes, adapters, channels, and growth factors, along with their receptors. Zn deficiency or excessive Zn absorption disrupts Zn homeostasis and affects growth, morphogenesis, and immune response, as well as neurosensory and endocrine functions. Zn levels must be adjusted properly to maintain the cellular processes and biological responses necessary for life. Zn transporters regulate Zn levels by controlling Zn influx and efflux between extracellular and intracellular compartments, thus, modulating the Zn concentration and distribution. Although the physiological functions of the Zn transporters remain to be clarified, there is growing evidence that Zn transporters are related to human diseases, and that Zn transporter-mediated Zn ion acts as a signaling factor, called "Zinc signal". Here we describe critical roles of Zn transporters in the body and their contribution at the molecular, biochemical, and genetic levels, and review recently reported disease-related mutations in the Zn transporter genes.
Collapse
Affiliation(s)
- Takafumi Hara
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima, Japan
| | - Taka-Aki Takeda
- Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Teruhisa Takagishi
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima, Japan
| | - Kazuhisa Fukue
- Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Taiho Kambe
- Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kyoto, Japan.
| | - Toshiyuki Fukada
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima, Japan.
- Division of Pathology, Department of Oral Diagnostic Sciences, School of Dentistry, Showa University, Tokyo, Japan.
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan.
| |
Collapse
|
40
|
Hojyo S, Fukada T. Zinc transporters and signaling in physiology and pathogenesis. Arch Biochem Biophys 2016; 611:43-50. [DOI: 10.1016/j.abb.2016.06.020] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 06/23/2016] [Accepted: 06/28/2016] [Indexed: 12/11/2022]
|
41
|
Syring KE, Boortz KA, Oeser JK, Ustione A, Platt KA, Shadoan MK, McGuinness OP, Piston DW, Powell DR, O'Brien RM. Combined Deletion of Slc30a7 and Slc30a8 Unmasks a Critical Role for ZnT8 in Glucose-Stimulated Insulin Secretion. Endocrinology 2016; 157:4534-4541. [PMID: 27754787 PMCID: PMC5133349 DOI: 10.1210/en.2016-1573] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Polymorphisms in the SLC30A8 gene, which encodes the ZnT8 zinc transporter, are associated with altered susceptibility to type 2 diabetes (T2D), and SLC30A8 haploinsufficiency is protective against the development of T2D in obese humans. SLC30A8 is predominantly expressed in pancreatic islet β-cells, but surprisingly, multiple knockout mouse studies have shown little effect of Slc30a8 deletion on glucose tolerance or glucose-stimulated insulin secretion (GSIS). Multiple other Slc30a isoforms are expressed at low levels in pancreatic islets. We hypothesized that functional compensation by the Slc30a7 isoform, which encodes ZnT7, limits the impact of Slc30a8 deletion on islet function. We therefore analyzed the effect of Slc30a7 deletion alone or in combination with Slc30a8 on in vivo glucose metabolism and GSIS in isolated islets. Deletion of Slc30a7 alone had complex effects in vivo, impairing glucose tolerance and reducing the glucose-stimulated increase in plasma insulin levels, hepatic glycogen levels, and pancreatic insulin content. Slc30a7 deletion also affected islet morphology and increased the ratio of islet α- to β-cells. However, deletion of Slc30a7 alone had no effect on GSIS in isolated islets, whereas combined deletion of Slc30a7 and Slc30a8 abolished GSIS. These data demonstrate that the function of ZnT8 in islets can be unmasked by removal of ZnT7 and imply that ZnT8 may affect T2D susceptibility through actions in other tissues where it is expressed at low levels rather than through effects on pancreatic islet function.
Collapse
Affiliation(s)
- Kristen E Syring
- Department of Molecular Physiology and Biophysics (K.E.S., K.A.B., J.K.O., O.P.M., R.M.O.), Vanderbilt University Medical School, Nashville, Tennessee 37232; Department of Cell Biology and Physiology (A.U., D.W.P.), Washington University School of Medicine, St. Louis, Missouri 63110; and Lexicon Pharmaceuticals Incorporated (K.A.P., M.K.S., D.R.P.), The Woodlands, Texas 77381
| | - Kayla A Boortz
- Department of Molecular Physiology and Biophysics (K.E.S., K.A.B., J.K.O., O.P.M., R.M.O.), Vanderbilt University Medical School, Nashville, Tennessee 37232; Department of Cell Biology and Physiology (A.U., D.W.P.), Washington University School of Medicine, St. Louis, Missouri 63110; and Lexicon Pharmaceuticals Incorporated (K.A.P., M.K.S., D.R.P.), The Woodlands, Texas 77381
| | - James K Oeser
- Department of Molecular Physiology and Biophysics (K.E.S., K.A.B., J.K.O., O.P.M., R.M.O.), Vanderbilt University Medical School, Nashville, Tennessee 37232; Department of Cell Biology and Physiology (A.U., D.W.P.), Washington University School of Medicine, St. Louis, Missouri 63110; and Lexicon Pharmaceuticals Incorporated (K.A.P., M.K.S., D.R.P.), The Woodlands, Texas 77381
| | - Alessandro Ustione
- Department of Molecular Physiology and Biophysics (K.E.S., K.A.B., J.K.O., O.P.M., R.M.O.), Vanderbilt University Medical School, Nashville, Tennessee 37232; Department of Cell Biology and Physiology (A.U., D.W.P.), Washington University School of Medicine, St. Louis, Missouri 63110; and Lexicon Pharmaceuticals Incorporated (K.A.P., M.K.S., D.R.P.), The Woodlands, Texas 77381
| | - Kenneth A Platt
- Department of Molecular Physiology and Biophysics (K.E.S., K.A.B., J.K.O., O.P.M., R.M.O.), Vanderbilt University Medical School, Nashville, Tennessee 37232; Department of Cell Biology and Physiology (A.U., D.W.P.), Washington University School of Medicine, St. Louis, Missouri 63110; and Lexicon Pharmaceuticals Incorporated (K.A.P., M.K.S., D.R.P.), The Woodlands, Texas 77381
| | - Melanie K Shadoan
- Department of Molecular Physiology and Biophysics (K.E.S., K.A.B., J.K.O., O.P.M., R.M.O.), Vanderbilt University Medical School, Nashville, Tennessee 37232; Department of Cell Biology and Physiology (A.U., D.W.P.), Washington University School of Medicine, St. Louis, Missouri 63110; and Lexicon Pharmaceuticals Incorporated (K.A.P., M.K.S., D.R.P.), The Woodlands, Texas 77381
| | - Owen P McGuinness
- Department of Molecular Physiology and Biophysics (K.E.S., K.A.B., J.K.O., O.P.M., R.M.O.), Vanderbilt University Medical School, Nashville, Tennessee 37232; Department of Cell Biology and Physiology (A.U., D.W.P.), Washington University School of Medicine, St. Louis, Missouri 63110; and Lexicon Pharmaceuticals Incorporated (K.A.P., M.K.S., D.R.P.), The Woodlands, Texas 77381
| | - David W Piston
- Department of Molecular Physiology and Biophysics (K.E.S., K.A.B., J.K.O., O.P.M., R.M.O.), Vanderbilt University Medical School, Nashville, Tennessee 37232; Department of Cell Biology and Physiology (A.U., D.W.P.), Washington University School of Medicine, St. Louis, Missouri 63110; and Lexicon Pharmaceuticals Incorporated (K.A.P., M.K.S., D.R.P.), The Woodlands, Texas 77381
| | - David R Powell
- Department of Molecular Physiology and Biophysics (K.E.S., K.A.B., J.K.O., O.P.M., R.M.O.), Vanderbilt University Medical School, Nashville, Tennessee 37232; Department of Cell Biology and Physiology (A.U., D.W.P.), Washington University School of Medicine, St. Louis, Missouri 63110; and Lexicon Pharmaceuticals Incorporated (K.A.P., M.K.S., D.R.P.), The Woodlands, Texas 77381
| | - Richard M O'Brien
- Department of Molecular Physiology and Biophysics (K.E.S., K.A.B., J.K.O., O.P.M., R.M.O.), Vanderbilt University Medical School, Nashville, Tennessee 37232; Department of Cell Biology and Physiology (A.U., D.W.P.), Washington University School of Medicine, St. Louis, Missouri 63110; and Lexicon Pharmaceuticals Incorporated (K.A.P., M.K.S., D.R.P.), The Woodlands, Texas 77381
| |
Collapse
|
42
|
Analysis of Zinc-Exporters Expression in Prostate Cancer. Sci Rep 2016; 6:36772. [PMID: 27833104 PMCID: PMC5105060 DOI: 10.1038/srep36772] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 10/20/2016] [Indexed: 01/15/2023] Open
Abstract
Maintaining optimal intracellular zinc (Zn) concentration is crucial for critical cellular functions. Depleted Zn has been associated with prostate cancer (PCa) progression. Solute carrier family 30 (SLC30A) proteins maintain cytoplasmic Zn balance by exporting Zn out to the extracellular space or by sequestering cytoplasmic Zn into intracellular compartments. In this study, we determined the involvement of Zn-exporters, SLC30A 1-10 in PCa, in the context of racial health disparity in human PCa samples obtained from European-American (EA) and African-American (AA) populations. We also analyzed the levels of Zn-exporters in a panel of PCa cells derived from EA and AA populations. We further explored the expression profile of Zn-exporters in PCa using Oncomine database. Zn-exporters were found to be differentially expressed at the mRNA level, with a significant upregulation of SLC30A1, SLC30A9 and SLC30A10, and downregulation of SLC30A5 and SLC30A6 in PCa, compared to benign prostate. Moreover, Ingenuity Pathway analysis revealed several interactions of Zn-exporters with certain tumor suppressor and promoter proteins known to be modulated in PCa. Our study provides an insight regarding Zn-exporters in PCa, which may open new avenues for future studies aimed at enhancing the levels of Zn by modulating Zn-transporters via pharmacological means.
Collapse
|
43
|
Roles of Zinc Signaling in the Immune System. J Immunol Res 2016; 2016:6762343. [PMID: 27872866 PMCID: PMC5107842 DOI: 10.1155/2016/6762343] [Citation(s) in RCA: 163] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 08/11/2016] [Indexed: 02/07/2023] Open
Abstract
Zinc (Zn) is an essential micronutrient for basic cell activities such as cell growth, differentiation, and survival. Zn deficiency depresses both innate and adaptive immune responses. However, the precise physiological mechanisms of the Zn-mediated regulation of the immune system have been largely unclear. Zn homeostasis is tightly controlled by the coordinated activity of Zn transporters and metallothioneins, which regulate the transport, distribution, and storage of Zn. There is growing evidence that Zn behaves like a signaling molecule, facilitating the transduction of a variety of signaling cascades in response to extracellular stimuli. In this review, we highlight the emerging functional roles of Zn and Zn transporters in immunity, focusing on how crosstalk between Zn and immune-related signaling guides the normal development and function of immune cells.
Collapse
|
44
|
Aydemir TB, Troche C, Kim MH, Cousins RJ. Hepatic ZIP14-mediated Zinc Transport Contributes to Endosomal Insulin Receptor Trafficking and Glucose Metabolism. J Biol Chem 2016; 291:23939-23951. [PMID: 27703010 DOI: 10.1074/jbc.m116.748632] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 09/12/2016] [Indexed: 12/12/2022] Open
Abstract
Zinc influences signaling pathways through controlled targeted zinc transport. Zinc transporter Zip14 KO mice display a phenotype that includes impaired intestinal barrier function with low grade chronic inflammation, hyperinsulinemia, and increased body fat, which are signatures of diet-induced diabetes (type 2 diabetes) and obesity in humans. Hyperglycemia in type 2 diabetes and obesity is caused by insulin resistance. Insulin resistance results in inhibition of glucose uptake by liver and other peripheral tissues, principally adipose and muscle and with concurrently higher hepatic glucose production. Therefore, modulation of hepatic glucose metabolism is an important target for antidiabetic treatment approaches. We demonstrate that during glucose uptake, cell surface abundance of zinc transporter ZIP14 and mediated zinc transport increases. Zinc is distributed to multiple sites in hepatocytes through sequential translocation of ZIP14 from plasma membrane to early and late endosomes. Endosomes from Zip14 KO mice were zinc-deficient because activities of the zinc-dependent insulin-degrading proteases insulin-degrading enzyme and cathepsin D were impaired; hence insulin receptor activity increased. Transient increases in cytosolic zinc levels are concurrent with glucose uptake and suppression of glycogen synthesis. In contrast, Zip14 KO mice exhibited greater hepatic glycogen synthesis and impaired gluconeogenesis and glycolysis related to low cytosolic zinc levels. We can conclude that ZIP14-mediated zinc transport contributes to regulation of endosomal insulin receptor activity and glucose homeostasis in hepatocytes. Therefore, modulation of ZIP14 transport activity presents a new target for management of diabetes and other glucose-related disorders.
Collapse
Affiliation(s)
- Tolunay Beker Aydemir
- From the Food Science and Human Nutrition Department and Center for Nutritional Sciences College of Agricultural and Life Sciences and
| | - Catalina Troche
- From the Food Science and Human Nutrition Department and Center for Nutritional Sciences College of Agricultural and Life Sciences and
| | - Min-Hyun Kim
- From the Food Science and Human Nutrition Department and Center for Nutritional Sciences College of Agricultural and Life Sciences and
| | - Robert J Cousins
- From the Food Science and Human Nutrition Department and Center for Nutritional Sciences College of Agricultural and Life Sciences and .,the Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, Florida 32611
| |
Collapse
|
45
|
The zinc transporter ZNT3 co-localizes with insulin in INS-1E pancreatic beta cells and influences cell survival, insulin secretion capacity, and ZNT8 expression. Biometals 2016; 29:287-98. [DOI: 10.1007/s10534-016-9915-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 02/05/2016] [Indexed: 01/21/2023]
|
46
|
Tepaamorndech S, Kirschke CP, Pedersen TL, Keyes WR, Newman JW, Huang L. Zinc transporter 7 deficiency affects lipid synthesis in adipocytes by inhibiting insulin-dependent Akt activation and glucose uptake. FEBS J 2015; 283:378-94. [PMID: 26524605 DOI: 10.1111/febs.13582] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 10/21/2015] [Accepted: 10/28/2015] [Indexed: 01/19/2023]
Abstract
Mice deficient for zinc transporter 7 protein (ZnT7) are mildly zinc deficient with low body weight gain and body fat accumulation. To investigate the underlying mechanism of ZnT7 deficiency in body adiposity, we examined fatty acid composition and insulin sensitivity in visceral (epididymal) and subcutaneous fat pads from Znt7 knockout and control mice. We showed that ZnT7 deficiency had adverse effects on fatty acid metabolism and insulin action in subcutaneous fat but not in epididymal fat in mice, consistent with the ZnT7 protein expression pattern in adipose tissues. Importantly, we found that the expression of ZnT7 protein was induced by lipogenic differentiation and reached a peak when the adipocyte was fully differentiated in mouse 3T3-L1 adipocytes. We demonstrated, using Znt7 knockdown (Znt7KD) 3T3-L1 adipocytes, that reduction in Znt7 expression blunted activations of the signal transduction pathways that regulated both basal and insulin-stimulated glucose uptake in adipocytes, resulting in low glucose uptake and lipid accumulation. The expression of the signaling mediators critical for the initiation of pre-adipocyte differentiation, including Pparγ and C/Ebpα, appeared not to be affected by Znt7KD in 3T3-L1 adipocytes. These findings strongly suggest a role for ZnT7 in adipocyte lipogenesis.
Collapse
Affiliation(s)
- Surapun Tepaamorndech
- Integrative Genetics and Genomics Graduate Group, University of California Davis, CA, USA
| | - Catherine P Kirschke
- Obesity and Metabolism Research Unit, USDA/ARS/Western Human Nutrition Research Center, Davis, CA, USA
| | - Theresa L Pedersen
- Obesity and Metabolism Research Unit, USDA/ARS/Western Human Nutrition Research Center, Davis, CA, USA
| | - William R Keyes
- Obesity and Metabolism Research Unit, USDA/ARS/Western Human Nutrition Research Center, Davis, CA, USA
| | - John W Newman
- Obesity and Metabolism Research Unit, USDA/ARS/Western Human Nutrition Research Center, Davis, CA, USA.,Department of Nutrition, University of California Davis, CA, USA
| | - Liping Huang
- Integrative Genetics and Genomics Graduate Group, University of California Davis, CA, USA.,Obesity and Metabolism Research Unit, USDA/ARS/Western Human Nutrition Research Center, Davis, CA, USA.,Department of Nutrition, University of California Davis, CA, USA
| |
Collapse
|
47
|
Wang X, Wang M, Li H, Lan X, Liu L, Li J, Li Y, Li J, Yi J, Du X, Yan J, Han Y, Zhang F, Liu M, Lu S, Li D. Upregulation of miR-497 induces hepatic insulin resistance in E3 rats with HFD-MetS by targeting insulin receptor. Mol Cell Endocrinol 2015; 416:57-69. [PMID: 26300412 DOI: 10.1016/j.mce.2015.08.021] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Revised: 07/31/2015] [Accepted: 08/20/2015] [Indexed: 12/20/2022]
Abstract
OBJECTIVE The study aims to find regulatory microRNA(s) responsible for down-regulated insulin receptor (InsR) in the liver of HFD-MetS E3 rats with insulin resistance. METHODS Firstly, hepatic insulin resistance in HFD-MetS E3 rats was evaluated by RT-qPCR, western blotting, immunohistochemistry and PAS staining. Secondly, the candidate miRNAs targeting rat InsR were predicted through online softwares and detected in the liver of HFD-MetS E3 rats with insulin resistance. Then, the expression of InsR, phosphorylated IRS-1 (pIRS-1) at Tyr632, phosphorylated AKTs (pAKTs) at Ser473 and Thr308, phosphorylated GSK-3β (p GSK-3β) at Ser9, phosphorylated GS (pGS) at Ser641 and the glycogen content were detected in CBRH-7919 cells treated with 100 nM insulin for different time periods by western blotting or PAS staining respectively, after transient transfection with miR-497 mimics or inhibitors for 24 h. Lastly, the relation between miR-497 and InsR was further determined using dual luciferase reporter assay. RESULTS Elevated miR-497 was negatively related with down-regulated InsR in the liver of HFD-MetS E3 rats with insulin resistance. Comparing with the mNC group, glycogen content and the expression of InsR, pIRS-1 (Tyr632), pAKTs (Ser473 and Thr308) and pGSK-3β (Ser9) decreased significantly in CBRH-7919 cells, while pGS (Ser641) increased significantly, after transient transfection with miR-497 mimics for 24 h and treatment with 100 nM insulin for corresponding time periods, counter to those results in CBRH-7919 cells after similar procedures with miR-497 inhibitors and insulin. In addition, dual luciferase reporter assay further confirmed that miR-497 can bind to the 3'UTR of rat InsR. CONCLUSION Insulin receptor is the target gene of miR-497, and elevated miR-497 might induce hepatic insulin resistance in HFD-MetS E3 Rats through inhibiting the expression of insulin receptor and confining the activation of IRS-1/PI3K/Akt/GSK-3β/GS pathway to insulin.
Collapse
Affiliation(s)
- Xuan Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an, Shaanxi 710061, PR China; Department of Endocrinology, Jinshan Hospital of Fudan University, Shanghai 201508, PR China
| | - Meichen Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an, Shaanxi 710061, PR China
| | - Hongmin Li
- School of Life Sciences, Northwest University, Xi'an, Shaanxi 710061, PR China
| | - Xi Lan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an, Shaanxi 710061, PR China
| | - Li Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an, Shaanxi 710061, PR China
| | - Jiaxi Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an, Shaanxi 710061, PR China
| | - Yue Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an, Shaanxi 710061, PR China
| | - Jing Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an, Shaanxi 710061, PR China
| | - Jing Yi
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an, Shaanxi 710061, PR China
| | - Xiaojuan Du
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an, Shaanxi 710061, PR China
| | - Jidong Yan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an, Shaanxi 710061, PR China
| | - Yan Han
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an, Shaanxi 710061, PR China
| | - Fujun Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an, Shaanxi 710061, PR China
| | - Min Liu
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Institute, University of Cincinnati, Cincinnati, OH 45237, USA
| | - Shemin Lu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an, Shaanxi 710061, PR China
| | - Dongmin Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an, Shaanxi 710061, PR China.
| |
Collapse
|
48
|
Kambe T, Tsuji T, Hashimoto A, Itsumura N. The Physiological, Biochemical, and Molecular Roles of Zinc Transporters in Zinc Homeostasis and Metabolism. Physiol Rev 2015; 95:749-84. [DOI: 10.1152/physrev.00035.2014] [Citation(s) in RCA: 556] [Impact Index Per Article: 55.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Zinc is involved in a variety of biological processes, as a structural, catalytic, and intracellular and intercellular signaling component. Thus zinc homeostasis is tightly controlled at the whole body, tissue, cellular, and subcellular levels by a number of proteins, with zinc transporters being particularly important. In metazoan, two zinc transporter families, Zn transporters (ZnT) and Zrt-, Irt-related proteins (ZIP) function in zinc mobilization of influx, efflux, and compartmentalization/sequestration across biological membranes. During the last two decades, significant progress has been made in understanding the molecular properties, expression, regulation, and cellular and physiological roles of ZnT and ZIP transporters, which underpin the multifarious functions of zinc. Moreover, growing evidence indicates that malfunctioning zinc homeostasis due to zinc transporter dysfunction results in the onset and progression of a variety of diseases. This review summarizes current progress in our understanding of each ZnT and ZIP transporter from the perspective of zinc physiology and pathogenesis, discussing challenging issues in their structure and zinc transport mechanisms.
Collapse
Affiliation(s)
- Taiho Kambe
- Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Tokuji Tsuji
- Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Ayako Hashimoto
- Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Naoya Itsumura
- Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| |
Collapse
|
49
|
Hagmeyer S, Haderspeck JC, Grabrucker AM. Behavioral impairments in animal models for zinc deficiency. Front Behav Neurosci 2015; 8:443. [PMID: 25610379 PMCID: PMC4285094 DOI: 10.3389/fnbeh.2014.00443] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 12/08/2014] [Indexed: 01/12/2023] Open
Abstract
Apart from teratogenic and pathological effects of zinc deficiency such as the occurrence of skin lesions, anorexia, growth retardation, depressed wound healing, altered immune function, impaired night vision, and alterations in taste and smell acuity, characteristic behavioral changes in animal models and human patients suffering from zinc deficiency have been observed. Given that it is estimated that about 17% of the worldwide population are at risk for zinc deficiency and that zinc deficiency is associated with a variety of brain disorders and disease states in humans, it is of major interest to investigate, how these behavioral changes will affect the individual and a putative course of a disease. Thus, here, we provide a state of the art overview about the behavioral phenotypes observed in various models of zinc deficiency, among them environmentally produced zinc deficient animals as well as animal models based on a genetic alteration of a particular zinc homeostasis gene. Finally, we compare the behavioral phenotypes to the human condition of mild to severe zinc deficiency and provide a model, how zinc deficiency that is associated with many neurodegenerative and neuropsychological disorders might modify the disease pathologies.
Collapse
Affiliation(s)
- Simone Hagmeyer
- WG Molecular Analysis of Synaptopathies, Neurology Department, Neurocenter of Ulm University Ulm, Germany
| | - Jasmin Carmen Haderspeck
- WG Molecular Analysis of Synaptopathies, Neurology Department, Neurocenter of Ulm University Ulm, Germany
| | - Andreas Martin Grabrucker
- WG Molecular Analysis of Synaptopathies, Neurology Department, Neurocenter of Ulm University Ulm, Germany ; Institute for Anatomy and Cell Biology, Ulm University Ulm, Germany
| |
Collapse
|
50
|
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
|