1
|
Tao Y, Pan Q, Cai T, Lu ZH, Haque M, Dottorini T, Colvin LA, Smith BH, Meng W. A genome-wide association study identifies novel genetic variants associated with neck or shoulder pain in the UK biobank (N = 430,193). Pain Rep 2025; 10:e1267. [PMID: 40291381 PMCID: PMC12026381 DOI: 10.1097/pr9.0000000000001267] [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: 08/19/2024] [Revised: 01/11/2025] [Accepted: 02/09/2025] [Indexed: 04/30/2025] Open
Abstract
Introduction Neck and shoulder pain are prevalent musculoskeletal disorders that significantly affect the quality of life for a substantial portion of the global population. Studies have shown that women are more susceptible than men. Objective This study aims to discover genetic variants associated with neck or shoulder pain through a genome-wide association study (GWAS), using data from 430,193 participants in the UK Biobank. Methods A genome-wide association study was performed adjusting for age, sex, BMI, and 8 population principal components. Significant and independent genetic variants were replicated by FinnGen. Results The primary GWAS revealed 5 significant genetic loci (including 2 novel) associated with neck or shoulder pain, with the most significant single nucleotide polymorphism (SNP) being rs9889282 (P = 2.63 × 10-12) near CA10 on chromosome 17. Two novel significant associations were detected on chromosomes 18 and 14, with the top SNPs being rs4608411 (P = 8.20 × 10-9) near TCF4 and rs370565192 (P = 3.80 × 10-8) in DCAF5, respectively. Our secondary GWAS identified a single novel genetic locus in SLC24A3 among males and 2 genetic loci (including one novel near LINC02770) among females. In the replication stage, the SLC39A8 locus was weakly supported by the FinnGen cohort. The tissue expression analysis revealed a significant association between brain tissues and neck or shoulder pain. Conclusion In summary, this study has identified novel genetic variants for neck or shoulder pain. Sex-stratified GWAS also suggested that sex played a role in the occurrence of the phenotype.
Collapse
Affiliation(s)
- Yiwen Tao
- Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute, University of Nottingham Ningbo China, Ningbo, China
| | - Qi Pan
- Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute, University of Nottingham Ningbo China, Ningbo, China
| | - Tengda Cai
- Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute, University of Nottingham Ningbo China, Ningbo, China
| | - Zen Huat Lu
- PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, Bandar Seri Begawan, Brunei Darussalam
| | - Mainul Haque
- School of Mathematical Sciences, University of Nottingham Ningbo China, Ningbo, China
| | - Tania Dottorini
- School of Veterinary Medicine and Science, University of Nottingham, Nottingham, United Kingdom
| | - Lesley A. Colvin
- Division of Population Health and Genomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom
| | - Blair H. Smith
- Division of Population Health and Genomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom
| | - Weihua Meng
- Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute, University of Nottingham Ningbo China, Ningbo, China
- Division of Population Health and Genomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom
- Center for Public Health, Faculty of Medicine, Health and Life Sciences, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, United Kingdom
| |
Collapse
|
2
|
Ahn JH, da Silva Pedrosa M, Lopez LR, Tibbs TN, Jeyachandran JN, Vignieri EE, Rothemich A, Cumming I, Irmscher AD, Haswell CJ, Zamboni WC, Yu YRA, Ellermann M, Denson LA, Arthur JC. Intestinal E. coli-produced yersiniabactin promotes profibrotic macrophages in Crohn's disease. Cell Host Microbe 2025; 33:71-88.e9. [PMID: 39701098 DOI: 10.1016/j.chom.2024.11.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 11/11/2024] [Accepted: 11/22/2024] [Indexed: 12/21/2024]
Abstract
Inflammatory bowel disease (IBD)-associated fibrosis causes significant morbidity. Mechanisms are poorly understood but implicate the microbiota, especially adherent-invasive Escherichia coli (AIEC). We previously demonstrated that AIEC producing the metallophore yersiniabactin (Ybt) promotes intestinal fibrosis in an IBD mouse model. Since macrophages interpret microbial signals and influence inflammation/tissue remodeling, we hypothesized that Ybt metal sequestration disrupts this process. Here, we show that macrophages are abundant in human IBD-fibrosis tissue and mouse fibrotic lesions, where they co-localize with AIEC. Ybt induces profibrotic gene expression in macrophages via stabilization and nuclear translocation of hypoxia-inducible factor 1-alpha (HIF-1α), a metal-dependent immune regulator. Importantly, Ybt-producing AIEC deplete macrophage intracellular zinc and stabilize HIF-1α through inhibition of zinc-dependent HIF-1α hydroxylation. HIF-1α+ macrophages localize to sites of disease activity in human IBD-fibrosis strictures and mouse fibrotic lesions, highlighting their physiological relevance. Our findings reveal microbiota-mediated metal sequestration as a profibrotic trigger targeting macrophages in the inflamed intestine.
Collapse
Affiliation(s)
- Ju-Hyun Ahn
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Marlus da Silva Pedrosa
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Lacey R Lopez
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Taylor N Tibbs
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Joanna N Jeyachandran
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Emily E Vignieri
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Aaron Rothemich
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Ian Cumming
- Department of Pulmonary and Critical Care Medicine, Duke University, Durham, NC 27710, USA
| | - Alexander D Irmscher
- UNC Advanced Translational Pharmacology and Analytical Chemistry Lab, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Corey J Haswell
- UNC Advanced Translational Pharmacology and Analytical Chemistry Lab, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - William C Zamboni
- UNC Advanced Translational Pharmacology and Analytical Chemistry Lab, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yen-Rei A Yu
- Department of Pulmonary and Critical Care Medicine, Duke University, Durham, NC 27710, USA; Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Melissa Ellermann
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Lee A Denson
- Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Janelle C Arthur
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
3
|
Gao C, Zhang H, Wang Y, Wang S, Guo X, Han Y, Zhao H, An X. Global Transcriptomic and Characteristics Comparisons between Mouse Fetal Liver and Bone Marrow Definitive Erythropoiesis. Cells 2024; 13:1149. [PMID: 38995000 PMCID: PMC11240549 DOI: 10.3390/cells13131149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 06/30/2024] [Accepted: 07/02/2024] [Indexed: 07/13/2024] Open
Abstract
Erythropoiesis occurs first in the yolk sac as a transit "primitive" form, then is gradually replaced by the "definitive" form in the fetal liver (FL) during fetal development and in the bone marrow (BM) postnatally. While it is well known that differences exist between primitive and definitive erythropoiesis, the similarities and differences between FL and BM definitive erythropoiesis have not been studied. Here we performed comprehensive comparisons of erythroid progenitors and precursors at all maturational stages sorted from E16.5 FL and adult BM. We found that FL cells at all maturational stages were larger than their BM counterparts. We further found that FL BFU-E cells divided at a faster rate and underwent more cell divisions than BM BFU-E. Transcriptome comparison revealed that genes with increased expression in FL BFU-Es were enriched in cell division. Interestingly, the expression levels of glucocorticoid receptor Nr3c1, Myc and Myc downstream target Ccna2 were significantly higher in FL BFU-Es, indicating the role of the Nr3c1-Myc-Ccna2 axis in the enhanced proliferation/cell division of FL BFU-E cells. At the CFU-E stage, the expression of genes associated with hemoglobin biosynthesis were much higher in FL CFU-Es, indicating more hemoglobin production. During terminal erythropoiesis, overall temporal patterns in gene expression were conserved between the FL and BM. While biological processes related to translation, the tricarboxylic acid cycle and hypoxia response were upregulated in FL erythroblasts, those related to antiviral signal pathway were upregulated in BM erythroblasts. Our findings uncovered previously unrecognized differences between FL and BM definitive erythropoiesis and provide novel insights into erythropoiesis.
Collapse
Affiliation(s)
- Chengjie Gao
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; (C.G.)
- Laboratory of Membrane Biology, New York Blood Center, New York, NY 10065, USA
| | - Huan Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; (C.G.)
| | - Yaomei Wang
- Laboratory of Membrane Biology, New York Blood Center, New York, NY 10065, USA
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China
| | - Shihui Wang
- Institute of Hematology, People’s Hospital of Zhengzhou University, Zhengzhou 450003, China
| | - Xinhua Guo
- Laboratory of Membrane Biology, New York Blood Center, New York, NY 10065, USA
| | - Yongshuai Han
- Laboratory of Membrane Biology, New York Blood Center, New York, NY 10065, USA
| | - Huizhi Zhao
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; (C.G.)
| | - Xiuli An
- Laboratory of Membrane Biology, New York Blood Center, New York, NY 10065, USA
| |
Collapse
|
4
|
Mazgaj R, Lipiński P, Starzyński RR. Iron Supplementation of Pregnant Sows to Prevent Iron Deficiency Anemia in Piglets: A Procedure of Questionable Effectiveness. Int J Mol Sci 2024; 25:4106. [PMID: 38612915 PMCID: PMC11012493 DOI: 10.3390/ijms25074106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 04/03/2024] [Accepted: 04/05/2024] [Indexed: 04/14/2024] Open
Abstract
In pigs, iron deficiency anemia (IDA) is a common disorder that occurs during the early postnatal period, leading to the stunted growth and increased mortality of piglets. The main cause of IDA is low iron stores in the liver of newborn piglets; these stores constitute the main source of iron needed to satisfy the erythropoietic requirements of the piglets in their first weeks of life. Insufficient iron stores in piglets are usually due to the inadequate placental iron transfer from the sow to the fetuses. Therefore, iron supplementation in pregnant sows has been implemented to enhance placental iron transfer and increase iron accumulation in the liver of the fetuses. Over the years, several oral and parenteral approaches have been attempted to supplement sows with various iron preparations, and consequently, to improve piglets' red blood cell indices. However, there is debate with regard to the effectiveness of iron supplementation in pregnant sows for preventing IDA in newborn piglets. Importantly, this procedure should be carried out with caution to avoid iron over-supplementation, which can lead to iron toxicity. This article aims to critically review and evaluate the use of iron supplementation in pregnant sows as a procedure for preventing IDA in piglets.
Collapse
Affiliation(s)
- Rafał Mazgaj
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology Polish Academy of Sciences, 05-552 Magdalenka, Poland;
- Laboratory of Metalloprotein Biology, Institute of Biochemistry and Biophysics Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Paweł Lipiński
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology Polish Academy of Sciences, 05-552 Magdalenka, Poland;
| | - Rafał R. Starzyński
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology Polish Academy of Sciences, 05-552 Magdalenka, Poland;
| |
Collapse
|
5
|
Nebert DW. Gene-Environment Interactions: My Unique Journey. Annu Rev Pharmacol Toxicol 2024; 64:1-26. [PMID: 37788491 DOI: 10.1146/annurev-pharmtox-022323-082311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
I am deeply honored to be invited to write this scientific autobiography. As a physician-scientist, pediatrician, molecular biologist, and geneticist, I have authored/coauthored more than 600 publications in the fields of clinical medicine, biochemistry, biophysics, pharmacology, drug metabolism, toxicology, molecular biology, cancer, standardized gene nomenclature, developmental toxicology and teratogenesis, mouse genetics, human genetics, and evolutionary genomics. Looking back, I think my career can be divided into four distinct research areas, which I summarize mostly chronologically in this article: (a) discovery and characterization of the AHR/CYP1 axis, (b) pharmacogenomics and genetic prediction of response to drugs and other environmental toxicants, (c) standardized drug-metabolizing gene nomenclature based on evolutionary divergence, and (d) discovery and characterization of the SLC39A8 gene encoding the ZIP8 metal cation influx transporter. Collectively, all four topics embrace gene-environment interactions, hence the title of my autobiography.
Collapse
Affiliation(s)
- Daniel W Nebert
- Department of Environmental and Public Health Sciences and Center for Environmental Genetics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Department of Pediatrics and Molecular Developmental Biology, Division of Human Genetics, Cincinnati Children's Hospital, Cincinnati, Ohio, USA;
| |
Collapse
|
6
|
Pan S, Hale AT, Lemieux ME, Raval DK, Garton TP, Sadler B, Mahaney KB, Strahle JM. Iron homeostasis and post-hemorrhagic hydrocephalus: a review. Front Neurol 2024; 14:1287559. [PMID: 38283681 PMCID: PMC10811254 DOI: 10.3389/fneur.2023.1287559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Accepted: 11/21/2023] [Indexed: 01/30/2024] Open
Abstract
Iron physiology is regulated by a complex interplay of extracellular transport systems, coordinated transcriptional responses, and iron efflux mechanisms. Dysregulation of iron metabolism can result in defects in myelination, neurotransmitter synthesis, and neuronal maturation. In neonates, germinal matrix-intraventricular hemorrhage (GMH-IVH) causes iron overload as a result of blood breakdown in the ventricles and brain parenchyma which can lead to post-hemorrhagic hydrocephalus (PHH). However, the precise mechanisms by which GMH-IVH results in PHH remain elusive. Understanding the molecular determinants of iron homeostasis in the developing brain may lead to improved therapies. This manuscript reviews the various roles iron has in brain development, characterizes our understanding of iron transport in the developing brain, and describes potential mechanisms by which iron overload may cause PHH and brain injury. We also review novel preclinical treatments for IVH that specifically target iron. Understanding iron handling within the brain and central nervous system may provide a basis for preventative, targeted treatments for iron-mediated pathogenesis of GMH-IVH and PHH.
Collapse
Affiliation(s)
- Shelei Pan
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - Andrew T. Hale
- Department of Neurosurgery, University of Alabama at Birmingham School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Mackenzie E. Lemieux
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - Dhvanii K. Raval
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - Thomas P. Garton
- Department of Neurology, Johns Hopkins University School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Brooke Sadler
- Department of Pediatrics, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
- Department of Hematology and Oncology, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - Kelly B. Mahaney
- Department of Neurosurgery, Stanford University School of Medicine, Stanford University, Palo Alto, CA, United States
| | - Jennifer M. Strahle
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
- Department of Pediatrics, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
- Department of Orthopedic Surgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| |
Collapse
|
7
|
Sangkhae V, Fisher AL, Ganz T, Nemeth E. Iron Homeostasis During Pregnancy: Maternal, Placental, and Fetal Regulatory Mechanisms. Annu Rev Nutr 2023; 43:279-300. [PMID: 37253681 PMCID: PMC10723031 DOI: 10.1146/annurev-nutr-061021-030404] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Pregnancy entails a large negative balance of iron, an essential micronutrient. During pregnancy, iron requirements increase substantially to support both maternal red blood cell expansion and the development of the placenta and fetus. As insufficient iron has long been linked to adverse pregnancy outcomes, universal iron supplementation is common practice before and during pregnancy. However, in high-resource countries with iron fortification of staple foods and increased red meat consumption, the effects of too much iron supplementation during pregnancy have become a concern because iron excess has also been linked to adverse pregnancy outcomes. In this review, we address physiologic iron homeostasis of the mother, placenta, and fetus and discuss perturbations in iron homeostasis that result in pathological pregnancy. As many mechanistic regulatory systems have been deduced from animal models, we also discuss the principles learned from these models and how these may apply to human pregnancy.
Collapse
Affiliation(s)
- Veena Sangkhae
- Center for Iron Disorders, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA;
| | - Allison L Fisher
- Endocrine Unit and Nephrology Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Tomas Ganz
- Center for Iron Disorders, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA;
| | - Elizabeta Nemeth
- Center for Iron Disorders, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA;
| |
Collapse
|
8
|
Pasquadibisceglie A, Bonaccorsi di Patti MC, Musci G, Polticelli F. Membrane Transporters Involved in Iron Trafficking: Physiological and Pathological Aspects. Biomolecules 2023; 13:1172. [PMID: 37627237 PMCID: PMC10452680 DOI: 10.3390/biom13081172] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/25/2023] [Accepted: 07/26/2023] [Indexed: 08/27/2023] Open
Abstract
Iron is an essential transition metal for its involvement in several crucial biological functions, the most notable being oxygen storage and transport. Due to its high reactivity and potential toxicity, intracellular and extracellular iron levels must be tightly regulated. This is achieved through transport systems that mediate cellular uptake and efflux both at the level of the plasma membrane and on the membranes of lysosomes, endosomes and mitochondria. Among these transport systems, the key players are ferroportin, the only known transporter mediating iron efflux from cells; DMT1, ZIP8 and ZIP14, which on the contrary, mediate iron influx into the cytoplasm, acting on the plasma membrane and on the membranes of lysosomes and endosomes; and mitoferrin, involved in iron transport into the mitochondria for heme synthesis and Fe-S cluster assembly. The focus of this review is to provide an updated view of the physiological role of these membrane proteins and of the pathologies that arise from defects of these transport systems.
Collapse
Affiliation(s)
| | | | - Giovanni Musci
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy;
| | - Fabio Polticelli
- Department of Sciences, University Roma Tre, 00146 Rome, Italy;
- National Institute of Nuclear Physics, Roma Tre Section, 00146 Rome, Italy
| |
Collapse
|
9
|
Yu S, Zhao N. The Regulation of ZIP8 by Dietary Manganese in Mice. Int J Mol Sci 2023; 24:ijms24065962. [PMID: 36983036 PMCID: PMC10056016 DOI: 10.3390/ijms24065962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/18/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
ZIP8 is a newly identified manganese transporter. A lack of functional ZIP8 results in severe manganese deficiency in both humans and mice, indicating that ZIP8 plays a crucial role in maintaining body manganese homeostasis. Despite a well-acknowledged connection between ZIP8 and manganese metabolism, how ZIP8 is regulated under high-manganese conditions remains unclear. The primary goal of this study was to examine the regulation of ZIP8 by high-manganese intake. We used both neonatal and adult mouse models in which mice were supplied with dietary sources containing either a normal or a high level of manganese. We discovered that high-manganese intake caused a reduction in liver ZIP8 protein in young mice. Since a decrease in hepatic ZIP8 leads to reduced manganese reabsorption from the bile, our study identified a novel mechanism for the regulation of manganese homeostasis under high-manganese conditions: high dietary manganese intake results in a decrease in ZIP8 in the liver, which in turn decreases the reabsorption of manganese from the bile to prevent manganese overload in the liver. Interestingly, we found that a high-manganese diet did not cause a decrease in hepatic ZIP8 in adult animals. To determine the potential reason for this age-dependent variation, we compared the expressions of liver ZIP8 in 3-week-old and 12-week-old mice. We found that liver ZIP8 protein content in 12-week-old mice decreases when compared with that of 3-week-old mice under normal conditions. Overall, results from this study provide novel insights to facilitate the understanding of ZIP8's function in regulating manganese metabolism.
Collapse
Affiliation(s)
- Suetmui Yu
- School of Nutritional Sciences and Wellness, The University of Arizona, Tucson, AZ 85721, USA
| | - Ningning Zhao
- School of Nutritional Sciences and Wellness, The University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
10
|
Cao C, Fleming MD. Loss of the placental iron exporter ferroportin 1 causes embryonic demise in late-gestation mouse pregnancy. Development 2022; 149:285826. [PMID: 36398730 DOI: 10.1242/dev.201160] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 11/07/2022] [Indexed: 11/19/2022]
Abstract
Fetal development relies on adequate iron supply by the placenta. The placental syncytiotrophoblasts (SCTB) express high levels of iron transporters, including ferroportin1 (Fpn1). Whether they are essential in the placenta has not been tested directly, mainly due to the lack of gene manipulation tools in SCTB. Here, we aimed to generate a SCTB-specific Cre mouse and use it to determine the role of placental Fpn1. Using CRISPR/Cas9 technology, we created a syncytin b (Synb) Cre line (SynbCre) targeting the fetal-facing SCTB layer in mouse placental labyrinth. SynbCre deleted Fpn1 in late gestation mouse placentas reliably with high efficiency. Embryos without placental Fpn1 were pale and runted, and died before birth. Fpn1 null placentas had reduced transferrin receptor expression, increased oxidative stress and detoxification responses, and accumulated ferritin in the SCTB instead of the fetal endothelium. In summary, we demonstrate that SynbCre is an effective and specific tool to investigate placental gene function in vivo. The loss of Fpn1 in late gestation mouse placenta is embryonically lethal, providing direct evidence for an essential role of Fpn1 in placental iron transport.
Collapse
Affiliation(s)
- Chang Cao
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Mark D Fleming
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA
| |
Collapse
|
11
|
The schizophrenia-associated missense variant rs13107325 regulates dendritic spine density. Transl Psychiatry 2022; 12:361. [PMID: 36056013 PMCID: PMC9440106 DOI: 10.1038/s41398-022-02137-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 08/17/2022] [Accepted: 08/23/2022] [Indexed: 11/26/2022] Open
Abstract
The missense variant rs13107325 (C/T, p.Ala391Thr) in SLC39A8 consistently showed robust association with schizophrenia in recent genome-wide association studies (GWASs), suggesting the potential pathogenicity of this non-synonymous risk variant. Nevertheless, how this missense variant confers schizophrenia risk remains unknown. Here we constructed a knock-in mouse model (by introducing a threonine at the 393th amino acid of mouse SLC39A8 (SLC39A8-p.393T), which corresponds to rs13107325 (p.Ala391Thr) of human SLC39A8) to explore the potential roles and biological effects of this missense variant in schizophrenia pathogenesis. We assessed multiple phenotypes and traits (associated with rs13107325) of the knock-in mice, including body and brain weight, concentrations of metal ions (including cadmium, zinc, manganese, and iron) transported by SLC39A8, blood lipids, proliferation and migration of neural stem cells (NSCs), cortical development, behaviors and cognition, transcriptome, dendritic spine density, and synaptic transmission. Many of the tested phenotypes did not show differences in SLC39A8-p.393T knock-in and wild-type mice. However, we found that zinc concentration in brain and blood of SLC39A8-p.393T knock-in mice was dysregulated compared with wild-types, validating the functionality of rs13107325. Further analysis indicated that cortical dendritic spine density of the SLC39A8-p.393T knock-in mice was significantly decreased compared with wild-types, indicating the important role of SLC39A8-p.393T in dendritic spine morphogenesis. These results indicated that SLC39A8-p.393T knock-in resulted in decreased dendritic spine density, thus mimicking the dendritic spine pathology observed in schizophrenia. Our study indicates that rs13107325 might confer schizophrenia risk by regulating zinc concentration and dendritic spine density, a featured characteristic that was frequently reported to be decreased in schizophrenia.
Collapse
|
12
|
Cheng W, Zhang L, Sa P, Luo J, Li M. Transcriptomic analysis reveals the effects of maternal selenium deficiency on placental transport, hormone synthesis, and immune response in mice. METALLOMICS : INTEGRATED BIOMETAL SCIENCE 2022; 14:6674774. [PMID: 36002020 DOI: 10.1093/mtomcs/mfac062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 08/19/2022] [Indexed: 11/14/2022]
Abstract
Selenium deficiency has been considered to increase the risk of gestational complications. Our previous work showed that maternal selenium deficiency suppressed proliferation, induced autophagy dysfunction and apoptosis in the placenta of mice. However, other effects of maternal selenium deficiency on the placenta and the underlying mechanisms remain unclear. In the present study, dietary selenium deficiency in dams significantly suppressed glutathione peroxidase (GSH-Px) activity, total antioxidant capacity (T-AOC), and increased malondialdehyde (MDA) content in the placentae, confirming the oxidative stress in the placenta. By transcriptome sequencing analysis, the DEGs were involved in many biological processes, including ion transport, lipid metabolic process, immune response, transmembrane transport, and others. According to the KEGG analysis, the DEGs were primarily enriched in metabolic pathways, PI3K-Akt signaling pathway, and others. Among these, the steroid hormone biosynthesis pathway enriched the most DEGs. Hsd3b1, an ER enzyme involved in progesterone synthesis, was validated downregulated. Consistently, the progesterone content in the serum of the selenium-deficient group was decreased. Ion transporters and transmembrane transporters, such as Heph, Trf, Slc39a8, Slc23a1, Atp7b, and Kcnc1, were reduced in the selenium-deficient placentae. Immune response-related genes, including Ccl3, Ccl8, Cxcl10, and Cxcl14, were increased in the selenium-deficient placentae, along with an increase in macrophage number. These results suggested that maternal selenium deficiency may impair progesterone biosynthesis, reduce nutrient transporters expression, and promote immune response by increasing the oxidative stress of the placentae. This present study provides a novel insight into the possible cause of placenta disorder during pregnancy.
Collapse
Affiliation(s)
- Wanpeng Cheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Lantian Zhang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.,Department of Anatomy, Basic Medical College, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Peiyue Sa
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.,Department of Anatomy, Basic Medical College, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Jing Luo
- Department of Clinical Medicine, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Mengdi Li
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.,Department of Anatomy, Basic Medical College, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| |
Collapse
|
13
|
Iron Metabolism and Ferroptosis in Physiological and Pathological Pregnancy. Int J Mol Sci 2022; 23:ijms23169395. [PMID: 36012659 PMCID: PMC9409111 DOI: 10.3390/ijms23169395] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 08/16/2022] [Accepted: 08/17/2022] [Indexed: 12/13/2022] Open
Abstract
Iron is a vital element in nearly every living organism. During pregnancy, optimal iron concentration is essential for both maternal health and fetal development. As the barrier between the mother and fetus, placenta plays a pivotal role in mediating and regulating iron transport. Imbalances in iron metabolism correlate with severe adverse pregnancy outcomes. Like most other nutrients, iron exhibits a U-shaped risk curve. Apart from iron deficiency, iron overload is also dangerous since labile iron can generate reactive oxygen species, which leads to oxidative stress and activates ferroptosis. In this review, we summarized the molecular mechanism and regulation signals of placental iron trafficking under physiological conditions. In addition, we revealed the role of iron metabolism and ferroptosis in the view of preeclampsia and gestational diabetes mellitus, which may bring new insight to the pathogenesis and treatment of pregnancy-related diseases.
Collapse
|
14
|
Clinical and Molecular Aspects of Iron Metabolism in Failing Myocytes. LIFE (BASEL, SWITZERLAND) 2022; 12:life12081203. [PMID: 36013382 PMCID: PMC9409945 DOI: 10.3390/life12081203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/15/2022] [Accepted: 07/22/2022] [Indexed: 11/16/2022]
Abstract
Heart failure (HF) is a common disease that causes significant limitations on the organism's capacity and, in extreme cases, leads to death. Clinically, iron deficiency (ID) plays an essential role in heart failure by deteriorating the patient's condition and is a prognostic marker indicating poor clinical outcomes. Therefore, in HF patients, supplementation of iron is recommended. However, iron treatment may cause adverse effects by increasing iron-related apoptosis and the production of oxygen radicals, which may cause additional heart damage. Furthermore, many knowledge gaps exist regarding the complex interplay between iron deficiency and heart failure. Here, we describe the current, comprehensive knowledge about the role of the proteins involved in iron metabolism. We will focus on the molecular and clinical aspects of iron deficiency in HF. We believe that summarizing the new advances in the translational and clinical research regarding iron deficiency in heart failure should broaden clinicians' awareness of this comorbidity.
Collapse
|
15
|
Abstract
Virus-induced cell death has long been thought of as a double-edged sword in the inhibition or exacerbation of viral infections. The vital role of iron, an essential element for various enzymes in the maintenance of cellular physiology and efficient viral replication, places it at the crossroads and makes it a micronutrient of competition between the viruses and the host. Viruses can interrupt iron uptake and the antioxidant response system, while others can utilize iron transporter proteins as receptors. Interestingly, the unavailability of iron facilitates certain viral infections and causes cell death characterized by lipid peroxide accumulation and malfunction of the antioxidant system. In this review, we discuss how iron uptake, regulation and metabolism, including the redistribution of iron in the host defense system during viral infection, can induce ferroptosis. Fenton reactions, a central characteristic of ferroptosis, are caused by the increased iron content in the cell. Therefore, viral infections that increase cellular iron content or intestinal iron absorption are likely to cause ferroptosis. In addition, we discuss the hijacking of the iron regulatoy pathway and the antioxidant response, both of which are typical in viral infections. Understanding the potential signaling mechanisms of ferroptosis in viral infections will aid in the development of new therapeutic agents.
Collapse
|
16
|
Willekens J, Runnels LW. Impact of Zinc Transport Mechanisms on Embryonic and Brain Development. Nutrients 2022; 14:2526. [PMID: 35745255 PMCID: PMC9231024 DOI: 10.3390/nu14122526] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/09/2022] [Accepted: 06/09/2022] [Indexed: 12/04/2022] Open
Abstract
The trace element zinc (Zn) binds to over ten percent of proteins in eukaryotic cells. Zn flexible chemistry allows it to regulate the activity of hundreds of enzymes and influence scores of metabolic processes in cells throughout the body. Deficiency of Zn in humans has a profound effect on development and in adults later in life, particularly in the brain, where Zn deficiency is linked to several neurological disorders. In this review, we will summarize the importance of Zn during development through a description of the outcomes of both genetic and early dietary Zn deficiency, focusing on the pathological consequences on the whole body and brain. The epidemiology and the symptomology of Zn deficiency in humans will be described, including the most studied inherited Zn deficiency disease, Acrodermatitis enteropathica. In addition, we will give an overview of the different forms and animal models of Zn deficiency, as well as the 24 Zn transporters, distributed into two families: the ZIPs and the ZnTs, which control the balance of Zn throughout the body. Lastly, we will describe the TRPM7 ion channel, which was recently shown to contribute to intestinal Zn absorption and has its own significant impact on early embryonic development.
Collapse
Affiliation(s)
| | - Loren W. Runnels
- Department of Pharmacology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA;
| |
Collapse
|
17
|
Verouti SN, Pujol-Giménez J, Bermudez-Lekerika P, Scherler L, Bhardwaj R, Thomas A, Lenglet S, Siegrist M, Hofstetter W, Fuster DG, Hediger MA, Escher G, Vogt B. The Allelic Variant A391T of Metal Ion Transporter ZIP8 (SLC39A8) Leads to Hypotension and Enhanced Insulin Resistance. Front Physiol 2022; 13:912277. [PMID: 35784893 PMCID: PMC9240775 DOI: 10.3389/fphys.2022.912277] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 05/18/2022] [Indexed: 12/12/2022] Open
Abstract
The metal ion transporter ZIP8 (SLC39A8) mediates cellular uptake of vital divalent metal ions. Genome-wide association studies (GWAS) showed that the single-nucleotide polymorphism (SNP) variant A391T (rs13107325) is associated with numerous human traits, including reduced arterial blood pressure, increased body mass index and hyperlipidemia. We analyzed in vitro the transport properties of mutant ZIP8 A391T and investigated in vivo in mice the physiological effects of this polymorphism. In vitro, the intrinsic transport properties of mutant ZIP8 were similar to those of wild type ZIP8, but cellular uptake of zinc, cadmium and iron was attenuated due to reduced ZIP8 plasma membrane expression. We then generated the ZIP8 A393T mice (ZIP8KI) that carry the corresponding polymorphism and characterized their phenotype. We observed lower protein expression in lung and kidney membrane extracts in ZIP8KI mice. The ZIP8KI mice exhibited striking changes in metal ion composition of the tissues, including cobalt, palladium, mercury and platinum. In agreement with GWAS, ZIP8KI mice showed reduced arterial blood pressure. Body weight and plasma lipid composition remained unchanged, although these features were reported to be increased in GWAS. ZIP8KI mice also exhibited remarkable insulin resistance and were protected from elevated blood glucose when challenged by dietary sucrose supplementation. We showed that increased hepatic insulin receptor expression and decreased ZnT8 (slc30a8) metal ion transporter mRNA expression are associated with this phenotypic change. In conclusion, our data reveal that ZIP8 plays an important role in blood pressure regulation and glucose homeostasis.
Collapse
Affiliation(s)
- Sophia N. Verouti
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department of Nephrology and Hypertension, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Jonai Pujol-Giménez
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department of Nephrology and Hypertension, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Paola Bermudez-Lekerika
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department of Nephrology and Hypertension, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Laeticia Scherler
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department of Nephrology and Hypertension, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Rajesh Bhardwaj
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department of Nephrology and Hypertension, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, NIH, Durham, NC, United States
| | - Aurélien Thomas
- Forensic Toxicology and Chemistry Unit, CURML, Lausanne University Hospital, Geneva University, Geneva, Switzerland
- Faculty Unit of Toxicology, CURML, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Sébastien Lenglet
- Forensic Toxicology and Chemistry Unit, CURML, Lausanne University Hospital, Geneva University, Geneva, Switzerland
| | - Mark Siegrist
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Willy Hofstetter
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Daniel G. Fuster
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department of Nephrology and Hypertension, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Matthias A. Hediger
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department of Nephrology and Hypertension, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- *Correspondence: Matthias A. Hediger, ; Geneviève Escher, ; Bruno Vogt,
| | - Geneviève Escher
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department of Nephrology and Hypertension, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- *Correspondence: Matthias A. Hediger, ; Geneviève Escher, ; Bruno Vogt,
| | - Bruno Vogt
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department of Nephrology and Hypertension, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- *Correspondence: Matthias A. Hediger, ; Geneviève Escher, ; Bruno Vogt,
| |
Collapse
|
18
|
Zinc transporters ZIPT-2.4 and ZIPT-15 are required for normal C. elegans fecundity. J Assist Reprod Genet 2022; 39:1261-1276. [PMID: 35501415 DOI: 10.1007/s10815-022-02495-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 04/11/2022] [Indexed: 10/18/2022] Open
Abstract
PURPOSE The requirement of zinc for the development and maturation of germ lines and reproductive systems is deeply conserved across evolution. The nematode Caenorhabditis elegans offers a tractable platform to study the complex system of distributing zinc to the germ line. We investigated several zinc importers to investigate how zinc transporters play a role in the reproductive system in nematodes, as well as establish a platform to study zinc transporter biology in germline and reproductive development. METHODS Previous high throughput transcriptional datasets as well as phylogenetic analysis identified several putative zinc transporters that have a function in reproduction in worms. Phenotypic analysis of CRISPR-generated knockouts and tags included characterization of offspring output, gonad development, and protein localization. Light and immunofluorescence microscopy allowed for visualization of physiological and molecular effects of zinc transporter mutations. RESULTS Disruption of two zinc transporters, ZIPT-2.4 and ZIPT-15, was shown to lead to defects in reproductive output. A mutation in zipt-2.4 has subtle effects on reproduction, while a mutation in zipt-15 has a clear impact on gonad and germline development that translates into a more pronounced defect in fecundity. Both transporters have germline expression, as well as additional expression in other cell types. CONCLUSIONS Two ZIP-family zinc transporter orthologs of human ZIP6/10 and ZIP1/2/3 proteins are important for full reproductive fecundity and participate in development of the gonad. Notably, these zinc transporters are present in gut and reproductive tissues in addition to the germ line, consistent with a complex zinc trafficking network important for reproductive success.
Collapse
|
19
|
Helman SL, Wilkins SJ, McKeating DR, Perkins AV, Whibley PE, Cuffe JSM, Simmons DG, Fuqua BK, Vulpe CD, Wallace DF, O'Callaghan JL, Pelzer ES, Anderson GJ, Frazer DM. The Placental Ferroxidase Zyklopen Is Not Essential for Iron Transport to the Fetus in Mice. J Nutr 2021; 151:2541-2550. [PMID: 34114013 DOI: 10.1093/jn/nxab174] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/09/2021] [Accepted: 05/07/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND The ferroxidase zyklopen (Zp) has been implicated in the placental transfer of iron to the fetus. However, the evidence for this is largely circumstantial. OBJECTIVES This study aimed to determine whether Zp is essential for placental iron transfer. METHODS A model was established using 8- to 12-wk-old pregnant C57BL/6 mice on standard rodent chow in which Zp was knocked out in the fetus and fetal components of the placenta. Zp was also disrupted in the entire placenta using global Zp knockout mice. Inductively coupled plasma MS was used to measure total fetal iron, an indicator of the amount of iron transferred by the placenta to the fetus, at embryonic day 18.5 of gestation. Iron transporter expression in the placenta was measured by Western blotting, and the expression of Hamp1, the gene encoding the iron regulatory hormone hepcidin, was determined in fetal liver by real-time PCR. RESULTS There was no change in the amount of iron transferred to the fetus when Zp was disrupted in either the fetal component of the placenta or the entire placenta. No compensatory changes in the expression of the iron transport proteins transferrin receptor 1 or ferroportin were observed, nor was there any change in fetal liver Hamp1 mRNA. Hephl1, the gene encoding Zp, was expressed mainly in the maternal decidua of the placenta and not in the nutrient-transporting syncytiotrophoblast. Disruption of Zp in the whole placenta resulted in a 26% increase in placental size (P < 0.01). CONCLUSIONS Our data indicate that Zp is not essential for the efficient transfer of iron to the fetus in mice and is localized predominantly in the maternal decidua. The increase in placental size observed when Zp is knocked out in the entire placenta suggests that this protein may play a role in placental development.
Collapse
Affiliation(s)
- Sheridan L Helman
- Molecular Nutrition Laboratory, QIMR Berghofer Medical Research Institute, Herston, Australia.,Faculty of Medicine, The University of Queensland, St. Lucia, Australia
| | - Sarah J Wilkins
- Iron Metabolism Laboratory, QIMR Berghofer Medical Research Institute, Herston, Australia
| | - Daniel R McKeating
- School of Medical Science, Griffith University, Gold Coast Campus, Southport, Australia
| | - Anthony V Perkins
- School of Medical Science, Griffith University, Gold Coast Campus, Southport, Australia
| | - Page E Whibley
- Molecular Nutrition Laboratory, QIMR Berghofer Medical Research Institute, Herston, Australia
| | - James S M Cuffe
- School of Biomedical Sciences, The University of Queensland, St. Lucia, Australia
| | - David G Simmons
- School of Biomedical Sciences, The University of Queensland, St. Lucia, Australia
| | - Brie K Fuqua
- David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Christopher D Vulpe
- Department of Physiological Sciences, University of Florida, Gainesville, FL, USA
| | - Daniel F Wallace
- School of Biomedical Sciences, Queensland University of Technology, Gardens Point, Australia
| | - Jessica L O'Callaghan
- School of Biomedical Sciences, Queensland University of Technology, Gardens Point, Australia
| | - Elise S Pelzer
- School of Biomedical Sciences, Queensland University of Technology, Gardens Point, Australia
| | - Gregory J Anderson
- Iron Metabolism Laboratory, QIMR Berghofer Medical Research Institute, Herston, Australia.,School of Chemistry and Molecular Bioscience, The University of Queensland, St. Lucia, Australia
| | - David M Frazer
- Molecular Nutrition Laboratory, QIMR Berghofer Medical Research Institute, Herston, Australia.,School of Biomedical Sciences, The University of Queensland, St. Lucia, Australia.,School of Biomedical Sciences, Queensland University of Technology, Gardens Point, Australia
| |
Collapse
|
20
|
De R, Prakash KU, Edison ES. Complex Interactions in Regulation of Haematopoiesis-An Unexplored Iron Mine. Genes (Basel) 2021; 12:genes12081270. [PMID: 34440444 PMCID: PMC8391430 DOI: 10.3390/genes12081270] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/10/2021] [Accepted: 07/13/2021] [Indexed: 12/14/2022] Open
Abstract
Iron is one of the most abundant metals on earth and is vital for the growth and survival of life forms. It is crucial for the functioning of plants and animals as it is an integral component of the photosynthetic apparatus and innumerable proteins and enzymes. It plays a pivotal role in haematopoiesis and affects the development and differentiation of different haematopoietic lineages, apart from its obvious necessity in erythropoiesis. A large amount of iron stores in humans is diverted towards the latter process, as iron is an indispensable component of haemoglobin. This review summarises the important players of iron metabolism and homeostasis that have been discovered in recent years and highlights the overall significance of iron in haematopoiesis. Its role in maintenance of haematopoietic stem cells, influence on differentiation of varied haematopoietic lineages and consequences of iron deficiency/overloading on development and maturation of different groups of haematopoietic cells have been discussed.
Collapse
|
21
|
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
|
22
|
Kang JA, Kwak JS, Park SH, Sim KY, Kim SK, Shin Y, Jung IJ, Yang JI, Chun JS, Park SG. ZIP8 exacerbates collagen-induced arthritis by increasing pathogenic T cell responses. Exp Mol Med 2021; 53:560-571. [PMID: 33795795 PMCID: PMC8102558 DOI: 10.1038/s12276-021-00591-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 12/20/2020] [Accepted: 01/08/2021] [Indexed: 02/01/2023] Open
Abstract
Zinc is a trace element that is essential for immune responses. Therefore, changes in cellular zinc levels in specific immune cells may influence inflammatory autoimmune diseases, such as rheumatoid arthritis (RA). However, the regulation of zinc mobilization in immune cells and its role in the pathogenesis of RA are not fully understood. Thus, we investigated the roles of zinc transporters in RA pathogenesis. We demonstrated that ZIP8 was specifically upregulated in CD4+ T cells that infiltrated the inflamed joint and that ZIP8 deficiency in CD4+ T cells abrogated collagen-induced arthritis. ZIP8 deficiency dramatically affected zinc influx in effector T cells and profoundly reduced T cell receptor (TCR)-mediated signaling, including NF-κB and MAPK signaling, which are pathways that are involved in T helper (Th) 17 cell differentiation. Taken together, our findings suggest that ZIP8 depletion in CD4+ T cells attenuates TCR signaling due to insufficient cellular zinc, thereby reducing the function of effector CD4+ T cells, including Th17 cells. Our results also suggest that targeting ZIP8 may be a useful strategy to inhibit RA development and pathogenesis.
Collapse
Affiliation(s)
- Jung-Ah Kang
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
- Cell Logistics Research Center, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
- Infectious Disease Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Ji-Sun Kwak
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
- National Creative Research Initiatives Center for Osteoarthritis Pathogenesis, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Sang-Heon Park
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
- Cell Logistics Research Center, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
- College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
| | - Kyu-Young Sim
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
- Cell Logistics Research Center, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
- College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
| | - Seul Ki Kim
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
- National Creative Research Initiatives Center for Osteoarthritis Pathogenesis, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Youngnim Shin
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
- National Creative Research Initiatives Center for Osteoarthritis Pathogenesis, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - In Jung Jung
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
- National Creative Research Initiatives Center for Osteoarthritis Pathogenesis, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Jeong-In Yang
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
- National Creative Research Initiatives Center for Osteoarthritis Pathogenesis, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Jang-Soo Chun
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea.
- Cell Logistics Research Center, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea.
- National Creative Research Initiatives Center for Osteoarthritis Pathogenesis, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea.
| | - Sung-Gyoo Park
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea.
- Cell Logistics Research Center, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea.
- College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
23
|
Cao C, Prado MA, Sun L, Rockowitz S, Sliz P, Paulo JA, Finley D, Fleming MD. Maternal Iron Deficiency Modulates Placental Transcriptome and Proteome in Mid-Gestation of Mouse Pregnancy. J Nutr 2021; 151:1073-1083. [PMID: 33693820 PMCID: PMC8112763 DOI: 10.1093/jn/nxab005] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/26/2020] [Accepted: 01/06/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Maternal iron deficiency (ID) is associated with poor pregnancy and fetal outcomes. The effect is thought to be mediated by the placenta but there is no comprehensive assessment of placental responses to maternal ID. Additionally, whether the influence of maternal ID on the placenta differs by fetal sex is unknown. OBJECTIVES To identify gene and protein signatures of ID mouse placentas at mid-gestation. A secondary objective was to profile the expression of iron genes in mouse placentas across gestation. METHODS We used a real-time PCR-based array to determine the mRNA expression of all known iron genes in mouse placentas at embryonic day (E) 12.5, E14.5, E16.5, and E19.5 (n = 3 placentas/time point). To determine the effect of maternal ID, we performed RNA sequencing and proteomics in male and female placentas from ID and iron-adequate mice at E12.5 (n = 8 dams/diet). RESULTS In female placentas, 6 genes, including transferrin receptor (Tfrc) and solute carrier family 11 member 2, were significantly changed by maternal ID. An additional 154 genes were altered in male ID placentas. A proteomic analysis quantified 7662 proteins in the placenta. Proteins translated from iron-responsive element (IRE)-containing mRNA were altered in abundance; ferritin and ferroportin 1 decreased, while TFRC increased in ID placentas. Less than 4% of the significantly altered genes in ID placentas occurred both at the transcriptional and translational levels. CONCLUSIONS Our data demonstrate that the impact of maternal ID on placental gene expression in mice is limited in scope and magnitude at mid-gestation. We provide strong evidence for IRE-based transcriptional and translational coordination of iron gene expression in the mouse placenta. Finally, we discover sexually dimorphic effects of maternal ID on placental gene expression, with more genes and pathways altered in male compared with female mouse placentas.
Collapse
Affiliation(s)
- Chang Cao
- Address correspondence to CC (e-mail: )
| | - Miguel A Prado
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Liang Sun
- Computational Health Informatics Program, Boston Children's Hospital, Boston, MA, USA,The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA, USA
| | - Shira Rockowitz
- Computational Health Informatics Program, Boston Children's Hospital, Boston, MA, USA,The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA, USA
| | - Piotr Sliz
- Computational Health Informatics Program, Boston Children's Hospital, Boston, MA, USA,The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA, USA,Division of Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Daniel Finley
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Mark D Fleming
- Department of Pathology, Boston Children's Hospital, Boston, MA, USA
| |
Collapse
|
24
|
Schizophrenia-associated SLC39A8 polymorphism is a loss-of-function allele altering glutamate receptor and innate immune signaling. Transl Psychiatry 2021; 11:136. [PMID: 33608496 PMCID: PMC7895948 DOI: 10.1038/s41398-021-01262-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 02/02/2021] [Indexed: 11/08/2022] Open
Abstract
Schizophrenia is a complex and heterogenous disease that presents with abnormalities in glutamate signaling and altered immune and inflammatory signals. Genome-wide association studies have indicated specific genes and pathways that may contribute to schizophrenia. We assessed the impact of the functional missense variant SLC39A8 (ZIP8)-A391T (ZIP8A391T) on zinc transport, glutamate signaling, and the neuroinflammatory response. The ZIP8A391T mutation resulted in reduced zinc transport into the cell, suggesting a loss in the tight control of zinc in the synaptic cleft. Electrophysiological recordings from perturbed neurons revealed a significant reduction in NMDA- and AMPA-mediated spontaneous EPSCs (sEPSCs) and a reduction in GluN2A and GluA1/2/3 receptor surface expression. All phenotypes were rescued by re-expression of wild-type ZIP8 (ZIP8WT) or application of the membrane-impermeable zinc chelator ZX1. ZIP8 reduction also resulted in decreased BBB integrity, increased IL-6/IL-1β protein expression, and increased NFκB following TNFα stimulation, indicating that ZIP8 loss-of-function may exacerbate immune and inflammatory signals. Together, our findings demonstrate that the A391T missense mutation results in alterations in glutamate and immune function and provide novel therapeutic targets relevant to schizophrenia.
Collapse
|
25
|
Hildebrand S, Löwa N, Paysen H, Fratila RM, Reverte-Salisa L, Trakoolwilaiwan T, Niu Z, Kasparis G, Preuss SF, Kosch O, M de la Fuente J, Thanh NTK, Wiekhorst F, Pfeifer A. Quantification of Lipoprotein Uptake in Vivo Using Magnetic Particle Imaging and Spectroscopy. ACS NANO 2021; 15:434-446. [PMID: 33306343 DOI: 10.1021/acsnano.0c03229] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Lipids are a major source of energy for most tissues, and lipid uptake and storage is therefore crucial for energy homeostasis. So far, quantification of lipid uptake in vivo has primarily relied on radioactive isotope labeling, exposing human subjects or experimental animals to ionizing radiation. Here, we describe the quantification of in vivo uptake of chylomicrons, the primary carriers of dietary lipids, in metabolically active tissues using magnetic particle imaging (MPI) and magnetic particle spectroscopy (MPS). We show that loading artificial chylomicrons (ACM) with iron oxide nanoparticles (IONPs) enables rapid and highly sensitive post hoc detection of lipid uptake in situ using MPS. Importantly, by utilizing highly magnetic Zn-doped iron oxide nanoparticles (ZnMNPs), we generated ACM with MPI tracer properties superseding the current gold-standard, Resovist, enabling quantification of lipid uptake from whole-animal scans. We focused on brown adipose tissue (BAT), which dissipates heat and can consume a large part of nutrient lipids, as a model for tightly regulated and inducible lipid uptake. High BAT activity in humans correlates with leanness and improved cardiometabolic health. However, the lack of nonradioactive imaging techniques is an important hurdle for the development of BAT-centered therapies for metabolic diseases such as obesity and type 2 diabetes. Comparison of MPI measurements with iron quantification by inductively coupled plasma mass spectrometry revealed that MPI rivals the performance of this highly sensitive technique. Our results represent radioactivity-free quantification of lipid uptake in metabolically active tissues such as BAT.
Collapse
Affiliation(s)
- Staffan Hildebrand
- Institut für Pharmakologie und Toxikologie, Rheinische Friedrich-Wilhelms-Universität Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Norbert Löwa
- 8.23 Metrology for Magnetic Nanoparticles, Physikalisch-Technische Bundesanstalt, Abbestraße 2-12, 10587 Berlin, Germany
| | - Hendrik Paysen
- 8.23 Metrology for Magnetic Nanoparticles, Physikalisch-Technische Bundesanstalt, Abbestraße 2-12, 10587 Berlin, Germany
| | - Raluca M Fratila
- INMA - Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, 50009 Zaragoza, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
| | - Laia Reverte-Salisa
- Institut für Pharmakologie und Toxikologie, Rheinische Friedrich-Wilhelms-Universität Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Thithawat Trakoolwilaiwan
- Biophysics Group, Department of Physics and Astronomy, University College London, London WC1E 6BT, U.K
- UCL Healthcare Biomagnetics Laboratories, 21 Albemarle Street, London W1S 4BS, U.K
| | - Zheming Niu
- Institut für Pharmakologie und Toxikologie, Rheinische Friedrich-Wilhelms-Universität Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Georgios Kasparis
- Biophysics Group, Department of Physics and Astronomy, University College London, London WC1E 6BT, U.K
- UCL Healthcare Biomagnetics Laboratories, 21 Albemarle Street, London W1S 4BS, U.K
| | - Stephanie Franziska Preuss
- Institut für Pharmakologie und Toxikologie, Rheinische Friedrich-Wilhelms-Universität Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Olaf Kosch
- 8.23 Metrology for Magnetic Nanoparticles, Physikalisch-Technische Bundesanstalt, Abbestraße 2-12, 10587 Berlin, Germany
| | - Jesus M de la Fuente
- INMA - Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, 50009 Zaragoza, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
| | - Nguyen Thi Kim Thanh
- Biophysics Group, Department of Physics and Astronomy, University College London, London WC1E 6BT, U.K
- UCL Healthcare Biomagnetics Laboratories, 21 Albemarle Street, London W1S 4BS, U.K
| | - Frank Wiekhorst
- 8.23 Metrology for Magnetic Nanoparticles, Physikalisch-Technische Bundesanstalt, Abbestraße 2-12, 10587 Berlin, Germany
| | - Alexander Pfeifer
- Institut für Pharmakologie und Toxikologie, Rheinische Friedrich-Wilhelms-Universität Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| |
Collapse
|
26
|
Manganese homeostasis at the host-pathogen interface and in the host immune system. Semin Cell Dev Biol 2021; 115:45-53. [PMID: 33419608 DOI: 10.1016/j.semcdb.2020.12.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/17/2020] [Accepted: 12/18/2020] [Indexed: 02/07/2023]
Abstract
Manganese serves as an indispensable catalytic center and the structural core of various enzymes that participate in a plethora of biological processes, including oxidative phosphorylation, glycosylation, and signal transduction. In pathogenic microorganisms, manganese is required for survival by maintaining basic biochemical activity and virulence; in contrast, the host utilizes a process known as nutritional immunity to sequester manganese from invading pathogens. Recent epidemiological and animal studies have shown that manganese increases the immune response in a wide range of vertebrates, including humans, rodents, birds, and fish. On the other hand, excess manganese can cause neurotoxicity and other detrimental effects. Here, we review recent data illustrating the essential role of manganese homeostasis at the host-pathogen interface and in the host immune system. We also discuss the accumulating body of evidence that manganese modulates various signaling pathways in immune processes. Finally, we discuss the key molecular players involved in manganese's immune regulatory function, as well as the clinical implications with respect to cancer immunotherapy.
Collapse
|
27
|
Iron and manganese transport in mammalian systems. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1868:118890. [PMID: 33091506 DOI: 10.1016/j.bbamcr.2020.118890] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/24/2020] [Accepted: 10/08/2020] [Indexed: 12/23/2022]
Abstract
Studies in recent years have significantly expanded, refined, and redefined the repertoire of transporters and other proteins involved in iron and manganese (Mn) transport and homeostasis. In this review, we discuss highlights of the recent literature on iron and Mn transport, focusing on the roles of membrane transporters and related proteins. Studies are considered from the vantage point of main organs, tissues, and cell types that actively control whole-body iron or Mn homeostasis, with emphasis on studies in which in vivo metal transport was measured directly or implicated by using knockout mouse models. Overviews of whole-body and cellular iron and Mn homeostasis are also provided to give physiological context for key transporters and to highlight how they participate in the uptake, intracellular trafficking, and efflux of each metal. Important similarities and differences in iron and Mn transport are noted, and future research opportunities and challenges are identified.
Collapse
|
28
|
Sunuwar L, Frkatović A, Sharapov S, Wang Q, Neu HM, Wu X, Haritunians T, Wan F, Michel S, Wu S, Donowitz M, McGovern D, Lauc G, Sears C, Melia J. Pleiotropic ZIP8 A391T implicates abnormal manganese homeostasis in complex human disease. JCI Insight 2020; 5:140978. [PMID: 32897876 PMCID: PMC7605523 DOI: 10.1172/jci.insight.140978] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 08/26/2020] [Indexed: 12/15/2022] Open
Abstract
ZIP8 is a metal transporter with a role in manganese (Mn) homeostasis. A common genetic variant in ZIP8 (rs13107325; A391T) ranks in the top 10 of pleiotropic SNPs identified in GWAS; A391T has associations with an increased risk of schizophrenia, obesity, Crohn’s disease, and reduced blood Mn. Here, we used CRISPR/Cas9-mediated knockin (KI) to generate a mouse model of ZIP8 A391T (Zip8 393T-KI mice). Recapitulating the SNP association with blood Mn, blood Mn was reduced in Zip8 393T-KI mice. There was restricted abnormal tissue Mn homeostasis, with decreases in liver and kidney Mn and a reciprocal increase in biliary Mn, providing in vivo evidence of hypomorphic Zip8 function. Upon challenge in a chemically induced colitis model, male Zip8 393T-KI mice exhibited enhanced disease susceptibility. ZIP8 391-Thr associated with reduced triantennary plasma N-glycan species in a population-based cohort to define a genotype-specific glycophenotype hypothesized to be linked to Mn-dependent glycosyltransferase activity. This glycophenotype was maintained in a cohort of patients with Crohn’s disease. These data and the pleiotropic disease associations with ZIP8 391-Thr suggest underappreciated roles of Mn homeostasis in complex human disease. Abnormal manganese homeostasis is implicated by a GWAS disease-associated SNP, rs13107325 (ZIP8 A391T), studied in a knockin mouse model and human N-glycome analyses.
Collapse
Affiliation(s)
- Laxmi Sunuwar
- Department of Medicine, Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Sodbo Sharapov
- Laboratory of Glycogenomics, Institute of Cytology and Genetics of Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Qinchuan Wang
- Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Heather M Neu
- University of Maryland School of Pharmacy, University of Maryland, Baltimore, Maryland, USA
| | - Xinqun Wu
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Talin Haritunians
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Fengyi Wan
- Department of Biochemistry and Molecular Biology and.,Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Sarah Michel
- University of Maryland School of Pharmacy, University of Maryland, Baltimore, Maryland, USA
| | - Shaoguang Wu
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Mark Donowitz
- Department of Medicine, Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Dermot McGovern
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Gordan Lauc
- Genos Glycoscience Research Laboratory, Zagreb, Croatia
| | - Cynthia Sears
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Joanna Melia
- Department of Medicine, Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
29
|
Chen X, Yu C, Kang R, Tang D. Iron Metabolism in Ferroptosis. Front Cell Dev Biol 2020; 8:590226. [PMID: 33117818 PMCID: PMC7575751 DOI: 10.3389/fcell.2020.590226] [Citation(s) in RCA: 565] [Impact Index Per Article: 113.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 09/17/2020] [Indexed: 01/12/2023] Open
Abstract
Ferroptosis is a form of regulated cell death that is characterized by iron-dependent oxidative damage and subsequent plasma membrane ruptures and the release of damage-associated molecular patterns. Due to the role of iron in mediating the production of reactive oxygen species and enzyme activity in lipid peroxidation, ferroptosis is strictly controlled by regulators involved in many aspects of iron metabolism, such as iron uptake, storage, utilization, and efflux. Translational and transcriptional regulation of iron homeostasis provide an integrated network to determine the sensitivity of ferroptosis. Impaired ferroptosis is implicated in various iron-related pathological conditions or diseases, such as cancer, neurodegenerative diseases, and ischemia-reperfusion injury. Understanding the molecular mechanisms underlying the regulation of iron metabolism during ferroptosis may provide effective strategies for the treatment of ferroptosis-associated diseases. Indeed, iron chelators effectively prevent the occurrence of ferroptosis, which may provide new approaches for the treatment of iron-related disorders. In this review, we summarize recent advances in the theoretical modeling of iron-dependent ferroptosis, and highlight the therapeutic implications of iron chelators in diseases.
Collapse
Affiliation(s)
- Xin Chen
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, The Third Affiliated Hospital, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China.,Department of Surgery, UT Southwestern Medical Center, Dallas, TX, United States
| | - Chunhua Yu
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, United States
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, United States
| | - Daolin Tang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, The Third Affiliated Hospital, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China.,Department of Surgery, UT Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
30
|
Altamura S, Marques O, Colucci S, Mertens C, Alikhanyan K, Muckenthaler MU. Regulation of iron homeostasis: Lessons from mouse models. Mol Aspects Med 2020; 75:100872. [DOI: 10.1016/j.mam.2020.100872] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 06/28/2020] [Accepted: 07/02/2020] [Indexed: 12/13/2022]
|
31
|
Mealer RG, Jenkins BG, Chen CY, Daly MJ, Ge T, Lehoux S, Marquardt T, Palmer CD, Park JH, Parsons PJ, Sackstein R, Williams SE, Cummings RD, Scolnick EM, Smoller JW. The schizophrenia risk locus in SLC39A8 alters brain metal transport and plasma glycosylation. Sci Rep 2020; 10:13162. [PMID: 32753748 PMCID: PMC7403432 DOI: 10.1038/s41598-020-70108-9] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 07/20/2020] [Indexed: 12/15/2022] Open
Abstract
A common missense variant in SLC39A8 is convincingly associated with schizophrenia and several additional phenotypes. Homozygous loss-of-function mutations in SLC39A8 result in undetectable serum manganese (Mn) and a Congenital Disorder of Glycosylation (CDG) due to the exquisite sensitivity of glycosyltransferases to Mn concentration. Here, we identified several Mn-related changes in human carriers of the common SLC39A8 missense allele. Analysis of structural brain MRI scans showed a dose-dependent change in the ratio of T2w to T1w signal in several regions. Comprehensive trace element analysis confirmed a specific reduction of only serum Mn, and plasma protein N-glycome profiling revealed reduced complexity and branching. N-glycome profiling from two individuals with SLC39A8-CDG showed similar but more severe alterations in branching that improved with Mn supplementation, suggesting that the common variant exists on a spectrum of hypofunction with potential for reversibility. Characterizing the functional impact of this variant will enhance our understanding of schizophrenia pathogenesis and identify novel therapeutic targets and biomarkers.
Collapse
Affiliation(s)
- Robert G Mealer
- Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- The Stanley Center for Psychiatric Research at Broad Institute of Harvard/MIT, Cambridge, MA, USA.
- National Center for Functional Glycomics, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| | - Bruce G Jenkins
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Chia-Yen Chen
- Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- The Stanley Center for Psychiatric Research at Broad Institute of Harvard/MIT, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Mark J Daly
- The Stanley Center for Psychiatric Research at Broad Institute of Harvard/MIT, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Tian Ge
- Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- The Stanley Center for Psychiatric Research at Broad Institute of Harvard/MIT, Cambridge, MA, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Sylvain Lehoux
- National Center for Functional Glycomics, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Thorsten Marquardt
- Klinik und Poliklinik für Kinder- und Jugendmedizin-Allgemeine Pädiatrie, Universitätsklinikum Münster, Münster, Germany
| | - Christopher D Palmer
- Laboratory of Inorganic and Nuclear Chemistry, Wadsworth Center, New York State Department of Health, Albany, NY, USA
- Department of Environmental Health Sciences, School of Public Health, University at Albany, Albany, NY, USA
| | - Julien H Park
- Klinik und Poliklinik für Kinder- und Jugendmedizin-Allgemeine Pädiatrie, Universitätsklinikum Münster, Münster, Germany
| | - Patrick J Parsons
- Laboratory of Inorganic and Nuclear Chemistry, Wadsworth Center, New York State Department of Health, Albany, NY, USA
- Department of Environmental Health Sciences, School of Public Health, University at Albany, Albany, NY, USA
| | - Robert Sackstein
- Department of Translational Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Sarah E Williams
- Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- National Center for Functional Glycomics, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Richard D Cummings
- National Center for Functional Glycomics, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Edward M Scolnick
- The Stanley Center for Psychiatric Research at Broad Institute of Harvard/MIT, Cambridge, MA, USA
| | - Jordan W Smoller
- Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- The Stanley Center for Psychiatric Research at Broad Institute of Harvard/MIT, Cambridge, MA, USA
| |
Collapse
|
32
|
Cross JH, Prentice AM, Cerami C. Hepcidin, Serum Iron, and Transferrin Saturation in Full-Term and Premature Infants during the First Month of Life: A State-of-the-Art Review of Existing Evidence in Humans. Curr Dev Nutr 2020; 4:nzaa104. [PMID: 32793848 PMCID: PMC7413980 DOI: 10.1093/cdn/nzaa104] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 06/04/2020] [Accepted: 06/11/2020] [Indexed: 02/07/2023] Open
Abstract
Neonates regulate iron at birth and in early postnatal life. We reviewed literature from PubMed and Ovid Medline containing data on umbilical cord and venous blood concentrations of hepcidin and iron, and transferrin saturation (TSAT), in human neonates from 0 to 1 mo of age. Data from 59 studies were used to create reference ranges for hepcidin, iron, and TSAT for full-term-birth (FTB) neonates over the first month of life. In FTB neonates, venous hepcidin increases 100% over the first month of life (to reach 61.1 ng/mL; 95% CI: 20.1, 102.0 ng/mL) compared with umbilical cord blood (29.7 ng/mL; 95% CI: 21.1, 38.3 ng/mL). Cord blood has a high concentration of serum iron (28.4 μmol/L; 95% CI: 26.0, 31.1 μmol/L) and levels of TSAT (51.7%; 95% CI: 46.5%, 56.9%). After a short-lived immediate postnatal hypoferremia, iron and TSAT rebounded to approximately half the levels in the cord by the end of the first month. There were insufficient data to formulate reference ranges for preterm neonates.
Collapse
Affiliation(s)
- James H Cross
- Epidemiology and Population Health, Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Fajara, Banjul, The Gambia
| | - Andrew M Prentice
- Epidemiology and Population Health, Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Fajara, Banjul, The Gambia
| | - Carla Cerami
- Epidemiology and Population Health, Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Fajara, Banjul, The Gambia
| |
Collapse
|
33
|
Winslow JW, Limesand KH, Zhao N. The Functions of ZIP8, ZIP14, and ZnT10 in the Regulation of Systemic Manganese Homeostasis. Int J Mol Sci 2020; 21:ijms21093304. [PMID: 32392784 PMCID: PMC7246657 DOI: 10.3390/ijms21093304] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 05/04/2020] [Accepted: 05/05/2020] [Indexed: 12/27/2022] Open
Abstract
As an essential nutrient, manganese is required for the regulation of numerous cellular processes, including cell growth, neuronal health, immune cell function, and antioxidant defense. However, excess manganese in the body is toxic and produces symptoms of neurological and behavioral defects, clinically known as manganism. Therefore, manganese balance needs to be tightly controlled. In the past eight years, mutations of genes encoding metal transporters ZIP8 (SLC39A8), ZIP14 (SLC39A14), and ZnT10 (SLC30A10) have been identified to cause dysregulated manganese homeostasis in humans, highlighting the critical roles of these genes in manganese metabolism. This review focuses on the most recent advances in the understanding of physiological functions of these three identified manganese transporters and summarizes the molecular mechanisms underlying how the loss of functions in these genes leads to impaired manganese homeostasis and human diseases.
Collapse
|
34
|
Pascoal C, Francisco R, Ferro T, Dos Reis Ferreira V, Jaeken J, Videira PA. CDG and immune response: From bedside to bench and back. J Inherit Metab Dis 2020; 43:90-124. [PMID: 31095764 DOI: 10.1002/jimd.12126] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 05/13/2019] [Accepted: 05/15/2019] [Indexed: 12/20/2022]
Abstract
Glycosylation is an essential biological process that adds structural and functional diversity to cells and molecules, participating in physiological processes such as immunity. The immune response is driven and modulated by protein-attached glycans that mediate cell-cell interactions, pathogen recognition and cell activation. Therefore, abnormal glycosylation can be associated with deranged immune responses. Within human diseases presenting immunological defects are congenital disorders of glycosylation (CDG), a family of around 130 rare and complex genetic diseases. In this review, we have identified 23 CDG with immunological involvement, characterized by an increased propensity to-often life-threatening-infection. Inflammatory and autoimmune complications were found in 7 CDG types. CDG natural history(ies) and the mechanisms behind the immunological anomalies are still poorly understood. However, in some cases, alterations in pathogen recognition and intracellular signaling (eg, TGF-β1, NFAT, and NF-κB) have been suggested. Targeted therapies to restore immune defects are only available for PGM3-CDG and SLC35C1-CDG. Fostering research on glycoimmunology may elucidate the involved pathophysiological mechanisms and open new therapeutic avenues, thus improving CDG patients' quality of life.
Collapse
Affiliation(s)
- Carlota Pascoal
- Portuguese Association for CDG, Lisbon, Portugal
- CDG & Allies - Professionals and Patient Associations International Network (CDG & Allies - PPAIN), Caparica, Portugal
- UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Rita Francisco
- Portuguese Association for CDG, Lisbon, Portugal
- CDG & Allies - Professionals and Patient Associations International Network (CDG & Allies - PPAIN), Caparica, Portugal
- UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Tiago Ferro
- CDG & Allies - Professionals and Patient Associations International Network (CDG & Allies - PPAIN), Caparica, Portugal
- UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Vanessa Dos Reis Ferreira
- Portuguese Association for CDG, Lisbon, Portugal
- CDG & Allies - Professionals and Patient Associations International Network (CDG & Allies - PPAIN), Caparica, Portugal
| | - Jaak Jaeken
- CDG & Allies - Professionals and Patient Associations International Network (CDG & Allies - PPAIN), Caparica, Portugal
- Center for Metabolic Diseases, Department of Development and Regeneration, UZ and KU Leuven, Leuven, Belgium
| | - Paula A Videira
- Portuguese Association for CDG, Lisbon, Portugal
- CDG & Allies - Professionals and Patient Associations International Network (CDG & Allies - PPAIN), Caparica, Portugal
- UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| |
Collapse
|
35
|
Nebert DW, Liu Z. SLC39A8 gene encoding a metal ion transporter: discovery and bench to bedside. Hum Genomics 2019; 13:51. [PMID: 31521203 PMCID: PMC6744627 DOI: 10.1186/s40246-019-0233-3] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 08/14/2019] [Indexed: 01/08/2023] Open
Abstract
SLC39A8 is an evolutionarily highly conserved gene that encodes the ZIP8 metal cation transporter in all vertebrates. SLC39A8 is ubiquitously expressed, including pluripotent embryonic stem cells; SLC39A8 expression occurs in every cell type examined. Uptake of ZIP8-mediated Mn2+, Zn2+, Fe2+, Se4+, and Co2+ represents endogenous functions-moving these cations into the cell. By way of mouse genetic differences, the phenotype of "subcutaneous cadmium-induced testicular necrosis" was assigned to the Cdm locus in the 1970s. This led to identification of the mouse Slc39a8 gene, its most closely related Slc39a14 gene, and creation of Slc39a8-overexpressing, Slc39a8(neo/neo) knockdown, and cell type-specific conditional knockout mouse lines; the Slc39a8(-/-) global knockout mouse is early-embryolethal. Slc39a8(neo/neo) hypomorphs die between gestational day 16.5 and postnatal day 1-exhibiting severe anemia, dysregulated hematopoiesis, hypoplastic spleen, dysorganogenesis, stunted growth, and hypomorphic limbs. Not surprisingly, genome-wide association studies subsequently revealed human SLC39A8-deficiency variants exhibiting striking pleiotropy-defects correlated with clinical disorders in virtually every organ, tissue, and cell-type: numerous developmental and congenital disorders, the immune system, cardiovascular system, kidney, lung, liver, coagulation system, central nervous system, musculoskeletal system, eye, and gastrointestinal tract. Traits with which SLC39A8-deficiency variants are currently associated include Mn2+-deficient hypoglycosylation; numerous birth defects; Leigh syndrome-like mitochondrial redox deficiency; decreased serum high-density lipoprotein-cholesterol levels; increased body mass index; greater risk of coronary artery disease, hypotension, cardiovascular death, allergy, ischemic stroke, schizophrenia, Parkinson disease, inflammatory bowel disease, Crohn disease, myopia, and adolescent idiopathic scoliosis; systemic lupus erythematosus with primary Sjögren syndrome; decreased height; and inadvertent participation in the inflammatory progression of osteoarthritis.
Collapse
Affiliation(s)
- Daniel W Nebert
- Department of Environmental Health and Center for Environmental Genetics, University of Cincinnati Medical Center, Cincinnati, OH, 45267-0056, USA.
- Division of Human Genetics, Department of Pediatrics & Molecular Developmental Biology, Cincinnati Children's Hospital, Cincinnati, OH, 45229-2899, USA.
| | - Zijuan Liu
- Department of Biological Sciences, Oakland University, Rochester, MI, 48309, USA
| |
Collapse
|
36
|
Geng X, Liu L, Banes-Berceli A, Yang Z, Kang P, Shen J, Tsai KJ, Liu Z. Role of ZIP8 in regulating cell morphology and NF-κB/Snail2 signaling. Metallomics 2019; 10:953-964. [PMID: 29927450 DOI: 10.1039/c8mt00079d] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
ZIP8 is a recently identified membrane transporter which facilitates uptake of many substrates including both essential and toxic divalent metals (e.g. zinc, manganese, iron, cadmium) and inorganic selenium. Many ZIP8 regulated downstream signals and pathways remain to be elucidated. In this study, we investigated ZIP8 regulatory roles in downstream targets in ZIP8-gain and loss cells and in ZIP8 overexpressed lungs. Our results show that the overexpression of ZIP8 in mouse fibroblast cells (MEF) induces significant morphological change and re-organization of filament actin (F-actin), along with increased cell proliferation and migration rate. In ZIP8 knockout chronic myelogenous leukemia HAP1 cells, significant clonal morphological change with increased cell-cell adhesion was observed. In the ZIP8 overexpressed lung, F-actin was aberrantly enriched around the tracheal branch. In these ZIP8 gain and loss cell lines and ZIP8 transgenic lungs, we identified two relevant transcription factors, NF-κB and Snail2, whose activation is dependent on the ZIP8 level. They were both significantly upregulated in ZIP8 overexpressed cells and lungs. Expression of NF-κB and Snail2 targets, COL1A2 and E-cadherin, was also correspondingly elevated. Taken together, our results suggest that ZIP8 is a new regulator for cell morphology and cytoskeleton which involves NF-κB and Snail2.
Collapse
Affiliation(s)
- Xiangrong Geng
- Department of Biological Sciences, Oakland University, 2200 N. Squirrel Rd, Rochester, MI 48309, USA.
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Gordon SJV, Fenker DE, Vest KE, Padilla-Benavides T. Manganese influx and expression of ZIP8 is essential in primary myoblasts and contributes to activation of SOD2. Metallomics 2019; 11:1140-1153. [PMID: 31086870 PMCID: PMC6584035 DOI: 10.1039/c8mt00348c] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Trace elements such as copper (Cu), zinc (Zn), iron (Fe), and manganese (Mn) function as enzyme cofactors and second messengers in cell signaling. Trace elements are emerging as key regulators of differentiation and development of mammalian tissues including blood, brain, and skeletal muscle. We previously reported an influx of Cu and dynamic expression of metal transporters during differentiation of skeletal muscle cells. Here, we demonstrate that during differentiation of skeletal myoblasts an increase of Mn, Fe and Zn also occurs. Interestingly the Mn increase is concomitant with increased Mn-dependent SOD2 levels. To better understand the Mn import pathway in skeletal muscle cells, we probed the functional relevance of the closely related proteins ZIP8 and ZIP14, which are implicated in Zn, Mn, and Fe transport. Partial depletion of ZIP8 severely impaired growth of myoblasts and led to cell death under differentiation conditions, indicating that ZIP8-mediated metal transport is essential in skeletal muscle cells. Moreover, knockdown of Zip8 impaired activity of the Mn-dependent SOD2. Growth defects were partially rescued only by Mn supplementation to the medium, suggesting additional functions for ZIP8 in the skeletal muscle lineage. Restoring wild type Zip8 into the knockdown cells rescued the proliferation and differentiation phenotypes. On the other hand, knockdown of Zip14, had only a mild effect on myotube size, consistent with a role for ZIP14 in muscle hypertrophy. Simultaneous knockdown of both Zip8 and Zip14 further impaired differentiation and led cell death. This is the first report on the functional relevance of two members of the ZIP family of metal transporters in the skeletal muscle lineage, and further supports the paradigm that trace metal transporters are important modulators of mammalian tissue development.
Collapse
Affiliation(s)
- Shellaina J. V. Gordon
- Department of Biochemistry and Molecular Pharmacology,
University of Massachusetts Medical School, 394 Plantation St., Worcester, MA,
01605, USA
| | - Daniel E. Fenker
- Department of Molecular Genetics, Biochemistry &
Microbiology, University of Cincinnati School of Medicine, 231 Albert Sabin Way,
Cincinnati, OH, 45267, USA
| | - Katherine E. Vest
- Department of Molecular Genetics, Biochemistry &
Microbiology, University of Cincinnati School of Medicine, 231 Albert Sabin Way,
Cincinnati, OH, 45267, USA
| | - Teresita Padilla-Benavides
- Department of Biochemistry and Molecular Pharmacology,
University of Massachusetts Medical School, 394 Plantation St., Worcester, MA,
01605, USA
| |
Collapse
|
38
|
Generation and Molecular Characterization of Human Ring Sideroblasts: a Key Role of Ferrous Iron in Terminal Erythroid Differentiation and Ring Sideroblast Formation. Mol Cell Biol 2019; 39:MCB.00387-18. [PMID: 30670569 DOI: 10.1128/mcb.00387-18] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 01/16/2019] [Indexed: 12/20/2022] Open
Abstract
Ring sideroblasts are a hallmark of sideroblastic anemia, although little is known about their characteristics. Here, we first generated mutant mice by disrupting the GATA-1 binding motif at the intron 1 enhancer of the ALAS2 gene, a gene responsible for X-linked sideroblastic anemia (XLSA). Although heterozygous female mice showed an anemic phenotype, ring sideroblasts were not observed in their bone marrow. We next established human induced pluripotent stem cell-derived proerythroblast clones harboring the same ALAS2 gene mutation. Through coculture with sodium ferrous citrate, mutant clones differentiated into mature erythroblasts and became ring sideroblasts with upregulation of metal transporters (MFRN1, ZIP8, and DMT1), suggesting a key role for ferrous iron in erythroid differentiation. Interestingly, holo-transferrin (holo-Tf) did not induce erythroid differentiation as well as ring sideroblast formation, and mutant cells underwent apoptosis. Despite massive iron granule content, ring sideroblasts were less apoptotic than holo-Tf-treated undifferentiated cells. Microarray analysis revealed upregulation of antiapoptotic genes in ring sideroblasts, a profile partly shared with erythroblasts from a patient with XLSA. These results suggest that ring sideroblasts exert a reaction to avoid cell death by activating antiapoptotic programs. Our model may become an important tool to clarify the pathophysiology of sideroblastic anemia.
Collapse
|
39
|
Abstract
Most cells in the body acquire iron via receptor-mediated endocytosis of transferrin, the circulating iron transport protein. When cellular iron levels are sufficient, the uptake of transferrin decreases to limit further iron assimilation and prevent excessive iron accumulation. In iron overload conditions, such as hereditary hemochromatosis and thalassemia major, unregulated iron entry into the plasma overwhelms the carrying capacity of transferrin, resulting in non-transferrin-bound iron (NTBI), a redox-active, potentially toxic form of iron. Plasma NTBI is rapidly cleared from the circulation primarily by the liver and other organs (e.g., pancreas, heart, and pituitary) where it contributes significantly to tissue iron overload and related pathology. While NTBI is usually not detectable in the plasma of healthy individuals, it does appear to be a normal constituent of brain interstitial fluid and therefore likely serves as an important source of iron for most cell types in the CNS. A growing body of literature indicates that NTBI uptake is mediated by non-transferrin-bound iron transporters such as ZIP14, L-type and T-type calcium channels, DMT1, ZIP8, and TRPC6. This review provides an overview of NTBI uptake by various tissues and cells and summarizes the evidence for and against the roles of individual transporters in this process.
Collapse
Affiliation(s)
- Mitchell D Knutson
- Food Science and Human Nutrition Department, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
40
|
Sangkhae V, Nemeth E. Placental iron transport: The mechanism and regulatory circuits. Free Radic Biol Med 2019; 133:254-261. [PMID: 29981833 PMCID: PMC7059975 DOI: 10.1016/j.freeradbiomed.2018.07.001] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 06/30/2018] [Accepted: 07/03/2018] [Indexed: 02/06/2023]
Abstract
As the interface between the fetal and maternal circulation, the placenta facilitates both nutrient and waste exchange for the developing fetus. Iron is essential for healthy pregnancy, and transport of iron across the placenta is required for fetal growth and development. Perturbation of this transfer can lead to adverse pregnancy outcomes. Despite its importance, our understanding of how a large amount of iron is transported across placental membranes, how this process is regulated, and which iron transporter proteins function in different placental cells remains rudimentary. Mechanistic studies in mouse models, including placenta-specific deletion or overexpression of iron-related proteins will be essential to make progress. This review summarizes our current understanding about iron transport across the syncytiotrophoblast under physiological conditions and identifies areas for further investigation.
Collapse
Affiliation(s)
- Veena Sangkhae
- Center for Iron Disorders, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, 10833 LeConte Ave, CHS 37-131, Los Angeles, CA 90095, USA.
| | - Elizabeta Nemeth
- Center for Iron Disorders, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, 10833 LeConte Ave, CHS 37-131, Los Angeles, CA 90095, USA.
| |
Collapse
|
41
|
Xu W, Wu H, Shang L. Identification of novel candidate indicators for assessing zinc status during pregnancy in mice from microarray data. BMC Pharmacol Toxicol 2019; 20:12. [PMID: 30770772 PMCID: PMC6377758 DOI: 10.1186/s40360-019-0288-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Accepted: 01/29/2019] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND This study aimed to identify potential zinc status indicators and to clarify the mechanisms underlying zinc deficiency-induced organ damage and mortality in mice. METHODS The dataset GSE97112, including placental tissues of mice fed diets containing normal and low concentrations of zinc, was downloaded and preprocessed. Differentially expressed genes (DEGs) were calculated and identified for zinc deficiency-related gene clusters by using the weighed gene co-expression network analysis (WGCNA) algorithm. The Gene Ontology (GO)-Biological Process (BP) and KEGG pathway of genes in the zinc deficiency-related WGCNA modules were analyzed, and the protein-protein interaction (PPI) network was constructed. In addition, modules of the PPI network were identified, and transcription factors (TFs) and miRNAs regulating DEGs were predicted. Finally, drug-gene interactions were selected. RESULTS A total of 1055 DEGs containing 586 up- and 469 down-regulated genes were obtained. Three modules based on WGCNA had high correlation with degree of zinc deficiency. Annexin A1 (ANXA1), C-C motif chemokine receptor 3 (CCR3), C-X-C motif chemokine receptor 2 (CXCR2), and interleukin 2 (IL-2) were hub nodes in the PPI network. Three modules in the PPI network were identified, including module 1 associated with olfactory conduction and module 2 associated with inflammatory response. ANXA1, CCR3, and IL-2 were regulated by TFs. In addition, CXCR2, ANXA, and IL-2 were drug targets. CONCLUSION CXCR2, ANXA1, and CCR3 as well as olfactory receptor-related genes (proteins) may be used as biomarkers to assess zinc status in mice.
Collapse
Affiliation(s)
- Wan Xu
- The Fourth Military Medical University, Xi'an, Shaanxi Province, 710032, China
- Department of Obstetrics and Gynecology, PLA Army General Hospital, No.5 Nanmencang, Dongcheng District, Beijing, 100700, China
| | - Hongyan Wu
- Department of Obstetrics and Gynecology, PLA Army General Hospital, No.5 Nanmencang, Dongcheng District, Beijing, 100700, China
| | - Lixin Shang
- Department of Obstetrics and Gynecology, PLA Army General Hospital, No.5 Nanmencang, Dongcheng District, Beijing, 100700, China.
| |
Collapse
|
42
|
Using phenome-wide association to investigate the function of a schizophrenia risk locus at SLC39A8. Transl Psychiatry 2019; 9:45. [PMID: 30696806 PMCID: PMC6351652 DOI: 10.1038/s41398-019-0386-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 11/08/2018] [Accepted: 11/13/2018] [Indexed: 12/18/2022] Open
Abstract
While nearly all common genomic variants associated with schizophrenia have no known function, one corresponds to a missense variant associated with change in efficiency of a metal ion transporter, ZIP8, coded by SLC39A8. This variant has been linked to a range of phenotypes and is believed to be under recent selection pressure, but its impact on health is poorly understood. We sought to understand phenotypic implications of this variant in a large genomic biobank using an unbiased phenome-wide approach. Specifically, we generated 50 topics based on diagnostic codes using latent Dirichlet allocation, and examined them for association with the risk variant. Then, any significant topics were further characterized by examining association with individual diagnostic codes contributing to the topic. Among 50 topics, 1 was associated at an experiment-wide significance threshold (beta = 0.003, uncorrected p = 0.00049), comprising predominantly brain-related codes, including intracranial hemorrhage, cerebrovascular disease, and delirium/dementia. These results suggest that a functional variant previously associated with schizophrenia risk also increases liability to cerebrovascular disease. They further illustrate the utility of a topic-based approach to phenome-wide association.
Collapse
|
43
|
Iron Homeostasis in the Lungs-A Balance between Health and Disease. Pharmaceuticals (Basel) 2019; 12:ph12010005. [PMID: 30609678 PMCID: PMC6469191 DOI: 10.3390/ph12010005] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 12/23/2018] [Accepted: 12/25/2018] [Indexed: 12/15/2022] Open
Abstract
A strong mechanistic link between the regulation of iron homeostasis and oxygen sensing is evident in the lung, where both systems must be properly controlled to maintain lung function. Imbalances in pulmonary iron homeostasis are frequently associated with respiratory diseases, such as chronic obstructive pulmonary disease and with lung cancer. However, the underlying mechanisms causing alterations in iron levels and the involvement of iron in the development of lung disorders are incompletely understood. Here, we review current knowledge about the regulation of pulmonary iron homeostasis, its functional importance, and the link between dysregulated iron levels and lung diseases. Gaining greater knowledge on how iron contributes to the pathogenesis of these diseases holds promise for future iron-related therapeutic strategies.
Collapse
|
44
|
Role of ZIP8 in regulation of cisplatin sensitivity through Bcl-2. Toxicol Appl Pharmacol 2018; 362:52-58. [PMID: 30342059 DOI: 10.1016/j.taap.2018.10.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 09/12/2018] [Accepted: 10/16/2018] [Indexed: 02/06/2023]
Abstract
ZIP8 is a membrane transporter that facilitates the uptake of divalent metals (e.g., Zn, Mn, Fe, Cd) and the mineral selenite in anionic form. ZIP8 functionality has been recently reported to regulate cell proliferation, migration and cytoskeleton arrangement, exhibiting an essential role for normal physiology. In this study, we report a ZIP8 role in chemotherapy response. We show ZIP8 regulates cell sensitivity to the anti-cancer drug cisplatin. Overexpression of ZIP8 in mouse embryonic fibroblast (MEF) cells induces cisplatin sensitivity, while knockout of ZIP8 in leukemia HAP1 cells leads to cisplatin resistance. In ZIP8 altered cells and transgenic mice, we show cisplatin is not a direct ZIP8 substrate. Further studies demonstrate that ZIP8 regulates anti-apoptotic protein Bcl-2. ZIP8 overexpression decreases Bcl-2 levels in cultured cells, mice lung and liver tissue while loss of ZIP8 elevates Bcl-2 expression in HAP1 cells and liver tissue. We also observe that ZIP8 overexpression modulates cisplatin-induced cell apoptosis, manifested by the increased protein level of cleaved Caspase-3. Since Bcl-2 elevation was previously discovered to induce cisplatin drug resistance, our results suggest ZIP8 may modulate cisplatin drug responses as well as apoptosis through Bcl-2. We therefore conclude ZIP8 is a new molecule to be involved in cisplatin drug responses and is predicted as a genetic factor to be considered in cisplatin therapy.
Collapse
|
45
|
Essential Role of Zinc and Zinc Transporters in Myeloid Cell Function and Host Defense against Infection. J Immunol Res 2018; 2018:4315140. [PMID: 30417019 PMCID: PMC6207864 DOI: 10.1155/2018/4315140] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 09/27/2018] [Indexed: 12/19/2022] Open
Abstract
Zinc is an essential micronutrient known to play a vital role in host defense against pathogens. Diets that are deficient in zinc lead to impaired immunity and delayed recovery from and worse outcomes following infection. Sustained insufficient zinc intake leads to dysregulation of the innate immune response and increases susceptibility to infection whereas zinc supplementation in at-risk populations has been shown to restore host defense and reduce pathogen-related morbidity and mortality. Upon infection, zinc deficiency leads to increased pathology due to imbalance in key signaling networks that result in excessive inflammation and collateral tissue damage. In particular, zinc impacts macrophage function, a critical front-line cell in host defense, in addition to other immune cells. Deficits in zinc adversely impact macrophage function resulting in dysregulation of phagocytosis, intracellular killing, and cytokine production. An additional work in this field has revealed a vital role for several zinc transporter proteins that are required for proper bioredistribution of zinc within mononuclear cells to achieve an optimal immune response against invading microorganisms. In this review, we will discuss the most recent developments regarding zinc's role in innate immunity and protection against pathogen invasion.
Collapse
|
46
|
Haller G, McCall K, Jenkitkasemwong S, Sadler B, Antunes L, Nikolov M, Whittle J, Upshaw Z, Shin J, Baschal E, Cruchaga C, Harms M, Raggio C, Morcuende JA, Giampietro P, Miller NH, Wise C, Gray RS, Solnica-Krezel L, Knutson M, Dobbs MB, Gurnett CA. A missense variant in SLC39A8 is associated with severe idiopathic scoliosis. Nat Commun 2018; 9:4171. [PMID: 30301978 PMCID: PMC6177404 DOI: 10.1038/s41467-018-06705-0] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 08/30/2018] [Indexed: 01/09/2023] Open
Abstract
Genetic factors predictive of severe adolescent idiopathic scoliosis (AIS) are largely unknown. To identify genetic variation associated with severe AIS, we performed an exome-wide association study of 457 severe AIS cases and 987 controls. We find a missense SNP in SLC39A8 (p.Ala391Thr, rs13107325) associated with severe AIS (P = 1.60 × 10-7, OR = 2.01, CI = 1.54-2.62). This pleiotropic SNP was previously associated with BMI, blood pressure, cholesterol, and blood manganese level. We replicate the association in a second cohort (841 cases and 1095 controls) resulting in a combined P = 7.02 × 10-14, OR = 1.94, CI = 1.63-2.34. Clinically, the minor allele of rs13107325 is associated with greater spinal curvature, decreased height, increased BMI and lower plasma manganese in our AIS cohort. Functional studies demonstrate reduced manganese influx mediated by the SLC39A8 p.Ala391Thr variant and vertebral abnormalities, impaired growth, and decreased motor activity in slc39a8 mutant zebrafish. Our results suggest the possibility that scoliosis may be amenable to dietary intervention.
Collapse
Affiliation(s)
- Gabe Haller
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO, USA
| | - Kevin McCall
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO, USA
| | - Supak Jenkitkasemwong
- Food Science and Human Nutrition Department, University of Florida, Gainesville, FL, USA
| | - Brooke Sadler
- Department of Neurology, Washington University, St. Louis, MO, USA
| | - Lilian Antunes
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO, USA
| | - Momchil Nikolov
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO, USA
| | - Julia Whittle
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO, USA
| | - Zachary Upshaw
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO, USA
| | - Jimann Shin
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Erin Baschal
- Department of Orthopaedic Surgery, University of Colorado, Denver, CO, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University, St. Louis, MO, USA
| | - Matthew Harms
- Department of Neurology, Columbia University, New York, NY, USA
| | | | - Jose A Morcuende
- Department of Orthopaedics and Rehabilitation, University of Iowa, Iowa City, IA, USA
| | | | - Nancy H Miller
- Department of Orthopaedic Surgery, University of Colorado, Denver, CO, USA
| | - Carol Wise
- Sarah M. and Charles E. Seay Center for Musculoskeletal Research, Texas Scottish Rite Hospital for Children, Dallas, TX, USA
- Departments of Orthopaedic Surgery, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
- McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - Ryan S Gray
- Department of Pediatrics, Dell Pediatric Research Institute, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | - Lila Solnica-Krezel
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Mitchell Knutson
- Food Science and Human Nutrition Department, University of Florida, Gainesville, FL, USA
| | - Matthew B Dobbs
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO, USA
- Shriners Hospital for Children, St. Louis, MO, USA
| | - Christina A Gurnett
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO, USA.
- Department of Neurology, Washington University, St. Louis, MO, USA.
- Department of Pediatrics, Washington University, St. Louis, MO, USA.
| |
Collapse
|
47
|
Liu L, Geng X, Cai Y, Copple B, Yoshinaga M, Shen J, Nebert DW, Wang H, Liu Z. Hepatic ZIP8 deficiency is associated with disrupted selenium homeostasis, liver pathology, and tumor formation. Am J Physiol Gastrointest Liver Physiol 2018; 315:G569-G579. [PMID: 29927321 PMCID: PMC6230695 DOI: 10.1152/ajpgi.00165.2018] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 06/06/2018] [Accepted: 06/06/2018] [Indexed: 01/31/2023]
Abstract
Zrt/Irt-like protein 8 (ZIP8) (encoded by Slc39a8) is a multifunctional membrane transporter that influxes essential metal cations Zn2+, Mn2+, Fe2+, and nonmetal inorganic selenite (HSeO3-). Physiological roles of ZIP8 in different cell types and tissues remain to be elucidated. We aimed to investigate ZIP8 functions in liver. Two mouse models were used in this study: 1) 13- to 21-mo-old Slc39a8(+/neo) hypomorphs having diminished ZIP8 levels and 2) a liver-specific ZIP8 acute knockdown mouse (Ad-shZip8). Histology, immunohistochemistry, and Western blotting were used to investigate ZIP8-deficiency effects on hepatic injury, inflammatory changes, and oxidative stress. Selenium (Se) and zinc (Zn) were quantified in tissues by inductively coupled plasma-mass spectrophotometry. We found that ZIP8 is required to maintain normal liver function; moderate or acute decreases in ZIP8 activity resulted in hepatic pathology. Spontaneous liver neoplastic nodules appeared in ~50% of Slc39a8(+/neo) between 13 and 21 mo of age, exhibiting features of inflammation, fibrosis, and liver injury. In Ad-shZip8 mice, significant hepatomegaly was observed; histology showed ZIP8 deficiency was associated with hepatocyte injury, inflammation, and proliferation. Significant decreases in Se, but not Zn, were found in Ad-shZip8 liver. Consistent with this Se deficit, liver expression of selenoproteins glutathione peroxidases 1 and 2 was downregulated, along with decreases in antioxidant superoxide dismutases 1 and 2, consistent with increased oxidative stress. Thus, ZIP8 plays an important role in maintaining normal hepatic function, likely through regulating Se homeostasis and redox balance. Hepatic ZIP8 deficiency is associated with liver pathology, including oxidative stress, inflammation, proliferation, and hepatocellular injury. NEW & NOTEWORTHY Zrt/Irt-like protein 8 (ZIP8) is a multifunctional membrane transporter that facilitates biometal and mineral uptake. The role of ZIP8 in liver physiology has not been previously investigated. Liu et al. discovered unique ZIP8 functions, i.e., regulation of hepatic selenium content and association of ZIP8 deficiency in mouse liver with liver defects.
Collapse
Affiliation(s)
- Liu Liu
- Department of Biological Sciences, Oakland University , Rochester, Minnesota
| | - Xiangrong Geng
- Department of Biological Sciences, Oakland University , Rochester, Minnesota
| | - Yihong Cai
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Institute for Liver Disease of Anhui Medical University , Hefei, Anhui , People's Republic of China
| | - Bryan Copple
- Department of Pharmacology and Toxicology, Michigan State University , East Lansing, Michigan
| | - Masafumi Yoshinaga
- Department of Cellular Biology and Pharmacology, Herbert Wertheim College of Medicine, Florida International University , Miami, Florida
| | - Jian Shen
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center , Rochester, New York
| | - Daniel W Nebert
- Department of Environmental Health and Center for Environmental Genetics, University of Cincinnati Medical Center , Cincinnati, Ohio
| | - Hua Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Institute for Liver Disease of Anhui Medical University , Hefei, Anhui , People's Republic of China
| | - Zijuan Liu
- Department of Biological Sciences, Oakland University , Rochester, Minnesota
| |
Collapse
|
48
|
Tanimura N, Liao R, Wilson GM, Dent MR, Cao M, Burstyn JN, Hematti P, Liu X, Zhang Y, Zheng Y, Keles S, Xu J, Coon JJ, Bresnick EH. GATA/Heme Multi-omics Reveals a Trace Metal-Dependent Cellular Differentiation Mechanism. Dev Cell 2018; 46:581-594.e4. [PMID: 30122630 DOI: 10.1016/j.devcel.2018.07.022] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 06/01/2018] [Accepted: 07/19/2018] [Indexed: 01/27/2023]
Abstract
By functioning as an enzyme cofactor, hemoglobin component, and gene regulator, heme is vital for life. One mode of heme-regulated transcription involves amplifying the activity of GATA-1, a key determinant of erythrocyte differentiation. To discover biological consequences of the metal cofactor-transcription factor mechanism, we merged GATA-1/heme-regulated sectors of the proteome and transcriptome. This multi-omic analysis revealed a GATA-1/heme circuit involving hemoglobin subunits, ubiquitination components, and proteins not implicated in erythrocyte biology, including the zinc exporter Slc30a1. Though GATA-1 induced expression of Slc30a1 and the zinc importer Slc39a8, Slc39a8 dominantly increased intracellular zinc, which conferred erythroblast survival. Subsequently, a zinc transporter switch, involving decreased importer and sustained exporter expression, reduced intracellular zinc during terminal differentiation. Downregulating Slc30a1 increased intracellular zinc and, strikingly, accelerated differentiation. This analysis established a conserved paradigm in which a GATA-1/heme circuit controls trace metal transport machinery and trace metal levels as a mechanism governing cellular differentiation.
Collapse
Affiliation(s)
- Nobuyuki Tanimura
- UW-Madison Blood Research Program, Department of Cell and Regenerative Biology, Wisconsin Institutes for Medical Research, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; UW Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Ruiqi Liao
- UW-Madison Blood Research Program, Department of Cell and Regenerative Biology, Wisconsin Institutes for Medical Research, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; UW Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Gary M Wilson
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Matthew R Dent
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Miao Cao
- UW-Madison Blood Research Program, Department of Cell and Regenerative Biology, Wisconsin Institutes for Medical Research, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; UW Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Judith N Burstyn
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Peiman Hematti
- UW Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Xin Liu
- Children's Medical Center Research Institute, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Yuannyu Zhang
- Children's Medical Center Research Institute, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ye Zheng
- Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53706, USA
| | - Sunduz Keles
- Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53706, USA
| | - Jian Xu
- Children's Medical Center Research Institute, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Joshua J Coon
- Department of Biomolecular Chemistry, University of Wisconsin School of Medicine, Madison, WI 53706, USA; Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; Morgridge Institute for Research, Madison, WI 53715, USA; Genome Center of Wisconsin, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Emery H Bresnick
- UW-Madison Blood Research Program, Department of Cell and Regenerative Biology, Wisconsin Institutes for Medical Research, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; UW Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA.
| |
Collapse
|
49
|
Cheng G, Chang FJ, You PH, Lin J, Huang XY, Wu HY, Yan L, Deng JZ, You HJ, Sun CF. ZIP8 induces monocyte adhesion to the aortas ex-vivo by regulating zinc influx. Int Immunopharmacol 2018; 62:203-211. [PMID: 30015240 DOI: 10.1016/j.intimp.2018.06.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 06/08/2018] [Accepted: 06/12/2018] [Indexed: 01/11/2023]
Abstract
Monocytes recruited and adhering to the inflamed arteries are crucial for atherosclerosis development. Here, we report the role of zinc (Zn2+) homeostasis in monocyte adhesion and recruitment. By comparing the expression levels of Zn2+ transporters between non-adhering and adhering monocytes, we found that the Zn2+ importer ZIP8 was specifically upregulated in monocytes adhering to the aortas ex-vivo. Although the overexpression of ZIP8 increased the absorption of Zn2+, Fe2+ and Cd2+ in monocytes, only Zn2+ supplementation was demonstrated capable of promoting the adhesion of monocytes to endothelial monolayers in vitro. In addition, we confirmed the role of ZIP8-dependent Zn2+ influx in promoting monocyte adhesion to the aortas ex-vivo. More importantly, the enforced expression of ZIP8 increased monocyte adhesion and recruitment to the nascent atherosclerotic lesions in ApoE-/- mice. Overall, our results suggest that the Zn2+ influx in monocytes regulated by ZIP8 is a novel factor determining their adhesion and recruitment to atherosclerotic lesions, and that targeting ZIP8 or Zn2+ homeostasis may represent a novel strategy to interfere these activities.
Collapse
Affiliation(s)
- Gong Cheng
- Department of Cardiology, The First Affiliated Hospital of Xi'an Jiaotong University, China
| | - Feng-Jun Chang
- Department of Cardiology, Shaanxi Provincial People's Hospital, China
| | - Peng-Hua You
- Department of Cardiology, Shaanxi Provincial People's Hospital, China
| | - Jing Lin
- Department of Cardiology, Shaanxi Provincial People's Hospital, China
| | - Xiao-Yan Huang
- Department of Central Laboratory, Shaanxi Provincial People's Hospital, China
| | - Hao-Yu Wu
- Department of Cardiology, Shaanxi Provincial People's Hospital, China
| | - Li Yan
- Department of Cardiology, Shaanxi Provincial People's Hospital, China
| | - Ji-Zhao Deng
- Department of Cardiology, Shaanxi Provincial People's Hospital, China
| | - Hong-Jun You
- Department of Cardiology, Shaanxi Provincial People's Hospital, China
| | - Chao-Feng Sun
- Department of Cardiology, The First Affiliated Hospital of Xi'an Jiaotong University, China.
| |
Collapse
|
50
|
Chen J, Gálvez-Peralta M, Zhang X, Deng J, Liu Z, Nebert DW. In utero gene expression in the Slc39a8(neo/neo) knockdown mouse. Sci Rep 2018; 8:10703. [PMID: 30013175 PMCID: PMC6048144 DOI: 10.1038/s41598-018-29109-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 07/05/2018] [Indexed: 12/13/2022] Open
Abstract
Slc39a8 encodes ZIP8, a divalent cation/bicarbonate symporter expressed in pluripotent mouse embryonic stem cells, and therefore ubiquitous in adult tissues; ZIP8 influxes Zn2+, Mn2+ and Fe2+. Slc39a8(neo/neo) knockdown mice exhibit 10-15% of wild-type ZIP8 mRNA and protein levels, and show pleiotropic phenotype of stunted growth, neonatal lethality, multi-organ dysmorphogenesis, and dysregulated hematopoiesis manifested as severe anemia. Herein we performed RNA-seq analysis of gestational day (GD)13.5 yolk sac and placenta, and GD16.5 liver, kidney, lung, heart and cerebellum, comparing Slc39a8(neo/neo) with Slc39a8(+/+) wild-type. Meta-data analysis of differentially-expressed genes revealed 29 unique genes from all tissues - having enriched GO categories associated with hematopoiesis and hypoxia and KEGG categories of complement, response to infection, and coagulation cascade - consistent with dysregulated hematopoietic stem cell fate. Based on transcription factor (TF) profiles in the JASPAR database, and searching for TF-binding sites enriched by Pscan, we identified numerous genes encoding zinc-finger and other TFs associated with hematopoietic stem cell functions. We conclude that, in this mouse model, deficient ZIP8-mediated divalent cation transport affects zinc-finger (e.g. GATA proteins) and other TFs interacting with GATA proteins (e.g. TAL1), predominantly in yolk sac. These data strongly support the phenotype of dysmorphogenesis and anemia seen in Slc39a8(neo/neo) mice in utero.
Collapse
Affiliation(s)
- Jing Chen
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229, USA
| | - Marina Gálvez-Peralta
- Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, Ohio, 45267, USA.,Department of Pharmaceutical Sciences, West Virginia University Medical Center, Morgantown, WV, 26506, USA
| | - Xiang Zhang
- Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, Ohio, 45267, USA
| | - Jingyuan Deng
- Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, Ohio, 45267, USA.,Amazon.com, Inc., Seattle, WA, 98101, USA
| | - Zijuan Liu
- Department of Biological Sciences, Oakland University, Rochester, MI, 48309, USA
| | - Daniel W Nebert
- Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, Ohio, 45267, USA.
| |
Collapse
|