Zinc transporter ZIP12

Last updated
SLC39A12
Identifiers
Aliases SLC39A12 , LZT-Hs8, ZIP-12, bA570F3.1, solute carrier family 39 member 12, ZIP12
External IDs OMIM: 608734 MGI: 2139274 HomoloGene: 17654 GeneCards: SLC39A12
Orthologs
SpeciesHumanMouse
Entrez
Ensembl
UniProt
RefSeq (mRNA)

NM_152725
NM_001145195
NM_001282733
NM_001282734

NM_001012305

RefSeq (protein)

NP_001138667
NP_001269662
NP_001269663
NP_689938

NP_001012305

Location (UCSC)n/a Chr 2: 14.39 – 14.5 Mb
PubMed search [2] [3]
Wikidata
View/Edit Human View/Edit Mouse

Solute carrier family 39 member 12 is a protein that in humans is encoded by the SLC39A12 gene. [4]

Contents

Function

Zinc is an essential cofactor for hundreds of enzymes. It is involved in protein, nucleic acid, carbohydrate, and lipid metabolism, as well as in the control of gene transcription, growth, development, and differentiation. ZIP12 belongs to a subfamily of proteins that show structural characteristics of zinc transporters. [5]

Basic properties

Zinc transporter ZIP12 is a protein that is encoded by the solute carrier 39 member 12 (SLC39A12) gene. [5] [6] ZIP12 is part of a family of Zrt-like, IRT-like proteins (ZIPs) that transport metals. ZIP12 is most closely related to a similar transporter, ZIP4, which is mutated in the genetic disorder acrodermatitis enteropathica. [7] [8] Human ZIP12 shares 31 percent of its amino acids with human ZIP4 between their conserved regions. [9] There are two main splice variants of ZIP12 in humans, which are 691 and 654 amino acids long. [9] The difference in the lengths of these 2 variants of ZIP12 are due to the inclusion or exclusion of an in-frame exon. [9]

The ZIP12 protein contains many elements that are conserved across other ZIP transporters in vertebrates (including mammals and humans). [9] ZIP12 has eight transmembrane domains and contains histidine residues within transmembrane regions four and five that are believed to be necessary for zinc transport across cellular membranes. [5] [6] [9] ZIP12 is present at the plasma membrane and can transport zinc ions from the outside of the cell to the inside. [10] [11]

The SLC39A12 gene is conserved across vertebrates, including humans, non-human primates like rhesus monkeys, cats, dogs, rodents including rats and mice, birds such as chickens, and frogs such as Xenopus laevis and Xenopus tropicalis. [9] The SLC39A12 gene is present in some fish such as Japanese medaka, Nile tilapia, and European seabass, but the SLC39A12 gene is not present in zebrafish. [9] ZIP12 has been shown to transport zinc, and there is currently no evidence that ZIP12 can transport metals other than zinc. ZIP12 is expressed in many tissues and is particularly high in the brain and eye. [9] [10] In mice, ZIP12 mRNA is not detected in pancreas. [10]

Role in neurite extension and mitochondria in mouse neural cells

In mouse Neuro-2a cells and primary mouse neurons, ZIP12 is necessary for neurite extension. [10] Neurites are projections from the cell body of a neural cell during differentiation, and neurites can refer to either axons or dendrites. To study how ZIP12 is important for a neural cell to extend neurites out from the cell body, researchers used short hairpin RNA (shRNA) to induce RNA interference to degrade ZIP12 mRNA and reduce ZIP12 protein. [10] In Neuro-2a cells and primary mouse neurons transfected with shRNA specifically targeting ZIP12, the neural cells have shorter neurites. [10] Increasing intracellular zinc with a zinc ionophore that can cross the cellular membrane while bypassing ZIP12 can restore neurite extension in cells with targeted ZIP12 depletion. [10]

In a subsequent study, Neuro-2a cells with targeted ZIP12 mutations using CRISPR-mediated genome editing also have shorter neurites during differentiation and mitochondrial dysfunction. [12] In addition, ZIP12-deleted cells have reduced cellular respiration, [12] which is a measure of mitochondrial function. Neurite extension of Neuro-2a is more affected by rotenone and sodium azide, [12] which are inhibitors of the electron transport chain of the mitochondria, in cells without ZIP12. ZIP12-deleted cells also have increased superoxide generation and higher oxidative damage, [12] which are consistent with impaired mitochondrial function. Exposing ZIP12-deleted cells to antioxidants such as alpha-tocopherol (vitamin E), MitoQ, or MitoTEMPO can restore neurite length, which indicates that the oxidative damage present in cells without ZIP12 leads to stunted neurites. [12]

Role in early nervous system development of Xenopus tropicalis

ZIP12 is present in the forebrain, midbrain, and eye of Xenopus tropicalis in nervous system development. [10] ZIP12 is also present at the anterior neuropore during closure of the neural tube. [10] ZIP12 mRNA is concentrated in the neural tube, and ZIP12 expression is higher in the neural tube compared to the rest of the embryo. To study how ZIP12 is necessary for Xenopus tropicalis embryo development, the researchers injected embryos with antisense morpholino oligonucleotides that deplete the embryos of ZIP12. [10] In embryos injected with morpholinos targeting the translation start site of ZIP12, the embryos have incomplete neural tube closure at the anterior neuropore, followed by embryonic death. [10] Embryos injected with morpholinos that alter ZIP12 splicing and impair its function have slower neural tube closure, often lack eyes (called anopia), and undergo embryonic death shortly after neural tube closure. [10]

Impact on human brain MRI patterns

Genome-wide association studies (GWAS) and exome sequencing from subjects in the UK Biobank show that gene polymorphism and mutations in ZIP12 are associated with altered susceptibility weighted imaging intensity and T1 FAST magnetic resonance imaging (MRI) in the human brain. [13] [14] Polymorphisms (rs10430577, rs10430578) near SLC39A12 are the lead single nucleotide polymorphisms (SNPs) most associated with altered swMRI intensity in the caudate, putamen, and pallidum and T1 FAST MRI in the putamen. [13] Susceptibility weighted magnetic resonance imaging is sensitive to metal content in the tissues analyzed. Associated missense ZIP12 mutations (rs10764176, rs72778328) have reduced zinc transport activity when measured in Chinese hamster ovary (CHO) cells. [12] However, the impact of the changes in the human brain caused by ZIP12 polymorphisms and mutations is currently unknown.

Role in hypoxia-induced pulmonary hypertension

Hypoxia induces the expression of ZIP12 in the endothelium of mammalian pulmonary vessels. The induction of ZIP12 results in the proliferation and thickening of pulmonary vascular smooth muscle cells, which leads to pulmonary hypertension. Zhao et al. [15] identified ZIP12 as the responsible gene through congenic breeding between Fisher 344 (F344) rats, which are resistant to hypoxia-induced pulmonary hypertension, and susceptible Wistar Kyoto (WKY) rats. Resistant F344 rats crossed with non-resistant WKY rats produce subcongenic strains, and quantitative trait loci (QTL) analysis was used to determine which genes co-segregate with the hypoxic response by the pulmonary vessels and sensitivity to pulmonary hypertension. [15] A ZIP12 frameshift mutation in F344 rats truncates the protein and reduces cellular zinc uptake by pulmonary endothelial smooth muscle cells. [15] Additional support for ZIP12 as the responsible gene was shown when a similar resistance to hypoxia-induced pulmonary hypertension was observed in rats with targeted deletion of the SLC39A12 (ZIP12) gene by zinc finger nucleases. [15] In addition to rats, cattle and humans also show increased ZIP12 protein when housed in hypoxic environments, which implies that response of increased ZIP12 protein to hypoxia is found across different mammals. [15] A hypoxia response element (HRE) is present within a SLC39A12 intron, which can increase ZIP12 expression under hypoxic conditions. [15] In a separate study using human vascular endothelial and smooth muscle cells, ZIP12 expression increased after intracellular zinc chelation by TPEN. [16]

Association with schizophrenia

Associations between ZIP12 and schizophrenia have been reported. A non-coding polymorphism in ZIP12 has been described as being more prevalent in patients with schizophrenia, [17] although this finding has not yet been replicated in other studies. In another study using genome-wide microarrays and post-mortem brain tissue, researchers found higher abundance of ZIP12 mRNA in frontal lobe, superior frontal gyrus, and inferior frontal gyrus of brains from schizophrenic subjects. [11] Higher expression of both splice variants of ZIP12 was detected in the brains of patients with schizophrenia. [11]

Mutations and copy number variations in SLC39A12 have been reported for autism, although it is unclear whether genetic variability contributes towards autism risk. In one study assessing copy number variations in Han Chinese subjects with autism, one person had a heterozygous deletion in SLC39A12. [18] In another study, a premature stop codon was detected in one copy of SLC39A12 for one autistic subject. [19]

Possible associations with cancer

Altered expression and mutations in ZIP12 have been detected in various cancers. In 145 patients with esophageal adenocarcinoma, whole exome sequencing found that 12 patients had ZIP12 missense mutations in tumors negative for microsatellite instability. [20] Coding mutations in ZIP12 were also detected in a separate study on esophageal adenocarcinoma. [21] Differences in ZIP12 expression has been reported in different cancers. ZIP12 mRNA was elevated in non-small cell lung cancer biopsied tissues from at least half of tested patients. [22] ZIP12 protein abundance was lower in the breast cancer lines T47D and MDA-MB-231 when compared to non-malignant mammary cell line MCF10A. [23]

Other associations or functions of ZIP12

Associations of ZIP12 with additional diseases or physiological functions have been reported. In broiler male chicks, ZIP12 mRNA expression in the duodenum, a region of the small intestine, decreases in response to an oral challenge with Salmonella. [24] ZIP12 mRNA and protein increased in lung and liver of chickens after ascites syndrome by intravenous cellulose microparticle injection. [25] The restoration of zinc to zinc-deficient T-cells induces ZIP12 expression, [26] which may promote cytokine production by the immune system. Using quantitative trait loci (QTL) mapping in 2 different cow strains, SLC39A12 (ZIP12) may be a candidate gene that affects fertility in female Chinese and Nordic Holstein cows. [27] ZIP12 mRNA is more abundant in mouse oocytes compared to cumulus cells, which indicates that ZIP12 may play a role in reproduction and fertility. [28] A genome-wide association study (GWAS) in horses has linked an intronic polymorphism in SLC39A12 to endurance racing performance in Arabian horses. [29] One study reported that fasting glucose is associated with two polymorphisms in the SLC39A12 gene, [30] although these findings have not been confirmed in other studies and ZIP12 expression has not been detected in the pancreas. [10]

Notes

Related Research Articles

<span class="mw-page-title-main">Functional genomics</span> Field of molecular biology

Functional genomics is a field of molecular biology that attempts to describe gene functions and interactions. Functional genomics make use of the vast data generated by genomic and transcriptomic projects. Functional genomics focuses on the dynamic aspects such as gene transcription, translation, regulation of gene expression and protein–protein interactions, as opposed to the static aspects of the genomic information such as DNA sequence or structures. A key characteristic of functional genomics studies is their genome-wide approach to these questions, generally involving high-throughput methods rather than a more traditional "candidate-gene" approach.

<span class="mw-page-title-main">L1 (protein)</span> Mammalian protein found in Homo sapiens

L1, also known as L1CAM, is a transmembrane protein member of the L1 protein family, encoded by the L1CAM gene. This protein, of 200-220 kDa, is a neuronal cell adhesion molecule with a strong implication in cell migration, adhesion, neurite outgrowth, myelination and neuronal differentiation. It also plays a key role in treatment-resistant cancers due to its function. It was first identified in 1984 by M. Schachner who found the protein in post-mitotic mice neurons.

<span class="mw-page-title-main">Ferroportin</span> Protein

Ferroportin-1, also known as solute carrier family 40 member 1 (SLC40A1) or iron-regulated transporter 1 (IREG1), is a protein that in humans is encoded by the SLC40A1 gene. Ferroportin is a transmembrane protein that transports iron from the inside of a cell to the outside of the cell. Ferroportin is the only known iron exporter.

<span class="mw-page-title-main">T-box transcription factor T</span> Protein-coding gene in the species Homo sapiens

T-box transcription factor T, also known as Brachyury protein, is encoded for in humans by the TBXT gene. Brachyury functions as a transcription factor within the T-box family of genes. Brachyury homologs have been found in all bilaterian animals that have been screened, as well as the freshwater cnidarian Hydra.

<span class="mw-page-title-main">ABCB11</span> Protein-coding gene in the species Homo sapiens

ATP-binding cassette, sub-family B member 11 (ABCB11), also known as the bile salt export pump (BSEP), is a protein which in humans is encoded by the ABCB11 gene.

<span class="mw-page-title-main">HMGA2</span> Protein-coding gene in the species Homo sapiens

High-mobility group AT-hook 2, also known as HMGA2, is a protein that, in humans, is encoded by the HMGA2 gene.

<span class="mw-page-title-main">ERCC1</span> Protein-coding gene in the species Homo sapiens

DNA excision repair protein ERCC-1 is a protein that in humans is encoded by the ERCC1 gene. Together with ERCC4, ERCC1 forms the ERCC1-XPF enzyme complex that participates in DNA repair and DNA recombination.

<span class="mw-page-title-main">ABCC1</span> Protein-coding gene in the species Homo sapiens

Multidrug resistance-associated protein 1 (MRP1) is a protein that in humans is encoded by the ABCC1 gene.

<span class="mw-page-title-main">ELAC2</span> Protein-coding gene in the species Homo sapiens

Zinc phosphodiesterase ELAC protein 2 is an enzyme that in humans is encoded by the ELAC2 gene. on chromosome 17. It is an endonuclease thought to be involved in mitochondrial tRNA maturation,

<span class="mw-page-title-main">SNAI2</span> Protein

Zinc finger protein SNAI2 is a transcription factor that in humans is encoded by the SNAI2 gene. It promotes the differentiation and migration of certain cells and has roles in initiating gastrulation.

<span class="mw-page-title-main">Zinc transporter ZIP1</span> Protein found in humans

Zinc transporter ZIP1 is a protein that in humans is encoded by the SLC39A1 gene.

<span class="mw-page-title-main">Zinc transporter ZIP6</span> Protein found in humans

Zinc transporter ZIP6 is a protein that in humans is encoded by the SLC39A6 gene.

<span class="mw-page-title-main">PLAGL2</span> Protein-coding gene in the species Homo sapiens

Zinc finger protein PLAGL2 is a protein that in humans is encoded by the PLAGL2 gene.

<span class="mw-page-title-main">TFAP2B</span> Protein that in humans and is encoded by the TFAP2B gene

Transcription factor AP-2 beta also known as AP2-beta is a protein that in humans is encoded by the TFAP2B gene.

<span class="mw-page-title-main">Knockout rat</span> Type of genetically engineered rat

A knockout rat is a genetically engineered rat with a single gene turned off through a targeted mutation used for academic and pharmaceutical research. Knockout rats can mimic human diseases and are important tools for studying gene function and for drug discovery and development. The production of knockout rats was not economically or technically feasible until 2008.

miR-137

In molecular biology, miR-137 is a short non-coding RNA molecule that functions to regulate the expression levels of other genes by various mechanisms. miR-137 is located on human chromosome 1p22 and has been implicated to act as a tumor suppressor in several cancer types including colorectal cancer, squamous cell carcinoma and melanoma via cell cycle control.

<span class="mw-page-title-main">EGR4</span> Protein-coding gene in the species Homo sapiens

Early growth response protein 4 (EGR-4), also known as AT133, is a protein that in humans is encoded by the EGR4 gene.

<span class="mw-page-title-main">Cancer epigenetics</span> Field of study in cancer research

Cancer epigenetics is the study of epigenetic modifications to the DNA of cancer cells that do not involve a change in the nucleotide sequence, but instead involve a change in the way the genetic code is expressed. Epigenetic mechanisms are necessary to maintain normal sequences of tissue specific gene expression and are crucial for normal development. They may be just as important, if not even more important, than genetic mutations in a cell's transformation to cancer. The disturbance of epigenetic processes in cancers, can lead to a loss of expression of genes that occurs about 10 times more frequently by transcription silencing than by mutations. As Vogelstein et al. points out, in a colorectal cancer there are usually about 3 to 6 driver mutations and 33 to 66 hitchhiker or passenger mutations. However, in colon tumors compared to adjacent normal-appearing colonic mucosa, there are about 600 to 800 heavily methylated CpG islands in the promoters of genes in the tumors while these CpG islands are not methylated in the adjacent mucosa. Manipulation of epigenetic alterations holds great promise for cancer prevention, detection, and therapy. In different types of cancer, a variety of epigenetic mechanisms can be perturbed, such as the silencing of tumor suppressor genes and activation of oncogenes by altered CpG island methylation patterns, histone modifications, and dysregulation of DNA binding proteins. There are several medications which have epigenetic impact, that are now used in a number of these diseases.

<span class="mw-page-title-main">LINE1</span> Group of transposable elements

LINE1 is a family of related class I transposable elements in the DNA of some organisms, classified with the long interspersed elements (LINEs). L1 transposons comprise approximately 17% of the human genome. These active L1s can interrupt the genome through insertions, deletions, rearrangements, and copy number variations. L1 activity has contributed to the instability and evolution of genomes and is tightly regulated in the germline by DNA methylation, histone modifications, and piRNA. L1s can further impact genome variation through mispairing and unequal crossing over during meiosis due to its repetitive DNA sequences.

<span class="mw-page-title-main">Zinc transporter ZIP9</span> Protein found in humans

Zinc transporter ZIP9, also known as Zrt- and Irt-like protein 9 (ZIP9) and solute carrier family 39 member 9, is a protein that in humans is encoded by the SLC39A9 gene. This protein is the 9th member out of 14 ZIP family proteins, which is a membrane androgen receptor (mAR) coupled to G proteins, and also classified as a zinc transporter protein. ZIP family proteins transport zinc metal from the extracellular environment into cells through cell membrane.

References

  1. 1 2 3 GRCm38: Ensembl release 89: ENSMUSG00000036949 - Ensembl, May 2017
  2. "Human PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  3. "Mouse PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  4. "Entrez Gene: Solute carrier family 39 member 12" . Retrieved June 15, 2017.
  5. 1 2 3 Taylor KM, Nicholson RI (April 2003). "The LZT proteins; the LIV-1 subfamily of zinc transporters". Biochimica et Biophysica Acta (BBA) - Biomembranes. 1611 (1–2): 16–30. doi: 10.1016/s0005-2736(03)00048-8 . PMID   12659941.
  6. 1 2 Eide DJ (February 2004). "The SLC39 family of metal ion transporters". Pflügers Archiv. 447 (5): 796–800. doi:10.1007/s00424-003-1074-3. PMID   12748861. S2CID   11765308.
  7. Küry S, Dréno B, Bézieau S, Giraudet S, Kharfi M, Kamoun R, Moisan JP (July 2002). "Identification of SLC39A4, a gene involved in acrodermatitis enteropathica". Nature Genetics. 31 (3): 239–40. doi:10.1038/ng913. PMID   12068297. S2CID   34586490.
  8. Wang K, Zhou B, Kuo YM, Zemansky J, Gitschier J (July 2002). "A novel member of a zinc transporter family is defective in acrodermatitis enteropathica". American Journal of Human Genetics. 71 (1): 66–73. doi:10.1086/341125. PMC   419995 . PMID   12032886.
  9. 1 2 3 4 5 6 7 8 Chowanadisai W (2014). "Comparative genomic analysis of slc39a12/ZIP12: insight into a zinc transporter required for vertebrate nervous system development". PLOS ONE. 9 (11): e111535. Bibcode:2014PLoSO...9k1535C. doi: 10.1371/journal.pone.0111535 . PMC   4222902 . PMID   25375179.
  10. 1 2 3 4 5 6 7 8 9 10 11 12 13 Chowanadisai W, Graham DM, Keen CL, Rucker RB, Messerli MA (June 2013). "Neurulation and neurite extension require the zinc transporter ZIP12 (slc39a12)". Proceedings of the National Academy of Sciences of the United States of America. 110 (24): 9903–8. Bibcode:2013PNAS..110.9903C. doi: 10.1073/pnas.1222142110 . PMC   3683776 . PMID   23716681.
  11. 1 2 3 Scarr E, Udawela M, Greenough MA, Neo J, Suk Seo M, Money TT, et al. (2016). "Increased cortical expression of the zinc transporter SLC39A12 suggests a breakdown in zinc cellular homeostasis as part of the pathophysiology of schizophrenia". npj Schizophrenia. 2: 16002. doi:10.1038/npjschz.2016.2. PMC   4898896 . PMID   27336053.
  12. 1 2 3 4 5 6 Strong MD, Hart MD, Tang TZ, Ojo BA, Wu L, Nacke MR, et al. (September 2020). "Role of zinc transporter ZIP12 in susceptibility-weighted brain magnetic resonance imaging (MRI) phenotypes and mitochondrial function". FASEB Journal. 34 (9): 10702–12725. doi: 10.1096/fj.202000772R . PMID   32716562.
  13. 1 2 Elliott LT, Sharp K, Alfaro-Almagro F, Shi S, Miller KL, Douaud G, et al. (October 2018). "Genome-wide association studies of brain imaging phenotypes in UK Biobank". Nature. 562 (7726): 210–216. Bibcode:2018Natur.562..210E. doi:10.1038/s41586-018-0571-7. PMC   6786974 . PMID   30305740.
  14. Cirulli ET, White S, Read RW, Elhanan G, Metcalf WJ, Tanudjaja F, et al. (January 2020). "Genome-wide rare variant analysis for thousands of phenotypes in over 70,000 exomes from two cohorts". Nature Communications. 11 (1): 542. Bibcode:2020NatCo..11..542C. doi:10.1038/s41467-020-14288-y. PMC   6987107 . PMID   31992710.
  15. 1 2 3 4 5 6 Zhao L, Oliver E, Maratou K, Atanur SS, Dubois OD, Cotroneo E, et al. (August 2015). "The zinc transporter ZIP12 regulates the pulmonary vascular response to chronic hypoxia". Nature. 524 (7565): 356–60. Bibcode:2015Natur.524..356Z. doi:10.1038/nature14620. PMC   6091855 . PMID   26258299.
  16. Abdo AI, Tran HB, Hodge S, Beltrame JF, Zalewski PD (June 2021). "Zinc Homeostasis Alters Zinc Transporter Protein Expression in Vascular Endothelial and Smooth Muscle Cells". Biological Trace Element Research. 199 (6): 2158–2171. doi:10.1007/s12011-020-02328-z. PMID   32776265. S2CID   221101181.
  17. Bly M (January 2006). "Examination of the zinc transporter gene, SLC39A12". Schizophrenia Research. 81 (2–3): 321–2. doi:10.1016/j.schres.2005.07.039. PMID   16311021. S2CID   42890790.
  18. Gazzellone MJ, Zhou X, Lionel AC, Uddin M, Thiruvahindrapuram B, Liang S, et al. (2014). "Copy number variation in Han Chinese individuals with autism spectrum disorder". Journal of Neurodevelopmental Disorders. 6 (1): 34. doi: 10.1186/1866-1955-6-34 . PMC   4147384 . PMID   25170348.
  19. Krumm N, Turner TN, Baker C, Vives L, Mohajeri K, Witherspoon K, et al. (June 2015). "Excess of rare, inherited truncating mutations in autism". Nature Genetics. 47 (6): 582–8. doi:10.1038/ng.3303. PMC   4449286 . PMID   25961944.
  20. Dulak AM, Stojanov P, Peng S, Lawrence MS, Fox C, Stewart C, et al. (May 2013). "Exome and whole-genome sequencing of esophageal adenocarcinoma identifies recurrent driver events and mutational complexity". Nature Genetics. 45 (5): 478–86. doi:10.1038/ng.2591. PMC   3678719 . PMID   23525077.
  21. Murugaesu N, Wilson GA, Birkbak NJ, Watkins T, McGranahan N, Kumar S, et al. (August 2015). "Tracking the genomic evolution of esophageal adenocarcinoma through neoadjuvant chemotherapy". Cancer Discovery. 5 (8): 821–831. doi:10.1158/2159-8290.CD-15-0412. PMC   4529488 . PMID   26003801.
  22. Huang C, Cui X, Sun X, Yang J, Li M (October 2016). "Zinc transporters are differentially expressed in human non-small cell lung cancer". Oncotarget. 7 (41): 66935–66943. doi:10.18632/oncotarget.11884. PMC   5341848 . PMID   27611948.
  23. Chandler P, Kochupurakkal BS, Alam S, Richardson AL, Soybel DI, Kelleher SL (January 2016). "Subtype-specific accumulation of intracellular zinc pools is associated with the malignant phenotype in breast cancer". Molecular Cancer. 15: 2. doi: 10.1186/s12943-015-0486-y . PMC   4700748 . PMID   26728511.
  24. Wu A, Bai S, Ding X, Wang J, Zeng Q, Peng H, et al. (July 2020). "The Systemic Zinc Homeostasis Was Modulated in Broilers Challenged by Salmonella". Biological Trace Element Research. 196 (1): 243–251. doi:10.1007/s12011-019-01921-1. PMC   7289780 . PMID   31641975.
  25. Cui H, Liu J, Xu G, Ren X, Li Z, Li Y, Ning Z (February 2019). "Altered Expression of Zinc Transporter ZIP12 in Broilers of Ascites Syndrome Induced by Intravenous Cellulose Microparticle Injection". Biochemical Genetics. 57 (1): 159–169. doi:10.1007/s10528-018-9876-3. PMID   30073576. S2CID   51906880.
  26. Daaboul D, Rosenkranz E, Uciechowski P, Rink L (October 2012). "Repletion of zinc in zinc-deficient cells strongly up-regulates IL-1β-induced IL-2 production in T-cells". Metallomics. 4 (10): 1088–97. doi: 10.1039/c2mt20118f . PMID   22983538.
  27. Liu A, Wang Y, Sahana G, Zhang Q, Liu L, Lund MS, Su G (August 2017). "Genome-wide Association Studies for Female Fertility Traits in Chinese and Nordic Holsteins". Scientific Reports. 7 (1): 8487. Bibcode:2017NatSR...7.8487L. doi:10.1038/s41598-017-09170-9. PMC   5559619 . PMID   28814769.
  28. Lisle RS, Anthony K, Randall MA, Diaz FJ (April 2013). "Oocyte-cumulus cell interactions regulate free intracellular zinc in mouse oocytes". Reproduction. 145 (4): 381–90. doi: 10.1530/REP-12-0338 . PMID   23404848.
  29. Ricard A, Robert C, Blouin C, Baste F, Torquet G, Morgenthaler C, et al. (2017). "Endurance Exercise Ability in the Horse: A Trait with Complex Polygenic Determinism". Frontiers in Genetics. 8: 89. doi: 10.3389/fgene.2017.00089 . PMC   5488500 . PMID   28702049.
  30. Ge S, Wang Y, Song M, Li X, Yu X, Wang H, et al. (July 2018). "Type 2 Diabetes Mellitus: Integrative Analysis of Multiomics Data for Biomarker Discovery". Omics. 22 (7): 514–523. doi:10.1089/omi.2018.0053. PMID   30004843.

Further reading

This article incorporates text from the United States National Library of Medicine, which is in the public domain.