Abstract
Zinc (Zn) is an essential trace element for numerous biological events in mammals. Zn functions as a signaling mediator, leading to the regulation of physiological cell actions and, therefore, has therapeutic potential. Recent breast cancer research has shown that Zn transporters contribute to malignancy processes; thus, elucidating the roles of Zn and Zn transporters in breast cancer may lead to the development of novel strategies for breast cancer diagnosis and therapy. The Zn transporter ZIP6 mediates the acquisition of malignant phenotypes such as hypoxic resistance and epithelial-mesenchymal transition (EMT), which determine breast tumor grade and prognosis. ZIP6 expression contributes to the efficacy of anticancer therapy through Zn-induced autophagy. The maintenance of breast cancer stem-like cells requires Zn modulation through the cooperative function of ZIP6 and ZIP7. These findings suggest that the ZIP6-mediated Zn network is a potent driving force toward malignancy. In this review, we summarize recent progress in understanding the emerging roles of Zn and ZIP6 in the regulation of malignant characteristics related to hypoxic adaptive response, drug therapy, and stemness. We also discuss the possibility and future challenges of innovative breast cancer therapies using ZIP6 and Zn-related molecules.
Introduction
Breast cancer is the most commonly occurring cancer in women, with both morbidity and mortality increasing on a yearly basis [1,2]. Recent breast cancer studies have elucidated the molecular mechanisms of its pathological conditions, and evidence-based theoretical treatment protocols including pharmacotherapy using molecular target drugs are selected according to the molecular features of breast cancer cells [3,4]. However, breast cancer cells acquire malignant phenotypes aggressively during treatment, even in the early stages of growth, causing metastasis, recurrence, and resistance to therapy [5,6]. Thus, the range of current therapies is limited, and little is known about the regulatory machinery of breast cancer malignant transformation.
In recent years, a growing body of evidence has indicated that zinc (Zn) and Zn transporters are closely related to the malignant processes of breast cancer, presenting an attractive potential approach for novel therapeutic strategies [7-11]. As a trace element, Zn is essential for human survival; intracellular Zn2+ levels are spatiotemporally regulated by ZRT IRT-like proteins (ZIPs; SLC39A, 14 members) to facilitate the influx of Zn into the cytosol, and zinc transporters (ZnT; SLC30A, 9 members) mediate Zn efflux from the cytosol [12,13]. These transporters contribute to maintaining Zn homeostasis within cells and throughout the body by strictly managing the cellular Zn balance according to the distributions of their molecular species in a tissue-specific manner [8-13]. Zn has been shown to target molecules selectively as an intracellular and extracellular signaling mediator, such that the Zn signaling pathway spatiotemporally regulates cell functions and Zn transporter aberrations are linked to the onset and progression of diseases such as cancers [7-14], Alzheimer’s disease [15], diabetes [16-18], and other pathological processes [19-24] (Figure 1). Interestingly, zinc transporters in cancers have been reported from various fields including clinical analysis as well as molecular and structural biology, and only 5 ZnTs and almost all ZIP transporters are involved in a number of cancers [25]. Patterns of ZIP dysregulation differ among cancers, and indicate the complicated roles of Zn in various cancer types including prostate cancer [26,43], pancreatic cancer [27-29,44,59], hepatocellular carcinoma [30,45], lung cancer [31], glioma [32], oral squamous [33], nasopharyngeal [34], ovarian cancer [35,60], colorectal cancer [46], esophageal cancer [36,47], breast cancer [37-42,49-52,54-57], gastric adenocarcinoma [48], neuroblastoma [53], and bladder cancer [58], as well as in phenotypes associated with tumorigenesis, metastasis, and therapeutic resistance (Table 1). In metastatic cancers, ZIP14 is upregulated in skeletal muscle, promoting cachexia [61].
Zn levels in the breast tissue and serum of patients with breast cancer are higher and lower, respectively, than those of healthy subjects [62-65]. Both clinical and in vitro studies have demonstrated the potential roles of ZIP6 in estrogen receptor (ER)-positive breast cancer in about 70% of breast cancer patients [37-42], of ZIP7 in ER antagonist tamoxifen-resistant breast cancer [49-52], and of ZIP10 in breast cancer invasion and metastasis [55-57]. In particular, ZIP6 was identified in human breast cancer MCF-7 cells expressing substantial levels of ER [66], and the ZIP6-centered Zn network was found to have important implications for ER-positive breast cancer [8,11,37-42,67]. Decreased expression of ZIP6 in MCF-7 in response to stimuli in the environment surrounding cancer cells such as high glucose concentration and hypoxia induces epithelial-mesenchymal transition (EMT), promoting stemness and resistance to cell death [8,11,39]. In the clinical setting, patients with low ZIP6 expression in primary breast cancer sites have worse prognoses than those with high ZIP6 expression, suggesting that ZIP6 is negatively associated with prognosis [40]. Therefore, the cell culture model of ZIP6-knockdown MCF-7 mimics highly malignant breast cancer and is a potential tool for the investigation of breast cancer progression [8,11,39].
In this review, we summarize current progress in our understanding of the role of the main ZIP6 and ZIP6-related Zn transporters, focusing on breast cancer cell behaviors in hypoxia as a crucial factor in the tumor microenvironment, as well as on hormone therapy and breast cancer stem-like cells, which directly impact malignant progression and prognosis. We also discuss Zn transporters and Zn-targeted molecules as candidate markers for the malignant phenotype of breast cancer, which may represent a promising target for novel treatment strategies.

Table 1.
ZIP transporters and cancers.
Gene name |
Protein name |
Type of cancer cells |
Functional activity in cancer cells |
References |
SLA39a1 |
ZIP1 |
Prostate |
Expression down-regulated |
26 |
SLA39a2 |
ZIP2 |
Prostate |
Expression down-regulated |
26 |
SLA39a3 |
ZIP3 |
Prostate |
Expression down-regulated |
26 |
Pancreatic |
Expression down-regulated |
27 |
SLA39a4 |
ZIP4 |
Pancreatic |
Expression up-regulated |
28 |
Pancreatic |
Expression up-regulated in cancer cell-derived exosomes |
29 |
Hepatocellular |
Expression up-regulated |
30 |
Lung |
Expression up-regulated |
31 |
Glioma |
Expression up-regulated |
32 |
Oral squamous |
Expression up-regulated |
33 |
Nasopharyngeal |
Expression up-regulated |
34 |
Ovarian |
Expression up-regulated |
35 |
SLA39a5 |
ZIP5 |
Esophageal |
Expression up-regulated |
36 |
SLA39a6 |
ZIP6 |
Metastatic ER+breast |
Expression up-regulated |
37,67 |
ER+breast |
Expression down-regulated |
38-42 |
Prostate |
Expression up-regulated |
43 |
Pancreatic |
Expression up-regulated |
44 |
Hepatocellular |
Expression up-regulated |
45 |
Colorectal |
Expression up-regulated |
46 |
Esophageal squamous-cell |
Expression up-regulated |
47 |
Gastric |
Expression up-regulated |
48 |
SLA39a7 |
ZIP7 |
Anti-hormone resistance in ER+breast |
Expression up-regulated |
49-52,92 |
SLA39a8 |
ZIP8 |
Neuroblastoma |
Proliferation inhibited by ZIP8 knockdown |
53 |
SLA39a9 |
ZIP9 |
Prostate, breast |
Apoptosis induced by ZIP9 activation |
54 |
SLA39a10 |
ZIP10 |
Metastatic breast |
Expression up-regulated |
55-57 |
SLA39a11 |
ZIP11 |
Renal cell |
ZIP11 variant as risk factor |
58 |
Bladder |
ZIP11 variant as risk factor |
58 |
Pancreatic |
Expression up-regulated |
59 |
SLA39a13 |
ZIP13 |
Ovarian |
Expression up-regulated |
60 |
SLA39a14 |
ZIP14 |
Cachexia in metastatic cancer |
Expression up-regulated in skeletal muscle |
61 |
ZIP6 and hypoxic adaptive response of breast cancer cells
Hypoxia occurs in internal solid tumor structures, including breast cancer, as a result of rapid tumor cell growth [68]. This hypoxic region has been reported to contribute to tumor plasticity and heterogeneity, and to promote more aggressive phenotypes such as angiogenesis, invasion, EMT, stemness, and resistance to chemotherapy and radiotherapy [69-71]. To clarify the hypoxic profile in breast cancer cells, we cultured MCF-7 in three dimensions using a soft agar colony formation assay [8,11]. MCF-7 formed colonies, and colony interior was confirmed to be hypoxic according to strong red fluorescence of the hypoxia-sensing probe LOX-1. The specific membrane-permeable Zn chelator N,N,N',N'-tetrakis (2-pyridylmethyl) ethylenediamine (TPEN) significantly inhibited colony formation, and this inhibition was rescued by increasing the level of intracellular free Zn using zinc ionophore pyrithion. These findings suggest that intracellular Zn is required for survival under hypoxia and tumorigenic potential. Hypoxia-inducible factor 1α (HIF-1α) is a pivotal regulator in the process of hypoxic adaptive response [72,73]. MCF-7 cultured under hypoxic conditions showed higher viability than normal cells, depending on the activity of HIF-1α, and HIF-1α activity was positively correlated with intracellular Zn levels under hypoxic conditions. These data suggest that Zn-mediated HIF-1α activity promotes MCF-7 cell survival. Although the mechanisms underlying Zn-mediated HIF-1α activity in MCF-7 remains unknown, HIF-1α protein requires Zn to maintain its structure [74]. The knockdown of ZIP6, which plays an important role in maintaining intracellular Zn homeostasis in MCF-7, indicated that the disruption of intracellular Zn homeostasis, implying the presence of other zinc transporters, leads to increased intracellular free Zn concentration and higher survival rates under hypoxic conditions through apoptosis inhibition and EMT induction [39]. A zebrafish embryo study showed that a downstream factor of ZIP6 regulates signal transducer and activator of transcription 3 (STAT3) expression, which is linked to EMT [37]. ZIP6 expression levels were found to be decreased in high-grade primary tumor sites among patients with breast cancer [8,11,41], and low ZIP6 expression levels at primary breast cancer sites are correlated with shorter relapse-free survival periods [40]. Both experimental findings and clinical observations indicate that ZIP6 downregulation is deeply involved in the malignancy phenotype, suggesting that ZIP6 levels in primary breast tumors are potential biomarkers of cancer grade and poor prognosis in patients with breast cancer (Figure 2) [8,11,40,41]. Further analysis of the ZIP6 downregulation-switched zinc network, including the involvement of other Zn transporters and the behavior of intracellular and extracellular Zn in malignant progression under hypoxia, is expected to indicate future directions for the development of breast cancer diagnosis and treatment.
ZIP6 and pharmacotherapy in breast cancer
At least 70% of breast cancers are female hormone ER-positive and thus benefit from endocrine treatments including ER antagonist tamoxifen and aromatase inhibitors, which block estrogen production [75]. However, more than 25 % of all breast cancer patients show endocrine resistance, and even after 5 years of tamoxifen use, one in three patients with endocrine treatments still recur within 15 years; thus, the elucidation of endocrine-resistant mechanisms is an urgent clinical challenge [75,76]. High expression of Bcl-2, an anti-apoptotic molecule that regulates programmed cell death, has been reported to cause endocrine therapy resistance in breast cancer, and efficient inhibition of Bcl-2 function is expected to be a promising strategy to prevent endocrine resistance [76,77]. Great effort has been made to investigate the sensitivity of genetic or pharmacological inhibition of Bcl-2 in breast cancer [78]. YC137, a novel small molecule-specific inhibitor of Bcl-2, induces apoptosis in breast cancer cells that express high levels of Bcl-2, but does not attack normally functioning cells including hematopoietic progenitors, peripheral blood mononuclear cells, small intestine epithelial cells, and myoblasts [79]. Intriguingly, we found that the cytotoxicity of YC137 against estrogen-dependent human breast cancer cell, MCF-7, was markedly enhanced in combination with Zn chloride (ZnCl2). Treatment with ZnCl2 increased the concentration of labile Zn in MCF-7, and Zn supplementation triggered a switch from apoptotic cell death by YC137 (type I programmed cell death) to non-apoptotic cell death with autophagy (type II programmed cell death). Autophagy, in which autophagy-related protein-mediated autophagosomes form around misfolded proteins and damaged organelles to induce their degradation, is crucial for maintaining cell homeostasis and controlling cell death according to stress condition and breast cancer development stage [80,81]. Autophagy can be induced, and plays a cytoprotective or pro-survival role under most cancer therapies, whereas the collapse and excess of autophagy contributes to cytotoxicity in breast cancer cells [82]. During treatment with tamoxifen in MCF-7 cells, Zn accumulates in autophagosomes, resulting in autophagosome-mediated cell death [83]. Recent studies have demonstrated that Zn transporters ZIP4, ZIP8, ZIP14, ZnT2, ZnT4, and ZnT10, are located in endosomes, lysosomes, and autolysosomes, and have suggested that Zn contributes to autophagy [84,85]. The enhancement of YC137 cytotoxicity in combination with ZnCl2 was abolished in ZIP6-knockdown MCF-7 cells, indicating that Zn supplied by ZIP6 is required for increased sensitivity of YC137. Studies on the roles of Zn in breast cancer therapy have recently begun, and will need to take into account the relationship between ZIP6 and autophagy. The combination of YC137 and ZnCl2 does not induce damage to normal mammary gland cell model, human non-tumorigenic breast epithelial MCF-10A cells. As ZIP6 downregulation is involved in malignant progression in breast cancer [8,11,40,41], we propose that the combination of YC137 and ZnCl2 may be an effective cancer-specific anticancer method for preventing the generation of therapeutic resistance, especially in early breast cancer, where ZIP6 expression is high. Future studies are required to elucidate the in vivo response to this treatment and to determine the appropriate dosing regimen with Zn supplement as adjuvant.

ZIP6/ZIP7 and breast cancer stem-like cells (BCSCs)
BCSCs are a distinct group of breast cancer-initiating cells with the capacity for self-renewal and differentiation; they hide within tumor tissue or throughout in the body [86,87]. BCSCs contribute to the cellular origin, tumor maintenance, and progression of breast cancer [88]. Clinically, BCSCs are considered to be responsible for the development of treatment resistance and cancer recurrence due to their relative resistance to radiation, cytotoxic chemotherapy, and molecular targeted therapy [86-88]. However, the molecular mechanisms that regulate BCSC maintenance remain poorly understood, and evaluation is required to improve the clinical outcomes of patients with breast cancer [86-88].
Among three-dimensional in vitro cell culture models, the spheroid culture method has been widely used as a powerful tool to maintain and grow cells selectively with cancer stem cell characteristics [88]. When MCF-7 cells were sphere-cultured in serum-free medium under non-adherent conditions, they formed spherical aggregates (spheres) and showed high expression of stem cell markers, self-renewal ability, and high tumorigenicity in vivo, all of which are characteristics of BCSCs [89]. In a spheroid culture assay using MCF-7 cells, treatment with TPEN revealed strong inhibition of sphere-forming capacity and nuclear expression of PR domain zinc finger protein 14 (PRDM14), a cancer stem cell transcription factor essential for the maintenance of BCSCs [90]. This inhibition was neutralized by the addition of Zn through zinc ionophores, suggesting that intracellular Zn-dependent PRDM14 expression in nuclear is essential for sphere-forming capacity. ZIP6 knockdown in MCF-7 cells resulted in significantly higher sphere formation efficiency compared to control cells. The high Zn-dependent PRDM14-mediated sphere-forming capacity of ZIP6-knockdown MCF-7 cells was significantly suppressed by functional inhibition of the zinc transporter ZIP7, which transports Zn from the endoplasmic reticulum to the cytoplasm. ZIP7, which directly drives pro-survival signaling pathways such as AKT, mTOR, and MAPK for tumorigenesis [91], is increased in endocrine-resistant breast cancer [50]. Phosphorylated ZIP7, which is the Zn-transporting form, may be a good candidate marker of aggressive cancer [92]. ZIP7 expression is also strongly associated with poor outcomes in clinical breast cancer samples [93]. BCSCs are increased in ZIP7 activity against ZIP6, suggesting that ZIP6 and ZIP7 cooperatively regulate BCSC maintenance via Zn-dependent regulation of PRDM14 expression (Figure 3). PRDM14 is not expressed in normal tissue stem cells, whereas PRDM14 is highly expressed in BCSCs as an essential transcription factor [90]. Survival analysis of breast cancer patients at stage II or III revealed that patients with high PRDM14 expression had worse prognoses than those with low or undetectable PRDM14 expression [90]. These findings suggest that PRDM14 is a promising target for the development of definitive therapeutic strategies. However, it is difficult to target the PRDM14 molecule using antibody and small-molecule therapeutics, because PRDM14 is expressed and activated in the nucleus [94]. The elucidation of Zn-mediated BCSCs through PRDM14 could dramatically expand the development of new molecularly targeted therapies for BCSCs.

Conclusion and perspective
ZIP6 is a key molecule determining the fate of ER-positive breast cancer cells, and its expression is controlled by the surrounding environment and malignant progression (Figure 4). A deeper understanding of the Zn regulatory network originating from ZIP6 will help to clarify the malignant mechanism of breast cancer and promote the development of novel strategies involving Zn-related molecules. The role of Zn biochemistry at the molecular level in therapeutic strategies, and the biology of the target molecules of Zn signaling and mechanisms regulating ZIP6 gene expression, require further study. Interestingly, the construction of ZIP6 includes a PrP-like amino acid sequence in the extracellular NH2-terminal region, and two members of the LIV-1 subfamily (ZIP5 and 10) to which ZIP6 belongs have been reported to be involved in etiology of prion disease via processing [12,95-98]. The trafficking of ZIP6 to the plasma membrane is regulated by processing on the endoplasmic reticulum [12,37]; therefore, ZIP6 processing regulation may have important implications in breast cancer physiology, although the relationship between Zn status and processing must be considered [12]. The relationships between Zn transported via ZIP6 and Zn reservoirs (intracellular metallothionein or glutathione), the effects of other metals on Zn functions, and the roles of ZIPs other than ZIP6/ZIP7 and ZnTs in the cellular/extracellular Zn network of breast cancer also require further investigation. ZIP6 has been reported to function in conjunction with its close homolog ZIP10, which is located on the plasm membrane [55,56,99,100]. Zn transported via ZIP6 and ZIP10 was found to interact with NCAM1, which promotes cell detachment through the activation of glycogen synthase kinase 3 (GSK-3), which allows cells to migrate and become metastatic [100]. This finding is consistent with the high levels of ZIP6 found in patients with ER-positive breast cancer and lymph node metastasis [67]. A recent study also demonstrated that ZIP10 mRNA expression was significantly higher in the breast cancer tissues of patients with lymph node metastasis than in those without metastasis [57]. These data suggest that the role of ZIP6 differs between metastatic foci and primary tumors, and that ZIP6 and ZIP10 may cooperate with transporter functions during cell migration, although further studies are needed to clarify the involvement of a ZIP6 and ZIP10 heteromer [8,11,99,100]. For therapeutic applications, it is necessary to consider the regulation of ZIP6-mediated Zn signaling in a breast cancer phenotype-specific manner, in conjunction with drug delivery system research. The discovery and development of inhibitors and activators of ZIP6 and other Zn transporters may reveal promising anticancer therapies. A ZIP7 inhibitor (NVS-ZP7-4) was recently identified as the first reported ZIP-specific small molecule inhibitor using a phenotype-based high-throughput screening system that targeted the Notch pathway, although its function in breast cancer and in vivo remain poorly understood [101].
Finally, a further understanding of the role Zn in breast cancer is anticipated to reveal crucial molecular mechanisms that will facilitate the development of precise diagnostic and effective therapeutic strategies.
Acknowledgments
This work was supported in part by Japan Society for the Promotion of Science (JSPS) KAKENHI grants (nos. 24790099, 15K07955, 18K06676, and 21K06567 for T. T.-N.), the Naito Foundation (T.T.-N.), a Nagai Memorial Research Scholarship from the Pharmaceutical Society of Japan (to C. M.), and the Takeda Science Foundation (no. J20ZZ00356 to I.N.). We thank Ms. Chinami Ikushima, Dr. Reiko Yutani, and Dr. Haruki Torii for their helpful support.
Statements about COI
The authors declare no conflict of interest associated with this manuscript.
References
- [1]
World Cancer Research Fund (WCRF): “
Breast cancer statistics.
Breast cancer is the most common cancer in women worldwide”. WCRF,
https://www.wcrf.org/dietandcancer/breast-cancer-statistics/ (accessed December 29,
2021).
- [2]
Sung
H
,
Ferlay
J
,
Siegel
RL
,
Laversanne
M
,
Soerjomataram
I
,
Jemal
A
,
Bray
F
:
Global Cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries.
CA Cancer J Clin
71(
3):
209-
249,
2021.
- [3]
Lønning
PE
,
Sørlie
T
and
Børresen-Dale
AL
:
Genomics in breast cancer-therapeutic implications.
Nat Clin Pract Oncol
2(
1):
26-
33,
2005.
- [4]
Shimoi
T
,
Nagai
SE
,
Yoshinami
T
,
Takahashi
M
,
Arioka
H
,
Ishihara
M
,
Kikawa
Y
,
Koizumi
K
,
Kondo
N
,
Sagara
Y
,
Takada
M
,
Takano
T
,
Tsurutani
J
,
Naito
Y
,
Nakamura
R
,
Hattori
M
,
Hara
F
,
Hayashi
N
,
Mizuno
T
,
Miyashita
M
,
Yamashita
N
,
Yamanaka
T
,
Saji
S
,
Iwata
H
,
Toyama
T
:
The Japanese Breast Cancer Society Clinical Practice Guidelines for systemic treatment of breast cancer, 2018 edition.
Breast Cancer
27(
3):
322-
331,
2020.
- [5]
Riggio
AI
,
Varley
KE
,
Welm
AL
:
The lingering mysteries of metastatic recurrence in breast cancer.
Br J Cancer
124(
1):
13-
26,
2021.
- [6]
Early Breast Cancer Trialistsʼ Collaborative Group (EBCTCG)
:
Effects of chemotherapy and hormonal therapy for early breast cancer on recurrence and 15-year survival: an overview of the randomised trials.
Lancet
365(
9472):
1687-
1717,
2005.
- [7]
Hu
J
:
Toward unzipping the ZIP metal transporters: structure, evolution, and implications on drug discovery against cancer.
FEBS J
288(
20):
5805-
5825,
2021.
- [8]
Takatani-Nakase
T
:
Zinc Transporters and the progression of breast cancers.
Biol Pharm Bull.
41(
10):
1517-
1522,
2018.
- [9]
Hara
T
,
Takeda
TA
,
Takagishi
T
,
Fukue
K
,
Kambe
T
,
Fukada
T
:
Physiological roles of zinc transporters: molecular and genetic importance in zinc homeostasis.
J Physiol Sci
67(
2):
283-
301,
2017.
- [10]
Takagishi
T
,
Hara
T
,
Fukada
T
:
Recent advances in the role of SLC39A/ZIP zinc transporters in vivo.
Int J Mol Sci
18(
12):
2708,
2017.
- [11]
Takatani-Nakase
T
,
Matsui
C
,
Takahashi
K
:
Role of the LIV-1 subfamily of zinc transporters in the development and progression of breast cancers: A mini review.
Biomed Res Clin Prac
1(
3):
71-
75,
2016.
- [12]
Kambe
T
,
Tsuji
T
,
Hashimoto
A
,
Itsumura
N
:
The physiological, biochemical, and molecular roles of zinc transporters in zinc homeostasis and metabolism.
Physiol Rev
95(
3):
749-
784,
2015.
- [13]
Kambe
T
,
Taylor
KM
,
Fu
D
:
Zinc transporters and their functional integration in mammalian cells.
J Biol Chem
296:
100320,
2021.
- [14]
Wang
J
,
Zhao
H
,
Xu
Z
,
Cheng
X
:
Zinc dysregulation in cancers and its potential as a therapeutic target.
Cancer Biol Med
17(
3):
612-
625,
2020.
- [15]
Lyubartseva
G
,
Smith
JL
,
Markesbery
WR
,
Lovell
MA
:
Alterations of zinc transporter proteins ZnT-1, ZnT-4 and ZnT-6 in preclinical Alzheimerʼs disease brain.
Brain Pathol
20(
2):
343-
350,
2010.
- [16]
Sladek
R
,
Rocheleau
G
,
Rung
J
,
Dina
C
,
Shen
L
,
Serre
D
,
Boutin
P
,
vincent
D
,
Belisle
A
,
Hadjadj
S
,
Balkau
B
,
Heude
B
,
Charpentier
G
,
Hudson T
J
,
Montpetit
A
,
Pshezhetsky
AV
,
Prentki
M
,
Posner
BI
,
Balding
DJ
,
Meyre
D
,
Polychronakos
C
,
Froguel
P
:
A genome-wide association study identifies novel risk loci for type 2 diabetes.
Nature
445(
7130):
881-
885,
2007.
- [17]
Chimienti
F
,
Devergnas
S
,
Favier
A
,
Seve
M
:
Identification and cloning of a beta-cell-specific zinc transporter, ZnT-8, localized into insulin secretory granules.
Diabetes
53(
9):
2330-
2337,
2004.
- [18]
Wenzlau
JM
,
Juhl
K
,
Yu
L
,
Moua
O
,
Sarkar
SA
,
Gottlieb
P
,
Rewers
M
,
Eisenbarth
GS
,
Jensen
J
,
Davidson
HW
,
Hutton
JC
:
The cation efflux transporter ZnT8 (Slc30A8) is a major autoantigen in human type 1 diabetes.
Proc Natl Acad Sci USA
104(
43):
17040-
17045,
2007.
- [19]
Wang
K
,
Zhou
B
,
Kuo
YM
,
Zemansky
J
,
Gitschier
J
:
A novel member of a zinc transporter family is defective in acrodermatitis enteropathica.
Am J Hum Genet
71(
1):
66-
73,
2002.
- [20]
Fukada
T
,
Civic
N
,
Furuichi
T
,
Shimoda
S
,
Mishima
K
,
Higashiyama
H
,
Idaira
Y
,
Asada
Y
,
Kitamura
H
,
Yamasaki
S
,
Hojyo
S
,
Nakayama
M
,
Ohara
O
,
Koseki
H
,
Dos Santos
HG
,
Bonafe
L
,
Ha-Vinh
R
,
Zankl
A
,
Unger
S
,
Kraenzlin
ME
,
Beckmann
JS
,
Saito
I
,
Rivolta
C
,
Ikegawa
S
,
Superti-Furga
A
,
Hirano
T
:
The zinc transporter SLC39A13/ZIP13 is required for connective tissue development; its involvement in BMP/TGF-beta signaling pathways.
PLoS One
3(
11):
e3642,
2008.
- [21]
Ohashi
W
,
Kimura
S
,
Iwanaga
T
,
Furusawa
Y
,
Irié
T
,
Izumi
H
,
Watanabe
T
,
Hijikata
A
,
Hara
T
,
Ohara
O
,
Koseki
H
,
Sato
T
,
Robine
S
,
Mori
H
,
Hattori
Y
,
Watarai
H
,
Mishima
K
,
Ohno
H
,
Hase
K
,
Fukada
T
:
Zinc transporter SLC39A7/ZIP7 promotes intestinal epithelial self-renewal by resolving ER stress.
PLoS Genet
12(
10):
e1006349,
2016.
- [22]
Hojyo
S
,
Miyai
T
,
Fujishiro
H
,
Kawamura
M
,
Yasuda
T
,
Hijikata
A
,
Bin
BH
,
Irié
T
,
Tanaka
J
,
Atsumi
T
,
Murakami
M
,
Nakayama
M
,
Ohara
O
,
Himeno
S
,
Yoshida
H
,
Koseki
H
,
Ikawa
T
,
Mishima
K
,
Fukada
T
:
Zinc transporter SLC39A10/ZIP10 controls humoral immunity by modulating B-cell receptor signal strength.
Proc Natl Acad Sci USA
111(
32):
11786-
11791,
2014.
- [23]
Bin
BH
,
Bhin
J
,
Takaishi
M
,
Toyoshima
KE
,
Kawamata
S
,
Ito
K
,
Hara
T
,
Watanabe
T
,
Irié
T
,
Takagishi
T
,
Lee
SH
,
Jung
HS
,
Rho
S
,
Seo
J
,
Choi
DH
,
Hwang
D
,
Koseki
H
,
Ohara
O
,
Sano
S
,
Tsuji
T
,
Mishima
K
,
Fukada
T
:
Requirement of zinc transporter ZIP10 for epidermal development: Implication of the ZIP10-p63 axis in epithelial homeostasis.Proc Natl Acad Sci USA
114(
46):
12243-
12248,
2017.
- [24]
Fukunaka
A
,
Fukada
T
,
Bhin
J
,
Suzuki
L
,
Tsuzuki
T
,
Takamine
Y
,
Bin
BH
,
Yoshihara
T
,
Ichinoseki-Sekine
N
,
Naito
H
,
Miyatsuka
T
,
Takamiya
S
,
Sasaki
T
,
Inagaki
T
,
Kitamura
T
,
Kajimura
S
,
Watada
H
,
Fujitani
Y
:
Zinc transporter ZIP13 suppresses beige adipocyte biogenesis and energy expenditure by regulating C/EBP-β expression.
PLoS Genet
13(
8):
e1006950,
2017.
- [25]
Pan
Z
,
Choi
S
,
Ouadid-Ahidouch
H
,
Yang
JM
,
Beattie
JH
,
Korichneva
I
:
Zinc transporters and dysregulated channels in cancers.
Front Biosci (Landmark Ed)
22:
623-
643,
2017.
- [26]
Kolenko
V
,
Teper
E
,
Kutikov
A
,
Uzzo
R
:
Zinc and zinc transporters in prostate carcinogenesis.
Nat Rev Urol
10(
4):
219-
226,
2013.
- [27]
Costello
LC
,
Franklin
RB
:
A review of the current status and concept of the emerging implications of zinc and zinc transporters in the development of pancreatic cancer.
Pancreat Disord Ther
4:
002,
2013.
- [28]
Cui
X
,
Zhang
Y
,
Yang
J
,
Sun
X
,
Hagan
JP
,
Guha
S
,
Li
M
:
ZIP4 confers resistance to zinc deficiency-induced apoptosis in pancreatic cancer.
Cell Cycle
13(
7):
1180-
1186,
2014.
- [29]
Jin
H
,
Liu
P
,
Wu
Y
,
Meng
X
,
Wu
M
,
Han
J
,
Tan
X
:
Exosomal zinc transporter ZIP4 promotes cancer growth and is a novel diagnostic biomarker for pancreatic cancer.
Cancer Sci
109(
9):
2946-
2956,
2018.
- [30]
Weaver
BP
,
Zhang
Y
,
Hiscox
S
,
Guo
GL
,
Apte
U
,
Taylor
KM
,
Sheline
CT
,
Wang
L
,
Andrews
GK
:
Zip4 (Slc39a4) expression is activated in hepatocellular carcinomas and functions to repress apoptosis, enhance cell cycle and increase migration.
PLoS One
5(
10):
e13158,
2010.
- [31]
Huang
C
,
Cui
X
,
Sun
X
,
Yang
J
,
Li
M
:
Zinc transporters are differentially expressed in human non-small cell lung cancer.
Oncotarget
7(
41):
66935-
66943,
2016.
- [32]
Lin
Y
,
Chen
Y
,
Wang
Y
,
Yang
J
,
Zhu
VF
,
Liu
Y
,
Cui
X
,
Chen
L
,
Yan
W
,
Jiang
T
,
Hergenroeder
GW
,
Fletcher
SA
,
Levine
JM
,
Kim
DH
,
Tandon
N
,
Zhu
JJ
,
Li
M
:
ZIP4 is a novel molecular marker for glioma.
Neuro Oncol
15(
8):
1008-
1016,
2013.
- [33]
Ishida
S
,
Kasamatsu
A
,
Endo-Sakamoto
Y
,
Nakashima
D
,
Koide
N
,
Takahara
T
,
Shimizu
T
,
Iyoda
M
,
Shiiba
M
,
Tanzawa
H
,
Uzawa
K
:
Novel mechanism of aberrant ZIP4 expression with zinc supplementation in oral tumorigenesis.
Biochem Biophys Res Commun
483(
1):
339-
345,
2017.
- [34]
Zeng
Q
,
Liu
YM
,
Liu
J
,
Han
J
,
Guo
JX
,
Lu
S
,
Huang
XM
,
Yi
P
,
Lang
JY
,
Zhang
P
,
Wang
CT
:
Inhibition of ZIP4 reverses epithelial-to-mesenchymal transition and enhances the radiosensitivity in human nasopharyngeal carcinoma cells.
Cell Death Dis
10(
8):
588,
2019.
- [35]
Fan
Q
,
Cai
Q
,
Li
P
,
Wang
W
,
Wang
J
,
Gerry
E
,
Wang
TL
,
Shih
IM
,
Nephew
KP
,
Xu
Y
:
The novel ZIP4 regulation and its role in ovarian cancer.
Oncotarget
8(
52):
90090-
90107,
2017.
- [36]
Li
Q
,
Jin
J
,
Liu
J
,
Wang
L
,
He
Y
:
Knockdown of zinc transporter ZIP5 by RNA interference inhibits esophageal cancer growth in vivo.
Oncol Res
24(
3):
205-
214,
2016.
- [37]
Hogstrand
C
,
Kille
P
,
Ackland
ML
,
Hiscox
S
,
Taylor
KM
:
A mechanism for epithelial-mesenchymal transition and anoikis resistance in breast cancer triggered by zinc channel ZIP6 and STAT3 (signal transducer and activator of transcription 3)
Biochem J
455(
2):
229-
237,
2013.
- [38]
Lopez
V
,
Kelleher
SL
:
Zip6-attenuation promotes epithelial-to-mesenchymal transition in ductal breast tumor (T47D) cells.
Exp Cell Res
316(
3):
366-
375,
2010.
- [39]
Matsui
C
,
Takatani-Nakase
T
,
Hatano
Y
,
Kawahara
S
,
Nakase
I
,
Takahashi
K
:
Zinc and its transporter ZIP6 are key mediators of breast cancer cell survival under high glucose conditions.
FEBS Lett
591(
20):
3348-
3359,
2017.
- [40]
Kasper
G
,
Weiser
AA
,
Rump
A
,
Sparbier
K
,
Dahl
E
,
Wild
P
,
Schwidetzky
U
,
Castanos-Velez
E
,
Lehmann
K
:
Expression levels of the putative zinc transporter LIV-1 are associated with a better outcome of breast cancer patients.
Int J Cancer
117(
6):
961-
973,
2005.
- [41]
Taylor
KM
,
Morgan
HE
,
Smart
K
,
Zahari
NM
,
Pumford
S
,
Ellis
IO
,
Robertson
JFR
,
Nicholson
RI
:
The emerging role of the LIV-1 subfamily of zinc transporters in breast cancer.
Mol Med
13(
7-8):
396-
406,
2007.
- [42] Tozlu S, Girault I, Vacher S, Vendrell J, Andrieu C, Spyratos F, Cohen P, Lidereau R, Bieche I: Identification of novel genes that co-cluster with estrogen receptor alpha in breast tumor biopsy specimens, using a large-scale real-time reverse transcription-PCR approach. Endocr Relat Cancer 13(4): 1109-1120, 2006.
- [43]
Lue
HW
,
Yang
X
,
Wang
R
,
Qian
W
,
Xu
RZ
,
Lyles
R
,
Osunkoya
AO
,
Zhou
BP
,
Vessella
RL
,
Zayzafoon
M
,
Liu
ZR
,
Zhau
HE
,
Chung
LW
:
LIV-1 promotes prostate cancer epithelial-to-mesenchymal transition and metastasis through HB-EGF shedding and EGFR-mediated ERK signaling.
PLoS One
6(
11):
e27720,
2011.
- [44]
Unno
J
,
Satoh
K
,
Hirota
M
,
Kanno
A
,
Hamada
S
,
Ito
H
,
Masamune
A
,
Tsukamoto
N
,
Motoi
F
,
Egawa
S
,
Unno
M
,
Horii
A
,
Shimosegawa
T
:
LIV-1 enhances the aggressive phenotype through the induction of epithelial to mesenchymal transition in human pancreatic carcinoma cells.
Int J Oncol
35(
4):
813-
821,
2009.
- [45]
Shen
R
,
Xie
F
,
Shen
H
,
Liu
Q
,
Zheng
T
,
Kou
X
,
Wang
D
,
Yang
J
:
Negative correlation of LIV-1 and E-cadherin expression in hepatocellular carcinoma cells.
PLoS One
8(
2):
e56542,
2013.
- [46]
Barresi
V
,
Valenti
G
,
Spampinato
G
,
Musso
N
,
Castorina
S
,
Rizzarelli
E
,
Condorelli
DF
:
Transcriptome analysis reveals an altered expression profile of zinc transporters in colorectal cancer.
J Cell Biochem
119(
12):
9707-
9719,
2018.
- [47]
Wu
C
,
Li
D
,
Jia
W
,
Hu
Z
,
Zhou
Y
,
Yu
D
,
Tong
T
,
Wang
M
,
Lin
D
,
Qiao
Y
,
Zhou
Y
,
Chang
J
,
Zhai
K
,
Wang
M
,
Wei
L
,
Tan
W
,
Shen
H
,
Zeng
Y
,
Lin
D
:
Genome-wide association study identifies common variants in SLC39A6 associated with length of survival in esophageal squamous-cell carcinoma.
Nat Genet
45(
6):
632-
638,
2013.
- [48]
Gao
J
,
Ren
W
,
Xiao
C
,
Wang
L
,
Huang
Q
,
Zhang
Z
,
Dang
Y
,
Weng
P
,
Wang
H
,
Fang
X
,
Zhuang
M
,
Lin
L
,
Chen
S
:
Involvement of SLC39A6 in gastric adenocarcinoma and correlation of the SLC39A6 polymorphism rs1050631 with clinical outcomes after resection.
BMC Cancer
19(
1):
1069,
2019.
- [49]
Hogstrand
C
,
Kille
P
,
Nicholson
RI
,
Taylor
KM
:
Zinc transporters and cancer: a potential role for ZIP7 as a hub for tyrosine kinase activation.
Trends Mol Med
15(
3):
101-
111,
2009.
- [50]
Taylor
KM
,
Vichova
P
,
Jordan
N
,
Hiscox
S
,
Hendley
R
,
Nicholson
RI
:
ZIP7-mediated intracellular zinc transport contributes to aberrant growth factor signaling in antihormone-resistant breast cancer cells.
Endocrinology
149(
10):
4912-
4920,
2008.
- [51]
Taylor
KM
,
Hiscox
S
,
Nicholson
R
,
Hogstrand
C
,
Kille
P
:
Protein kinase CK2 triggers cytosolic zinc signaling pathways by phosphorylation of zinc channel ZIP7.
Sci Signal
5(
210):
ra11,
2012.
- [52]
Ziliotto
S
,
Gee
JMW
,
Ellis
IO
,
Green
AR
,
Finlay
P
,
Gobbato
A
,
Taylor
KM
:
Activated zinc transporter ZIP7 as an indicator of anti-hormone resistance in breast cancer.
Metallomics
11(
9):
1579-
1592,
2019.
- [53]
Mei
Z
,
Yan
P
,
Wang
Y
,
Liu
S
,
He
F
:
Knockdown of zinc transporter ZIP8 expression inhibits neuroblastoma progression and metastasis in vitro.
Mol Med Rep
18(
1):
477-
485,
2018.
- [54]
Thomas
P
,
Dong
J
:
(-)-Epicatechin acts as a potent agonist of the membrane androgen receptor, ZIP9 (SLC39A9), to promote apoptosis of breast and prostate cancer cells.
J Steroid Biochem Mol Biol
211:
105906,
2021.
- [55]
Takatani-Nakase
T
,
Matsui
C
,
Maeda
S
,
Kawahara
S
,
Takahashi
K
:
High glucose level promotes migration behavior of breast cancer cells through zinc and its transporters.
PLoS One
9(
2):
e90136,
2014.
- [56]
Takatani-Nakase
T
:
Migration behavior of breast cancer cells in the environment of high glucose level and the role of zinc and its transporter.
Yakugaku Zasshi
133(
11),
1195-
1199,
2013.
- [57]
Kagara
N
,
Tanaka
N
,
Noguchi
S
,
Hirano
T
:
Zinc and its transporter ZIP10 are involved in invasive behavior of breast cancer cells.
Cancer Sci
98(
5):
692-
697,
2007.
- [58]
Wu
L
,
Chaffee
KG
,
Parker
AS
,
Sicotte
H
,
Petersen
GM
.:
Zinc transporter genes and urological cancers: integrated analysis suggests a role for ZIP11 in bladder cancer.
Tumour Biol
36(
10):
7431-
7437,
2015.
- [59]
Zhu
B
,
Huo
R
,
Zhi
Q
,
Zhan
M
,
Chen
X
,
Hua
ZC
:
Increased expression of zinc transporter ZIP4, ZIP11, ZnT1, and ZnT6 predicts poor prognosis in pancreatic cancer.
J Trace Elem Med Biol
65:
126734,
2021.
- [60]
Cheng
X
,
Wang
J
,
Liu
C
,
Jiang
T
,
Yang
N
,
Liu
D
,
Zhao
H
,
Xu
Z
:
Zinc transporter SLC39A13/ZIP13 facilitates the metastasis of human ovarian cancer cells via activating Src/FAK signaling pathway.
J Exp Clin Cancer Res
40(
1):
199,
2021.
- [61]
Wang
G
,
Biswas
AK
,
Ma
W
,
Kandpal
M
,
Coker
C
,
Grandgenett
PM
,
Hollingsworth
MA
,
Jain
R
,
Tanji
K
,
Lόpez-Pintado
S
,
Borczuk
A
,
Hebert
D
,
Jenkitkasemwong
S
,
Hojyo
S
,
Davuluri
RV
,
Knutson
MD
,
Fukada
T
,
Acharyya
S
:
Metastatic cancers promote cachexia through ZIP14 upregulation in skeletal muscle.
Nat Med
24(
6):
770-
781,
2018.
- [62]
Jouybari
L
,
Kiani
F
,
Akbari
A
,
Sanagoo
A
,
Sayehmiri
F
,
Aaseth
J
,
Chartrand
MS
,
Sayehmiri
K
,
Chirumbolo
S
,
Bjørklund
G
:
A meta-analysis of zinc levels in breast cancer.
J Trace Elem Med Biol
56:
90-
99,
2019.
- [63]
Feng
Y
,
Zeng
JW
,
Ma
Q
,
Zhang
S
,
Tang
J
,
Feng
JF
:
Serum copper and zinc levels in breast cancer: A meta-analysis.
J Trace Elem Med Biol
62:
126629,
2020.
- [64]
Rusch
P
,
Hirner
AV
,
Schmitz
O
,
Kimmig
R
,
Hoffmann
O
,
Diel
M
:
Zinc distribution within breast cancer tissue of different intrinsic subtypes.
Arch Gynecol Obstet
303(
1):
195-
205,
2021.
- [65]
Gumulec
J
,
Masarik
M
,
Krizkova
S
,
Adam
V
,
Hubalek
J
,
Hrabeta
J
,
Eckschlager
T
,
Stiborova
M
,
Kizek
R
:
Insight to physiology and pathology of zinc(II) ions and their actions in breast and prostate carcinoma.
Curr Med Chem
18(
33):
5041-
5051,
2011.
- [66]
Manning
DL
,
Daly
RJ
,
Lord
PG
,
Kelly
KF
,
Green
CD
:
Effects of oestrogen on the expression of a 4.4 kb mRNA in the ZR-75-1 human breast cancer cell line.
Mol Cell Endocrinol
59(
3):
205-
212,
1988.
- [67]
Manning
DL
,
Robertson
JF
,
Ellis
IO
,
Elston
CW
,
McClelland
RA
,
Gee
JM
,
Jones
RJ
,
Green
CD
,
Cannon
P
,
Blamey
RW
,
Nicholson
RI
:
Oestrogen-regulated genes in breast cancer: association of pLIV1 with lymph node involvement.
Eur J Cancer
30A(
5):
675-
678,
1994.
- [68]
Muz
B
,
de la Puente
P
,
Azab
F
,
Azab
AK
:
The role of hypoxia in cancer progression, angiogenesis, metastasis, and resistance to therapy.
Hypoxia
3:
83-
92,
2015.
- [69]
Lundgren
K
,
Nordenskjold
B
,
Landberg
G
:
Hypoxia, Snail and incomplete epithelial-mesenchymal transition in breast cancer.
Br J Cancer
101(
10):
1769-
1781,
2009.
- [70]
Dave
B
,
Mitta
V
,
Tan
NM
,
Chang
JC
:
Epithelial-mesenchymal transition, cancer stem cells and treatment resistance.
Breast Cancer Res
14(
1):
202,
2012.
- [71]
Gao
T
,
Li
JZ
,
Lu
Y
,
Zhang
CY
,
Li
Q
,
Mao
J
,
Li
LH
:
The mechanism between epithelial mesenchymal transition in breast cancer and hypoxia microenvironment.
Biomed Pharmacother
80:
393-
405,
2016.
- [72]
McKeown
SR
:
Defining normoxia, physoxia and hypoxia in tumours—implications for treatment response.
Br J Radiol
87(
1035):
20130676,
2014.
- [73]
Kaelin
WG Jr
,
Ratcliffe
PJ
:
Oxygen sensing by metazoans: the central role of the HIF hydroxylase pathway.
Mol Cell
30(
4):
393-
402,
2008.
- [74]
Yoo
YG
,
Kong
G
,
Lee
MO
:
Metastasis-associated protein 1 enhances stability of hypoxia-inducible factor-1alpha protein by recruiting histone deacetylase 1.
EMBO J
25(
6):
1231-
1241,
2006.
- [75]
Early Breast Cancer Trialistsʼ Collaborative Group (EBCTCG);
Davies
C
,
Godwin
J
,
Gray
R
,
Clarke
M
,
Cutter
D
,
Darby
S
,
McGale
P
,
Pan
HC
,
Taylor
C
,
Wang
YC
,
Dowsett
M
,
Ingle
J
,
Peto
R
:
Relevance of breast cancer hormone receptors and other factors to the efficacy of adjuvant tamoxifen: patient-level meta-analysis of randomised trials.
Lancet
378(
9793):
771-
784,
2011.
- [76]
Musgrove
EA
,
Sutherland
RL
:
Biological determinants of endocrine resistance in breast cancer.
Nat Rev Cancer
9(
9):
631-
643,
2009.
- [77]
Yu
B
,
Sun
X
,
Shen
HY
,
Gao
F
,
Fan
YM
,
Sun
ZJ
:
Expression of the apoptosis-related genes BCL-2 and BAD in human breast carcinoma and their associated relationship with chemosensitivity.
J Exp Clin Cancer Res
29(
1):
107,
2010.
- [78]
Suvarna
V
,
Singh
V
,
Murahari
M
:
Current overview on the clinical update of Bcl-2 anti-apoptotic inhibitors for cancer therapy.
Eur J Pharmacol
862:
172655,
2019.
- [79]
Real
PJ
,
Cao
Y
,
Wang
R
,
Nikolovska-Coleska
Z
,
Sanz-Ortiz
J
,
Wang
S
,
Fernandez-Luna
JL
:
Breast cancer cells can evade apoptosis-mediated selective killing by a novel small molecule inhibitor of Bcl-2.
Cancer Res
64(
21):
7947-
7953,
2004.
- [80]
Wang
C
,
Hu
Q
,
Shen
HM
:
Pharmacological inhibitors of autophagy as novel cancer therapeutic agents.
Pharmacol Res
105:
164-
175,
2016.
- [81]
Lindqvist
LM
,
Simon
AK
,
Baehrecke
EH
:
Current questions and possible controversies in autophagy.
Cell Death Discov
1:
15036,
2015.
- [82]
Li
X
,
He
S
,
Ma
B
:
Autophagy and autophagy-related proteins in cancer.
Mol Cancer
19(
1):
12,
2020.
- [83]
Hwang
JJ
,
Kim
HN
,
Kim
J
,
Cho
DH
,
Kim
MJ
,
Kim
YS
,
Kim
Y
,
Park
SJ
,
Koh
JY
:
Zinc(II) ion mediates tamoxifen-induced autophagy and cell death in MCF-7 breast cancer cell line.
Biometals
23(
6):
997-
1013,
2010.
- [84]
Liuzzi
JP
,
Guo
L
,
Yoo
C
,
Stewart
TS
:
Zinc and autophagy.
Biometals
27(
6):
1087-
1096,
2014.
- [85]
Lopez
V
,
Foolad
F
,
Kelleher
SL
:
ZnT2-overexpression represses the cytotoxic effects of zinc hyper-accumulation in malignant metallothionein-null T47D breast tumor cells.
Cancer Lett
304(
1):
41-
51,
2011.
- [86]
Beck
B
,
Blanpain
C
:
Unravelling cancer stem cell potential.
Nat Rev Cancer
13(
10):
727-
738,
2013.
- [87]
Liu
S
,
Wicha
MS
:
Targeting breast cancer stem cells.
J Clin Oncol
28(
25):
4006-
4012,
2010.
- [88]
Kreso
A
,
Dick
JE
:
Evolution of the cancer stem cell model.
Cell Stem Cell
14(
3):
275-
291,
2014.
- [89]
Tang
T
,
Yang
Z
,
Zhu
Q
,
Wu
Y
,
Sun
K
,
Alahdal
M
,
Zhang
Y
,
Xing
Y
,
Shen
Y
,
Xia
T
,
Xi
T
,
Pan
Y
,
Jin
L
:
Up-regulation of miR-210 induced by a hypoxic microenvironment promotes breast cancer stem cells metastasis, proliferation, and self-renewal by targeting E-cadherin.
FASEB J
32:
6965-
6981,
2018.
- [90]
Taniguchi
H
,
Hoshino
D
,
Moriya
C
,
Zembutsu
H
,
Nishiyama
N
,
Yamamoto
H
,
Kataoka
K
,
Imai
K
:
Silencing PRDM14 expression by an innovative RNAi therapy inhibits stemness, tumorigenicity, and metastasis of breast cancer.
Oncotarget
8(
29):
46856-
46874,
2017.
- [91]
Nimmanon
T
,
Ziliotto
S
,
Morris
S
,
Flanagan
L
,
Taylor
KM
:
Phosphorylation of zinc channel ZIP7 drives MAPK, PI3K and mTOR growth and proliferation signalling.
Metallomics
9(
5):
471-
481,
2017.
- [92]
Ziliotto
S
,
Gee
JMW
,
Ellis
IO
,
Green
AR
,
Finlay
P
,
Gobbato
A
,
Taylor
KM
:
Activated zinc transporter ZIP7 as an indicator of anti-hormone resistance in breast cancer.
Metallomics
11(
9):
1579-
1592,
2019.
- [93]
Ziliotto
S
,
Ogle
O
,
Taylor
KM
:
Targeting Zinc(II) Signalling to Prevent Cancer. Astrid Sigel, Helmut Sigel, Eva Freisinger, Roland K.O. Sigel (ed): Metallo-Drugs: Development and Action of Anticancer Agents,
Met Ions Life Sci
18,
2018,
507-
530.
- [94]
Taniguchi
H
,
Imai
K
:
Silencing PRDM14 via oligonucleotide therapeutics suppresses tumorigenicity and metastasis of breast cancer.
Methods Mol Biol
1974:
233-
243,
2019.
- [95]
Ehsani
S
,
Huo
H
,
Salehzadeh
A
,
Pocanschi
CL
,
Watts
JC
,
Wille
H
,
Westaway
D
,
Rogaeva
E
,
St George-Hyslop
PH
,
Schmitt-Ulms
G
:
Family reunion--the ZIP/prion gene family.
Prog Neurobiol
93(
3):
405-
420,
2011.
- [96]
Ehsani
S
,
Salehzadeh
A
,
Huo
H
,
Reginold
W
,
Pocanschi
CL
,
Ren
H
,
Wang
H
,
So
K
,
Sato
C
,
Mehrabian
M
,
Strome
R
,
Trimble
WS
,
Hazrati
LN
,
Rogaeva
E
,
Westaway
D
,
Carlson
GA
,
Schmitt-Ulms
G
:
LIV-1 ZIP ectodomain shedding in prion-infected mice resembles cellular response to transition metal starvation.
J Mol Biol
422(
4):
556-
574,
2012.
- [97]
Pocanschi
CL
,
Ehsani
S
,
Mehrabian
M
,
Wille
H
,
Reginold
W
,
Trimble
WS
,
Wang
H
,
Yee
A
,
Arrowsmith
CH
,
Bozóky
Z
,
Kay
LE
,
Forman-Kay
JD
,
Rini
JM
,
Schmitt-Ulms
G
:
The ZIP5 ectodomain co-localizes with PrP and may acquire a PrP-like fold that assembles into a dimer.
PLoS One
8(
9):
e72446,
2013.
- [98]
Schmitt-Ulms
G
,
Ehsani
S
,
Watts
JC
,
Westaway
D
,
Wille
H
:
Evolutionary descent of prion genes from the ZIP family of metal ion transporters.
PLoS One
4(
9):
e7208,
2009.
- [99]
Taylor
KM
,
Muraina
IA
,
Brethour
D
,
Schmitt-Ulms
G
,
Nimmanon
T
,
Ziliotto
S
,
Kille
P
,
Hogstrand
C
:
Zinc transporter ZIP10 forms a heteromer with ZIP6 which regulates embryonic development and cell migration.
Biochem J
473(
16):
2531-
2544,
2016.
- [100]
Brethour
D
,
Mehrabian
M
,
Williams
D
,
Wang
X
,
Ghodrati
F
,
Ehsani
S
,
Rubie
EA
,
Woodgett
JR
,
Sevalle
J
,
Xi
Z
,
Rogaeva
E
,
Schmitt-Ulms
G
:
A ZIP6-ZIP10 heteromer controls NCAM1 phosphorylation and integration into focal adhesion complexes during epithelial-to-mesenchymal transition.
Sci Rep
7:
40313,
2017.
- [101]
Nolin
E
,
Gans
S
,
Llamas
L
,
Bandyopadhyay
S
,
Brittain
SM
,
Bernasconi-Elias
P
,
Carter
KP
,
Loureiro
JJ
,
Thomas
JR
,
Schirle
M
,
Yang
Y
,
Guo
N
,
Roma
G
,
Schuierer
S
,
Beibel
M
,
Lindeman
A
,
Sigoillot
F
,
Chen
A
,
Xie
KX
,
Ho
S
,
Reece-Hoyes
J
,
Weihofen
WA
,
Tyskiewicz
K
,
Hoepfner
D
,
McDonald
RI
,
Guthrie
N
,
Dogra
A
,
Guo
H
,
Shao
J
,
Ding
J
,
Canham
SM
,
Boynton
G
,
George
EL
,
Kang
ZB
,
Antczak
C
,
Porter
JA
,
Wallace
O
,
Tallarico
JA
,
Palmer
AE
,
Jenkins
JL
,
Jain
RK
,
Bushell
SM
,
Fryer
CJ
:
Discovery of a ZIP7 inhibitor from a Notch pathway screen.
Nat Chem Biol
15(
2):
179-
188,
2019.