Advances of Zinc Signaling Studies in Prostate Cancer
Abstract
:1. Introduction
2. Zinc Levels in Prostate Tissues and Sera of PCa Patients
3. Biological Functions of Zinc in PCa
3.1. Zinc and Its Anti-Proliferative Activities
3.2. Zinc and Its Role in Cell Death
3.3. Zinc and Its Anti-Metastasis Effects
4. Zinc-Associated Compounds and Their Functions in PCa
4.1. Zinc Transporters
4.2. Zinc Finger-Containing Transcription Factors
5. Clinical Applications of Zinc Signaling in PCa
6. Conclusions and Future Prospects
Author Contributions
Funding
Conflicts of Interest
Abbreviations
AAS | Atomic absorption spectrophotometry |
AP-N | Aminopeptidase N |
AR | Androgen receptor |
BAPTA | 1,2-bis(o-aminophenoxy)ethane-N,N,N′,N′-tetraacetic acid |
BAX | BCL2-associated X protein |
BCL2 | B cell leukemia/lymphoma 2 |
bFGF | Basic fibroblast growth factor |
CD164 | CD164 antigen, sialomucin |
DHT | Dihydrotestosterone |
ECM | Extracellular matrix |
EMT | Epithelial-to-mesenchymal transformation |
ERK1/2 | Extracellular signal-regulated kinase 1/2 |
FBL | Fibrillarin |
HDAC1 | Histone deacetylase 1 |
HIF-1α | Hypoxia inducible factor-1α |
ICAM-1 | Intercellular adhesion molecule 1 |
iCEST | Ion chemical exchange saturation transfer |
IGF-1 | Insulin like growth factor 1 |
IGFBP-3 | Insulin like growth factor binding protein 3 |
IHC | Immunohistochemistry |
IL-1 | Interleukin 1 |
JNK | c-Jun N-terminal kinase |
MAPKs | Mitogen activated protein kinases |
MDM2 | Murine double minute 2 |
MMP-9 | Matrix metallopeptidase 9 |
MRI | Magnetic resonance imaging |
mTOR | Mechanistic target of rapamycin kinase |
NCoR | Nuclear receptor corepressor 1 |
NF-κB | Nuclear factor kappa B |
PCa | Prostate cancer |
PCNA | Proliferating cell nuclear antigen |
PIAS1 | Protein inhibitor of activated STAT 1 |
PI3K | Phosphoinositide 3-kinase |
PKC | Protein kinase C |
PLZF | Promyelocytic leukemia zinc finger |
PrEC | Prostatic epithelial cell |
PSA | Prostate-specific antigen |
PTEN | Phosphatase and tensin homolog |
RREB-1 | Ras responsive element binding protein 1 |
SP1 | Specificity protein 1 |
TF-BAPTA | 5,5′,6,6′-tetrafluoro-BAPTA |
TFs | Transcription factors |
TGFβ | Transforming growth factor β |
TME | Tumor microenvironment |
TNF-α | Tumor necrosis factor α |
TPEN | N,N,N′,N′-tetrakis(2-pyridylmethyl)-ethylenediamine |
TRAMP | Transgenic adenocarcinoma of the mouse prostate |
Treg | Regulatory T cell |
UTR | Untranslated regions |
VEGF | Vascular endothelial growth factor |
VHR | Vaccinia H1-related phosphatase |
ZAP-70 | Zeta chain-associated protein-70 |
ZFs | Zinc fingers |
ZIPs | ZRT- and Irt-like proteins |
ZnTs | Zinc transporters |
ZP | Zn-pyrithione |
ZPP1 | Zinpyr (ZP) family of zinc probes |
References
- Connor, M.J.; Shah, T.T.; Horan, G.; Bevan, C.L.; Winkler, M.; Ahmed, H.U. Cytoreductive treatment strategies for de novo metastatic prostate cancer. Nat. Rev. Clin. Oncol. 2019. [Google Scholar] [CrossRef]
- Farashi, S.; Kryza, T.; Clements, J.; Batra, J. Post-GWAS in prostate cancer: From genetic association to biological contribution. Nat. Rev. Cancer 2019, 19, 46–59. [Google Scholar] [CrossRef]
- Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2019. CA Cancer J. Clin. 2019, 69, 7–34. [Google Scholar] [CrossRef] [Green Version]
- Huland, H.; Graefen, M. Changing Trends in Surgical Management of Prostate Cancer: The End of Overtreatment? Eur. Urol. 2015, 68, 175–178. [Google Scholar] [CrossRef] [PubMed]
- Barry, M.J.; Simmons, L.H. Prevention of Prostate Cancer Morbidity and Mortality: Primary Prevention and Early Detection. Med. Clin. N. Am. 2017, 101, 787–806. [Google Scholar] [CrossRef] [PubMed]
- Lilja, H.; Ulmert, D.; Vickers, A.J. Prostate-specific antigen and prostate cancer: Prediction, detection and monitoring. Nat. Rev. Cancer 2008, 8, 268–278. [Google Scholar] [CrossRef] [PubMed]
- Carlsson, S.V.; Kattan, M.W. Prostate cancer: Personalized risk—Stratified screening or abandoning it altogether? Nat. Rev. Clin. Oncol. 2016, 13, 140–142. [Google Scholar] [CrossRef]
- Eidelman, E.; Twum-Ampofo, J.; Ansari, J.; Siddiqui, M.M. The Metabolic Phenotype of Prostate Cancer. Front. Oncol. 2017, 7, 131. [Google Scholar] [CrossRef]
- James, N.D.; de Bono, J.S.; Spears, M.R.; Clarke, N.W.; Mason, M.D.; Dearnaley, D.P.; Ritchie, A.W.S.; Amos, C.L.; Gilson, C.; Jones, R.J.; et al. Abiraterone for Prostate Cancer Not Previously Treated with Hormone Therapy. N. Engl. J. Med. 2017, 377, 338–351. [Google Scholar] [CrossRef]
- Jani, A.B.; Hellman, S. Early prostate cancer: Clinical decision-making. Lancet 2003, 361, 1045–1053. [Google Scholar] [CrossRef]
- Martel, C.L.; Gumerlock, P.H.; Meyers, F.J.; Lara, P.N. Current strategies in the management of hormone refractory prostate cancer. Cancer Treat. Rev. 2003, 29, 171–187. [Google Scholar] [CrossRef]
- Kumar, S.; Singh, R.; Malik, S.; Manne, U.; Mishra, M. Prostate cancer health disparities: An immuno-biological perspective. Cancer Lett. 2018, 414, 153–165. [Google Scholar] [CrossRef] [PubMed]
- Pascual-Geler, M.; Urquiza-Salvat, N.; Cozar, J.M.; Robles-Fernandez, I.; Rivas, A.; Martinez-Gonzalez, L.J.; Ocana-Peinado, F.M.; Lorente, J.A.; Alvarez-Cubero, M.J. The influence of nutritional factors on prostate cancer incidence and aggressiveness. Aging Male 2018, 21, 31–39. [Google Scholar] [CrossRef] [PubMed]
- Ho, E.; Beaver, L.M.; Williams, D.E.; Dashwood, R.H. Dietary factors and epigenetic regulation for prostate cancer prevention. Adv. Nutr. 2011, 2, 497–510. [Google Scholar] [CrossRef] [Green Version]
- Han, Y.; Zhang, R.; Dong, C.; Cheng, F.; Guo, Y. Sensitive electrochemical sensor for nitrite ions based on rose-like AuNPs/MoS2/graphene composite. Biosens. Bioelectron. 2019, 142, 111529. [Google Scholar] [CrossRef]
- Yu, H.N.; Shen, S.R.; Yin, J.J. Effects of metal ions, catechins, and their interactions on prostate cancer. Crit. Rev. Food Sci. Nutr. 2007, 47, 711–719. [Google Scholar] [CrossRef]
- Krezel, A.; Maret, W. The biological inorganic chemistry of zinc ions. Arch. Biochem. Biophys. 2016, 611, 3–19. [Google Scholar] [CrossRef] [Green Version]
- Maywald, M.; Wessels, I.; Rink, L. Zinc Signals and Immunity. Int. J. Mol. Sci. 2017, 18, 2222. [Google Scholar] [CrossRef] [Green Version]
- Chasapis, C.T.; Loutsidou, A.C.; Spiliopoulou, C.A.; Stefanidou, M.E. Zinc and human health: An update. Arch. Toxicol. 2012, 86, 521–534. [Google Scholar] [CrossRef]
- Andreini, C.; Banci, L.; Bertini, I.; Rosato, A. Counting the zinc-proteins encoded in the human genome. J. Proteome Res. 2006, 5, 196–201. [Google Scholar] [CrossRef]
- Kambe, T.; Matsunaga, M.; Takeda, T.A. Understanding the Contribution of Zinc Transporters in the Function of the Early Secretory Pathway. Int. J. Mol. Sci. 2017, 18, 2179. [Google Scholar] [CrossRef] [Green Version]
- Zhao, T.; Huang, Q.; Su, Y.; Sun, W.; Huang, Q.; Wei, W. Zinc and its regulators in pancreas. Inflammopharmacology 2019, 27, 453–464. [Google Scholar] [CrossRef] [PubMed]
- Kolenko, V.; Teper, E.; Kutikov, A.; Uzzo, R. Zinc and zinc transporters in prostate carcinogenesis. Nat. Rev. Urol. 2013, 10, 219–226. [Google Scholar] [CrossRef] [Green Version]
- Portbury, S.D.; Adlard, P.A. Zinc Signal in Brain Diseases. Int. J. Mol. Sci. 2017, 18, 2506. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ho, E.; Ames, B.N. Low intracellular zinc induces oxidative DNA damage, disrupts p53, NFkappa B, and AP1 DNA binding, and affects DNA repair in a rat glioma cell line. Proc. Natl. Acad. Sci. USA 2002, 99, 16770–16775. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ziliotto, S.; Ogle, O.; Taylor, K.M. Targeting Zinc(II) Signalling to Prevent Cancer. Met. Ions Life Sci. 2018, 18. [Google Scholar] [CrossRef]
- Kambe, T.; Hashimoto, A.; Fujimoto, S. Current understanding of ZIP and ZnT zinc transporters in human health and diseases. Cell. Mol. Life Sci. 2014, 71, 3281–3295. [Google Scholar] [CrossRef]
- Pan, Z.; Choi, S.; Ouadid-Ahidouch, H.; Yang, J.M.; Beattie, J.H.; Korichneva, I. Zinc transporters and dysregulated channels in cancers. Front. Biosci. 2017, 22, 623–643. [Google Scholar] [CrossRef] [Green Version]
- Prasad, A.S. Discovery of human zinc deficiency: 50 years later. J. Trace Elem. Med. Biol. 2012, 26, 66–69. [Google Scholar] [CrossRef]
- Wakwe, V.C.; Odum, E.P.; Amadi, C. The impact of plasma zinc status on the severity of prostate cancer disease. Investig. Clin. Urol. 2019, 60, 162–168. [Google Scholar] [CrossRef]
- Aydin, A.; Arsova-Sarafinovska, Z.; Sayal, A.; Eken, A.; Erdem, O.; Erten, K.; Ozgok, Y.; Dimovski, A. Oxidative stress and antioxidant status in non-metastatic prostate cancer and benign prostatic hyperplasia. Clin. Biochem. 2006, 39, 176–179. [Google Scholar] [CrossRef] [PubMed]
- Christudoss, P.; Selvakumar, R.; Fleming, J.J.; Gopalakrishnan, G. Zinc status of patients with benign prostatic hyperplasia and prostate carcinoma. Indian J. Urol. 2011, 27, 14–18. [Google Scholar] [CrossRef] [PubMed]
- Darago, A.; Sapota, A.; Matych, J.; Nasiadek, M.; Skrzypinska-Gawrysiak, M.; Kilanowicz, A. The correlation between zinc and insulin-like growth factor 1 (IGF-1), its binding protein (IGFBP-3) and prostate-specific antigen (PSA) in prostate cancer. Clin. Chem. Lab. Med. 2011, 49, 1699–1705. [Google Scholar] [CrossRef] [PubMed]
- Kaba, M.; Pirincci, N.; Yuksel, M.B.; Gecit, I.; Gunes, M.; Ozveren, H.; Eren, H.; Demir, H. Serum levels of trace elements in patients with prostate cancer. Asian Pac. J. Cancer Prev. 2014, 15, 2625–2629. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Costello, L.C.; Franklin, R.B. A comprehensive review of the role of zinc in normal prostate function and metabolism; and its implications in prostate cancer. Arch. Biochem. Biophys. 2016, 611, 100–112. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhao, J.; Wu, Q.; Hu, X.; Dong, X.; Wang, L.; Liu, Q.; Long, Z.; Li, L. Comparative study of serum zinc concentrations in benign and malignant prostate disease: A Systematic Review and Meta-Analysis. Sci. Rep. 2016, 6, 25778. [Google Scholar] [CrossRef] [Green Version]
- Costello, L.C.; Franklin, R.B. Decreased zinc in the development and progression of malignancy: An important common relationship and potential for prevention and treatment of carcinomas. Expert Opin. Ther. Targets 2017, 21, 51–66. [Google Scholar] [CrossRef] [Green Version]
- Costello, L.C.; Franklin, R.B. Zinc is decreased in prostate cancer: An established relationship of prostate cancer! J. Biol. Inorg. Chem. 2011, 16, 3–8. [Google Scholar] [CrossRef]
- Franklin, R.B.; Feng, P.; Milon, B.; Desouki, M.M.; Singh, K.K.; Kajdacsy-Balla, A.; Bagasra, O.; Costello, L.C. hZIP1 zinc uptake transporter down regulation and zinc depletion in prostate cancer. Mol. Cancer 2005, 4, 32. [Google Scholar] [CrossRef] [Green Version]
- Clavijo Jordan, M.V.; Lo, S.T.; Chen, S.; Preihs, C.; Chirayil, S.; Zhang, S.; Kapur, P.; Li, W.H.; De Leon-Rodriguez, L.M.; Lubag, A.J.; et al. Zinc-sensitive MRI contrast agent detects differential release of Zn(II) ions from the healthy vs. malignant mouse prostate. Proc. Natl. Acad. Sci. USA 2016, 113, E5464–E5471. [Google Scholar] [CrossRef] [Green Version]
- Yuan, Y.; Wei, Z.; Chu, C.; Zhang, J.; Song, X.; Walczak, P.; Bulte, J.W.M. Development of Zinc-Specific iCEST MRI as an Imaging Biomarker for Prostate Cancer. Angew. Chem. Int. Ed. Engl. 2019, 58, 15512–15517. [Google Scholar] [CrossRef] [PubMed]
- Hoang, B.X.; Han, B.; Shaw, D.G.; Nimni, M. Zinc as a possible preventive and therapeutic agent in pancreatic, prostate, and breast cancer. Eur. J. Cancer Prev. 2016, 25, 457–461. [Google Scholar] [CrossRef] [PubMed]
- Singh, C.K.; Pitschmann, A.; Ahmad, N. Resveratrol-zinc combination for prostate cancer management. Cell Cycle 2014, 13, 1867–1874. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Franz, M.C.; Anderle, P.; Burzle, M.; Suzuki, Y.; Freeman, M.R.; Hediger, M.A.; Kovacs, G. Zinc transporters in prostate cancer. Mol. Asp. Med. 2013, 34, 735–741. [Google Scholar] [CrossRef] [Green Version]
- Zaichick, V.; Sviridova, T.V.; Zaichick, S.V. Zinc in the human prostate gland: Normal, hyperplastic and cancerous. Int. Urol. Nephrol. 1997, 29, 565–574. [Google Scholar] [CrossRef]
- Costello, L.C.; Feng, P.; Milon, B.; Tan, M.; Frankin, R.B. Role of zinc in the pathogenesis and treatment of prostate cancer: Critical issues to resolve. Prostate Cancer Prostatic Dis. 2004, 7, 111–117. [Google Scholar] [CrossRef] [Green Version]
- Takatani-Nakase, T. Zinc Transporters and the Progression of Breast Cancers. Biol. Pharm. Bull. 2018, 41, 1517–1522. [Google Scholar] [CrossRef] [Green Version]
- Yu, Z.; Yu, Z.; Chen, Z.; Yang, L.; Ma, M.; Lu, S.; Wang, C.; Teng, C.; Nie, Y. Zinc chelator TPEN induces pancreatic cancer cell death through causing oxidative stress and inhibiting cell autophagy. J. Cell. Physiol. 2019, 234, 20648–20661. [Google Scholar] [CrossRef]
- Ninsontia, C.; Phiboonchaiyanan, P.P.; Kiratipaiboon, C.; Chanvorachote, P. Zinc suppresses stem cell properties of lung cancer cells through protein kinase C-mediated beta-catenin degradation. Am. J. Physiol. Cell Physiol. 2017, 312, C487–C499. [Google Scholar] [CrossRef] [Green Version]
- Hosui, A.; Kimura, E.; Abe, S.; Tanimoto, T.; Onishi, K.; Kusumoto, Y.; Sueyoshi, Y.; Matsumoto, K.; Hirao, M.; Yamada, T.; et al. Long-Term Zinc Supplementation Improves Liver Function and Decreases the Risk of Developing Hepatocellular Carcinoma. Nutrients 2018, 10, 1955. [Google Scholar] [CrossRef] [Green Version]
- Mawson, C.A.; Fischer, M.I. The occurrence of zinc in the human prostate gland. Can. J. Med. Sci. 1952, 30, 336–339. [Google Scholar] [CrossRef] [PubMed]
- Costello, L.C.; Franklin, R.B. Novel role of zinc in the regulation of prostate citrate metabolism and its implications in prostate cancer. Prostate 1998, 35, 285–296. [Google Scholar] [CrossRef]
- Platz, E.A.; Helzlsouer, K.J. Selenium, zinc, and prostate cancer. Epidemiol. Rev. 2001, 23, 93–101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ogunlewe, J.O.; Osegbe, D.N. Zinc and cadmium concentrations in indigenous blacks with normal, hypertrophic, and malignant prostate. Cancer 1989, 63, 1388–1392. [Google Scholar] [CrossRef]
- Zaichick, V.Y.; Sviridova, T.V.; Zaichick, S.V. Zinc concentration in human prostatic fluid: Normal, chronic prostatitis, adenoma and cancer. Int. Urol. Nephrol. 1996, 28, 687–694. [Google Scholar] [CrossRef] [PubMed]
- Johnson, L.A.; Kanak, M.A.; Kajdacsy-Balla, A.; Pestaner, J.P.; Bagasra, O. Differential zinc accumulation and expression of human zinc transporter 1 (hZIP1) in prostate glands. Methods 2010, 52, 316–321. [Google Scholar] [CrossRef]
- Györkey, F.; Min, K.W.; Huff, J.A.; Györkey, P. Zinc and magnesium in human prostate gland: Normal, hyperplastic, and neoplastic. Cancer Res. 1967, 27, 1348–1353. [Google Scholar]
- Ghosh, S.K.; Kim, P.; Zhang, X.A.; Yun, S.H.; Moore, A.; Lippard, S.J.; Medarova, Z. A novel imaging approach for early detection of prostate cancer based on endogenous zinc sensing. Cancer Res. 2010, 70, 6119–6127. [Google Scholar] [CrossRef] [Green Version]
- Li, X.M.; Zhang, L.; Li, J.; Li, Y.; Wang, H.L.; Ji, G.Y.; Kuwahara, M.; Zhao, X.J. Measurement of serum zinc improves prostate cancer detection efficiency in patients with PSA levels between 4 ng/mL and 10 ng/mL. Asian J. Androl. 2005, 7, 323–328. [Google Scholar] [CrossRef]
- Onyema-iloh, B.O.; Meludu, S.C.; Iloh, E.; Nnodim, J.; Onyegbule, O.; Mykembata, B. Biochemical changes in some trace elements, antioxidant vitamins and their therapeutic importance in prostate cancer patients. Asian J. Med. Sci. 2015, 6, 95–97. [Google Scholar] [CrossRef] [Green Version]
- Chen, G.; Wu, L.; Wu, Y.; Zhu, J. Content change of zinc and cadmium in serum of patients with prostate cancer and its clinical significance. J. Clin. Urol. 2015, 30, 439–441. [Google Scholar] [CrossRef]
- Feustel, A.; Wennrich, R.; Schmidt, B. Serum-Zn-levels in prostatic cancer. Urol. Res. 1989, 17, 41–42. [Google Scholar] [CrossRef]
- Park, S.Y.; Wilkens, L.R.; Morris, J.S.; Henderson, B.E.; Kolonel, L.N. Serum zinc and prostate cancer risk in a nested case-control study: The multiethnic cohort. Prostate 2013, 73, 261–266. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bialkowska, K.; Marciniak, W.; Muszynska, M.; Baszuk, P.; Gupta, S.; Jaworska-Bieniek, K.; Sukiennicki, G.; Durda, K.; Gromowski, T.; Prajzendanc, K.; et al. Association of zinc level and polymorphism in MMP-7 gene with prostate cancer in Polish population. PLoS ONE 2018, 13, e0201065. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yao, D.; Johnes, A.; Fragmann, C. The content of serum zinc concentration in prostate disease. Int. J. Surg. 1977, 4, 225–227. [Google Scholar]
- Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [Green Version]
- Wang, W.; Li, D.; Sui, G. YY1 Is an Inducer of Cancer Metastasis. Crit. Rev. Oncog. 2017, 22, 1–11. [Google Scholar] [CrossRef]
- Liang, J.Y.; Liu, Y.Y.; Zou, J.; Franklin, R.B.; Costello, L.C.; Feng, P. Inhibitory effect of zinc on human prostatic carcinoma cell growth. Prostate 1999, 40, 200–207. [Google Scholar] [CrossRef] [Green Version]
- Yan, M.; Hardin, K.; Ho, E. Differential response to zinc-induced apoptosis in benign prostate hyperplasia and prostate cancer cells. J. Nutr. Biochem. 2010, 21, 687–694. [Google Scholar] [CrossRef] [Green Version]
- Banudevi, S.; Senthilkumar, K.; Sharmila, G.; Arunkumar, R.; Vijayababu, M.R.; Arunakaran, J. Effect of zinc on regulation of insulin-like growth factor signaling in human androgen-independent prostate cancer cells. Clin. Chim. Acta 2010, 411, 172–178. [Google Scholar] [CrossRef]
- Uzzo, R.G.; Crispen, P.L.; Golovine, K.; Makhov, P.; Horwitz, E.M.; Kolenko, V.M. Diverse effects of zinc on NF-kappaB and AP-1 transcription factors: Implications for prostate cancer progression. Carcinogenesis 2006, 27, 1980–1990. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wong, P.F.; Abubakar, S. High intracellular Zn2+ ions modulate the VHR, ZAP-70 and ERK activities of LNCaP prostate cancer cells. Cell. Mol. Biol. Lett. 2008, 13, 375–390. [Google Scholar] [CrossRef] [PubMed]
- Han, C.T.; Schoene, N.W.; Lei, K.Y. Influence of zinc deficiency on Akt-Mdm2-p53 and Akt-p21 signaling axes in normal and malignant human prostate cells. Am. J. Physiol. Cell Physiol. 2009, 297, C1188–C1199. [Google Scholar] [CrossRef] [PubMed]
- Zhang, P.; Li, Y.; Tang, X.; Guo, R.; Li, J.; Chen, Y.Y.; Guo, H.; Su, J.; Sun, L.; Liu, Y. Zinc enhances chemosensitivity to paclitaxel in PC3 prostate cancer cells. Oncol. Rep. 2018, 40, 2269–2277. [Google Scholar] [CrossRef]
- To, P.K.; Do, M.H.; Cho, Y.S.; Kwon, S.Y.; Kim, M.S.; Jung, C. Zinc Inhibits Expression of Androgen Receptor to Suppress Growth of Prostate Cancer Cells. Int. J. Mol. Sci. 2018, 19, 3062. [Google Scholar] [CrossRef] [Green Version]
- Hacioglu, C.; Kacar, S.; Kar, F.; Kanbak, G.; Sahinturk, V. Concentration-Dependent Effects of Zinc Sulfate on DU-145 Human Prostate Cancer Cell Line: Oxidative, Apoptotic, Inflammatory, and Morphological Analyzes. Biol. Trace Elem. Res. 2019. [Google Scholar] [CrossRef]
- Wong, P.F.; Abubakar, S. Comparative transcriptional study of the effects of high intracellular zinc on prostate carcinoma cells. Oncol. Rep. 2010, 23, 1501–1516. [Google Scholar] [CrossRef]
- Tikkanen, R.; Nikolic-Paterson, D.J. Mitogen-Activated Protein Kinases: Functions in Signal Transduction and Human Diseases. Int. J. Mol. Sci. 2019, 20, 4844. [Google Scholar] [CrossRef] [Green Version]
- Meister, M.; Tomasovic, A.; Banning, A.; Tikkanen, R. Mitogen-Activated Protein (MAP) Kinase Scaffolding Proteins: A Recount. Int. J. Mol. Sci. 2013, 14, 4854–4884. [Google Scholar] [CrossRef] [Green Version]
- Banudevi, S.; Elumalai, P.; Arunkumar, R.; Senthilkumar, K.; Gunadharini, D.N.; Sharmila, G.; Arunakaran, J. Chemopreventive effects of zinc on prostate carcinogenesis induced by N-methyl-N-nitrosourea and testosterone in adult male Sprague-Dawley rats. J. Cancer Res. Clin. Oncol. 2011, 137, 677–686. [Google Scholar] [CrossRef]
- Shah, M.R.; Kriedt, C.L.; Lents, N.H.; Hoyer, M.K.; Jamaluddin, N.; Klein, C.; Baldassare, J. Direct intra-tumoral injection of zinc-acetate halts tumor growth in a xenograft model of prostate cancer. J. Exp. Clin. Cancer Res. 2009, 28, 84. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Prasad, A.S.; Mukhtar, H.; Beck, F.W.; Adhami, V.M.; Siddiqui, I.A.; Din, M.; Hafeez, B.B.; Kucuk, O. Dietary zinc and prostate cancer in the TRAMP mouse model. J. Med. Food 2010, 13, 70–76. [Google Scholar] [CrossRef] [PubMed]
- Gu, J.; Wang, B.; Liu, Y.; Zhong, L.; Tang, Y.; Guo, H.; Jiang, T.; Wang, L.; Li, Y.; Cai, L. Murine double minute 2 siRNA and wild-type p53 gene therapy interact positively with zinc on prostate tumours in vitro and in vivo. Eur. J. Cancer 2014, 50, 1184–1194. [Google Scholar] [CrossRef] [PubMed]
- Fong, L.Y.; Jing, R.; Smalley, K.J.; Wang, Z.X.; Taccioli, C.; Fan, S.; Chen, H.; Alder, H.; Huebner, K.; Farber, J.L.; et al. Human-like hyperplastic prostate with low ZIP1 induced solely by Zn deficiency in rats. Proc. Natl. Acad. Sci. USA 2018, 115, E11091–E11100. [Google Scholar] [CrossRef] [Green Version]
- Song, Y.; Ho, E. Zinc and prostatic cancer. Curr. Opin. Clin. Nutr. Metab. Care 2009, 12, 640–645. [Google Scholar] [CrossRef] [Green Version]
- Feng, P.; Li, T.L.; Guan, Z.X.; Franklin, R.B.; Costello, L.C. Direct effect of zinc on mitochondrial apoptogenesis in prostate cells. Prostate 2002, 52, 311–318. [Google Scholar] [CrossRef] [Green Version]
- Feng, P.; Li, T.; Guan, Z.; Franklin, R.B.; Costello, L.C. The involvement of Bax in zinc-induced mitochondrial apoptogenesis in malignant prostate cells. Mol. Cancer 2008, 7, 25. [Google Scholar] [CrossRef] [Green Version]
- Hong, S.H.; Choi, Y.S.; Cho, H.J.; Lee, J.Y.; Kim, J.C.; Hwang, T.K.; Kim, S.W. Antiproliferative effects of zinc-citrate compound on hormone refractory prostate cancer. Chin. J. Cancer Res. 2012, 24, 124–129. [Google Scholar] [CrossRef] [Green Version]
- Feng, P.; Li, T.L.; Guan, Z.X.; Franklin, R.B.; Costello, L.C. Effect of zinc on prostatic tumorigenicity in nude mice. Ann. N. Y. Acad. Sci. 2003, 1010, 316–320. [Google Scholar] [CrossRef]
- Ku, J.H.; Seo, S.Y.; Kwak, C.; Kim, H.H. The role of survivin and Bcl-2 in zinc-induced apoptosis in prostate cancer cells. Urol. Oncol. 2012, 30, 562–568. [Google Scholar] [CrossRef]
- Yang, N.; Zhao, B.; Rasul, A.; Qin, H.; Li, J.; Li, X. PIAS1-modulated Smad2/4 complex activation is involved in zinc-induced cancer cell apoptosis. Cell Death Dis. 2013, 4, e811. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Carraway, R.E.; Dobner, P.R. Zinc pyrithione induces ERK- and PKC-dependent necrosis distinct from TPEN-induced apoptosis in prostate cancer cells. Biochim. Biophys. Acta 2012, 1823, 544–557. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Green, D.R. The Coming Decade of Cell Death Research: Five Riddles. Cell 2019, 177, 1094–1107. [Google Scholar] [CrossRef] [PubMed]
- Bersuker, K.; Hendricks, J.M.; Li, Z.; Magtanong, L.; Ford, B.; Tang, P.H.; Roberts, M.A.; Tong, B.; Maimone, T.J.; Zoncu, R.; et al. The CoQ oxidoreductase FSP1 acts parallel to GPX4 to inhibit ferroptosis. Nature 2019. [Google Scholar] [CrossRef] [PubMed]
- Doll, S.; Freitas, F.P.; Shah, R.; Aldrovandi, M.; da Silva, M.C.; Ingold, I.; Grocin, A.G.; Xavier da Silva, T.N.; Panzilius, E.; Scheel, C.H.; et al. FSP1 is a glutathione-independent ferroptosis suppressor. Nature 2019. [Google Scholar] [CrossRef] [PubMed]
- Gudipaty, S.A.; Conner, C.M.; Rosenblatt, J.; Montell, D.J. Unconventional Ways to Live and Die: Cell Death and Survival in Development, Homeostasis, and Disease. Annu. Rev. Cell Dev. Biol. 2018, 34, 311–332. [Google Scholar] [CrossRef]
- Hu, Y.; Zhang, H.R.; Dong, L.; Xu, M.R.; Zhang, L.; Ding, W.P.; Zhang, J.Q.; Lin, J.; Zhang, Y.J.; Qiu, B.S.; et al. Enhancing tumor chemotherapy and overcoming drug resistance through autophagy-mediated intracellular dissolution of zinc oxide nanoparticles. Nanoscale 2019, 11, 11789–11807. [Google Scholar] [CrossRef]
- Kang, W.; Hong, S.H.; Lee, H.M.; Kim, N.Y.; Lim, Y.C.; Le le, T.M.; Lim, B.; Kim, H.C.; Kim, T.Y.; Ashida, H.; et al. Structural and biochemical basis for the inhibition of cell death by APIP, a methionine salvage enzyme. Proc. Natl. Acad. Sci. USA 2014, 111, E54–E61. [Google Scholar] [CrossRef] [Green Version]
- Palmer, L.D.; Jordan, A.T.; Maloney, K.N.; Farrow, M.A.; Gutierrez, D.B.; Gant-Branum, R.; Burns, W.J.; Romer, C.E.; Tsui, T.; Allen, J.L.; et al. Zinc intoxication induces ferroptosis in A549 human lung cells. Metallomics 2019, 11, 982–993. [Google Scholar] [CrossRef]
- Jensen, A.R.; Adams, Y.; Hviid, L. Cerebral Plasmodium falciparum malaria: The role of PfEMP1 in its pathogenesis and immunity, and PfEMP1-based vaccines to prevent it. Immunol. Rev. 2019. [Google Scholar] [CrossRef] [Green Version]
- Li, C.; Yang, Z.; Zhai, C.; Qiu, W.; Li, D.; Yi, Z.; Wang, L.; Tang, J.; Qian, M.; Luo, J.; et al. Maslinic acid potentiates the anti-tumor activity of tumor necrosis factor alpha by inhibiting NF-kappaB signaling pathway. Mol. Cancer 2010, 9, 73. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Luan, Y.; Xu, W. The structure and main functions of aminopeptidase N. Curr. Med. Chem. 2007, 14, 639–647. [Google Scholar] [CrossRef] [PubMed]
- Amin, S.A.; Adhikari, N.; Jha, T. Design of Aminopeptidase N Inhibitors as Anti-cancer Agents. J. Med. Chem. 2018, 61, 6468–6490. [Google Scholar] [CrossRef] [PubMed]
- Ishii, K.; Usui, S.; Sugimura, Y.; Yoshida, S.; Hioki, T.; Tatematsu, M.; Yamamoto, H.; Hirano, K. Aminopeptidase N regulated by zinc in human prostate participates in tumor cell invasion. Int. J. Cancer 2001, 92, 49–54. [Google Scholar] [CrossRef]
- Ishii, K.; Otsuka, T.; Iguchi, K.; Usui, S.; Yamamoto, H.; Sugimura, Y.; Yoshikawa, K.; Hayward, S.W.; Hirano, K. Evidence that the prostate-specific antigen (PSA)/Zn2+ axis may play a role in human prostate cancer cell invasion. Cancer Lett. 2004, 207, 79–87. [Google Scholar] [CrossRef]
- Apte, R.S.; Chen, D.S.; Ferrara, N. VEGF in Signaling and Disease: Beyond Discovery and Development. Cell 2019, 176, 1248–1264. [Google Scholar] [CrossRef] [Green Version]
- Zhao, Y.; Adjei, A.A. Targeting Angiogenesis in Cancer Therapy: Moving Beyond Vascular Endothelial Growth Factor. Oncologist 2015, 20, 660–673. [Google Scholar] [CrossRef] [Green Version]
- Golovine, K.; Uzzo, R.G.; Makhov, P.; Crispen, P.L.; Kunkle, D.; Kolenko, V.M. Depletion of intracellular zinc increases expression of tumorigenic cytokines VEGF, IL-6 and IL-8 in prostate cancer cells via NF-kappaB-dependent pathway. Prostate 2008, 68, 1443–1449. [Google Scholar] [CrossRef] [Green Version]
- Nardinocchi, L.; Pantisano, V.; Puca, R.; Porru, M.; Aiello, A.; Grasselli, A.; Leonetti, C.; Safran, M.; Rechavi, G.; Givol, D.; et al. Zinc downregulates HIF-1alpha and inhibits its activity in tumor cells in vitro and in vivo. PLoS ONE 2010, 5, e15048. [Google Scholar] [CrossRef] [Green Version]
- Brahimi-Horn, C.; Berra, E.; Pouysségur, J. Hypoxia: The tumor’s gateway to progression along the angiogenic pathway. Trends Cell Biol. 2001, 11, S32–S36. [Google Scholar] [CrossRef]
- Kerkar, S.P.; Restifo, N.P. Cellular constituents of immune escape within the tumor microenvironment. Cancer Res. 2012, 72, 3125–3130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jarosz-Biej, M.; Smolarczyk, R.; Cichon, T.; Kulach, N. Tumor Microenvironment as A “Game Changer” in Cancer Radiotherapy. Int. J. Mol. Sci. 2019, 20, 3212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Luo, J.; Ok Lee, S.; Liang, L.; Huang, C.K.; Li, L.; Wen, S.; Chang, C. Infiltrating bone marrow mesenchymal stem cells increase prostate cancer stem cell population and metastatic ability via secreting cytokines to suppress androgen receptor signaling. Oncogene 2014, 33, 2768–2778. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jiang, X.; Wang, J.; Deng, X.; Xiong, F.; Ge, J.; Xiang, B.; Wu, X.; Ma, J.; Zhou, M.; Li, X.; et al. Role of the tumor microenvironment in PD-L1/PD-1-mediated tumor immune escape. Mol. Cancer 2019, 18, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mora, J.; Mertens, C.; Meier, J.K.; Fuhrmann, D.C.; Brune, B.; Jung, M. Strategies to Interfere with Tumor Metabolism through the Interplay of Innate and Adaptive Immunity. Cells 2019, 8, 445. [Google Scholar] [CrossRef] [Green Version]
- Meng, W.; Hao, Y.; He, C.; Li, L.; Zhu, G. Exosome-orchestrated hypoxic tumor microenvironment. Mol. Cancer 2019, 18, 57. [Google Scholar] [CrossRef] [Green Version]
- Talbot, L.J.; Bhattacharya, S.D.; Kuo, P.C. Epithelial-mesenchymal transition, the tumor microenvironment, and metastatic behavior of epithelial malignancies. Int. J. Biochem. Mol. Biol. 2012, 3, 117–136. [Google Scholar]
- Bao, B.; Thakur, A.; Li, Y.; Ahmad, A.; Azmi, A.S.; Banerjee, S.; Kong, D.; Ali, S.; Lum, L.G.; Sarkar, F.H. The immunological contribution of NF-kappaB within the tumor microenvironment: A potential protective role of zinc as an anti-tumor agent. Biochim. Biophys. Acta 2012, 1825, 160–172. [Google Scholar] [CrossRef] [Green Version]
- Ferrandino, F.; Grazioli, P.; Bellavia, D.; Campese, A.F.; Screpanti, I.; Felli, M.P. Notch and NF-kappaB: Coach and Players of Regulatory T-Cell Response in Cancer. Front. Immunol. 2018, 9, 2165. [Google Scholar] [CrossRef] [Green Version]
- Bao, B.; Prasad, A.S.; Beck, F.W.; Bao, G.W.; Singh, T.; Ali, S.; Sarkar, F.H. Intracellular free zinc up-regulates IFN-gamma and T-bet essential for Th1 differentiation in Con-A stimulated HUT-78 cells. Biochem. Biophys. Res. Commun. 2011, 407, 703–707. [Google Scholar] [CrossRef] [Green Version]
- Prasad, A.S.; Beck, F.W.; Grabowski, S.M.; Kaplan, J.; Mathog, R.H. Zinc deficiency: Changes in cytokine production and T-cell subpopulations in patients with head and neck cancer and in noncancer subjects. Proc. Assoc. Am. Physicians 1997, 109, 68–77. [Google Scholar] [PubMed]
- Wessels, I.; Maywald, M.; Rink, L. Zinc as a Gatekeeper of Immune Function. Nutrients 2017, 9, 1286. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Maret, W. Zinc in Cellular Regulation: The Nature and Significance of “Zinc Signals”. Int. J. Mol. Sci. 2017, 18, 2285. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fukunaka, A.; Fujitani, Y. Role of Zinc Homeostasis in the Pathogenesis of Diabetes and Obesity. Int. J. Mol. Sci. 2018, 19, 476. [Google Scholar] [CrossRef] [Green Version]
- Kambe, T.; Tsuji, T.; Hashimoto, A.; Itsumura, N. The Physiological, Biochemical, and Molecular Roles of Zinc Transporters in Zinc Homeostasis and Metabolism. Physiol. Rev. 2015, 95, 749–784. [Google Scholar] [CrossRef]
- Singh, C.K.; Malas, K.M.; Tydrick, C.; Siddiqui, I.A.; Iczkowski, K.A.; Ahmad, N. Analysis of Zinc-Exporters Expression in Prostate Cancer. Sci. Rep. 2016, 6, 36772. [Google Scholar] [CrossRef] [Green Version]
- Cousins, R.J.; Liuzzi, J.P.; Lichten, L.A. Mammalian zinc transport, trafficking, and signals. J. Biol. Chem. 2006, 281, 24085–24089. [Google Scholar] [CrossRef] [Green Version]
- Desouki, M.M.; Geradts, J.; Milon, B.; Franklin, R.B.; Costello, L.C. hZip2 and hZip3 zinc transporters are down regulated in human prostate adenocarcinomatous glands. Mol. Cancer 2007, 6, 37. [Google Scholar] [CrossRef] [Green Version]
- Chen, Q.G.; Zhang, Z.; Yang, Q.; Shan, G.Y.; Yu, X.Y.; Kong, C.Z. The role of zinc transporter ZIP4 in prostate carcinoma. Urol. Oncol. 2012, 30, 906–911. [Google Scholar] [CrossRef]
- Franklin, R.B.; Ma, J.; Zou, J.; Guan, Z.; Kukoyi, B.I.; Feng, P.; Costello, L.C. Human ZIP1 is a major zinc uptake transporter for the accumulation of zinc in prostate cells. J. Inorg. Biochem. 2003, 96, 435–442. [Google Scholar] [CrossRef] [Green Version]
- Huang, L.; Kirschke, C.P.; Zhang, Y. Decreased intracellular zinc in human tumorigenic prostate epithelial cells: A possible role in prostate cancer progression. Cancer Cell Int. 2006, 6, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Golovine, K.; Makhov, P.; Uzzo, R.G.; Shaw, T.; Kunkle, D.; Kolenko, V.M. Overexpression of the zinc uptake transporter hZIP1 inhibits nuclear factor-kappaB and reduces the malignant potential of prostate cancer cells in vitro and in vivo. Clin. Cancer Res. 2008, 14, 5376–5384. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zou, J.; Milon, B.C.; Desouki, M.M.; Costello, L.C.; Franklin, R.B. hZIP1 zinc transporter down-regulation in prostate cancer involves the overexpression of ras responsive element binding protein-1 (RREB-1). Prostate 2011, 71, 1518–1524. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mihelich, B.L.; Khramtsova, E.A.; Arva, N.; Vaishnav, A.; Johnson, D.N.; Giangreco, A.A.; Martens-Uzunova, E.; Bagasra, O.; Kajdacsy-Balla, A.; Nonn, L. miR-183-96-182 cluster is overexpressed in prostate tissue and regulates zinc homeostasis in prostate cells. J. Biol. Chem. 2011, 286, 44503–44511. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Palmiter, R.D.; Findley, S.D. Cloning and functional characterization of a mammalian zinc transporter that confers resistance to zinc. EMBO J. 1995, 14, 639–649. [Google Scholar] [CrossRef]
- Hasumi, M.; Suzuki, K.; Matsui, H.; Koike, H.; Ito, K.; Yamanaka, H. Regulation of metallothionein and zinc transporter expression in human prostate cancer cells and tissues. Cancer Lett. 2003, 200, 187–195. [Google Scholar] [CrossRef]
- Beck, F.W.; Prasad, A.S.; Butler, C.E.; Sakr, W.A.; Kucuk, O.; Sarkar, F.H. Differential expression of hZnT-4 in human prostate tissues. Prostate 2004, 58, 374–381. [Google Scholar] [CrossRef]
- Henshall, S.M.; Afar, D.E.; Rasiah, K.K.; Horvath, L.G.; Gish, K.; Caras, I.; Ramakrishnan, V.; Wong, M.; Jeffry, U.; Kench, J.G.; et al. Expression of the zinc transporter ZnT4 is decreased in the progression from early prostate disease to invasive prostate cancer. Oncogene 2003, 22, 6005–6012. [Google Scholar] [CrossRef] [Green Version]
- Tepaamorndech, S.; Huang, L.; Kirschke, C.P. A null-mutation in the Znt7 gene accelerates prostate tumor formation in a transgenic adenocarcinoma mouse prostate model. Cancer Lett. 2011, 308, 33–42. [Google Scholar] [CrossRef]
- Baumgart, S.J.; Nevedomskaya, E.; Haendler, B. Dysregulated Transcriptional Control in Prostate Cancer. Int. J. Mol. Sci. 2019. [Google Scholar] [CrossRef] [Green Version]
- Ulz, P.; Perakis, S.; Zhou, Q.; Moser, T.; Belic, J.; Lazzeri, I.; Wolfler, A.; Zebisch, A.; Gerger, A.; Pristauz, G.; et al. Inference of transcription factor binding from cell-free DNA enables tumor subtype prediction and early detection. Nat. Commun. 2019, 10, 4666. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lambert, S.A.; Jolma, A.; Campitelli, L.F.; Das, P.K.; Yin, Y.; Albu, M.; Chen, X.; Taipale, J.; Hughes, T.R.; Weirauch, M.T. The Human Transcription Factors. Cell 2018, 172, 650–665. [Google Scholar] [CrossRef] [PubMed]
- Lee, T.I.; Young, R.A. Transcriptional regulation and its misregulation in disease. Cell 2013, 152, 1237–1251. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mackeh, R.; Marr, A.K.; Fadda, A.; Kino, T. C2H2-Type Zinc Finger Proteins: Evolutionarily Old and New Partners of the Nuclear Hormone Receptors. Nucl. Recept. Signal. 2018, 15, 1550762918801071. [Google Scholar] [CrossRef]
- Lee, Y.M.; Lim, C. Factors controlling the reactivity of zinc finger cores. J. Am. Chem. Soc. 2011, 133, 8691–8703. [Google Scholar] [CrossRef]
- Klug, A. The discovery of zinc fingers and their development for practical applications in gene regulation and genome manipulation. Q. Rev. Biophys. 2010, 43, 1–21. [Google Scholar] [CrossRef] [Green Version]
- Iuchi, S. Three classes of C2H2 zinc finger proteins. Cell. Mol. Life Sci. 2001, 58, 625–635. [Google Scholar] [CrossRef]
- Collins, T.; Stone, J.R.; Williams, A.J. All in the family: The BTB/POZ, KRAB, and SCAN domains. Mol. Cell. Biol. 2001, 21, 3609–3615. [Google Scholar] [CrossRef] [Green Version]
- Oteiza, P.I.; Mackenzie, G.G. Zinc, oxidant-triggered cell signaling, and human health. Mol. Asp. Med. 2005, 26, 245–255. [Google Scholar] [CrossRef]
- Kroncke, K.D. Zinc finger proteins as molecular targets for nitric oxide-mediated gene regulation. Antioxid. Redox Signal. 2001, 3, 565–575. [Google Scholar] [CrossRef]
- Orlov, A.P.; Orlova, M.A.; Trofimova, T.P.; Kalmykov, S.N.; Kuznetsov, D.A. The role of zinc and its compounds in leukemia. J. Biol. Inorg. Chem. 2018, 23, 347–362. [Google Scholar] [CrossRef] [PubMed]
- Gottlieb, B.; Lehvaslaiho, H.; Beitel, L.K.; Lumbroso, R.; Pinsky, L.; Trifiro, M. The Androgen Receptor Gene Mutations Database. Nucleic Acids Res. 1998, 26, 234–238. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tan, M.H.; Li, J.; Xu, H.E.; Melcher, K.; Yong, E.L. Androgen receptor: Structure, role in prostate cancer and drug discovery. Acta Pharmacol. Sin. 2015, 36, 3–23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shaffer, P.L.; Jivan, A.; Dollins, D.E.; Claessens, F.; Gewirth, D.T. Structural basis of androgen receptor binding to selective androgen response elements. Proc. Natl. Acad. Sci. USA 2004, 101, 4758–4763. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mangelsdorf, D.J.; Thummel, C.; Beato, M.; Herrlich, P.; Schutz, G.; Umesono, K.; Blumberg, B.; Kastner, P.; Mark, M.; Chambon, P.; et al. The nuclear receptor superfamily: The second decade. Cell 1995, 83, 835–839. [Google Scholar] [CrossRef] [Green Version]
- Sadi, M.V.; Walsh, P.C.; Barrack, E.R. Immunohistochemical study of androgen receptors in metastatic prostate cancer. Comparison of receptor content and response to hormonal therapy. Cancer 1991, 67, 3057–3064. [Google Scholar] [CrossRef]
- Chodak, G.W.; Kranc, D.M.; Puy, L.A.; Takeda, H.; Johnson, K.; Chang, C. Nuclear localization of androgen receptor in heterogeneous samples of normal, hyperplastic and neoplastic human prostate. J. Urol. 1992, 147, 798–803. [Google Scholar] [CrossRef]
- Heinlein, C.A.; Chang, C. Androgen receptor in prostate cancer. Endocr. Rev. 2004, 25, 276–308. [Google Scholar] [CrossRef] [Green Version]
- Cao, S.; Zhan, Y.; Dong, Y. Emerging data on androgen receptor splice variants in prostate cancer. Endocr. Relat. Cancer 2016, 23, T199–T210. [Google Scholar] [CrossRef]
- Elshan, N.; Rettig, M.B.; Jung, M.E. Molecules targeting the androgen receptor (AR) signaling axis beyond the AR-Ligand binding domain. Med. Res. Rev. 2019, 39, 910–960. [Google Scholar] [CrossRef]
- Azoitei, A.; Merseburger, A.S.; Godau, B.; Hoda, M.R.; Schmid, E.; Cronauer, M.V. C-terminally truncated constitutively active androgen receptor variants and their biologic and clinical significance in castration-resistant prostate cancer. J. Steroid Biochem. Mol. Biol. 2017, 166, 38–44. [Google Scholar] [CrossRef] [PubMed]
- Quigley, D.A.; Dang, H.X.; Zhao, S.G.; Lloyd, P.; Aggarwal, R.; Alumkal, J.J.; Foye, A.; Kothari, V.; Perry, M.D.; Bailey, A.M.; et al. Genomic Hallmarks and Structural Variation in Metastatic Prostate Cancer. Cell 2018, 174, 758–769. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Paschalis, A.; Sharp, A.; Welti, J.C.; Neeb, A.; Raj, G.V.; Luo, J.; Plymate, S.R.; de Bono, J.S. Alternative splicing in prostate cancer. Nat. Rev. Clin. Oncol. 2018, 15, 663–675. [Google Scholar] [CrossRef]
- Lee, D.K.; Chang, C. Endocrine mechanisms of disease: Expression and degradation of androgen receptor: Mechanism and clinical implication. J. Clin. Endocrinol. Metab. 2003, 88, 4043–4054. [Google Scholar] [CrossRef] [PubMed]
- Rodriguez-Bravo, V.; Pippa, R.; Song, W.M.; Carceles-Cordon, M.; Dominguez-Andres, A.; Fujiwara, N.; Woo, J.; Koh, A.P.; Ertel, A.; Lokareddy, R.K.; et al. Nuclear Pores Promote Lethal Prostate Cancer by Increasing POM121-Driven E2F1, MYC, and AR Nuclear Import. Cell 2018, 174, 1200–1215. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mowszowicz, I.; Lee, H.J.; Chen, H.T.; Mestayer, C.; Portois, M.C.; Cabrol, S.; Mauvais-Jarvis, P.; Chang, C. A point mutation in the second zinc finger of the DNA-binding domain of the androgen receptor gene causes complete androgen insensitivity in two siblings with receptor-positive androgen resistance. Mol. Endocrinol. 1993, 7, 861–869. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sankpal, U.T.; Goodison, S.; Abdelrahim, M.; Basha, R. Targeting Sp1 transcription factors in prostate cancer therapy. Med. Chem. 2011, 7, 518–525. [Google Scholar] [CrossRef]
- Berg, J.M. Sp1 and the subfamily of zinc finger proteins with guanine-rich binding sites. Proc. Natl. Acad. Sci. USA 1992, 89, 11109–11110. [Google Scholar] [CrossRef] [Green Version]
- Kaczynski, J.; Cook, T.; Urrutia, R. Sp1- and Kruppel-like transcription factors. Genome Biol. 2003, 4, 206. [Google Scholar] [CrossRef] [Green Version]
- Deng, X.; Shao, G.; Zhang, H.T.; Li, C.; Zhang, D.; Cheng, L.; Elzey, B.D.; Pili, R.; Ratliff, T.L.; Huang, J.; et al. Protein arginine methyltransferase 5 functions as an epigenetic activator of the androgen receptor to promote prostate cancer cell growth. Oncogene 2017, 36, 1223–1231. [Google Scholar] [CrossRef] [Green Version]
- Shin, T.; Sumiyoshi, H.; Matsuo, N.; Satoh, F.; Nomura, Y.; Mimata, H.; Yoshioka, H. Sp1 and Sp3 transcription factors upregulate the proximal promoter of the human prostate-specific antigen gene in prostate cancer cells. Arch. Biochem. Biophys. 2005, 435, 291–302. [Google Scholar] [CrossRef] [PubMed]
- Faber, P.W.; van Rooij, H.C.; van der Korput, H.A.; Baarends, W.M.; Brinkmann, A.O.; Grootegoed, J.A.; Trapman, J. Characterization of the human androgen receptor transcription unit. J. Biol. Chem. 1991, 266, 10743–10749. [Google Scholar] [PubMed]
- Barna, M.; Merghoub, T.; Costoya, J.A.; Ruggero, D.; Branford, M.; Bergia, A.; Samori, B.; Pandolfi, P.P. Plzf mediates transcriptional repression of HoxD gene expression through chromatin remodeling. Dev. Cell 2002, 3, 499–510. [Google Scholar] [CrossRef] [Green Version]
- Ono, R.; Masuya, M.; Nakajima, H.; Enomoto, Y.; Miyata, E.; Nakamura, A.; Ishii, S.; Suzuki, K.; Shibata-Minoshima, F.; Katayama, N.; et al. Plzf drives MLL-fusion-mediated leukemogenesis specifically in long-term hematopoietic stem cells. Blood 2013, 122, 1271–1283. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hong, S.H.; David, G.; Wong, C.W.; Dejean, A.; Privalsky, M.L. SMRT corepressor interacts with PLZF and with the PML-retinoic acid receptor alpha (RARalpha) and PLZF-RARalpha oncoproteins associated with acute promyelocytic leukemia. Proc. Natl. Acad. Sci. USA 1997, 94, 9028–9033. [Google Scholar] [CrossRef] [Green Version]
- David, G.; Alland, L.; Hong, S.H.; Wong, C.W.; DePinho, R.A.; Dejean, A. Histone deacetylase associated with mSin3A mediates repression by the acute promyelocytic leukemia-associated PLZF protein. Oncogene 1998, 16, 2549–2556. [Google Scholar] [CrossRef] [Green Version]
- Jin, Y.; Nenseth, H.Z.; Saatcioglu, F. Role of PLZF as a tumor suppressor in prostate cancer. Oncotarget 2017, 8, 71317–71324. [Google Scholar] [CrossRef] [Green Version]
- Hoatlin, M.E.; Zhi, Y.; Ball, H.; Silvey, K.; Melnick, A.; Stone, S.; Arai, S.; Hawe, N.; Owen, G.; Zelent, A.; et al. A novel BTB/POZ transcriptional repressor protein interacts with the Fanconi anemia group C protein and PLZF. Blood 1999, 94, 3737–3747. [Google Scholar] [CrossRef]
- Cao, J.; Zhu, S.; Zhou, W.; Li, J.; Liu, C.; Xuan, H.; Yan, J.; Zheng, L.; Zhou, L.; Yu, J.; et al. PLZF mediates the PTEN/AKT/FOXO3a signaling in suppression of prostate tumorigenesis. PLoS ONE 2013, 8, e77922. [Google Scholar] [CrossRef] [Green Version]
- Xiao, G.Q.; Unger, P.; Yang, Q.; Kinoshita, Y.; Singh, K.; McMahon, L.; Nastiuk, K.; Sha, K.; Krolewski, J.; Burstein, D. Loss of PLZF expression in prostate cancer by immunohistochemistry correlates with tumor aggressiveness and metastasis. PLoS ONE 2015, 10, e0121318. [Google Scholar] [CrossRef]
- Hsieh, C.L.; Botta, G.; Gao, S.; Li, T.; Van Allen, E.M.; Treacy, D.J.; Cai, C.; He, H.H.; Sweeney, C.J.; Brown, M.; et al. PLZF, a tumor suppressor genetically lost in metastatic castration-resistant prostate cancer, is a mediator of resistance to androgen deprivation therapy. Cancer Res. 2015, 75, 1944–1948. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jin, Y.; Qu, S.; Tesikova, M.; Wang, L.; Kristian, A.; Maelandsmo, G.M.; Kong, H.; Zhang, T.; Jeronimo, C.; Teixeira, M.R.; et al. Molecular circuit involving KLK4 integrates androgen and mTOR signaling in prostate cancer. Proc. Natl. Acad. Sci. USA 2013, 110, E2572–E2581. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jiang, F.; Wang, Z. Identification and characterization of PLZF as a prostatic androgen-responsive gene. Prostate 2004, 59, 426–435. [Google Scholar] [CrossRef] [PubMed]
- Kikugawa, T.; Kinugasa, Y.; Shiraishi, K.; Nanba, D.; Nakashiro, K.; Tanji, N.; Yokoyama, M.; Higashiyama, S. PLZF regulates Pbx1 transcription and Pbx1-HoxC8 complex leads to androgen-independent prostate cancer proliferation. Prostate 2006, 66, 1092–1099. [Google Scholar] [CrossRef] [PubMed]
- Gallus, S.; Foschi, R.; Negri, E.; Talamini, R.; Franceschi, S.; Montella, M.; Ramazzotti, V.; Tavani, A.; Dal Maso, L.; La Vecchia, C. Dietary zinc and prostate cancer risk: A case-control study from Italy. Eur. Urol. 2007, 52, 1052–1056. [Google Scholar] [CrossRef] [PubMed]
- Leitzmann, M.F.; Stampfer, M.J.; Wu, K.; Colditz, G.A.; Willett, W.C.; Giovannucci, E.L. Zinc supplement use and risk of prostate cancer. J. Natl. Cancer Inst. 2003, 95, 1004–1007. [Google Scholar] [CrossRef] [Green Version]
- Banudevi, S.; Elumalai, P.; Sharmila, G.; Arunkumar, R.; Senthilkumar, K.; Arunakaran, J. Protective effect of zinc on N-methyl-N-nitrosourea and testosterone-induced prostatic intraepithelial neoplasia in the dorsolateral prostate of Sprague Dawley rats. Exp. Biol. Med. 2011, 236, 1012–1021. [Google Scholar] [CrossRef]
- Fitzsimons, N.J.; Sun, L.; Moul, J.W. Medical technologies for the diagnosis of prostate cancer. Expert Rev. Med. Devices 2007, 4, 227–239. [Google Scholar] [CrossRef]
- Hoeks, C.M.; Hambrock, T.; Yakar, D.; Hulsbergen-van de Kaa, C.A.; Feuth, T.; Witjes, J.A.; Futterer, J.J.; Barentsz, J.O. Transition zone prostate cancer: Detection and localization with 3-T multiparametric MR imaging. Radiology 2013, 266, 207–217. [Google Scholar] [CrossRef] [Green Version]
- Costa, D.N.; Pedrosa, I.; Donato, F., Jr.; Roehrborn, C.G.; Rofsky, N.M. MR Imaging-Transrectal US Fusion for Targeted Prostate Biopsies: Implications for Diagnosis and Clinical Management. Radiographics 2015, 35, 696–708. [Google Scholar] [CrossRef]
- Costello, L.C.; Franklin, R.B. Prostatic fluid electrolyte composition for the screening of prostate cancer: A potential solution to a major problem. Prostate Cancer Prostatic Dis. 2009, 12, 17–24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cortesi, M.; Chechik, R.; Breskin, A.; Vartsky, D.; Ramon, J.; Raviv, G.; Volkov, A.; Fridman, E. Evaluating the cancer detection and grading potential of prostatic-zinc imaging: A simulation study. Phys. Med. Biol. 2009, 54, 781–796. [Google Scholar] [CrossRef] [PubMed]
- Ma, Q.; Lin, Z.H.; Yang, N.; Li, Y.; Su, X.G. A novel carboxymethyl chitosan-quantum dot-based intracellular probe for Zn2+ ion sensing in prostate cancer cells. Acta Biomater. 2014, 10, 868–874. [Google Scholar] [CrossRef] [PubMed]
- Fu, S.; Wan, X.; Du, C.; Wang, H.; Zhou, J.; Wang, Z. A novel fluorescent probe for the early detection of prostate cancer based on endogenous zinc sensing. Prostate 2019, 79, 1378–1385. [Google Scholar] [CrossRef]
- Zhang, X.A.; Hayes, D.; Smith, S.J.; Friedle, S.; Lippard, S.J. New strategy for quantifying biological zinc by a modified zinpyr fluorescence sensor. J. Am. Chem. Soc. 2008, 130, 15788–15789. [Google Scholar] [CrossRef] [Green Version]
- Robert, C.; Ribas, A.; Schachter, J.; Arance, A.; Grob, J.J.; Mortier, L.; Daud, A.; Carlino, M.S.; McNeil, C.M.; Lotem, M.; et al. Pembrolizumab versus ipilimumab in advanced melanoma (KEYNOTE-006): Post-hoc 5-year results from an open-label, multicentre, randomised, controlled, phase 3 study. Lancet Oncol. 2019, 20, 1239–1251. [Google Scholar] [CrossRef]
- Zhang, H.; Christensen, C.L.; Dries, R.; Oser, M.G.; Deng, J.; Diskin, B.; Li, F.; Pan, Y.; Zhang, X.; Yin, Y.; et al. CDK7 Inhibition Potentiates Genome Instability Triggering Anti-Tumor Immunity in Small Cell Lung Cancer. Cancer Cell 2019. [Google Scholar] [CrossRef]
- Goldberg, M.S. Improving cancer immunotherapy through nanotechnology. Nat. Rev. Cancer 2019, 19, 587–602. [Google Scholar] [CrossRef]
- Bryant, G.; Wang, L.; Mulholland, D.J. Overcoming Oncogenic Mediated Tumor Immunity in Prostate Cancer. Int. J. Mol. Sci. 2017, 18, 1542. [Google Scholar] [CrossRef]
- Read, S.A.; Obeid, S.; Ahlenstiel, C.; Ahlenstiel, G. The Role of Zinc in Antiviral Immunity. Adv. Nutr. 2019, 10, 696–710. [Google Scholar] [CrossRef] [Green Version]
- Shankar, A.H.; Prasad, A.S. Zinc and immune function: The biological basis of altered resistance to infection. Am. J. Clin. Nutr. 1998, 68, 447s–463s. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Read, S.A.; O’Connor, K.S.; Suppiah, V.; Ahlenstiel, C.L.E.; Obeid, S.; Cook, K.M.; Cunningham, A.; Douglas, M.W.; Hogg, P.J.; Booth, D.; et al. Zinc is a potent and specific inhibitor of IFN-lambda3 signalling. Nat. Commun. 2017, 8, 15245. [Google Scholar] [CrossRef] [PubMed]
- Prasad, A.S.; Bao, B. Molecular Mechanisms of Zinc as a Pro-Antioxidant Mediator: Clinical Therapeutic Implications. Antioxidants 2019, 8, 164. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mezzaroba, L.; Alfieri, D.F.; Colado Simao, A.N.; Vissoci Reiche, E.M. The role of zinc, copper, manganese and iron in neurodegenerative diseases. Neurotoxicology 2019, 74, 230–241. [Google Scholar] [CrossRef] [PubMed]
- Miyai, T.; Hojyo, S.; Ikawa, T.; Kawamura, M.; Irie, T.; Ogura, H.; Hijikata, A.; Bin, B.H.; Yasuda, T.; Kitamura, H.; et al. Zinc transporter SLC39A10/ZIP10 facilitates antiapoptotic signaling during early B-cell development. Proc. Natl. Acad. Sci. USA 2014, 111, 11780–11785. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kitamura, H.; Morikawa, H.; Kamon, H.; Iguchi, M.; Hojyo, S.; Fukada, T.; Yamashita, S.; Kaisho, T.; Akira, S.; Murakami, M.; et al. Toll-like receptor-mediated regulation of zinc homeostasis influences dendritic cell function. Nat. Immunol. 2006, 7, 971–977. [Google Scholar] [CrossRef]
- Yu, M.; Lee, W.W.; Tomar, D.; Pryshchep, S.; Czesnikiewicz-Guzik, M.; Lamar, D.L.; Li, G.; Singh, K.; Tian, L.; Weyand, C.M.; et al. Regulation of T cell receptor signaling by activation-induced zinc influx. J. Exp. Med. 2011, 208, 775–785. [Google Scholar] [CrossRef] [Green Version]
- Anzilotti, C.; Swan, D.J.; Boisson, B.; Deobagkar-Lele, M.; Oliveira, C.; Chabosseau, P.; Engelhardt, K.R.; Xu, X.; Chen, R.; Alvarez, L.; et al. An essential role for the Zn(2+) transporter ZIP7 in B cell development. Nat. Immunol. 2019, 20, 350–361. [Google Scholar] [CrossRef] [Green Version]
- Hojyo, S.; Miyai, T.; Fujishiro, H.; Kawamura, M.; Yasuda, T.; Hijikata, A.; Bin, B.H.; Irie, T.; Tanaka, J.; Atsumi, T.; et al. Zinc transporter SLC39A10/ZIP10 controls humoral immunity by modulating B-cell receptor signal strength. Proc. Natl. Acad. Sci. USA 2014, 111, 11786–11791. [Google Scholar] [CrossRef] [Green Version]
- Sanna, A.; Firinu, D.; Zavattari, P.; Valera, P. Zinc Status and Autoimmunity: A Systematic Review and Meta-Analysis. Nutrients 2018, 10, 68. [Google Scholar] [CrossRef] [Green Version]
- Rosenkranz, E.; Hilgers, R.D.; Uciechowski, P.; Petersen, A.; Plumakers, B.; Rink, L. Zinc enhances the number of regulatory T cells in allergen-stimulated cells from atopic subjects. Eur. J. Nutr. 2017, 56, 557–567. [Google Scholar] [CrossRef] [PubMed]
- Barnett, J.B.; Dao, M.C.; Hamer, D.H.; Kandel, R.; Brandeis, G.; Wu, D.; Dallal, G.E.; Jacques, P.F.; Schreiber, R.; Kong, E.; et al. Effect of zinc supplementation on serum zinc concentration and T cell proliferation in nursing home elderly: A randomized, double-blind, placebo-controlled trial. Am. J. Clin. Nutr. 2016, 103, 942–951. [Google Scholar] [CrossRef] [PubMed]
- Prasad, A.S.; Kucuk, O. Zinc in cancer prevention. Cancer Metastasis Rev. 2002, 21, 291–295. [Google Scholar] [CrossRef] [PubMed]
- Miyahira, A.K.; Sharp, A.; Ellis, L.; Jones, J.; Kaochar, S.; Larman, H.B.; Quigley, D.A.; Ye, H.; Simons, J.W.; Pienta, K.J.; et al. Prostate cancer research: The next generation; report from the 2019 Coffey-Holden Prostate Cancer Academy Meeting. Prostate 2020, 80, 113–132. [Google Scholar] [CrossRef] [PubMed]
- Yunger, S.; Bar El, A.; Zeltzer, L.A.; Fridman, E.; Raviv, G.; Laufer, M.; Schachter, J.; Markel, G.; Itzhaki, O.; Besser, M.J. Tumor-infiltrating lymphocytes from human prostate tumors reveal anti-tumor reactivity and potential for adoptive cell therapy. Oncoimmunology 2019, 8, e1672494. [Google Scholar] [CrossRef] [PubMed] [Green Version]
PCa/Control (Serum Zinc Level, µg/dL) | PCa/Control (Number) | Testing Assay | Cohort Sources/Populations | Age Medians or Ranges (PCa/Control) | References |
---|---|---|---|---|---|
61.60 ± 19.75/ 99.59 ± 29.23 | 220/220 | AAS | Nigeria | 69.73 ± 6.32/ 68.97 ± 5.76 | Wakwe et al. 2019 [30] |
576 ± 102/ 711 ± 164 | 25/24 | AAS | Turkey | 67.5 ± 8.8/ 65.0 ± 6.0 | Aydin et al. 2006 [31] |
63.40 ± 6.40/ 86.50 ± 15.20 | 18/20 | AAS | India | 55−85/ 30−50 | Christudoss et al. 2011 [32] |
4.66 ± 2.22/ 19.26 ± 3.26 | 30/32 | AAS | Turkey | 65.4 ± 4.2/ 62.8 ± 5.8 | Kaba et al. 2014 [34] |
8300 ± 213/ 9780 ± 257 | 42/101 | AAS | China | 70.1 ± 1.32/ 67.8 ± 0.85 | Li et al. 2005 [59] |
147.75 ± 42.05/ 168.78 ± 59.80 | 50/50 | AAS | Nigeria | 50−70/ 50−70 | Onyema-iloh et al. 2015 [60] |
63.92 ± 19.10/ 103.61 ± 32.43 | 85/90 | AAS | China | 64.7 ± 9.2/ 65.9 ± 8.4 | Chen et al. 2015 [61] |
91.55 ± 12.42/ 90.89 ± 12.42 | 50/10 | AAS | German | 68.6/65.9 | Feustel et al. 1989 [62] |
94.09 ± 20.40/ 93.9 ± 17.60 | 392/783 | AAS | America | 69.1 ± 7.1/ 68.9 ± 7.2 | Park et al. 2013 [63] |
89.89 ± 1.20/ 85.66 ± 1.31 | 197/197 | AAS | Poland | 72/72 | Białkowska et al. 2018 [64] |
112.93 ± 18.10/ 98.12 ± 8.24 | 40/28 | AAS | China | ND/ND | Yao et al. 1977 [65] |
© 2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Li, D.; Stovall, D.B.; Wang, W.; Sui, G. Advances of Zinc Signaling Studies in Prostate Cancer. Int. J. Mol. Sci. 2020, 21, 667. https://doi.org/10.3390/ijms21020667
Li D, Stovall DB, Wang W, Sui G. Advances of Zinc Signaling Studies in Prostate Cancer. International Journal of Molecular Sciences. 2020; 21(2):667. https://doi.org/10.3390/ijms21020667
Chicago/Turabian StyleLi, Dangdang, Daniel B. Stovall, Wenmeng Wang, and Guangchao Sui. 2020. "Advances of Zinc Signaling Studies in Prostate Cancer" International Journal of Molecular Sciences 21, no. 2: 667. https://doi.org/10.3390/ijms21020667
APA StyleLi, D., Stovall, D. B., Wang, W., & Sui, G. (2020). Advances of Zinc Signaling Studies in Prostate Cancer. International Journal of Molecular Sciences, 21(2), 667. https://doi.org/10.3390/ijms21020667