51
|
Baldari S, Garufi A, Granato M, Cuomo L, Pistritto G, Cirone M, D'Orazi G. Hyperglycemia triggers HIPK2 protein degradation. Oncotarget 2018; 8:1190-1203. [PMID: 27901482 PMCID: PMC5352047 DOI: 10.18632/oncotarget.13595] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 11/08/2016] [Indexed: 12/23/2022] Open
Abstract
Homeodomain interacting protein kinase-2 (HIPK2) is an evolutionary conserved kinase that modulates several key molecular pathways to restrain tumor growth and induce p53-depending apoptotic cell-death in response to anticancer therapies. HIPK2 silencing in cancer cells leads to chemoresistance and cancer progression, in part due to p53 inhibition. Recently, hyperglycemia has been shown to reduce p53 phosphorylation at serine 46 (Ser46), the target residue of HIPK2, thus impairing p53 apoptotic function. Here we asked whether hyperglycemia could, upstream of p53, target HIPK2. We focused on the effect of high glucose (HG) on HIPK2 protein stability and the underlying mechanisms. We found that HG reduced HIPK2 protein levels, therefore impairing HIPK2-induced p53 apoptotic activity. HG-triggered HIPK2 protein downregulation was rescued by both proteasome inhibitor MG132 and by protein phosphatase inhibitors Calyculin A (CL-A) and Okadaic Acid (OA). Looking for the phosphatase involved, we found that protein phosphatase 2A (PP2A) induced HIPK2 degradation, as evidenced by directly activating PP2A with FTY720 or by silencing PP2A with siRNA in HG condition. The effect of PP2A on HIPK2 protein degradation could be in part due to hypoxia-inducible factor-1 (HIF-1) activity which has been previously shown to induce HIPK2 proteasomal degradation through several ubiquitin ligases. Validation analysed performed with HIF-1α dominant negative or with silencing of Siah2 ubiquitin ligase clearly showed rescue of HG-induced HIPK2 degradation. These findings demonstrate how hyperglycemia, through a complex protein cascade, induced HIPK2 downregulation and consequently impaired p53 apoptotic activity, revealing a novel link between diabetes/obesity and tumor resistance to therapies.
Collapse
Affiliation(s)
- Silvia Baldari
- Department of Research, Advanced Diagnostics, and Technological Innovation, Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Alessia Garufi
- Department of Research, Advanced Diagnostics, and Technological Innovation, Regina Elena National Cancer Institute, 00144 Rome, Italy.,Department of Medical Sciences, Tumor Biology Unit, University 'G. d'Annunzio', 66013 Chieti, Italy
| | - Marisa Granato
- Department of Experimental Medicine, Pasteur-Fondazione Cenci Bolognetti Institute, Sapienza University, 00100 Rome, Italy
| | - Laura Cuomo
- U.O.C. Clinical Pathology, A.C.O., San Filippo Neri Hospital, 00100 Rome, Italy
| | - Giuseppa Pistritto
- Department of Systems Medicine, University Tor Vergata, 00133 Rome, Italy
| | - Mara Cirone
- Department of Experimental Medicine, Pasteur-Fondazione Cenci Bolognetti Institute, Sapienza University, 00100 Rome, Italy
| | - Gabriella D'Orazi
- Department of Research, Advanced Diagnostics, and Technological Innovation, Regina Elena National Cancer Institute, 00144 Rome, Italy.,Department of Medical Sciences, Tumor Biology Unit, University 'G. d'Annunzio', 66013 Chieti, Italy
| |
Collapse
|
52
|
Kheirouri S, Naghizadeh S, Alizadeh M. Zinc supplementation does not influence serum levels of VEGF, BDNF, and NGF in diabetic retinopathy patients: a randomized controlled clinical trial. Nutr Neurosci 2018; 22:718-724. [PMID: 29421993 DOI: 10.1080/1028415x.2018.1436236] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Objectives: This study was aimed to evaluate the effects of zinc (Zn) supplementation on serum levels of vascular endothelial growth factor (VEGF), brain-derived neurotrophic factor (BDNF), and nerve growth factor (NGF) in patients with diabetic retinopathy (DR). Methods: In this randomized clinical trial, 50 patients with DR were allocated into the Zn (n = 25) and placebo (n = 25) groups to receive 30 mg Zn gluconate or maltose dextrin per day, respectively, for three months. Metabolic parameters and blood pressure were measured. Serum levels of Zn were assessed by atomic absorption spectrophotometry and serum levels of VEGF, BDNF and NGF by ELISA. Results: Forty-five patients completed the intervention. Levels of VEGF, BDNF and NGF were not affected by the Zn supplementation. Levels of VEGF correlated negatively with levels of Zn and positively with BDNF and NGF. There was also a positive correlation between BDNF and NGF. Serum levels of VEGF, BDNF and NGF were negatively correlated with serum levels of the diabetic parameters measured. Conclusions: Strong positive relationship between the growth factors and their inverse association with metabolic factors is possibly suggesting the contribution of these factors in the pathogenesis of DR through acting in a same biological pathway.
Collapse
Affiliation(s)
- Sorayya Kheirouri
- Department of Nutrition, Tabriz University of Medical Sciences , Tabriz , Iran
| | - Siamak Naghizadeh
- Department of Nutrition, Tabriz University of Medical Sciences , Tabriz , Iran
| | - Mohammad Alizadeh
- Department of Nutrition, Tabriz University of Medical Sciences , Tabriz , Iran
| |
Collapse
|
53
|
Mousavi SN, Faghihi A, Motaghinejad M, Shiasi M, Imanparast F, Amiri HL, Shidfar F. Zinc and Selenium Co-supplementation Reduces Some Lipid Peroxidation and Angiogenesis Markers in a Rat Model of NAFLD-Fed High Fat Diet. Biol Trace Elem Res 2018; 181:288-295. [PMID: 28577233 DOI: 10.1007/s12011-017-1059-2] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2016] [Accepted: 11/07/2016] [Indexed: 12/13/2022]
Abstract
Studies have shown that non-alcoholic fatty liver disease (NAFLD) patients are more prone to cardiovascular disease (CVD). Zinc and selenium deficiency are common in NAFLD. But the effects of zinc and selenium co-supplementation before and/or after disease progression on CVD markers are not clear in NAFLD patients. This study aimed to compare the effects of zinc and selenium co-supplementation before and/or after disease progression on some of the CVD markers in an experimental model of NAFLD. Forty male Sprague Dawley rats (197 ± 4 g) were randomly assigned into four dietary groups: control group (C; received 9% of calorie as fat), model group (M; received 82% of calorie as fat), and supplementation before (BS) or after (AS) disease progression. Animals were fed diets for 20 weeks in all groups. Fasting plasma glucose (FPG), insulin, HOMA-IR, ALT, AST, lipid profile, malondialdehyde (MDA) and vascular endothelial growth factor (VEGF) levels were measured as CVD indices. Serum ALT, AST, FPG, insulin, MDA, VEGF and HOMA-IR were significantly higher in the M than C group. Co-supplementation reduced serum ALT and AST levels in the BS and AS groups compared with the M group. FPG, insulin, HOMA-IR, VEGF, MDA, LDL/HDL-c and TC/HDL-c ratio were significantly reduced in the AS compared with the M group. TG/HDL-c ratio was significantly reduced in the BS and AS compared with the M group. Serum MDA, VEGF, Insulin and HOMA-IR were significantly lowered in the AS than BS group (p < 0.05). Zinc and selenium co-supplementation after NAFLD progression reduced CVD risk indices in an experimental model.
Collapse
Affiliation(s)
- Seyedeh Neda Mousavi
- Iranian National Science Foundation, Tehran, Iran
- Department of Biochemistry and Nutrition, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Amirhosein Faghihi
- Iranian National Science Foundation, Tehran, Iran
- Colorectal Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Majid Motaghinejad
- Iranian National Science Foundation, Tehran, Iran
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Shiasi
- Iranian National Science Foundation, Tehran, Iran
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Imanparast
- Iranian National Science Foundation, Tehran, Iran
- Department of Laboratory Medicine, Khomein University of Medical Sciences, Khomein, Iran
| | - Hamid Lorvand Amiri
- Iranian National Science Foundation, Tehran, Iran
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Farzad Shidfar
- Iranian National Science Foundation, Tehran, Iran.
- Colorectal Research Center, Iran University of Medical Sciences, Tehran, Iran.
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
54
|
Himoto T, Masaki T. Associations between Zinc Deficiency and Metabolic Abnormalities in Patients with Chronic Liver Disease. Nutrients 2018; 10:nu10010088. [PMID: 29342898 PMCID: PMC5793316 DOI: 10.3390/nu10010088] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 01/04/2018] [Accepted: 01/05/2018] [Indexed: 02/06/2023] Open
Abstract
Zinc (Zn) is an essential trace element which has favorable antioxidant, anti-inflammatory, and apoptotic effects. The liver mainly plays a crucial role in maintaining systemic Zn homeostasis. Therefore, the occurrence of chronic liver diseases, such as chronic hepatitis, liver cirrhosis, or fatty liver, results in the impairment of Zn metabolism, and subsequently Zn deficiency. Zn deficiency causes plenty of metabolic abnormalities, including insulin resistance, hepatic steatosis and hepatic encephalopathy. Inversely, metabolic abnormalities like hypoalbuminemia in patients with liver cirrhosis often result in Zn deficiency. Recent studies have revealed the putative mechanisms by which Zn deficiency evokes a variety of metabolic abnormalities in chronic liver disease. Zn supplementation has shown beneficial effects on such metabolic abnormalities in experimental models and actual patients with chronic liver disease. This review summarizes the pathogenesis of metabolic abnormalities deriving from Zn deficiency and the favorable effects of Zn administration in patients with chronic liver disease. In addition, we also highlight the interactions between Zn and other trace elements, vitamins, amino acids, or hormones in such patients.
Collapse
Affiliation(s)
- Takashi Himoto
- Department of Medical Technology, Kagawa Prefectural University of Health Sciences, 281-1, Hara, Mure-Cho, Takamatsu, Kagawa 761-0123, Japan.
| | - Tsutomu Masaki
- Department of Gastroenterology and Neurology, Kagawa University School of Medicine, Kagawa 761-0123, Japan.
| |
Collapse
|
55
|
Wetherell D, Baldwin GS, Shulkes A, Bolton D, Ischia J, Patel O. Zinc ion dyshomeostasis increases resistance of prostate cancer cells to oxidative stress via upregulation of HIF1α. Oncotarget 2018; 9:8463-8477. [PMID: 29492208 PMCID: PMC5823553 DOI: 10.18632/oncotarget.23893] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 11/14/2017] [Indexed: 12/16/2022] Open
Abstract
Zinc ions (Zn2+) are known to influence cell survival and proliferation. However the homeostatic regulation of Zn2+ and their role in prostate cancer (PC) progression is poorly understood. Therefore the subcellular distribution and uptake of Zn2+ in PC cells were investigated. Inductively coupled plasma mass spectroscopy and fluorescent microscopy with the Zn2+-specific fluorescent probe FluoZin-3 were used to quantify total and free Zn2+, respectively, in the normal prostate epithelial cell line (PNT1A) and three human PC cell lines (PC3, DU145 and LNCaP). The effects of Zn2+ treatment on proliferation and survival were measured in vitro using MTT assays and in vivo using mouse xenografts. The ability of Zn2+ to protect against oxidative stress via a HIF1α-dependent mechanism was investigated using a HIF1α knock-down PC3 model. Our results demonstrate that the total Zn2+ concentration in normal PNT1A and PC cells is similar, but PC3 cells contain significantly higher free Zn2+ than PNT1A cells (p < 0.01). PNT1A cells can survive better in the presence of high concentrations of Zn2+ than PC3 cells. Exposure to 10 µM Zn2+ over 72 hours significantly reduces PC3 cell proliferation in vitro but not in vivo. Zn2+ increases PC3 cell survival up to 2.3-fold under oxidative stress, and this protective effect is not seen in PNT1A cells or in a HIF1α-KD PC3 cell model. A state of Zn2+ dyshomeostasis exists in PC. HIF1α is an integral component of a Zn2+-dependent protective mechanism present in PC3 cells. This pathway may be clinically significant through its contribution to castrate-resistant PC survival.
Collapse
Affiliation(s)
- David Wetherell
- Department of Surgery, University of Melbourne, Austin Health, Heidelberg, Victoria, 3084, Australia.,Department of Urology, Austin Health, Heidelberg, Victoria, 3084, Australia
| | - Graham S Baldwin
- Department of Surgery, University of Melbourne, Austin Health, Heidelberg, Victoria, 3084, Australia
| | - Arthur Shulkes
- Department of Surgery, University of Melbourne, Austin Health, Heidelberg, Victoria, 3084, Australia
| | - Damien Bolton
- Department of Surgery, University of Melbourne, Austin Health, Heidelberg, Victoria, 3084, Australia.,Department of Urology, Austin Health, Heidelberg, Victoria, 3084, Australia
| | - Joseph Ischia
- Department of Surgery, University of Melbourne, Austin Health, Heidelberg, Victoria, 3084, Australia.,Department of Urology, Austin Health, Heidelberg, Victoria, 3084, Australia
| | - Oneel Patel
- Department of Surgery, University of Melbourne, Austin Health, Heidelberg, Victoria, 3084, Australia
| |
Collapse
|
56
|
Wang Z, Deng X, Xiong R, Xiong S, Liu J, Cao X, Lei X, Chen Y, Zheng X, Tang G. Design, synthesis and biological evaluation of 3',4',5'-trimethoxy flavonoid benzimidazole derivatives as potential anti-tumor agents. MEDCHEMCOMM 2017; 9:305-315. [PMID: 30108924 DOI: 10.1039/c7md00578d] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 12/10/2017] [Indexed: 11/21/2022]
Abstract
A series of 3',4',5'-trimethoxy flavonoids with benzimidazole linked by different chain alkanes have been designed and synthesized. The potential activity of these compounds as anti-tumor agents was evaluated by cytotoxicity assay in MGC-803 (human gastric cancer), MCF-7 (human breast cancer), HepG-2 (human hepatoma) and MFC (mouse gastric cancer) tumor cell lines. Among them, compound 15 7-(3-(2-chloro-1H-benzo[d]imidazol-1-yl)propoxy)-2-(3,4,5-trimethoxyphenyl)-4H-chromen-4-one displayed the most potent antiproliferative activity, with IC50 values of 20.47 ± 2.07, 43.42 ± 3.56, 35.45 ± 2.03 μM and 23.47 ± 3.59 μM, respectively. The flow cytometry (FCM) results showed that compound 15 caused the cell cycle to be arrested in G1 phase and induced apoptosis of MFC cells in a dose-dependent manner. In addition, compound 15 exhibited a significant inhibitory effect on tumor growth in vivo. All the results outlined the great potential of compound 15 for further exploitation as anti-tumor agent.
Collapse
Affiliation(s)
- Zhe Wang
- Institute of Pharmacy and Pharmacology , Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study , University of South China , Hengyang , China . ;
| | - Xiangping Deng
- Institute of Pharmacy and Pharmacology , Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study , University of South China , Hengyang , China . ;
| | - Runde Xiong
- Institute of Pharmacy and Pharmacology , Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study , University of South China , Hengyang , China . ;
| | - Shujuan Xiong
- Institute of Pharmacy and Pharmacology , Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study , University of South China , Hengyang , China . ;
| | - Juan Liu
- Institute of Pharmacy and Pharmacology , Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study , University of South China , Hengyang , China . ;
| | - Xuan Cao
- Institute of Pharmacy and Pharmacology , Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study , University of South China , Hengyang , China . ;
| | - Xiaoyong Lei
- Institute of Pharmacy and Pharmacology , Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study , University of South China , Hengyang , China . ;
| | - Yanming Chen
- Mu Dan Jiang You Bo Pharmaceutical Co. Ltd. , Mudanjiang , China
| | - Xing Zheng
- Institute of Pharmacy and Pharmacology , Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study , University of South China , Hengyang , China . ;
| | - Guotao Tang
- Institute of Pharmacy and Pharmacology , Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study , University of South China , Hengyang , China . ;
| |
Collapse
|
57
|
Wang Z, Deng X, Xiong S, Xiong R, Liu J, Zou L, Lei X, Cao X, Xie Z, Chen Y, Liu Y, Zheng X, Tang G. Design, synthesis and biological evaluation of chrysin benzimidazole derivatives as potential anticancer agents. Nat Prod Res 2017; 32:2900-2909. [PMID: 29063798 DOI: 10.1080/14786419.2017.1389940] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
A series of chrysin benzimidazole derivatives were synthesised and evaluated for their anticancer activity in the search for potential anticancer agents. Among them, compound 18 displayed the most potent anti-proliferative activity against MFC cells with IC50 values of 25.72 ± 3.95 μM. The flow cytometry results displayed that compound 18 induced apoptosis of MFC cells in a dose-dependent manner and caused the cell cycle to be arrested in the G0/G1 phase. Furthermore, the preliminary anticancer activity in vivo was also studied in tumour-bearing mice, and the compound 18 exerted good inhibition effect on tumour growth. These results suggested that compound 18 had good anticancer activity, which could be a potential anticancer agent after further optimisation and evaluation.
Collapse
Affiliation(s)
- Zhe Wang
- a Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study , University of South China , Hengyang , China
| | - Xiangping Deng
- a Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study , University of South China , Hengyang , China
| | - Shujuan Xiong
- a Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study , University of South China , Hengyang , China
| | - Runde Xiong
- a Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study , University of South China , Hengyang , China
| | - Juan Liu
- a Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study , University of South China , Hengyang , China
| | - Liu Zou
- a Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study , University of South China , Hengyang , China
| | - Xiaoyong Lei
- a Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study , University of South China , Hengyang , China
| | - Xuan Cao
- a Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study , University of South China , Hengyang , China
| | - Zhizhong Xie
- a Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study , University of South China , Hengyang , China
| | - Yanming Chen
- b Mu Dan Jiang You Bo Pharmacertical Co. Ltd , Mudanjiang , China
| | - Yunmei Liu
- a Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study , University of South China , Hengyang , China
| | - Xing Zheng
- a Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study , University of South China , Hengyang , China
| | - Guotao Tang
- a Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study , University of South China , Hengyang , China
| |
Collapse
|
58
|
Rao K, Sethi K, Ischia J, Gibson L, Galea L, Xiao L, Yim M, Chang M, Papa N, Bolton D, Shulkes A, Baldwin GS, Patel O. Protective effect of zinc preconditioning against renal ischemia reperfusion injury is dose dependent. PLoS One 2017; 12:e0180028. [PMID: 28686686 PMCID: PMC5501469 DOI: 10.1371/journal.pone.0180028] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 06/08/2017] [Indexed: 11/25/2022] Open
Abstract
Objectives Ischemia-reperfusion injury (IRI) is a major cause of acute kidney injury and chronic kidney disease. Two promising preconditioning methods for the kidney, intermittent arterial clamping (IC) and treatment with the hypoxia mimetic cobalt chloride, have never been directly compared. Furthermore, the protective efficacy of the chemically related transition metal Zn2+ against renal IRI is unclear. Although Co2+ ions have been shown to protect the kidney via hypoxia inducible factor (HIF), the effect of Zn2+ ions on the induction of HIF1α, HIF2α and HIF3α has not been investigated previously. Materials and methods The efficacy of different preconditioning techniques was assessed using a Sprague-Dawley rat model of renal IRI. Induction of HIF proteins following Zn2+ treatment of the human kidney cell lines HK-2 (immortalized normal tubular cells) and ACHN (renal cancer) was measured using Western Blot. Results Following 40 minutes of renal ischemia in rats, cobalt preconditioning offered greater protection against renal IRI than IC as evidenced by lower peak serum creatinine and urea concentrations. ZnCl2 (10 mg/kg) significantly lowered the creatinine and urea concentrations compared to saline-treated control rats following a clinically relevant 60 minutes of ischemia. Zn2+ induced expression of HIF1α and HIF2α but not HIF3α in HK-2 and ACHN cells. Conclusion ZnCl2 preconditioning protects against renal IRI in a dose-dependent manner. Further studies are warranted to determine the possible mechanisms involved, and to assess the benefit of ZnCl2 preconditioning for clinical applications.
Collapse
Affiliation(s)
- Kenny Rao
- Department of Surgery, The University of Melbourne Victoria, Australia
- Department of Urology Austin Health, Victoria, Australia
| | - Kapil Sethi
- Department of Surgery, The University of Melbourne Victoria, Australia
- Department of Urology Austin Health, Victoria, Australia
| | - Joseph Ischia
- Department of Surgery, The University of Melbourne Victoria, Australia
- Department of Urology Austin Health, Victoria, Australia
| | - Luke Gibson
- Department of Surgery, The University of Melbourne Victoria, Australia
- Department of Urology Austin Health, Victoria, Australia
| | - Laurence Galea
- Department of Anatomical Pathology, Austin Health, Victoria, Australia
| | - Lin Xiao
- Department of Surgery, The University of Melbourne Victoria, Australia
| | - Mildred Yim
- Department of Surgery, The University of Melbourne Victoria, Australia
| | - Mike Chang
- Department of Surgery, The University of Melbourne Victoria, Australia
| | - Nathan Papa
- Department of Urology Austin Health, Victoria, Australia
| | - Damien Bolton
- Department of Surgery, The University of Melbourne Victoria, Australia
- Department of Urology Austin Health, Victoria, Australia
| | - Arthur Shulkes
- Department of Surgery, The University of Melbourne Victoria, Australia
| | - Graham S. Baldwin
- Department of Surgery, The University of Melbourne Victoria, Australia
| | - Oneel Patel
- Department of Surgery, The University of Melbourne Victoria, Australia
- * E-mail:
| |
Collapse
|
59
|
Lee SY, Mustafa S, Ching YW, Shafee N. Zinc induces normoxic accumulation of transcriptionally active hypoxia-inducible factor 1-alpha in mammary epithelial cells. Mol Biol 2017. [DOI: 10.1134/s0026893317010113] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
60
|
Uribe D, Torres Á, Rocha JD, Niechi I, Oyarzún C, Sobrevia L, San Martín R, Quezada C. Multidrug resistance in glioblastoma stem-like cells: Role of the hypoxic microenvironment and adenosine signaling. Mol Aspects Med 2017; 55:140-151. [PMID: 28223127 DOI: 10.1016/j.mam.2017.01.009] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 12/29/2016] [Accepted: 01/08/2017] [Indexed: 12/11/2022]
Abstract
Glioblastoma multiforme (GBM) is considered the most common and aggressive tumour of the central nervous system and is characterized for being highly chemoresistant. This property is mainly due to the activation of Multiple Drug Resistance (MDR) mechanisms that protect cancer cells from structurally and morphologically different drugs. Overexpression and increased ABC transporters activity is one of the most important MDR mechanisms at the clinical level, and both its expression and activity are elevated in GBM cells. Within the tumour, there is a subpopulation called glioblastoma stem-like cells (GSCs), which due to its high tumourigenic capacity and chemoresistance, have been postulated as the main responsible for tumour recurrence. The GSCs inhabit hypoxic tumour zones, niches that apart from maintaining and promoting stem phenotype have also been correlated with high chemoresistance. Of the signalling pathways activated during hypoxia, purinergic signalling has been highly associated to the induction of MDR mechanisms. Through its receptors, the nucleoside adenosine has been shown to promotes the chemoresistance mediated by ABC transporters. Therefore, targeting its components is a promising alternative for GBM treatment. In this review, we will discuss chemoresistance in GSCs and the effect of the hypoxic microenvironment and adenosine on MDR mechanisms.
Collapse
Affiliation(s)
- Daniel Uribe
- Molecular Pathology Laboratory, Institute of Biochemistry and Microbiology, Science Faculty, Universidad Austral de Chile, Valdivia, Chile
| | - Ángelo Torres
- Molecular Pathology Laboratory, Institute of Biochemistry and Microbiology, Science Faculty, Universidad Austral de Chile, Valdivia, Chile
| | - José Dellis Rocha
- Molecular Pathology Laboratory, Institute of Biochemistry and Microbiology, Science Faculty, Universidad Austral de Chile, Valdivia, Chile
| | - Ignacio Niechi
- Molecular Pathology Laboratory, Institute of Biochemistry and Microbiology, Science Faculty, Universidad Austral de Chile, Valdivia, Chile
| | - Carlos Oyarzún
- Molecular Pathology Laboratory, Institute of Biochemistry and Microbiology, Science Faculty, Universidad Austral de Chile, Valdivia, Chile
| | - Luis Sobrevia
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; Department of Physiology, Faculty of Pharmacy, Universidad de Sevilla, Seville E-41012, Spain; University of Queensland Centre for Clinical Research (UQCCR), Faculty of Medicine and Biomedical Sciences, University of Queensland, Herston QLD 4029, Queensland, Australia
| | - Rody San Martín
- Molecular Pathology Laboratory, Institute of Biochemistry and Microbiology, Science Faculty, Universidad Austral de Chile, Valdivia, Chile
| | - Claudia Quezada
- Molecular Pathology Laboratory, Institute of Biochemistry and Microbiology, Science Faculty, Universidad Austral de Chile, Valdivia, Chile.
| |
Collapse
|
61
|
Himoto T, Fujita K, Nomura T, Tani J, Miyoshi H, Morishita A, Yoneyama H, Kubota S, Haba R, Suzuki Y, Masaki T. Roles of Copper in Hepatocarcinogenesis via the Activation of Hypoxia-Inducible Factor-1α. Biol Trace Elem Res 2016; 174:58-64. [PMID: 27121973 DOI: 10.1007/s12011-016-0702-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Accepted: 04/11/2016] [Indexed: 01/31/2023]
Abstract
Hypoxia-inducible factor-1α (HIF-1α) is involved in the pathogenesis of hepatocellular carcinoma (HCC). However, the roles of trace elements in the activation of HIF-1α during hepatocarcinogenesis have been unclear. We investigated whether copper (Cu) and zinc (Zn) participated in the activation of HIF-1α in the process of hepatocarcinogenesis or not. Nine patients with chronic hepatitis (CH), five with liver cirrhosis (LC), 12 with HCC, and nine normal healthy controls were enrolled in this study. Their serum HIF-1α, Cu, and Zn levels were determined in the enrolled patients. Hepatic HIF-1α expression was evaluated, using an immunohistochemical procedure. The HCC patients had significantly higher serum HIF-1α levels than the CH patients (6.47 ± 1.57 vs. 5.09 ± 1.22 ng/ml, p = 0.0344). The serum Cu level in the HCC patients was also significantly higher than those in the CH and LC patients (137 ± 24 vs. 107 ± 15 μg/dl, 114 ± 24 μg/dl). Interestingly, a positive correlation was observed between serum HIF-1α and Cu levels in the enrolled patients (r = 0.425, p = 0.0137). In contrast, no significant differences in serum Zn levels were present between the HCC patients and the CH or LC patients. The serum HIF-1α was not positively correlated with the serum Zn level in the enrolled patients, either. Immunohistochemical analysis revealed that two of the five HCC patients had HIF-1α expression in the tumor tissues, whereas none of CH and LC had hepatic HIF-1α expression in the liver tissues. These data suggest that the activation of HIF-1α derived from a Cu accumulation in the liver may cause hepatocarcinogenesis.
Collapse
Affiliation(s)
- Takashi Himoto
- Department of Medical Technology, Kagawa Prefectural University of Health Sciences, 281-1, Hara, Mure-cho, Takamatsu, Kagawa, 761-0123, Japan.
| | - Koji Fujita
- Department of Gastroenterology and Neurology, Kagawa, Japan
| | - Takako Nomura
- Department of Gastroenterology and Neurology, Kagawa, Japan
| | - Joji Tani
- Department of Gastroenterology and Neurology, Kagawa, Japan
| | | | | | | | | | - Reiji Haba
- Department of Diagnosis Pathology, Osaka, Japan
| | - Yasuyuki Suzuki
- Department of Gastroenterological Surgery, Kagawa University School of Medicine, Kagawa, Japan
| | - Tsutomu Masaki
- Department of Gastroenterology and Neurology, Kagawa, Japan
| |
Collapse
|
62
|
Zhang X, Liang D, Fan J, Lian X, Zhao Y, Wang X, Chi ZH, Zhang P. Zinc Attenuates Tubulointerstitial Fibrosis in Diabetic Nephropathy Via Inhibition of HIF Through PI-3K Signaling. Biol Trace Elem Res 2016; 173:372-83. [PMID: 26956696 DOI: 10.1007/s12011-016-0661-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Accepted: 02/23/2016] [Indexed: 10/22/2022]
Abstract
Evidence has demonstrated that hypoxia may have a central pathogenic mechanism in the development of diabetic nephropathy (DN). Epithelial-to-mesenchymal transition (EMT) of mature tubular epithelial cells in kidney is a contributor to the renal accumulation of matrix protein in DN and is highly associated with the progression of tubulointerstitial fibrosis. Zinc (Zn) has anti-fibrosis effects in liver and lungs. In the present study, we aimed to investigate the effect of Zn on renal tubulointerstitial fibrosis especially under hypoxic conditions and its association with DN. We found that Zn treatment blockaded tubular EMT and attenuated renal tubulointerstitial fibrosis by downregulation of hypoxia-inducible factor alpha (HIF-1α) in the kidneys of diabetic streptozotocin-treated mice. High glucose (HG)/hypoxic conditions stimulated EMT in renal tubular cells as indicated by the significant decrease in epithelial marker E-cadherin and ZO-1 while the increase in mesenchymal markers α-smooth muscle actin (α-SMA). Zn supplement mainly prevented HG/hypoxic-induced HIF-1α accumulation and EMT marker changes. In co-treatment Zn with PI3K/Akt/GSK-3β signaling pathway, inhibitor LY294002 prevented HG/hypoxic-induced HIF-1α increase and EMT changes, suggesting that Zn may mediate HG/hypoxic-induced EMT through PI3K/Akt/GSK-3β pathway. Therefore, we concluded that Zn had an important anti-fibrosis role under HG/hypoxic conditions, and a novel mechanism contributing to Zn protection on renal tubular epithelial cells from HG/hypoxia-induced EMT through activation of PI3K/Akt/GSK-3β signaling pathway, which subsequently leads to the downregulation of the expression of HIF-1α.
Collapse
Affiliation(s)
- Xiuli Zhang
- Key Laboratory of Medical Cell Biology, Ministry of Education, Shenyang, Liaoning, 110001, People's Republic of China
- Department of Nephrology, Benxi Center Hospital, Benxi, Liaoning, 117000, People's Republic of China
| | - Dan Liang
- Troops of 95935 Unit, Harbin, Heilongjiang, 158000, People's Republic of China
| | - Jingyue Fan
- Department of Cardiology, Shuangcheng District People's Hospital, Harbin, 150100, People's Republic of China
| | - Xu Lian
- Department of Endocrinology, The Affiliated Hospital of Mudanjiang Medical University, Mudanjiang, Heilongjiang, 157000, People's Republic of China
| | - Yue Zhao
- Key Laboratory of Medical Cell Biology, Ministry of Education, Shenyang, Liaoning, 110001, People's Republic of China
| | - Xuemei Wang
- Key Laboratory of Medical Cell Biology, Ministry of Education, Shenyang, Liaoning, 110001, People's Republic of China
| | - Zhi-Hong Chi
- Key Laboratory of Medical Cell Biology, Ministry of Education, Shenyang, Liaoning, 110001, People's Republic of China.
- Department of Histology and Embryology, Liaoning Medical University, Jinzhou, 121001, People's Republic of China.
| | - Ping Zhang
- Department of Pathophysiology, China Medical University, No. 77, Puhe Road, Shenbei New Area, Shenyang, Liaoning, 110001, People's Republic of China
| |
Collapse
|
63
|
Pearce SC, Sanz Fernandez MV, Torrison J, Wilson ME, Baumgard LH, Gabler NK. Dietary organic zinc attenuates heat stress-induced changes in pig intestinal integrity and metabolism. J Anim Sci 2016; 93:4702-13. [PMID: 26523563 DOI: 10.2527/jas.2015-9018] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Dietary zinc (inorganic and organic or zinc AA complex forms) is essential for normal intestinal barrier function and regeneration of intestinal epithelium. Given that heat stress (HS) exposure can negatively affect intestinal integrity and caloric intake, possible nutritional mitigation strategies are needed to improve health, performance, and well-being. Therefore, our objective was to evaluate 2 dietary zinc sources and reduced caloric intake on intestinal integrity in growing pigs subjected to 12 h of HS. A total of 36 pigs were fed 1 of 2 diets: 1) a control diet (CON; 120 mg/kg of zinc from zinc sulfate) or 2) 60 mg/kg from zinc sulfate and 60 mg/kg from zinc AA complex (ZnAA). After 17 d, the CON pigs were then exposed to thermal neutral (TN) conditions with ad libitum intake (TN-CON), HS (37°C) with ad libitum intake (HS-CON), or pair-fed to HS intake under TN conditions (PFTN); the ZnAA pigs were exposed to only HS (HS-ZnAA). All pigs were sacrificed after 12 h of environmental exposure, and blood and tissue bioenergetics stress markers and ex vivo ileum and colon integrity were assessed. Compared with TN-CON, HS significantly ( < 0.05) increased rectal temperatures and respiration rates. Ileum villus and crypt morphology was reduced by both pair-feeding and HS. Both PFTN and HS-CON pigs also had reduced ileum integrity (dextran flux and transepithelial resistance) compared with the TN-CON pigs. However, ZnAA tended to mitigate the HS-induced changes in ileum integrity. Ileum mucin 2 protein abundance was increased due to HS and pair-feeding. Colonic integrity did not differ due to HS or PFTN treatments. Compared with the HS-CON, HS-ZnAA pigs tended to have reduced blood endotoxin concentrations. In conclusion, HS and reduced feed intake compromised intestinal integrity in pigs, and zinc AA complex source mitigates some of these negative effects.
Collapse
|
64
|
Garufi A, Trisciuoglio D, Cirone M, D'Orazi G. ZnCl2 sustains the adriamycin-induced cell death inhibited by high glucose. Cell Death Dis 2016; 7:e2280. [PMID: 27362798 PMCID: PMC5108333 DOI: 10.1038/cddis.2016.178] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 05/14/2016] [Accepted: 05/30/2016] [Indexed: 12/15/2022]
Abstract
Hyperglycemia, the condition of high blood glucose, is typical of diabetes and obesity and represents a significant clinical problem. The relationship between hyperglycemia and cancer risk has been established by several studies. Moreover, hyperglycemia has been shown to reduce cancer cell response to therapies, conferring resistance to drug-induced cell death. Therefore, counteracting the negative effects of hyperglycemia may positively improve the cancer cell death induced by chemotherapies. Recent studies showed that zinc supplementation may have beneficial effects on glycemic control. Here we aimed at evaluating whether ZnCl2 could counteract the high-glucose (HG) effects and consequently restore the drug-induced cancer cell death. At the molecular level we found that the HG-induced expression of genes known to be involved in chemoresistance (such as HIF-1α, GLUT1, and HK2 glycolytic genes, as well as NF-κB activity) was reduced by ZnCl2 treatment. In agreement, the adryamicin (ADR)-induced apoptotic cancer cell death was significantly impaired by HG and efficiently re-established by ZnCl2 cotreatment. Mechanistically, the ADR-induced c-Jun N-terminal kinase/stress-activated protein kinase (JNK/SAPK) phosphorylation, inhibited by HG, was efficiently restored by ZnCl2. The JNK involvement in apoptotic cell death was assessed by the use of JNK dominant-negative expression vector that indeed impaired the ZnCl2 ability to restore drug-induced cell death in HG condition. Altogether, these findings indicate that ZnCl2 supplementation efficiently restored the drug-induced cancer cell death, inhibited by HG, by both sustaining JNK activation and counteracting the glycolytic pathway.
Collapse
Affiliation(s)
- A Garufi
- Department of Research, Advanced Diagnostics, and Technological Innovation, Regina Elena National Cancer Institute, Rome, Italy.,Department of Medical, Oral and Biotechnological Sciences, Tumor Biology Section, University 'G. d'Annunzio', Chieti, Italy
| | - D Trisciuoglio
- Department of Research, Advanced Diagnostics, and Technological Innovation, Regina Elena National Cancer Institute, Rome, Italy
| | - M Cirone
- Department of Experimental Medicine, Pasteur-Fondazione Cenci Bolognetti Institute, Sapienza University, Rome, Italy
| | - G D'Orazi
- Department of Research, Advanced Diagnostics, and Technological Innovation, Regina Elena National Cancer Institute, Rome, Italy.,Department of Medical, Oral and Biotechnological Sciences, Tumor Biology Section, University 'G. d'Annunzio', Chieti, Italy
| |
Collapse
|
65
|
l-carnosine dipeptide overcomes acquired resistance to 5-fluorouracil in HT29 human colon cancer cells via downregulation of HIF1-alpha and induction of apoptosis. Biochimie 2016; 127:196-204. [PMID: 27234614 DOI: 10.1016/j.biochi.2016.05.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 05/16/2016] [Indexed: 01/19/2023]
Abstract
Hypoxia-inducible factor (HIF-1α) protein is over-expressed in many human cancers and is a major cause of resistance to drugs. HIF-1α up-regulation decreases the effectiveness of several anticancer agents, including 5-fluorouracil (5-FU), because it induces the expression of drug efflux transporters, alters DNA repair mechanisms and modifies the balance between pro- and antiapoptotic factors. These findings suggest that inhibition of HIF-1α activity may sensitize cancer cells to cytotoxic drugs. We previously reported that l-carnosine reduces HIF-1α expression by inhibiting the proliferation of colon cancer cells. In the present study we investigated the effect of l-carnosine on HT29 colon cancer cells with acquired resistance to 5-FU. We found that l-carnosine reduces colon cancer cell viability, decreases HIF-1α and multi-drug resistant protein MDR1-pg expression, and induces apoptosis. Moreover, the l-carnosine/5-FU combination lowers the expression of some chemoresistance markers. The combination index evaluated in vitro on the HT29-5FU cell line by median drug effect analysis reveals a significant synergistic effect.
Collapse
|
66
|
Liao W, Lai T, Chen L, Fu J, Sreenivasan ST, Yu Z, Ren J. Synthesis and Characterization of a Walnut Peptides-Zinc Complex and Its Antiproliferative Activity against Human Breast Carcinoma Cells through the Induction of Apoptosis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2016; 64:1509-1519. [PMID: 26878665 DOI: 10.1021/acs.jafc.5b04924] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
The walnut peptides and zinc ions were combined to generate a walnut peptides-zinc complex (WP1-Zn) with enhanced antiproliferative ability as well as reduced toxicity. The result indicated that Zn ions were successfully combined with WP1 through Zn-N and Zn-O covalent bonds. WP1-Zn compounds exhibited strong antiproliferative ability against the selected human cell lines, especially MCF-7 cells, whose survival rate reduced to 20.02% after exposure to 300 μg/mL of WP1-Zn for 48 h. WP1-Zn inhibited MCF-7 cell proliferation through inducing cell apoptosis and cell cycle arrest. The results indicated that WP1-Zn induced MCF-7 cell apoptosis via the ROS triggered mitochondrial-mediated pathway and cell surface receptor-mediated pathway. Our work is the first attempt to elucidate the synergic effect of novel walnut peptides and Zn and with the hope of better understanding the antiproliferative action of bioactive peptides and a zinc complex and support the potential application of WP1-Zn as a functional food ingredient or complementary medicine.
Collapse
Affiliation(s)
- Wenzhen Liao
- Department of Food Science and Technology, South China University of Technology , Wushan Road 381, Guangzhou 510640, China
| | - Ting Lai
- Department of Food Science and Technology, South China University of Technology , Wushan Road 381, Guangzhou 510640, China
| | - Luying Chen
- Department of Food Science and Technology, South China University of Technology , Wushan Road 381, Guangzhou 510640, China
| | - Junning Fu
- Department of Food Science and Engineering, Jinan University , Guangzhou 510623, China
| | - Sreeprasad T Sreenivasan
- Department of Civil and Environmental Engineering, Rice University , 6100 Main Street, Houston, Texas 77005, United States
| | - Zhiqiang Yu
- School of Pharmaceutical Science, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University , Guangzhou, Guangdong 510515, China
| | - Jiaoyan Ren
- Department of Food Science and Technology, South China University of Technology , Wushan Road 381, Guangzhou 510640, China
| |
Collapse
|
67
|
Kang X, Chen R, Zhang J, Li G, Dai PG, Chen C, Wang HJ. Expression Profile Analysis of Zinc Transporters (ZIP4, ZIP9, ZIP11, ZnT9) in Gliomas and their Correlation with IDH1 Mutation Status. Asian Pac J Cancer Prev 2016; 16:3355-60. [PMID: 25921144 DOI: 10.7314/apjcp.2015.16.8.3355] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Zinc transporters have been considered as essential regulators in many cancers; however, their mechanisms remain unknown, especially in gliomas. Isocitrate dehydrogenase 1(IDH1) mutation is crucial to glioma. This study aimed to investigate whether zinc transporters are correlated with glioma grade and IDH1 mutation status. MATERIALS AND METHODS IDH1 mutation status and mRNA expression of four zinc transporters (ZIP4, ZIP9, ZIP11, and ZnT9) were determined by subjecting a panel of 74 glioma tissue samples to quantitative real-time PCR and pyrosequencing. The correlations between the expression levels of these zinc transporter genes and the grade of glioma, as well as IDH1 mutation status, were investigated. RESULTS Among the four zinc transporter genes, high ZIP4 expression and low ZIP11 expression were significantly associated with higher grade (grades III and IV) tumors compared with lower grade (grades I and II) counterparts (p<0.0001). However, only ZIP11 exhibited weak correlation with IDH1 mutation status (p=0.045). Samples with mutations in IDH1 displayed higher ZIP11 expression than those without IDH1 mutations. CONCLUSIONS This finding indicated that zinc transporters may interact with IDH1 mutation by direct modulation or action in some shared pathways or genes to promote the development of glioma. Zinc transporters may play an important role in glioma. ZIP4 and ZIP11 are promising molecular diagnostic markers and novel therapeutic targets. Nevertheless, the detailed biological function of zinc transporters and the mechanism of the potential interaction between ZIP11 and IDH1 mutation in gliomagenesis should be further investigated.
Collapse
Affiliation(s)
- Xing Kang
- School of Life Sciences, Northwest University, Xi'an, China E-mail : ,
| | | | | | | | | | | | | |
Collapse
|
68
|
Cheng CC, Guan SS, Yang HJ, Chang CC, Luo TY, Chang J, Ho AS. Blocking heme oxygenase-1 by zinc protoporphyrin reduces tumor hypoxia-mediated VEGF release and inhibits tumor angiogenesis as a potential therapeutic agent against colorectal cancer. J Biomed Sci 2016; 23:18. [PMID: 26822586 PMCID: PMC4730655 DOI: 10.1186/s12929-016-0219-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 01/11/2016] [Indexed: 01/30/2023] Open
Abstract
Background Hypoxia in tumor niche is one of important factors to start regeneration of blood vessels, leading to increase survival, proliferation, and invasion in cancer cells. Under hypoxia microenvironment, furthermore, steadily increased hypoxia-inducible factor-1α (HIF-1α) is observed, and can increase vascular endothelial growth factor (VEGF) expression and promote angiogenesis. Zinc protoporphyrin (ZnPP), a heme oxygenase-1 (HO-1) inhibitor, is potential to inhibit tumor proliferation and progression. However, the mechanism of ZnPP in inhibition of tumor is not completely clear. We hypothesize that ZnPP may modulate HIF-1α through inhibiting HO-1, and then inhibit angiogenesis and tumor progression. This study aimed to dissect the mechanism of ZnPP in tumor suppression. Results We observed the amount of VEGF was increased in the sera of the colorectal cancer (CRC) patients (n = 34, p < 0.05). Furthermore, increased VEGF expression was also measured in colorectal cancer cells, HCT-15, culturing under mimicking hypoxic condition. It suggested that hypoxia induced VEGF production from cancer cells. VEGF production was significantly reduced from HCT-15 cells after exposure to HIF-1α inhibitor KC7F2, suggesting that HIF-1α regulated VEGF production. Moreover, we observed that the HO-1inhibitor ZnPP inhibited the expressions of HIF-1α and VEGF coupled with cell proliferations of HCT-15 cells, suggesting that ZnPP blocked HIF-1α expression, and then inhibited the consequent VEGF production. In the xenograft model, we also observed that the animals exposed to ZnPP displayed much smaller tumor nodules and less degree of angiogenesis with decreased expression of the angiogenesis marker, αvβ3 integrin, compared to that in normal control. Conclusions This study demonstrated that VEGF level in serum was elevated in the patients with CRC. The HO-1 inhibitor, ZnPP, possessed the properties of anti-tumor agent by decreasing HIF-1α levels, blocking VEGF production, impairing tumor angiogenesis, and inhibiting tumor growth.
Collapse
Affiliation(s)
- Chun-Chia Cheng
- Institute of Nuclear Energy Research, Atomic Energy Council, Taoyuan, Taiwan.,Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Siao-Syun Guan
- Institute of Nuclear Energy Research, Atomic Energy Council, Taoyuan, Taiwan
| | - Hao-Jhih Yang
- Institute of Nuclear Energy Research, Atomic Energy Council, Taoyuan, Taiwan
| | - Chun-Chao Chang
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei, Taiwan.,Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Tsai-Yueh Luo
- Institute of Nuclear Energy Research, Atomic Energy Council, Taoyuan, Taiwan
| | - Jungshan Chang
- Graduate Institute of Medical Sciences, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| | - Ai-Sheng Ho
- Division of Gastroenterology, Cheng Hsin General Hospital, Taipei, Taiwan. .,Nursing Department, Kang-Ning University, Taipei, Taiwan.
| |
Collapse
|
69
|
GARUFI ALESSIA, D'ORAZI VALERIO, CRISPINI ALESSANDRA, D'ORAZI GABRIELLA. Zn(II)-curc targets p53 in thyroid cancer cells. Int J Oncol 2015; 47:1241-1248. [PMID: 26314369 PMCID: PMC4583539 DOI: 10.3892/ijo.2015.3125] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 07/13/2015] [Indexed: 02/06/2023] Open
Abstract
TP53 mutation is a common event in many cancers, including thyroid carcinoma. Defective p53 activity promotes cancer resistance to therapies and a more malignant phenotype, acquiring oncogenic functions. Rescuing the function of mutant p53 (mutp53) protein is an attractive anticancer therapeutic strategy. Zn(II)-curc is a novel small molecule that has been shown to target mutp53 protein in several cancer cells, but its effect in thyroid cancer cells remains unclear. Here, we investigated whether Zn(II)-curc could affect p53 in thyroid cancer cells with both p53 mutation (R273H) and wild-type p53. Zn(II)-curc induced mutp53H273 downregulation and reactivation of wild-type functions, such as binding to canonical target promoters and target gene transactivation. This latter effect was similar to that induced by PRIMA-1. In addition, Zn(II)-curc triggered p53 target gene expression in wild-type p53-carrying cells. In combination treatments, Zn(II)-curc enhanced the antitumor activity of chemotherapeutic drugs, in both mutant and wild-type-carrying cancer cells. Taken together, our data indicate that Zn(II)-curc promotes the reactivation of p53 in thyroid cancer cells, providing in vitro evidence for a potential therapeutic approach in thyroid cancers.
Collapse
Affiliation(s)
- ALESSIA GARUFI
- Department of Experimental Oncology, Regina Elena National Cancer Institute, Rome, Italy
- Department of Medical, Oral and Biotechnological Sciences, University ‘G. d’Annunzio’, Chieti, Italy
| | - VALERIO D'ORAZI
- Department of Surgical Sciences, Sapienza University, Rome, Italy
| | - ALESSANDRA CRISPINI
- Department of Chemistry and Technologic Chemistry, University of Calabria, Cosenza, Italy
| | - GABRIELLA D'ORAZI
- Department of Experimental Oncology, Regina Elena National Cancer Institute, Rome, Italy
- Department of Medical, Oral and Biotechnological Sciences, University ‘G. d’Annunzio’, Chieti, Italy
| |
Collapse
|
70
|
Garufi A, Ubertini V, Mancini F, D'Orazi V, Baldari S, Moretti F, Bossi G, D'Orazi G. The beneficial effect of Zinc(II) on low-dose chemotherapeutic sensitivity involves p53 activation in wild-type p53-carrying colorectal cancer cells. J Exp Clin Cancer Res 2015; 34:87. [PMID: 26297485 PMCID: PMC4546314 DOI: 10.1186/s13046-015-0206-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 08/12/2015] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Activation of wild-type p53 in response to genotoxic stress occurs through different mechanisms including protein conformation, posttranslational modifications, and nuclear localization, leading to DNA binding to sequence-specific promoters. Zinc ion plays a crucial role in stabilizing p53/DNA binding to induce canonical target genes. Mutant p53 proteins undergo protein misfolding that can be counteracted by zinc. However, whether zinc supplementation might have a beneficial antitumor effect in wild-type p53-carrying cells in combination with drugs, has not been addressed so far. METHODS In this study we compared the effect of two antitumor treatments: on the one hand wild-type p53-carrying colon cancer cells were treated with low and high doses of chemotherapeutic agent Adriamycin and, on the other hand, Adriamycin was used in combination with ZnCl2. Biochemical and molecular analyses were applied to evaluate p53 activity and biological outcomes in this setting. Finally, the effect of the different combination treatments were applied to assess tumor growth in vivo in tumor xenografts. RESULTS We found that low-dose Adriamycin did not induce p53 activation in wtp53-carrying colon cancer cells, unless in combination with ZnCl2. Mechanistically, ZnCl2 was a key determinant in inducing wtp53/DNA binding and transactivation of target genes in response to low-dose Adriamycin that used alone did not achieve such effects. Finally, in vivo studies, in a model of wtp53 colon cancer xenograft, show that low-dose Adriamycin did not induce tumor regression unless in combination with ZnCl2 that activated endogenous wtp53. CONCLUSIONS These results provide evidence that ZnCl2 might be a valuable adjuvant in chemotherapeutic regimens of colorectal cancer harboring wild-type p53, able to both activate p53 and reduce the amount of drugs for antitumor purposes.
Collapse
Affiliation(s)
- Alessia Garufi
- Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio", 66013, Chieti, Italy.
- Translational Research Department, Regina Elena National Cancer Institute, 00158, Rome, Italy.
| | - Valentina Ubertini
- Translational Research Department, Regina Elena National Cancer Institute, 00158, Rome, Italy.
| | - Francesca Mancini
- Institute of Cell Biology and Neurobiology, National Research Council of Italy (CNR), Roma, Italy.
| | - Valerio D'Orazi
- Department of Surgical Sciences, Sapienza University, 00100, Rome, Italy.
| | - Silvia Baldari
- Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio", 66013, Chieti, Italy.
- Translational Research Department, Regina Elena National Cancer Institute, 00158, Rome, Italy.
| | - Fabiola Moretti
- Institute of Cell Biology and Neurobiology, National Research Council of Italy (CNR), Roma, Italy.
| | - Gianluca Bossi
- Laboratory of Medical Physics and Expert Systems, Regina Elena National Cancer Institute, 00158, Rome, Italy.
| | - Gabriella D'Orazi
- Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio", 66013, Chieti, Italy.
- Translational Research Department, Regina Elena National Cancer Institute, 00158, Rome, Italy.
| |
Collapse
|
71
|
Porru M, Zappavigna S, Salzano G, Luce A, Stoppacciaro A, Balestrieri ML, Artuso S, Lusa S, De Rosa G, Leonetti C, Caraglia M. Medical treatment of orthotopic glioblastoma with transferrin-conjugated nanoparticles encapsulating zoledronic acid. Oncotarget 2015; 5:10446-59. [PMID: 25431953 PMCID: PMC4279385 DOI: 10.18632/oncotarget.2182] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 07/08/2014] [Indexed: 01/12/2023] Open
Abstract
Glioblastomas are highly aggressive adult brain tumors with poor clinical outcome. In the central nervous system (CNS) the blood-brain barrier (BBB) is the most important limiting factor for both development of new drugs and drug delivery. Here, we propose a new strategy to treat glioblastoma based on transferrin (Tf)-targeted self-assembled nanoparticles (NPs) incorporating zoledronic acid (ZOL) (NPs-ZOL-Tf). NPs-ZOL-Tf have been assessed on the glioblastoma cell line U373MG-LUC that showed a refractoriness in vitro to temozolomide (TMZ) and fotemustine (FTM). NPs-ZOL-Tf treatment resulted in higher in vitro cytotoxic activity than free ZOL. However, the potentiation of anti-proliferative activity of NPs-ZOL-Tf was superimposable to that one induced by NPs-ZOL (not armed with Tf). On the other hand, NPs-ZOL-Tf showed a higher antitumor efficacy if compared with that one caused by NPs-ZOL in immunosuppressed mice intramuscularly bearing U373MG-LUC xenografts, inducing a significant tumor weight inhibition (TWI). The experiments performed on mice with intracranial U373MG-LUC xenografts confirmed the efficacy of NPs-ZOL-Tf. These effects were paralleled by a higher intratumour localization of fluorescently-labeled-NPs-Tf both in intramuscular and intracranial xenografts. In conclusion, our results demonstrate that the encapsulation of ZOL increases the antitumor efficacy of this drug in glioblastoma through the acquisition of ability to cross the BBB.
Collapse
Affiliation(s)
- Manuela Porru
- Experimental Chemotherapy Laboratory, Regina Elena National Cancer Institute, Via delle Messi d'oro, Rome, Italy
| | - Silvia Zappavigna
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Via Costantinopoli, Naples, Italy
| | - Giuseppina Salzano
- Department of Pharmacy, University of Naples Federico II, Via Montesano, Naples, Italy
| | - Amalia Luce
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Via Costantinopoli, Naples, Italy
| | - Antonella Stoppacciaro
- Experimental Chemotherapy Laboratory, Regina Elena National Cancer Institute, Via delle Messi d'oro, Rome, Italy
| | - Maria Luisa Balestrieri
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Via Costantinopoli, Naples, Italy
| | - Simona Artuso
- Experimental Chemotherapy Laboratory, Regina Elena National Cancer Institute, Via delle Messi d'oro, Rome, Italy
| | - Sara Lusa
- Department of Pharmacy, University of Naples Federico II, Via Montesano, Naples, Italy
| | - Giuseppe De Rosa
- Department of Pharmacy, University of Naples Federico II, Via Montesano, Naples, Italy
| | - Carlo Leonetti
- Experimental Chemotherapy Laboratory, Regina Elena National Cancer Institute, Via delle Messi d'oro, Rome, Italy
| | - Michele Caraglia
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Via Costantinopoli, Naples, Italy
| |
Collapse
|
72
|
Womeldorff M, Gillespie D, Jensen RL. Hypoxia-inducible factor-1 and associated upstream and downstream proteins in the pathophysiology and management of glioblastoma. Neurosurg Focus 2015; 37:E8. [PMID: 25581937 DOI: 10.3171/2014.9.focus14496] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Glioblastoma multiforme (GBM) is a highly aggressive brain tumor with an exceptionally poor patient outcome despite aggressive therapy including surgery, radiation, and chemotherapy. This aggressive phenotype may be associated with intratumoral hypoxia, which probably plays a key role in GBM tumor growth, development, and angiogenesis. A key regulator of cellular response to hypoxia is the protein hypoxia-inducible factor–1 (HIF-1). An examination of upstream hypoxic and nonhypoxic regulation of HIF-1 as well as a review of the downstream HIF-1– regulated proteins may provide further insight into the role of this transcription factor in GBM pathophysiology. Recent insights into upstream regulators that intimately interact with HIF-1 could provide potential therapeutic targets for treatment of this tumor. The same is potentially true for HIF-1–mediated pathways of glycolysis-, angiogenesis-, and invasion-promoting proteins. Thus, an understanding of the relationship between HIF-1, its upstream protein regulators, and its downstream transcribed genes in GBM pathogenesis could provide future treatment options for the care of patients with these tumors.
Collapse
|
73
|
Functional role of inorganic trace elements in angiogenesis-Part II: Cr, Si, Zn, Cu, and S. Crit Rev Oncol Hematol 2015; 96:143-55. [PMID: 26088455 DOI: 10.1016/j.critrevonc.2015.05.011] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 04/15/2015] [Accepted: 05/12/2015] [Indexed: 02/07/2023] Open
Abstract
Trace elements play critical roles in angiogenesis events. The effects of nitrogen, iron, selenium, phosphorus, gold, and calcium were discussed in part I. In part II, we evaluated the effect of chromium, silicon, zinc, copper, and sulfur on different aspects of angiogenesis, with critical roles in healing and regeneration processes, and undeniable roles in tumor growth and cancer therapy. This review is the second of series that serves as an overview of the role of inorganic elements in regulation of angiogenesis and vascular function. The methods of exposure, structure, mechanism, and potential activity of these trace elements are briefly discussed. An electronic search was performed on the role of these trace elements in angiogenesis from January 2005 to April 2014. The recent aspects of the relationship between five different trace elements and their role in regulation of angiogenesis, and homeostasis of pro- and anti-angiogenic factors were assessed. Many studies have investigated the effects and importance of these elements in angiogenesis events. Both stimulatory and inhibitory effects on angiogenesis are observed for the evaluated elements. Chromium can promote angiogenesis in pathological manners. Silicon as silica nanoparticles is anti-angiogenic, while in calcium silicate extracts and bioactive silicate glasses promote angiogenesis. Zinc is an anti-angiogenic agent acting on important genes and growth factors. Copper and sulfur compositions have pro-angiogenic functions by activating pro-angiogenic growth factors and promoting endothelial cells migration, growth, and tube formation. Thus, utilization of these elements may provide a unique opportunity to modulate angiogenesis under various setting.
Collapse
|
74
|
Luwor RB, Stylli SS, Kaye AH. Using bioluminescence imaging in glioma research. J Clin Neurosci 2015; 22:779-84. [DOI: 10.1016/j.jocn.2014.11.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 11/03/2014] [Indexed: 01/02/2023]
|
75
|
Holubova M, Axmanova M, Gumulec J, Raudenska M, Sztalmachova M, Babula P, Adam V, Kizek R, Masarik M. KRAS NF-κB is involved in the development of zinc resistance and reduced curability in prostate cancer. Metallomics 2015; 6:1240-53. [PMID: 24927480 DOI: 10.1039/c4mt00065j] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Zinc(II) ions are important components of many proteins and are involved in numerous cellular processes such as apoptosis or drug resistance. Prostate cancer has a unique relationship with zinc(II) ions. However, the relationship was examined only in short-term zinc(II) treatments. Therefore, the aim of this study was to create zinc-resistant prostatic cell lines at various stages of the disease (22Rv1 and PC-3) and a normal prostate epithelium (PNT1A) using a long-term zinc exposure. Consequently, the expression profile of the following genes was analyzed: BAX, Bcl-2, Beclin-1, CFLAR, HIF1α, KRAS, mTOR, MT1A, MT2A, NF-κB1, p53, survivin, ZIP1, ZnT-1. The resistance was verified using the MTT test; on average a 1.35-fold lower zinc(II) toxicity (higher IC50) was determined in zinc(II)-resistant cells. The associated resistance to cisplatin was also determined; IC50 for cisplatin was 1.52-fold higher. With regard to the gene expression profiles, our results indicate that differential mechanisms participate in the short-term zinc toxicity regulation and long-term resistance; the short-term treatment was associated with MT2A (p < 0.001), ZnT-1 (p < 0.001), and MT1A (p < 0.03) and the long-term resistance was associated particularly with NF-κB1 (p < 0.001), CFLAR (p < 0.001), KRAS (p < 0.001), p53 (p < 0.002), survivin (p = 0.02), ZIP1 (p = 0.002), BAX (p = 0.005), and HIF1α (p = 0.05). Therefore, the KRAS-PI3K-NF-κB pathway is expected to play a crucial role in the regulation of zinc resistance. In summary, compared to previous studies, identical mechanisms of resistance were demonstrated on multiple cell lines, both non-tumor and tumorous, derived both from primary and advanced secondary sites.
Collapse
Affiliation(s)
- Monika Holubova
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, CZ-625 00 Brno, Czech Republic.
| | | | | | | | | | | | | | | | | |
Collapse
|
76
|
Gerber PA, Bellomo EA, Hodson DJ, Meur G, Solomou A, Mitchell RK, Hollinshead M, Chimienti F, Bosco D, Hughes SJ, Johnson PRV, Rutter GA. Hypoxia lowers SLC30A8/ZnT8 expression and free cytosolic Zn2+ in pancreatic beta cells. Diabetologia 2014; 57:1635-44. [PMID: 24865615 PMCID: PMC4079946 DOI: 10.1007/s00125-014-3266-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Accepted: 04/23/2014] [Indexed: 12/16/2022]
Abstract
AIMS/HYPOTHESIS Hypoxic damage complicates islet isolation for transplantation and may contribute to beta cell failure in type 2 diabetes. Polymorphisms in the SLC30A8 gene, encoding the secretory granule zinc transporter 8 (ZnT8), influence type 2 diabetes risk, conceivably by modulating cytosolic Zn(2+) levels. We have therefore explored the role of ZnT8 and cytosolic Zn(2+) in the response to hypoxia of pancreatic islet cells. METHODS Human, mouse or rat islets were isolated and exposed to varying O2 tensions. Cytosolic free zinc was measured using the adenovirally expressed recombinant targeted zinc probe eCALWY4. Gene expression was measured using quantitative (q)RT-PCR, western (immuno-) blotting or immunocytochemistry. Beta cells were identified by insulin immunoreactivity. RESULTS Deprivation of O2 (1% vs 5% or 21%) for 24 h lowered free cytosolic Zn(2+) concentrations by ~40% (p < 0.05) and ~30% (p < 0.05) in mouse and human islet cells, respectively. Hypoxia similarly decreased SLC30A8 mRNA expression in islets, and immunoreactivity in beta cells. Implicating lowered ZnT8 levels in the hypoxia-induced fall in cytosolic Zn(2+), genetic ablation of Slc30a8 from mouse islets lowered cytosolic Zn(2+) by ~40% (p < 0.05) and decreased the induction of metallothionein (Mt1, Mt2) genes. Cell survival in the face of hypoxia was enhanced in small islets of older (>12 weeks) Slc30a8 null mice vs controls, but not younger animals. CONCLUSIONS/INTERPRETATION The response of pancreatic beta cells to hypoxia is characterised by decreased SLC30A8 expression and lowered cytosolic Zn(2+) concentrations. The dependence on ZnT8 of hypoxia-induced changes in cell survival may contribute to the actions of SLC30A8 variants on diabetes risk in humans.
Collapse
Affiliation(s)
- Philipp A. Gerber
- Section of Cell Biology, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College London, London, W12 ONN UK
- Division of Endocrinology, Diabetes and Clinical Nutrition, University Hospital Zurich, Zurich, Switzerland
| | - Elisa A. Bellomo
- Section of Cell Biology, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College London, London, W12 ONN UK
| | - David J. Hodson
- Section of Cell Biology, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College London, London, W12 ONN UK
| | - Gargi Meur
- Section of Cell Biology, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College London, London, W12 ONN UK
| | - Antonia Solomou
- Section of Cell Biology, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College London, London, W12 ONN UK
| | - Ryan K. Mitchell
- Section of Cell Biology, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College London, London, W12 ONN UK
| | - Michael Hollinshead
- Section of Microscopy, Department of Medicine, Imperial College London, London, UK
| | | | - Domenico Bosco
- Cell Isolation and Transplantation Centre, Department of Surgery, Geneva University Hospital, Geneva, Switzerland
| | - Stephen J. Hughes
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
- DRWF Human Islet Isolation Facility, Oxford Centre for Diabetes, Endocrinology and Metabolism, Oxford, UK
- Oxford NIHR Biomedical Research Centre, Oxford, UK
| | - Paul R. V. Johnson
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
- DRWF Human Islet Isolation Facility, Oxford Centre for Diabetes, Endocrinology and Metabolism, Oxford, UK
- Oxford NIHR Biomedical Research Centre, Oxford, UK
| | - Guy A. Rutter
- Section of Cell Biology, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College London, London, W12 ONN UK
| |
Collapse
|
77
|
Alkamal I, Ikromov O, Tölle A, Fuller TF, Magheli A, Miller K, Krause H, Kempkensteffen C. An epigenetic screen unmasks metallothioneins as putative contributors to renal cell carcinogenesis. Urol Int 2014; 94:99-110. [PMID: 24662736 DOI: 10.1159/000357282] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Accepted: 11/13/2013] [Indexed: 11/19/2022]
Abstract
OBJECTIVE Functional epigenetic studies aimed to re-express transcriptionally silenced genes in renal cell carcinoma (RCC) may facilitate the ongoing search for appropriate markers supporting clinical decision-making. METHODS The RCC cell line A-498 was treated with the DNA methyltransferase inhibitor zebularine under low-cytotoxicity conditions. RNA chip analyses revealed several upregulated transcripts that were further validated by qPCR on 49 matched pairs of human kidney tissues to identify suitable marker candidates. RESULTS Members of the metallothionein (MT) group were remarkably downregulated in tumor tissues. MT1G and MT1H expression was decreased in 98% of cases, whereas MT2A expression was downregulated in 73% of all cases. Comparison of 308 reactivated transcripts upregulated more than 1.5-fold to published data revealed a high number of shared candidates, which supports the consistency of this experimental approach. CONCLUSION MTs were found to be transcriptionally inactivated in human RCC. Our observations support the hypothesis of a possible involvement of these metalloproteins in renal cell carcinogenesis. Additional functional studies of these genes may provide clues for understanding renal cancers as essentially metabolic diseases.
Collapse
Affiliation(s)
- Imad Alkamal
- Klinik für Urologie, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | | | | | | | | | | | | | | |
Collapse
|
78
|
Wang S, Hannafon BN, Wolf RF, Zhou J, Avery JE, Wu J, Lind SE, Ding WQ. Characterization of docosahexaenoic acid (DHA)-induced heme oxygenase-1 (HO-1) expression in human cancer cells: the importance of enhanced BTB and CNC homology 1 (Bach1) degradation. J Nutr Biochem 2014; 25:515-25. [PMID: 24613086 DOI: 10.1016/j.jnutbio.2013.12.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Revised: 12/18/2013] [Accepted: 12/31/2013] [Indexed: 10/25/2022]
Abstract
The effect of docosahexaenoic acid (DHA) on heme oxygenase-1 (HO-1) expression in cancer cells has never been characterized. This study examines DHA-induced HO-1 expression in human cancer cell model systems. DHA enhanced HO-1 gene expression in a time- and concentration-dependent manner, with maximal induction at 21 h of treatment. This induction of HO-1 expression was confirmed in vivo using a xenograft nude mouse model fed a fish-oil-enriched diet. The increase in HO-1 gene transcription induced by DHA was significantly attenuated by the antioxidant N-acetyl cysteine, suggesting the involvement of oxidative stress. This was supported by direct measurement of lipid peroxide levels after DHA treatment. Using a human HO-1 gene promoter reporter construct, we identified two antioxidant response elements (AREs) that mediate the DHA-induced increase in HO-1 gene transcription. Knockdown of nuclear factor (erythroid-derived 2)-like 2 (Nrf2) expression compromised the DHA-induced increase in HO-1 gene transcription, indicating the importance of the Nrf2 pathway in this event. However, the nuclear protein levels of Nrf2 remained unchanged upon DHA treatment. Further studies demonstrated that DHA reduces nuclear Bach1 protein expression by promoting its degradation and attenuates Bach1 binding to the AREs in the HO-1 gene promoter. In contrast, DHA enhanced Nrf2 binding to the AREs without affecting nuclear Nrf2 expression levels, indicating a new cellular mechanism that mediates DHA's induction of HO-1 gene transcription. To our knowledge, this is the first characterization of DHA-induced HO-1 expression in human malignant cells.
Collapse
Affiliation(s)
- Shuai Wang
- Department of Pathology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 411A, Oklahoma City, OK 73104, USA
| | - Bethany N Hannafon
- Department of Pathology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 411A, Oklahoma City, OK 73104, USA
| | - Roman F Wolf
- Department of Pathology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 411A, Oklahoma City, OK 73104, USA
| | - Jundong Zhou
- Department of Radio-Oncology, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, P. R. China
| | - Jori E Avery
- Department of Pathology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 411A, Oklahoma City, OK 73104, USA
| | - Jinchang Wu
- Department of Radio-Oncology, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, P. R. China
| | - Stuart E Lind
- Departments of Pathology and Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Wei-Qun Ding
- Department of Pathology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 411A, Oklahoma City, OK 73104, USA.
| |
Collapse
|
79
|
Kim SY, Lee MJ, Na YR, Kim SY, Yang EG. Visualization of Hypoxia-Inducible Factor 1α-p300 Interactions in Live Cells by Fluorescence Resonance Energy Transfer. J Cell Biochem 2013; 115:271-80. [DOI: 10.1002/jcb.24659] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Accepted: 08/14/2013] [Indexed: 11/09/2022]
Affiliation(s)
- So Yeon Kim
- Center for Theragnosis, Biomedical Research Institute; Korea Institute of Science and Technology; Seoul 136-791 Republic of Korea
| | - Myong Jin Lee
- Center for Theragnosis, Biomedical Research Institute; Korea Institute of Science and Technology; Seoul 136-791 Republic of Korea
| | - Yu-Ran Na
- Center for Theragnosis, Biomedical Research Institute; Korea Institute of Science and Technology; Seoul 136-791 Republic of Korea
| | - Sang Yoon Kim
- Center for Theragnosis, Biomedical Research Institute; Korea Institute of Science and Technology; Seoul 136-791 Republic of Korea
| | - Eun Gyeong Yang
- Center for Theragnosis, Biomedical Research Institute; Korea Institute of Science and Technology; Seoul 136-791 Republic of Korea
| |
Collapse
|
80
|
Garufi A, Trisciuoglio D, Porru M, Leonetti C, Stoppacciaro A, D’Orazi V, Avantaggiati ML, Crispini A, Pucci D, D’Orazi G. A fluorescent curcumin-based Zn(II)-complex reactivates mutant (R175H and R273H) p53 in cancer cells. J Exp Clin Cancer Res 2013; 32:72. [PMID: 24220325 PMCID: PMC3851540 DOI: 10.1186/1756-9966-32-72] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 10/04/2013] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Mutations of the p53 oncosuppressor gene are amongst the most frequent aberration seen in human cancer. Some mutant (mt) p53 proteins are prone to loss of Zn(II) ion that is bound to the wild-type (wt) core, promoting protein aggregation and therefore unfolding. Misfolded p53 protein conformation impairs wtp53-DNA binding and transactivation activities, favouring tumor growth and resistance to antitumor therapies. Screening studies, devoted to identify small molecules that reactivate mtp53, represent therefore an attractive anti-cancer therapeutic strategy. Here we tested a novel fluorescent curcumin-based Zn(II)-complex (Zn-curc) to evaluate its effect on mtp53 reactivation in cancer cells. METHODS P53 protein conformation was examined after Zn-curc treatment by immunoprecipitation and immunofluorescence assays, using conformation-specific antibodies. The mtp53 reactivation was evaluated by chromatin-immunoprecipitation (ChIP) and semi-quantitative RT-PCR analyses of wild-type p53 target genes. The intratumoral Zn-curc localization was evaluated by immunofluorescence analysis of glioblastoma tissues of an ortothopic mice model. RESULTS The Zn-curc complex induced conformational change in p53-R175H and -R273H mutant proteins, two of the most common p53 mutations. Zn-curc treatment restored wtp53-DNA binding and transactivation functions and induced apoptotic cell death. In vivo studies showed that the Zn-curc complex reached glioblastoma tissues of an ortothopic mice model, highlighting its ability to crossed the blood-tumor barrier. CONCLUSIONS Our results demonstrate that Zn-curc complex may reactivate specific mtp53 proteins and that may cross the blood-tumor barrier, becoming a promising compound for the development of drugs to halt tumor growth.
Collapse
Affiliation(s)
- Alessia Garufi
- Department of Experimental Oncology, Regina Elena National Cancer Institute, 00159 Rome, Italy
| | - Daniela Trisciuoglio
- Department of Experimental Oncology, Regina Elena National Cancer Institute, 00159 Rome, Italy
| | - Manuela Porru
- Department of Experimental Oncology, Regina Elena National Cancer Institute, 00159 Rome, Italy
| | - Carlo Leonetti
- Department of Experimental Oncology, Regina Elena National Cancer Institute, 00159 Rome, Italy
| | | | - Valerio D’Orazi
- Department of Surgical Sciences, Sapienza University, 00100 Rome, Italy
| | - Maria Laura Avantaggiati
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Alessandra Crispini
- Centro di Eccellenza CEMIF.CAL-LASCAMM, CR-INSTM, Department of Chimica e Tecnologie Chimiche, University of Calabria, 87100 Cosenza, Italy
| | - Daniela Pucci
- Centro di Eccellenza CEMIF.CAL-LASCAMM, CR-INSTM, Department of Chimica e Tecnologie Chimiche, University of Calabria, 87100 Cosenza, Italy
| | - Gabriella D’Orazi
- Department of Experimental Oncology, Regina Elena National Cancer Institute, 00159 Rome, Italy
- Department of Medical, Oral and Biotechnological Sciences, University “G. d’Annunzio”, 66013 Chieti, Italy
| |
Collapse
|
81
|
Xue J, Wang S, Wu J, Hannafon BN, Ding WQ. Zinc at sub-cytotoxic concentrations induces heme oxygenase-1 expression in human cancer cells. Cell Physiol Biochem 2013; 32:100-10. [PMID: 23868099 DOI: 10.1159/000350128] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/10/2013] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND/AIMS This study investigated the effects of zinc on heme oxygenase-1 (HO-1) expression in human cancer cells. METHODS/RESULTS Zinc at sub-cytotoxic concentrations (50-100 μM) induces HO-1 expression in the MDA-MB-231 (human breast cancer) and A2780 (human ovarian cancer) cell lines in a concentration- and time-dependent manner. The induction of HO-1 by zinc was detected after 4-6 hours of treatment, reached maximal level at 8 hours, and declined thereafter. Using a human HO-1 gene promoter reporter construct, we identified two antioxidant response elements (AREs) that mediated the zinc-induced increase in HO-1 gene transcription, indicating that the nuclear factor (erythroid-derived 2)-like 2 (Nrf2) signaling pathway is involved in this event. This assumption was supported by the observations that knockdown of Nrf2 expression compromised the zinc-induced increase in HO-1 gene transcription, and that zinc increased Nrf2 protein expression and the Nrf2 binding to the AREs. Additionally, we found that the zinc-induced HO-1 gene transcription can be enhanced by clioquinol, a zinc ionophore, and reversed by pretreatment with TPEN, a known zinc chelator, indicating that an increase in intracellular zinc levels is responsible for this induction. CONCLUSION These findings demonstrate that zinc at sub-cytotoxic concentrations induces HO-1 expression in human cancer cells. The biological significance of this induction merits further investigation.
Collapse
Affiliation(s)
- Jing Xue
- School of Radiation Medicine and Protection, Soochow University, Suzhou, P. R. China
| | | | | | | | | |
Collapse
|
82
|
Glucose restriction induces cell death in parental but not in homeodomain-interacting protein kinase 2-depleted RKO colon cancer cells: molecular mechanisms and implications for tumor therapy. Cell Death Dis 2013; 4:e639. [PMID: 23703384 PMCID: PMC3674370 DOI: 10.1038/cddis.2013.163] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Tumor cell tolerance to nutrient deprivation can be an important factor for tumor progression, and may depend on deregulation of both oncogenes and oncosuppressor proteins. Homeodomain-interacting protein kinase 2 (HIPK2) is an oncosuppressor that, following its activation by several cellular stress, induces cancer cell death via p53-dependent or -independent pathways. Here, we used genetically matched human RKO colon cancer cells harboring wt-HIPK2 (HIPK2+/+) or stable HIPK2 siRNA interference (siHIPK2) to investigate in vitro whether HIPK2 influenced cell death in glucose restriction. We found that glucose starvation induced cell death, mainly due to c-Jun NH2-terminal kinase activation, in HIPK2+/+cells compared with siHIPK2 cells that did not die. 1H-nuclear magnetic resonance quantitative metabolic analyses showed a marked glycolytic activation in siHIPK2 cells. However, treatment with glycolysis inhibitor 2-deoxy-𝒟-glucose induced cell death only in HIPK2+/+ cells but not in siHIPK2 cells. Similarly, siGlut-1 interference did not re-establish siHIPK2 cell death under glucose restriction, whereas marked cell death was reached only after zinc supplementation, a condition known to reactivate misfolded p53 and inhibit the pseudohypoxic phenotype in this setting. Further siHIPK2 cell death was reached with zinc in combination with autophagy inhibitor. We propose that the metabolic changes acquired by cells after HIPK2 silencing may contribute to induce resistance to cell death in glucose restriction condition, and therefore be directly relevant for tumor progression. Moreover, elimination of such a tolerance might serve as a new strategy for cancer therapy.
Collapse
|
83
|
Pan R, Chen C, Liu WL, Liu KJ. Zinc promotes the death of hypoxic astrocytes by upregulating hypoxia-induced hypoxia-inducible factor-1alpha expression via poly(ADP-ribose) polymerase-1. CNS Neurosci Ther 2013; 19:511-20. [PMID: 23582235 DOI: 10.1111/cns.12098] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Revised: 02/18/2013] [Accepted: 02/25/2013] [Indexed: 01/01/2023] Open
Abstract
AIM Pathological release of excess zinc ions has been implicated in ischemic brain cell death. However, the underlying mechanisms remain to be elucidated. In stroke, ischemia-induced zinc release and hypoxia-inducible factor-1 (HIF-1) accumulation concurrently occur in the ischemic tissue. The present study tests the hypothesis that the presence of high intracellular zinc concentration is a major cause of modifications to PARP-1 and HIF-1α during hypoxia, which significantly contributes to cell death during ischemia. METHODS Primary cortical astrocytes and C8-D1A cells were exposed to different concentrations of zinc chloride. Cell death rate and protein expression of HIF-1 and Poly(ADP-ribose) polymerase (PARP)-1 were examined after 3-h hypoxic treatment. RESULTS Although 3-h hypoxia or 100 μM of zinc alone did not induce noticeable cytotoxicity, their combination led to a dramatic increase in astrocytic cell death in a zinc-concentration-dependent manner. Exposure of astrocytes to hypoxia for 3 h remarkably increased the levels of intracellular zinc and HIF-1α protein, which was further augmented by added exogenous zinc. Notably, HIF-1α knockdown blocked zinc-induced astrocyte death. Moreover, knockdown of PARP-1, another important protein in the response of hypoxia, attenuated the overexpression of HIF-1α and reduced the cell death rate. CONCLUSIONS Our studies show that zinc promotes hypoxic cell death through overexpression of the hypoxia response factor HIF-1α via the cell fate determine factor PARP-1 modification, which provides a novel mechanism for zinc-mediated ischemic brain injury.
Collapse
Affiliation(s)
- Rong Pan
- Department of Pharmaceutical Sciences, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | | | | | | |
Collapse
|
84
|
Kolenko V, Teper E, Kutikov A, Uzzo R. Zinc and zinc transporters in prostate carcinogenesis. Nat Rev Urol 2013; 10:219-26. [PMID: 23478540 DOI: 10.1038/nrurol.2013.43] [Citation(s) in RCA: 136] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The healthy human prostate accumulates the highest level of zinc of any soft tissue in the body. This unique property is retained in BPH, but is lost in prostatic malignancy, which implicates changes in zinc and its transporters in carcinogenesis. Indeed, zinc concentrations diminish early in the course of prostate carcinogenesis, preceding histopathological changes, and continue to decline during progression toward castration-resistant disease. Numerous studies suggest that increased zinc intake might protect against progression of prostatic malignancy. In spite of increased dietary intake, zinc accumulation might be limited by the diminished expression of zinc uptake transporters, resulting in decreased intratumoural zinc levels. This finding can explain the conflicting results of various epidemiological studies evaluating the role of zinc supplementation on primary and secondary prostate cancer prevention. Overall, more research into the mechanisms of zinc homeostasis are needed to fully understand its impact on prostate carcinogenesis. Only then can the potential of zinc and zinc transport proteins be harnessed in the diagnosis and treatment of men with prostate cancer.
Collapse
Affiliation(s)
- Vladimir Kolenko
- Fox Chase Cancer Center, Department of Surgical Oncology, Philadelphia, PA 19111-2497, USA.
| | | | | | | |
Collapse
|
85
|
Synthetic miRNA-mowers targeting miR-183-96-182 cluster or miR-210 inhibit growth and migration and induce apoptosis in bladder cancer cells. PLoS One 2012; 7:e52280. [PMID: 23284967 PMCID: PMC3524115 DOI: 10.1371/journal.pone.0052280] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Accepted: 11/12/2012] [Indexed: 02/07/2023] Open
Abstract
Background MicroRNAs (miRNAs) function as endogenous regulators of biological behaviors of human cancers. Several natural non-coding RNAs are reported to inhibit miRNAs by base-pairing interactions. These phenomena raise questions about the ability of artificial device to regulate miRNAs. The purpose of this study is to create synthetic devices that target a single miRNA or a miRNA cluster and to ascertain their therapeutic effects on the phenotypes of bladder cancer cells. Methodology/Principal Findings Tandem bulged miRNA binding sites were inserted into the 3′ untranslated region (UTR) of the SV-40 promoter-driven Renilla luciferase gene to construct two “miRNA-mowers” for suppression of miR-183-96-182 cluster or miR-210. A third device with tandem repeat sequences not complementary to any known miRNA was generated as an untargeted-control. In functional analyses, bladder cancer T24 and UM-UC-3 cells were transfected with each of the three devices, followed by assays for detection of their impacts. Luciferase assays indicated that the activities of the luciferase reporters in the miRNA-mowers were decreased to 30–50% of the untargeted-control. Using Real-Time qPCR, the expression levels of the target miRNAs were shown to be reduced 2-3-fold by the corresponding miRNA-mower. Cell growth, apoptosis, and migration were tested by MTT assay, flow cytometry assay, and in vitro scratch assay, respectively. Cell growth inhibition, increased apoptosis, and decreased motility were observed in miRNA-mowers-transfected bladder cancer cells. Conclusions/Significance Not only a single target miRNA but also the whole members of a target miRNA cluster can be blocked using this modular design strategy. Anti-cancer effects are induced by the synthetic miRNA-mowers in the bladder cancer cell lines. miR-183/96/182 cluster and miR-210 are shown to play oncogenic roles in bladder cancer. A potentially useful synthetic biology platform for miRNA loss-of-function study and cancer treatment has been established in this work.
Collapse
|
86
|
Deniro M, Al-Mohanna FA. Zinc transporter 8 (ZnT8) expression is reduced by ischemic insults: a potential therapeutic target to prevent ischemic retinopathy. PLoS One 2012; 7:e50360. [PMID: 23209723 PMCID: PMC3507680 DOI: 10.1371/journal.pone.0050360] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Accepted: 10/24/2012] [Indexed: 12/22/2022] Open
Abstract
The zinc (Zn++) transporter ZnT8 plays a crucial role in zinc homeostasis. It’s been reported that an acute decrease in ZnT8 levels impairs β cell function and Zn++ homeostasis, which contribute to the pathophysiology of diabetes mellitus (DM). Although ZnT8 expression has been detected in the retinal pigment epithelium (RPE), its expression profile in the retina has yet to be determined. Furthermore, the link between diabetes and ischemic retinopathy is well documented; nevertheless, the molecular mechanism(s) of such link has yet to be defined. Our aims were to; investigate the expression profile of ZnT8 in the retina; address the influence of ischemia on such expression; and evaluate the influence of YC-1; (3-(50-hydroxymethyl-20-furyl)-1-benzyl indazole), a hypoxia inducible factor-1 (HIF-1) inhibitor, on the status of ZnT8 expression. We used real-time RT-PCR, immunohistochemistry, and Western blot in the mouse model of oxygen-induced retinopathy (OIR) and Müller cells to evaluate the effects of ischemia/hypoxia and YC-1 on ZnT8 expression. Our data indicate that ZnT8 was strongly expressed in the outer nuclear layer (ONL), outer plexiform layer (OPL), ganglion cell layer (GCL), and nerve fiber layer (NFL), whereas the photoreceptor layer (PRL), inner nuclear layer (INL) and inner plexiform layer (IPL) showed moderate ZnT8 immunoreactivity. Furthermore, we demonstrate that retinal ischemic insult induces a significant downregulation of ZnT8 at the message and protein levels, YC-1 rescues the injured retina by restoring the ZnT8 to its basal homeostatic levels in the neovascular retinas. Our data indicate that ischemic retinopathy maybe mediated by aberrant Zn++ homeostasis caused by ZnT8 downregulation, whereas YC-1 plays a neuroprotective role against ischemic insult. Therefore, targeting ZnT8 provides a therapeutic strategy to combat neovascular eye diseases.
Collapse
Affiliation(s)
- Michael Deniro
- Research Department, King Khaled Eye Specialist Hospital (Affiliate of the Wilmer Eye Institute of the Johns Hopkins Medicine), Riyadh, Saudi Arabia.
| | | |
Collapse
|
87
|
Garufi A, Pistritto G, Ceci C, Di Renzo L, Santarelli R, Faggioni A, Cirone M, D’Orazi G. Targeting COX-2/PGE(2) pathway in HIPK2 knockdown cancer cells: impact on dendritic cell maturation. PLoS One 2012; 7:e48342. [PMID: 23144866 PMCID: PMC3492329 DOI: 10.1371/journal.pone.0048342] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Accepted: 09/24/2012] [Indexed: 02/04/2023] Open
Abstract
Background Homeodomain-interacting protein kinase 2 (HIPK2) is a multifunctional protein that exploits its kinase activity to modulate key molecular pathways in cancer to restrain tumor growth and induce response to therapies. For instance, HIPK2 knockdown induces upregulation of oncogenic hypoxia-inducible factor-1 (HIF-1) activity leading to a constitutive hypoxic and angiogenic phenotype with increased tumor growth in vivo. HIPK2 inhibition, therefore, releases pathways leading to production of pro-inflammatory molecules such as vascular endothelial growth factor (VEGF) or prostaglandin E2 (PGE2). Tumor-produced inflammatory mediators other than promote tumour growth and vascular development may permit evasion of anti-tumour immune responses. Thus, dendritic cells (DCs) dysfunction induced by tumor-produced molecules, may allow tumor cells to escape immunosurveillance. Here we evaluated the molecular mechanism of PGE2 production after HIPK2 depletion and how to modulate it. Methodology/Principal findings We show that HIPK2 knockdown in colon cancer cells resulted in cyclooxygenase-2 (COX-2) upregulation and COX-2-derived PGE2 generation. At molecular level, COX-2 upregulation depended on HIF-1 activity. We previously reported that zinc treatment inhibits HIF-1 activity. Here, zinc supplementation to HIPK2 depleted cells inhibited HIF-1-induced COX-2 expression and PGE2/VEGF production. At translational level, while conditioned media of both siRNA control and HIPK2 depleted cells inhibited DCs maturation, conditioned media of only zinc-treated HIPK2 depleted cells efficiently restored DCs maturation, seen as the expression of co-stimulatory molecules CD80 and CD86, cytokine IL-10 release, and STAT3 phosphorylation. Conclusion/Significance These findings show that: 1) HIPK2 knockdown induced COX-2 upregulation, mostly depending on HIF-1 activity; 2) zinc treatment downregulated HIF-1-induced COX-2 and inhibited PGE2/VEGF production; and 3) zinc treatment of HIPK2 depleted cells restored DCs maturation.
Collapse
Affiliation(s)
- Alessia Garufi
- Department of Experimental Oncology, Molecular Oncogenesis Laboratory, Regina Elena National Cancer Institute, Rome, Italy
| | - Giuseppa Pistritto
- Department of Neuroscience, Section of Pharmacology, University “Tor Vergata”, Rome, Italy
| | - Claudia Ceci
- Department of Neuroscience, Section of Pharmacology, University “Tor Vergata”, Rome, Italy
| | - Livia Di Renzo
- Department of Experimental Medicine, Institute Pasteur-Foundation Cenci Bolognetti, “Sapienza” University, Rome, Italy
| | - Roberta Santarelli
- Department of Experimental Medicine, Institute Pasteur-Foundation Cenci Bolognetti, “Sapienza” University, Rome, Italy
| | - Alberto Faggioni
- Department of Experimental Medicine, Institute Pasteur-Foundation Cenci Bolognetti, “Sapienza” University, Rome, Italy
| | - Mara Cirone
- Department of Experimental Medicine, Institute Pasteur-Foundation Cenci Bolognetti, “Sapienza” University, Rome, Italy
- * E-mail: (GD); (MC)
| | - Gabriella D’Orazi
- Department of Experimental Oncology, Molecular Oncogenesis Laboratory, Regina Elena National Cancer Institute, Rome, Italy
- Department of Medical, Oral and Biotechnological Sciences, University “G. d’Annunzio”, Chieti, Italy
- * E-mail: (GD); (MC)
| |
Collapse
|
88
|
Zhou J, Zhang S, Xue J, Avery J, Wu J, Lind SE, Ding WQ. Activation of peroxisome proliferator-activated receptor α (PPARα) suppresses hypoxia-inducible factor-1α (HIF-1α) signaling in cancer cells. J Biol Chem 2012; 287:35161-35169. [PMID: 22932900 DOI: 10.1074/jbc.m112.367367] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Activation of peroxisome proliferator-activated receptor α (PPARα) has been demonstrated to inhibit tumor growth and angiogenesis, yet the mechanisms behind these actions remain to be characterized. In this study, we examined the effects of PPARα activation on the hypoxia-inducible factor-1α (HIF-1α) signaling pathway in human breast (MCF-7) and ovarian (A2780) cancer cells under hypoxia. Incubation of cancer cells under 1% oxygen for 16 h significantly induced HIF-1α expression and activity as assayed by Western blotting and reporter gene analysis. Treatment of the cells with PPARα agonists, but not a PPARγ agonist, prior to hypoxia diminished hypoxia-induced HIF-1α expression and activity, and addition of a PPARα antagonist attenuated the suppression of HIF-1α signaling. Activation of PPARα attenuated hypoxia-induced HA-tagged HIF-1α protein expression without affecting the HA-tagged HIF-1α mutant protein level, indicating that PPARα activation promotes HIF-1α degradation in these cells. This was further confirmed using proteasome inhibitors, which reversed PPARα-mediated suppression of HIF-1α expression under hypoxia. Using the co-immunoprecipitation technique, we found that activation of PPARα enhances the binding of HIF-1α to von Hippel-Lindau tumor suppressor (pVHL), a protein known to mediate HIF-1α degradation through the ubiquitin-proteasome pathway. Following PPARα-mediated suppression of HIF-1α signaling, VEGF secretion from the cancer cells was significantly reduced, and tube formation by endothelial cells was dramatically impaired. Taken together, these findings demonstrate for the first time that activation of PPARα suppresses hypoxia-induced HIF-1α signaling in cancer cells, providing novel insight into the anticancer properties of PPARα agonists.
Collapse
Affiliation(s)
- Jundong Zhou
- Department of Radio-Oncology, Nanjing Medical University Affiliated Suzhou Hospital; Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104
| | - Shuyu Zhang
- School of Radiation Medicine and Protection, Soochow University, Suzhou 205123, China; Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104
| | - Jing Xue
- Department of Radio-Oncology, Nanjing Medical University Affiliated Suzhou Hospital; Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104
| | - Jori Avery
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104
| | - Jinchang Wu
- Department of Radio-Oncology, Nanjing Medical University Affiliated Suzhou Hospital
| | - Stuart E Lind
- Department of Pathology and Medicine, University of Colorado Denver, Denver, Colorado 80217
| | - Wei-Qun Ding
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104.
| |
Collapse
|
89
|
D'Orazi G, Rinaldo C, Soddu S. Updates on HIPK2: a resourceful oncosuppressor for clearing cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2012; 31:63. [PMID: 22889244 PMCID: PMC3432601 DOI: 10.1186/1756-9966-31-63] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Accepted: 07/27/2012] [Indexed: 02/04/2023]
Abstract
Homeodomain-interacting protein kinase 2 (HIPK2) is a multitalented protein that exploits its kinase activity to modulate key molecular pathways in cancer to restrain tumor growth and induce response to therapies. HIPK2 phosphorylates oncosuppressor p53 for apoptotic activation. In addition, also p53-independent apoptotic pathways are regulated by HIPK2 and can be exploited for anticancer purpose too. Therefore, HIPK2 activity is considered a central switch in targeting tumor cells toward apoptosis upon genotoxic damage and the preservation and/or restoration of HIPK2 function is crucial for an efficient tumor response to therapies. As a proof of principle, HIPK2 knockdown impairs p53 function, induces chemoresistance, angiogenesis, and tumor growth in vivo, on the contrary, HIPK2 overexpression activates apoptotic pathways, counteracts hypoxia, inhibits angiogenesis, and induces chemosensitivity both in p53-dependent and -independent ways. The role of HIPK2 in restraining tumor development was also confirmed by studies with HIPK2 knockout mice. Recent findings demonstrated that HIPK2 inhibitions do exist in tumors and depend by several mechanisms including HIPK2 cytoplasmic localization, protein degradation, and loss of heterozygosity (LOH), recapitulating the biological outcome obtained by RNA interference studies in tumor cells, such as p53 inactivation, resistance to therapies, apoptosis inhibition, and tumor progression. These findings may lead to new diagnostic and therapeutic approaches for treating cancer patients. This review will focus on the last updates about HIPK2 contribution in tumorigenesis and cancer treatment.
Collapse
Affiliation(s)
- Gabriella D'Orazi
- Department of Medical, Oral, and Biotechnological Sciences, University "G, d'Annunzio", Chieti 66013, Italy.
| | | | | |
Collapse
|
90
|
Sheffer M, Simon AJ, Jacob-Hirsch J, Rechavi G, Domany E, Givol D, D'Orazi G. Genome-wide analysis discloses reversal of the hypoxia-induced changes of gene expression in colon cancer cells by zinc supplementation. Oncotarget 2012; 2:1191-202. [PMID: 22202117 PMCID: PMC3282077 DOI: 10.18632/oncotarget.395] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Hypoxia-inducible factor 1 (HIF-1), the major transcription factor specifically activated during hypoxia, regulates genes involved in critical aspects of cancer biology, including angiogenesis, cell proliferation, glycolysis and invasion. The HIF-1a subunit is stabilized by low oxygen, genetic alteration and cobaltous ions, and its over-expression correlates with drug resistance and increased cancer mortality in various cancer types, therefore representing an important anticancer target. Zinc supplementation has been shown to counteract the hypoxic phenotype in cancer cells, in vitro and in vivo, hence, understanding the molecular pathways modulated by zinc under hypoxia may provide the basis for reprogramming signalling pathways for anticancer therapy. Here we performed genome-wide analyses of colon cancer cells treated with combinations of cobalt, zinc and anticancer drug and evaluated the effect of zinc on gene expression patterns. Using Principal Component Analysis we found that zinc markedly reverted the cobalt-induced changes of gene expression, with reactivation of the drug-induced transcription of pro-apoptotic genes. We conclude that the hypoxia pathway is a potential therapeutic target addressed by zinc that also influences tumor cell response to anticancer drug.
Collapse
Affiliation(s)
- Michal Sheffer
- Department of Physics of Complex Systems, Weizmann Institute of Science, Rehovot 76100, Israel
| | | | | | | | | | | | | |
Collapse
|
91
|
Nardinocchi L, Puca R, D'Orazi G. HIF-1α antagonizes p53-mediated apoptosis by triggering HIPK2 degradation. Aging (Albany NY) 2011; 3:33-43. [PMID: 21248371 PMCID: PMC3047137 DOI: 10.18632/aging.100254] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Many human diseases are characterized by the development of tissue hypoxia. Hypoxia-inducible factor (HIF) is a transcription factor that regulates fundamental cellular processes in response to changes in oxygen concentration, such as angiogenesis, survival, and alterations in metabolism. The levels of HIF-1α subunit are increased in most solid tumors not only by low oxygen but also by growth factors and oncogenes and correlate with patient prognosis and treatment failure. The link between HIF-1α and apoptosis, a major determinant of cancer progression and treatment outcome, is poorly understood. Here we show that HIF-1α protects against drug-induced apoptosis by antagonizing the function of the tumor suppressor p53. HIF-1α upregulation induced proteasomal degradation of homeodomain-interacting protein kinase-2 (HIPK2), the p53 apoptotic activator. Inhibition of HIF-1α by siRNA, HIF-1α-dominant negative or by zinc re-established the HIPK2 levels and the p53-mediated chemosensitivity in tumor cells. Our findings identify a novel circuitry between HIF-1α and p53, and provide a paradigm for HIPK2 dictating cell response to antitumor therapies.
Collapse
Affiliation(s)
- Lavinia Nardinocchi
- Department of Experimental Oncology, Molecular Oncogenesis Laboratory, National Cancer Institute "Regina Elena", Rome, Italy
| | | | | |
Collapse
|
92
|
Mimeault M, Batra SK. Frequent gene products and molecular pathways altered in prostate cancer- and metastasis-initiating cells and their progenies and novel promising multitargeted therapies. Mol Med 2011; 17:949-64. [PMID: 21607288 DOI: 10.2119/molmed.2011.00115] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2011] [Accepted: 05/19/2011] [Indexed: 12/14/2022] Open
Abstract
Recent gene expression profiling analyses and gain- and loss-of-function studies performed with distinct prostate cancer (PC) cell models indicated that the alterations in specific gene products and molecular pathways often occur in PC stem/progenitor cells and their progenies during prostate carcinogenesis and metastases at distant sites, including bones. Particularly, the sustained activation of epidermal growth factor receptor (EGFR), hedgehog, Wnt/β-catenin, Notch, hyaluronan (HA)/CD44 and stromal cell-derived factor-1 (SDF-1)/CXC chemokine receptor 4 (CXCR4) during the epithelial-mesenchymal transition (EMT) process may provide critical functions for PC progression to locally invasive, metastatic and androgen-independent disease states and treatment resistance. Moreover, an enhanced glycolytic metabolism in PC stem/progenitor cells and their progenies concomitant with the changes in their local microenvironment, including the induction of tumor hypoxia and release of diverse soluble factors by tumor myofibroblasts, also may promote the tumor growth, angiogenesis and metastases. More particularly, these molecular transforming events may cooperate to upregulate Akt, nuclear factor (NF)-κB, hypoxia-inducible factors (HIFs) and stemness gene products such as Oct3/4, Sox2, Nanog and Bmi-1 in PC cells that contribute to their acquisition of high self-renewal, tumorigenic and invasive capacities and survival advantages during PC progression. Consequently, the molecular targeting of these deregulated gene products in the PC- and metastasis-initiating cells and their progenies represent new promising therapeutic strategies of great clinical interest for eradicating the total PC cell mass and improving current antihormonal treatments and docetaxel-based chemotherapies, thereby preventing disease relapse and the death of PC patients.
Collapse
Affiliation(s)
- Murielle Mimeault
- Department of Biochemistry and Molecular Biology, College of Medicine, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | | |
Collapse
|