51
|
Astaxanthin Modulates Apoptotic Molecules to Induce Death of SKBR3 Breast Cancer Cells. Mar Drugs 2020; 18:md18050266. [PMID: 32438569 PMCID: PMC7281348 DOI: 10.3390/md18050266] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 05/12/2020] [Accepted: 05/14/2020] [Indexed: 12/22/2022] Open
Abstract
Astaxanthin (AST) is related to apoptosis but the details of the mechanism of how AST makes apoptosis is not clear. The present study investigated apoptotic effects of AST to SKBR3, a breast cancer cell line in detail. Cell viability assay showed cellular proliferation and morphological changes of the cells were observed under AST treatment. FACS analysis indicated that AST blocked cell cycle progression at G0/G1, suppressed proliferation dose-dependently, and induced apoptosis of the cells. The apoptosis of the cells by AST was further demonstrated through the decreased expression level of mutp53 and cleaved a PARP-1 fragment, respectively. In addition, AST induced the intrinsic apoptosis of the cells by activation of Bax/Bcl2, cleaved caspase-3, and cleaved caspase-9 as well as the phosphorylation of ERK1/2, JNK, and p38. Furthermore, AST decreased production of intracellular reactive oxygen species as well as modulated expressions of superoxide dismutases and Pontin, an anti-apoptotic factor. Co-immunoprecipitation assay revealed AST reduced interaction between Pontin and mutant p53. Taken together, these studies proved that AST regulates the expression of apoptotic molecules to induce intrinsic apoptosis of the cells, suggesting AST therapy might provide an alternative for improving the efficacies of other anti-cancer therapies for breast cancer.
Collapse
|
52
|
Huehne PS, Bhinija K, Srisomsap C, Chokchaichamnankit D, Weeraphan C, Svasti J, Mongkolsuk S. Detection of superoxide dismutase (Cu-Zn) isoenzymes in leaves and pseudobulbs of Bulbophyllum morphologlorum Kraenzl orchid by comparative proteomic analysis. Biochem Biophys Rep 2020; 22:100762. [PMID: 32395639 PMCID: PMC7210398 DOI: 10.1016/j.bbrep.2020.100762] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 02/26/2020] [Accepted: 02/27/2020] [Indexed: 11/28/2022] Open
Abstract
Typically, biological systems are protected from the toxic effect of free radicals by antioxidant defense. Extracts from orchids have been reported to show high levels of exogenous antioxidant activity including Bulbophyllum orchids but so far, there have been no reports on antioxidant enzymes. Therefore, differences in protein expression from leaves and pseudobulbs of Bulbophyllum morphologlorum Kraenzl and Dendrobium Sonia Earsakul were studied using two-dimensional gel electrophoresis and mass spectrometry (LC/MS/MS). Interestingly, the largest group of these stress response proteins were associated with antioxidant defense and temperature stress, including superoxide dismutase (Cu–Zn) and heat shock protein 70. The high expression of this antioxidant enzyme from Bulbophyllum morphologlorum Kraenzl was confirmed by activity staining on native-PAGE, and the two Cu/Zn-SODs isoenzymes were identified as Cu/Zn-SOD 1 and Cu/Zn-SOD 2 by LC/MS/MS. The results suggested that Bulbophyllum orchid can be a potential plant source for medicines and natural antioxidant supplements.
Collapse
Affiliation(s)
- Pattana S Huehne
- Laboratory of Biotechnology, Chulabhorn Research Institute, Bangkok, 10210, Thailand
| | - Kisana Bhinija
- Laboratory of Biotechnology, Chulabhorn Research Institute, Bangkok, 10210, Thailand
| | - Chantragan Srisomsap
- Laboratory of Biochemistry, Chulabhorn Research Institute, Bangkok, 10210, Thailand
| | | | - Churat Weeraphan
- Laboratory of Biochemistry, Chulabhorn Research Institute, Bangkok, 10210, Thailand
| | - Jisnuson Svasti
- Laboratory of Biochemistry, Chulabhorn Research Institute, Bangkok, 10210, Thailand.,Applied Biological Sciences Program, Chulabhorn Graduate Institute, Chulabhorn Royal Academy, Bangkok, 10210, Thailand
| | - Skorn Mongkolsuk
- Laboratory of Biotechnology, Chulabhorn Research Institute, Bangkok, 10210, Thailand.,Applied Biological Sciences Program, Chulabhorn Graduate Institute, Chulabhorn Royal Academy, Bangkok, 10210, Thailand
| |
Collapse
|
53
|
Hashemi SA, Karami M, Bathaie SZ. Saffron carotenoids change the superoxide dismutase activity in breast cancer: In vitro, in vivo and in silico studies. Int J Biol Macromol 2020; 158:845-853. [PMID: 32360463 DOI: 10.1016/j.ijbiomac.2020.04.063] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 02/27/2020] [Accepted: 04/07/2020] [Indexed: 01/26/2023]
Abstract
Superoxide dismutase (SOD) is an important member of the antioxidant defense system and is proposed as a therapeutic agent against the ROS-mediated diseases, and a therapeutic target for cancer treatment. Saffron carotenoids, crocin (Cro) and crocetin (Crt), are antioxidants with anticancer activity. In the present study, we investigated the effects of Cro/Crt on the SOD activity in both in vivo and in vitro models of breast cancer. Both Cro and Crt showed strong radical scavenging activity and SOD inhibition in the MCF-7 breast cancer cell line. The UVVis, circular dichroism and fluorometry studies proposed the binding of both Cro and Crt with SOD; the ΔG° of binding at 310 °K was -8.6 and -4.4 kcal/mol, respectively. The docking analysis predicted the Cro/Crt binding near the active site channel, but in different sites. According to the obtained data, Cro inhibits SOD activity by scavenging superoxide radical (O2), while Crt inhibits SOD by affecting the copper-binding site. In contrast to the in vitro data, both Cro and Crt effectively increased SOD activity in breast tumors of BALB/c mice, after one month of treatment. The mechanism that is important to compensate for the SOD decreased activity in cancer.
Collapse
Affiliation(s)
- S Ali Hashemi
- Department of Biochemistry, School of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Masoumeh Karami
- Department of Biochemistry, School of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - S Zahra Bathaie
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
54
|
Ge Y, Wang L, Li D, Zhao C, Li J, Liu T. Exploring the Extended Biological Functions of the Human Copper Chaperone of Superoxide Dismutase 1. Protein J 2020; 38:463-471. [PMID: 31140034 DOI: 10.1007/s10930-019-09824-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The human copper chaperone of SOD1 (designated as CCS) was discovered more than two decades ago. It is an important copper binding protein and a homolog of Saccharomyces cerevisiae LYS7. To date, no studies have systematically or specifically elaborated on the functional development of CCS. This review summarizes the essential information about CCS, such as its localization, 3D structure, and copper binding ability. An emphasis is placed on its interacting protein partners and its biological functions in vivo and in vitro. Three-dimensional structural analysis revealed that CCS is composed of three domains. Its primary molecular function is the delivery of copper to SOD1 and activation of SOD1. It has also been reported to bind to XIAP, Mia40, and X11α, and other proteins. Through these protein partners, CCS is implicated in several vital biological processes in vivo, such as copper homeostasis, apoptosis, angiogenesis and oxidative stress. This review is anticipated to assist scientists in systematically understanding the latest research developments of CCS for facilitating the development of new therapeutics targeting CCS in the future.
Collapse
Affiliation(s)
- Yan Ge
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of Antibiotics, Chengdu University, No. 168 Huaguan Road, Chenghua District, Chengdu, 610052, China.,International Phage Drug Research Center, Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu, China
| | - Lu Wang
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of Antibiotics, Chengdu University, No. 168 Huaguan Road, Chenghua District, Chengdu, 610052, China. .,International Phage Drug Research Center, Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu, China.
| | - Duanhua Li
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of Antibiotics, Chengdu University, No. 168 Huaguan Road, Chenghua District, Chengdu, 610052, China.,International Phage Drug Research Center, Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu, China
| | - Chen Zhao
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of Antibiotics, Chengdu University, No. 168 Huaguan Road, Chenghua District, Chengdu, 610052, China.,International Phage Drug Research Center, Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu, China
| | - Jinjun Li
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of Antibiotics, Chengdu University, No. 168 Huaguan Road, Chenghua District, Chengdu, 610052, China.,International Phage Drug Research Center, Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu, China
| | - Tao Liu
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of Antibiotics, Chengdu University, No. 168 Huaguan Road, Chenghua District, Chengdu, 610052, China.,International Phage Drug Research Center, Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu, China
| |
Collapse
|
55
|
Targeting Tumors Using Peptides. Molecules 2020; 25:molecules25040808. [PMID: 32069856 PMCID: PMC7070747 DOI: 10.3390/molecules25040808] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 02/07/2020] [Accepted: 02/10/2020] [Indexed: 12/16/2022] Open
Abstract
To penetrate solid tumors, low molecular weight (Mw < 10 KDa) compounds have an edge over antibodies: their higher penetration because of their small size. Because of the dense stroma and high interstitial fluid pressure of solid tumors, the penetration of higher Mw compounds is unfavored and being small thus becomes an advantage. This review covers a wide range of peptidic ligands—linear, cyclic, macrocyclic and cyclotidic peptides—to target tumors: We describe the main tools to identify peptides experimentally, such as phage display, and the possible chemical modifications to enhance the properties of the identified peptides. We also review in silico identification of peptides and the most salient non-peptidic ligands in clinical stages. We later focus the attention on the current validated ligands available to target different tumor compartments: blood vessels, extracelullar matrix, and tumor associated macrophages. The clinical advances and failures of these ligands and their therapeutic conjugates will be discussed. We aim to present the reader with the state-of-the-art in targeting tumors, by using low Mw molecules, and the tools to identify new ligands.
Collapse
|
56
|
Jiang X, Gray P, Patel M, Zheng J, Yin JJ. Crossover between anti- and pro-oxidant activities of different manganese oxide nanoparticles and their biological implications. J Mater Chem B 2020; 8:1191-1201. [PMID: 31967629 DOI: 10.1039/c9tb02524c] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Manganese oxide nanoparticles (MnOx NPs) have been suggested to possess several enzyme-like activities. However, studies often used either color change or fluorescence to determine the catalytic activity. Despite the simplicity and sensitivity of these probes, these methods may give distracting artifacts or not reflect the catalytic activities in biological systems. To address this issue, herein, we used electron spin resonance (ESR) spectroscopy, a technique proven effective in identifying and quantifying the free radicals produced/scavenged in nanomaterial-catalyzed reactions, to systematically evaluate the catalytic activities of three MnOx NPs (MnO2, Mn2O3, and Mn3O4 NPs) towards biologically relevant antioxidants (ascorbate and glutathione (GSH)) and reactive oxygen species (ROS) (hydrogen peroxide (H2O2), superoxide anion, and hydroxyl radical). We found that all three MnOx NPs possess both pro- and anti-oxidant activities, including oxidase-, catalase-, and superoxide dismutase (SOD)-like activities but without peroxidase-like or hydroxyl radical scavenging activity. In addition, there are differences among these MnOx NPs in their catalytic activities towards different reactions. Mn2O3 shows the strongest ascorbate oxidation activity, followed by MnO2 and Mn3O4, while Mn3O4 shows the strongest oxidation efficiency towards GSH compared to Mn2O3 and MnO2. In the catalyzed decomposition of H2O2, MnO2 NPs show higher efficiency in the generation of molecular oxygen than Mn2O3 or Mn3O4. Cellular studies indicate that all three MnOx NPs induced concentration-dependent decreases in the cell viability, with Mn3O4 > Mn3O2 > MnO2. At lower concentrations (<100 μM), consistent with the enzyme-like activities detected in solution, all three NPs significantly decreased H2O2-induced cytotoxicity in Caco-2 cells. Our study determined the multi-enzymatic activities of MnOx NPs and exhibited differences among MnOx NPs of different valences in their enzyme-like activities and their biological implications; these results provide valuable information for safe and efficient applications of MnOx NPs as ROS-scavenging biomedical nanomaterials.
Collapse
Affiliation(s)
- Xiumei Jiang
- Division of Analytical Chemistry and Division of Bioanalytical Chemistry, Office of Regulatory Science, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, College Park, Maryland 20740, USA.
| | - Patrick Gray
- Division of Analytical Chemistry and Division of Bioanalytical Chemistry, Office of Regulatory Science, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, College Park, Maryland 20740, USA.
| | - Mehulkumar Patel
- Division of Biology, Chemistry and Materials Science, Office of Science and Engineering Laboratories, Center for Devices and Radiological Health, U.S. Food and Drug Administration, Silver Spring, Maryland 20993, USA
| | - Jiwen Zheng
- Division of Biology, Chemistry and Materials Science, Office of Science and Engineering Laboratories, Center for Devices and Radiological Health, U.S. Food and Drug Administration, Silver Spring, Maryland 20993, USA
| | - Jun-Jie Yin
- Division of Analytical Chemistry and Division of Bioanalytical Chemistry, Office of Regulatory Science, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, College Park, Maryland 20740, USA.
| |
Collapse
|
57
|
Jouybari L, Kiani F, Akbari A, Sanagoo A, Sayehmiri F, Aaseth J, Chartrand MS, Sayehmiri K, Chirumbolo S, Bjørklund G. A meta-analysis of zinc levels in breast cancer. J Trace Elem Med Biol 2019; 56:90-99. [PMID: 31442959 DOI: 10.1016/j.jtemb.2019.06.017] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 06/07/2019] [Accepted: 06/19/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Breast cancer is the most commonly occurring neoplasm in females, comprising 16% of all female cancers worldwide. Various studies indicate some discrepancies regarding zinc (Zn) levels in various samples of breast cancer patients. OBJECTIVE The present study evaluated by meta-analysed the published data for Zn levels analyzed in breast tissue, plasma, serum, and hair samples and its relationship with breast cancer. METHODS The present meta-analysis included 36 studies, all of which were published in the years between 1984 to 2017 and selected by searching the databases MEDLINE, EMBASE, Cochrane Library, PubMed, Scopus, and the ISI Web of Knowledge. The articles were analyzed, and I² statistics were used to examine heterogeneity. The objective analysis was performed on data from the 36 studies, with total 1699 study subjects and 2009 controls. RESULTS Significant statistical differences overall were observed, based on a random effects model (SMD (95 % CI), -0.78[-1.40, -0.16], P = 0.014). Data from 19 of these studies indicated significant statistical differences between cancerous patients and controls with regard to serum and plasma Zn concentration (SMD [(95 %CI): -1.61(-2.43, -0.79)]. There was a significant statistical difference between the breast tissue and hair as regards Zn status (SMD (95%CI): 2.32(1.42, 3.21)) and (SMD (95v%CI): -1.80(-3.41, -0.20), respectively. Zn concentration levels typically decreased in blood and hair samples of patients with breast cancer, whereas it was elevated in tumor tissues. CONCLUSIONS There is a significant relationship between lowered serum Zn concentrations and risk of breast cancer onset or recurrences in women, but because of high heterogeneity, we recommend other primary studies.
Collapse
Affiliation(s)
- Leila Jouybari
- Nursing Research Center, Golestan University of Medical Sciences, Gorgan, Iran.
| | - Faezeh Kiani
- Student Research Committee, Ilam University of Medical Sciences, Ilam, Iran
| | - Akbar Akbari
- Department of Immunology, Abadan School of Medical Sciences, Abadan, Iran
| | - Akram Sanagoo
- Nursing Research Center, Golestan University of Medical Sciences, Gorgan, Iran.
| | - Fatemeh Sayehmiri
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Jan Aaseth
- Research Department, Innlandet Hospital Trust, Brumunddal, Norway; Inland Norway University of Applied Sciences, Elverum, Norway
| | | | - Kourosh Sayehmiri
- Prevention Center of Social Mental Injuries, School of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| | - Salvatore Chirumbolo
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy
| | - Geir Bjørklund
- Council for Nutritional and Environmental Medicine, Mo i Rana, Norway.
| |
Collapse
|
58
|
Thapa B, KC R, Bahniuk M, Schmitke J, Hitt M, Lavasanifar A, Kutsch O, Seol DW, Uludag H. Breathing New Life into TRAIL for Breast Cancer Therapy: Co-Delivery of pTRAIL and Complementary siRNAs Using Lipopolymers. Hum Gene Ther 2019; 30:1531-1546. [DOI: 10.1089/hum.2019.096] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- Bindu Thapa
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada
| | - Remant KC
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, Canada
| | - Markian Bahniuk
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, Canada
| | - Janine Schmitke
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, Canada
| | - Mary Hitt
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Afsaneh Lavasanifar
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada
| | - Olaf Kutsch
- Department of Medicine, University of Alabama, Birmingham, Alabama
| | - Dai-Wu Seol
- College of Pharmacy, Chung-Ang University, Seoul, South Korea
| | - Hasan Uludag
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, Canada
- Department of Biomedical Engineering, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| |
Collapse
|
59
|
Lin W, Han W, Wen K, Huang S, Tang Y, Lin Z, Han M. The Alterations of Copper and Zinc Homeostasis in Acute Appendicitis and the Clinical Significance. Biol Trace Elem Res 2019; 192:116-122. [PMID: 30771140 DOI: 10.1007/s12011-019-01661-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 02/04/2019] [Indexed: 02/05/2023]
Abstract
Copper (Cu) and zinc (Zn) are involved in inflammatory process. This study was to investigate the clinical significance of Cu and Zn homeostasis alterations in acute appendicitis (AA). One hundred twenty-two AA patients and 102 healthy controls were enrolled in this study. Of which, 85 patients' appendixes were collected after appendectomy. Another six appendixes from colon cancer patients were collected as tissue controls. The contents of Cu and Zn in serum or appendix were detected, and the Cu to Zn ratio (CZr) was calculated. The concentrations of serum ceruloplasmin (CP), Cu/Zn superoxide dismutase (SOD1), interleukin-6 (IL-6), and interleukin-22 in serum were measured, as well as the activity of CP and SOD1. The serum Zn concentration and SOD1 activity, appendix contents of Cu and Zn significantly decreased in AA patients, compared with those of controls, while serum CZr, concentrations of CP, SOD1, and IL-6, as well as CP activity increased significantly in AA patients. Additionally, serum concentrations of Zn, CP, CZr, or SOD1 activity varied in different pathological types of AA. Indicators such as serum SOD1 activity might serve as predictors for pathological classification before surgery. The serum Zn and CZr may be helpful for diagnosis of pure AA. The Cu and Zn homeostasis was altered in AA patients, which might contribute to inflammatory process of AA.
Collapse
Affiliation(s)
- Wenhao Lin
- Emergency Department of Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Wei Han
- Emergency Department of Shenzhen University General Hospital, Shenzhen, China
| | - Ke Wen
- Department of Microsurgery, Taihe Hospital, Shiyan, China
| | - Sunhua Huang
- Emergency Department of Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Yao Tang
- Emergency Department of Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Zhexuan Lin
- Bio-analytical Laboratory, Shantou University Medical College, Shantou, China.
| | - Ming Han
- Emergency Department of Shenzhen University General Hospital, Shenzhen, China.
| |
Collapse
|
60
|
Ge J, Zhang C, Sun YC, Zhang Q, Lv MW, Guo K, Li JL. Cadmium exposure triggers mitochondrial dysfunction and oxidative stress in chicken (Gallus gallus) kidney via mitochondrial UPR inhibition and Nrf2-mediated antioxidant defense activation. THE SCIENCE OF THE TOTAL ENVIRONMENT 2019; 689:1160-1171. [PMID: 31466156 DOI: 10.1016/j.scitotenv.2019.06.405] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 06/09/2019] [Accepted: 06/24/2019] [Indexed: 06/10/2023]
Abstract
Cadmium (Cd) is a widespread environmental pollutant that accumulates in living systems and represents a significant global health hazard. Cd poses a toxicity threat to both human and animal health, including that of birds. Further knowledge of Cd toxicology pathways will allow for a better understanding of Cd-induced nephrotoxicity. To evaluate Cd-induced nephrotoxicity through potential oxidative damage, male chickens were treated with 0 mg/kg, 35 mg/kg or 70 mg/kg CdCl2 in diet for 90 days. Markedly, histopathology indicated renal tubular epithelial cell swelling, renal function CREA content abnormalities, biochemical and morphologic indices indicative of Cd-induced kidney injury. Cd toxicity induced the up-regulation of Nrf2 and downstream target genes that relieve oxidative stress. Meanwhile, Cd disrupted the homeostasis of trace elements and promoted oxidative damage. Cd interfered with mitochondrial unfolded protein response (UPRmt)-related factors (SIRT1, SIRT3, PGC-1α, TFAM, Nrf1, and HTRA2) and disrupted the homeostasis of mitochondrial dynamics (OPA1, MFN1, MFN2, Fis1 and MFF), thereby exacerbating mitochondrial structural damage and mitochondrial dysfunction. In conclusion, our study demonstrated that the nephrotoxicity of Cd exposure results in oxidative stress and mitochondrial dysfunction by activating the Nrf2 signaling pathway and inhibiting UPRmt in the kidneys.
Collapse
Affiliation(s)
- Jing Ge
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Cong Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Yan-Chun Sun
- Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin 150070, PR China
| | - Qi Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Mei-Wei Lv
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Kai Guo
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Jin-Long Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin 150030, PR China; Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin 150030, PR China.
| |
Collapse
|
61
|
Petruzzelli R, Polishchuk RS. Activity and Trafficking of Copper-Transporting ATPases in Tumor Development and Defense against Platinum-Based Drugs. Cells 2019; 8:E1080. [PMID: 31540259 PMCID: PMC6769697 DOI: 10.3390/cells8091080] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 09/10/2019] [Accepted: 09/12/2019] [Indexed: 02/06/2023] Open
Abstract
Membrane trafficking pathways emanating from the Golgi regulate a wide range of cellular processes. One of these is the maintenance of copper (Cu) homeostasis operated by the Golgi-localized Cu-transporting ATPases ATP7A and ATP7B. At the Golgi, these proteins supply Cu to newly synthesized enzymes which use this metal as a cofactor to catalyze a number of vitally important biochemical reactions. However, in response to elevated Cu, the Golgi exports ATP7A/B to post-Golgi sites where they promote sequestration and efflux of excess Cu to limit its potential toxicity. Growing tumors actively consume Cu and employ ATP7A/B to regulate the availability of this metal for oncogenic enzymes such as LOX and LOX-like proteins, which confer higher invasiveness to malignant cells. Furthermore, ATP7A/B activity and trafficking allow tumor cells to detoxify platinum (Pt)-based drugs (like cisplatin), which are used for the chemotherapy of different solid tumors. Despite these noted activities of ATP7A/B that favor oncogenic processes, the mechanisms that regulate the expression and trafficking of Cu ATPases in malignant cells are far from being completely understood. This review summarizes current data on the role of ATP7A/B in the regulation of Cu and Pt metabolism in malignant cells and outlines questions and challenges that should be addressed to understand how ATP7A and ATP7B trafficking mechanisms might be targeted to counteract tumor development.
Collapse
Affiliation(s)
- Raffaella Petruzzelli
- Telethon Institute of Genetics and Medicine (TIGEM), Via Campi Flegrei 34, 80078 Pozzuoli (NA), Italy.
| | - Roman S Polishchuk
- Telethon Institute of Genetics and Medicine (TIGEM), Via Campi Flegrei 34, 80078 Pozzuoli (NA), Italy.
| |
Collapse
|
62
|
Ramírez-Expósito MJ, Mayas MD, Carrera-González MP, Martínez-Martos JM. Gender Differences in the Antioxidant Response to Oxidative Stress in Experimental Brain Tumors. Curr Cancer Drug Targets 2019; 19:641-654. [DOI: 10.2174/1568009618666181018162549] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Revised: 08/13/2018] [Accepted: 09/15/2018] [Indexed: 01/16/2023]
Abstract
Background:Brain tumorigenesis is related to oxidative stress and a decreased response of antioxidant defense systems. As it is well known that gender differences exist in the incidence and survival rates of brain tumors, it is important to recognize and understand the ways in which their biology can differ.Objective:To analyze gender differences in redox status in animals with chemically-induced brain tumors.Methods:Oxidative stress parameters, non-enzyme and enzyme antioxidant defense systems are assayed in animals with brain tumors induced by transplacental N-ethyl-N-nitrosourea (ENU) administration. Both tissue and plasma were analyzed to know if key changes in redox imbalance involved in brain tumor development were reflected systemically and could be used as biomarkers of the disease.Results:Several oxidative stress parameters were modified in tumor tissue of male and female animals, changes that were not reflected at plasma level. Regarding antioxidant defense system, only glutathione (GSH) levels were decreased in both brain tumor tissue and plasma. Superoxide dismutase (SOD) and catalase (CAT) activities were decreased in brain tumor tissue of male and female animals, but plasma levels were only altered in male animals. However, different protein and mRNA expression patterns were found for both enzymes. On the contrary, glutathione peroxidase (GPx) activity showed increased levels in brain tumor tissue without gender differences, being protein and gene expression also increased in both males and female animals. However, these changes in GPx were not reflected at plasma level.Conclusion:We conclude that brain tumorigenesis was related to oxidative stress and changes in brain enzyme and non-enzyme antioxidant defense systems with gender differences, whereas plasma did not reflect the main redox changes that occur at the brain level.
Collapse
Affiliation(s)
| | - María Dolores Mayas
- Department of Health Sciences, Faculty of Health Sciences, University of Jaen, Jaen, Spain
| | | | | |
Collapse
|
63
|
Kenny TC, Gomez ML, Germain D. Mitohormesis, UPR mt, and the Complexity of Mitochondrial DNA Landscapes in Cancer. Cancer Res 2019; 79:6057-6066. [PMID: 31484668 DOI: 10.1158/0008-5472.can-19-1395] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 07/25/2019] [Accepted: 08/21/2019] [Indexed: 12/20/2022]
Abstract
The discovery of the Warburg effect, the preference of cancer cells to generate ATP via glycolysis rather than oxidative phosphorylation, has fostered the misconception that cancer cells become independent of the electron transport chain (ETC) for survival. This is inconsistent with the need of ETC function for the generation of pyrimidines. Along with this misconception, a large body of literature has reported numerous mutations in mitochondrial DNA (mtDNA), further fueling the notion of nonfunctional ETC in cancer cells. More recent findings, however, suggest that cancers maintain oxidative phosphorylation capacity and that the role of mtDNA mutations in cancer is likely far more nuanced in light of the remarkable complexity of mitochondrial genetics. This review aims at describing the various model systems that were developed to dissect the role of mtDNA in cancer, including cybrids, and more recently mitochondrial-nuclear exchange and conplastic mice. Furthermore, we put forward the notion of mtDNA landscapes, where the surrounding nonsynonymous mutations and variants can enhance or repress the biological effect of specific mtDNA mutations. Notably, we review recent studies describing the ability of some mtDNA landscapes to activate the mitochondrial unfolded protein response (UPRmt) but not others. Furthermore, the role of the UPRmt in maintaining cancer cells in the mitohormetic zone to provide selective adaptation to stress is discussed. Among the genes activated by the UPRmt, we suggest that the dismutases SOD2 and SOD1 may play key roles in the establishment of the mitohormetic zone. Finally, we propose that using a UPRmt nuclear gene expression signature may be a more reliable readout than mtDNA landscapes, given their diversity and complexity.
Collapse
Affiliation(s)
- Timothy C Kenny
- Division of Hematology/Oncology, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Maria L Gomez
- Division of Hematology/Oncology, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Doris Germain
- Division of Hematology/Oncology, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
64
|
Moldogazieva NT, Mokhosoev IM, Mel'nikova TI, Porozov YB, Terentiev AA. Oxidative Stress and Advanced Lipoxidation and Glycation End Products (ALEs and AGEs) in Aging and Age-Related Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:3085756. [PMID: 31485289 PMCID: PMC6710759 DOI: 10.1155/2019/3085756] [Citation(s) in RCA: 291] [Impact Index Per Article: 48.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 06/27/2019] [Indexed: 01/24/2023]
Abstract
Oxidative stress is a consequence of the use of oxygen in aerobic respiration by living organisms and is denoted as a persistent condition of an imbalance between the generation of reactive oxygen species (ROS) and the ability of the endogenous antioxidant system (AOS) to detoxify them. The oxidative stress theory has been confirmed in many animal studies, which demonstrated that the maintenance of cellular homeostasis and biomolecular stability and integrity is crucial for cellular longevity and successful aging. Mitochondrial dysfunction, impaired protein homeostasis (proteostasis) network, alteration in the activities of transcription factors such as Nrf2 and NF-κB, and disturbances in the protein quality control machinery that includes molecular chaperones, ubiquitin-proteasome system (UPS), and autophagy/lysosome pathway have been observed during aging and age-related chronic diseases. The accumulation of ROS under oxidative stress conditions results in the induction of lipid peroxidation and glycoxidation reactions, which leads to the elevated endogenous production of reactive aldehydes and their derivatives such as glyoxal, methylglyoxal (MG), malonic dialdehyde (MDA), and 4-hydroxy-2-nonenal (HNE) giving rise to advanced lipoxidation and glycation end products (ALEs and AGEs, respectively). Both ALEs and AGEs play key roles in cellular response to oxidative stress stimuli through the regulation of a variety of cell signaling pathways. However, elevated ALE and AGE production leads to protein cross-linking and aggregation resulting in an alteration in cell signaling and functioning which causes cell damage and death. This is implicated in aging and various age-related chronic pathologies such as inflammation, neurodegenerative diseases, atherosclerosis, and vascular complications of diabetes mellitus. In the present review, we discuss experimental data evidencing the impairment in cellular functions caused by AGE/ALE accumulation under oxidative stress conditions. We focused on the implications of ALEs/AGEs in aging and age-related diseases to demonstrate that the identification of cellular dysfunctions involved in disease initiation and progression can serve as a basis for the discovery of relevant therapeutic agents.
Collapse
Affiliation(s)
- Nurbubu T. Moldogazieva
- I.M. Sechenov First Moscow State Medical University (Sechenov University), 8 Trubetskaya Street, Moscow, 119991, Russia
| | - Innokenty M. Mokhosoev
- I.M. Sechenov First Moscow State Medical University (Sechenov University), 8 Trubetskaya Street, Moscow, 119991, Russia
- N.I. Pirogov Russian National Research Medical University, 1 Ostrovityanov Street, Moscow, 117997, Russia
| | - Tatiana I. Mel'nikova
- I.M. Sechenov First Moscow State Medical University (Sechenov University), 8 Trubetskaya Street, Moscow, 119991, Russia
| | - Yuri B. Porozov
- I.M. Sechenov First Moscow State Medical University (Sechenov University), 8 Trubetskaya Street, Moscow, 119991, Russia
- Saint Petersburg National Research University of Information Technologies, Mechanics and Optics, 49 Kronverksky Prospect, St. Petersburg, 197101, Russia
| | - Alexander A. Terentiev
- N.I. Pirogov Russian National Research Medical University, 1 Ostrovityanov Street, Moscow, 117997, Russia
| |
Collapse
|
65
|
Banks CJ, Andersen JL. Mechanisms of SOD1 regulation by post-translational modifications. Redox Biol 2019; 26:101270. [PMID: 31344643 PMCID: PMC6658992 DOI: 10.1016/j.redox.2019.101270] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 06/28/2019] [Accepted: 07/03/2019] [Indexed: 12/18/2022] Open
Abstract
SOD1 is commonly known for its ROS scavenging activity, but recent work has uncovered additional roles in modulating metabolism, maintaining redox balance, and regulating transcription. This new paradigm of expanded SOD1 function raises questions regarding the regulation of SOD1 and the cellular partitioning of its biological roles. Despite decades of research on SOD1, much of which focuses on its pathogenic role in amyotrophic lateral sclerosis, relatively little is known about its regulation by post-translational modifications (PTMs). However, over the last decade, advancements in mass spectrometry have led to a boom in PTM discovery across the proteome, which has also revealed new mechanisms of SOD1 regulation by PTMs and an array of SOD1 PTMs with high likelihood of biological function. In this review, we address emerging mechanisms of SOD1 regulation by post-translational modifications, many of which begin to shed light on how the various functions of SOD1 are regulated within the cell.
Collapse
Affiliation(s)
- C J Banks
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, USA
| | - J L Andersen
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, USA.
| |
Collapse
|
66
|
Gomez ML, Shah N, Kenny TC, Jenkins EC, Germain D. SOD1 is essential for oncogene-driven mammary tumor formation but dispensable for normal development and proliferation. Oncogene 2019; 38:5751-5765. [PMID: 31222103 PMCID: PMC6639133 DOI: 10.1038/s41388-019-0839-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 12/19/2018] [Accepted: 03/23/2019] [Indexed: 12/11/2022]
Abstract
We previously reported that the dismutase SOD1 is overexpressed in breast cancer. However, whether SOD1 plays an active role in tumor formation in vivo has never been demonstrated. Further, as luminal cells of normal breast epithelial cells are enriched in SOD1, whether SOD1 is essential for normal mammary gland development has never been determined. We initiated this study to investigate the role of SOD1 in mammary gland tumorigenesis as well as in normal mammary gland development. We crossed the inducible erbB2 (MMTV-iErbB2) and Wnt (MMTV-Wnt) transgenic mice to the SOD1 heterozygote or knockout mice. Our results show that SOD1 is essential for oncogene-driven proliferation, but not normal proliferation of the mammary gland associated with pregnancy or other normal proliferative tissues such as skin and intestines. We show that activation of the oncogene ErbB2 is associated with increased ROS and that high ROS sub-population of ErbB2 cancer cells show elevated SOD1. In the same cells, decrease in SOD1 is associated with an elevation in both apoptosis as well as oncogene-induced senescence. Based on these results, we suggest that SOD1 carries a housekeeping function that maintains ROS levels below a threshold that supports oncogene-dependent proliferation, while allowing escape from oncogene-induced senescence, independently of the oncogene driving tumor formation. These results identify SOD1 as an ideal target for cancer therapy as SOD1 inhibitors hold the potential to prevent the growth of cancers cells of diverse genotypes, activate multiple modes of cell death therefore making acquired resistance more difficult, while sparing normal tissues.
Collapse
Affiliation(s)
- Maria L Gomez
- Department of Medicine, Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY, USA
| | - Nagma Shah
- Department of Medicine, Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY, USA
| | - Timothy C Kenny
- Department of Medicine, Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY, USA
| | - Edmund C Jenkins
- Department of Medicine, Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY, USA
| | - Doris Germain
- Department of Medicine, Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY, USA.
| |
Collapse
|
67
|
Luo P, Tan X, Luo S, Wang Z, Long L, Wang Y, Liao F, Chen L, Zhang C, He J, Huang Y, Liu Z, Gan Y, Chen Z, Wang Y, Liu Y, Wang Y, Shi C. An NIR‐Fluorophore‐Based Inhibitor of SOD1 Induces Apoptosis by Targeting Transcription Cofactor PC4. ADVANCED THERAPEUTICS 2019. [DOI: 10.1002/adtp.201800148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Peng Luo
- Institute of Rocket Force MedicineState Key Laboratory of TraumaBurns and Combined InjuryThird Military Medical University Chongqing 400038 China
| | - Xu Tan
- Institute of Rocket Force MedicineState Key Laboratory of TraumaBurns and Combined InjuryThird Military Medical University Chongqing 400038 China
| | - Shenglin Luo
- Institute of Rocket Force MedicineState Key Laboratory of TraumaBurns and Combined InjuryThird Military Medical University Chongqing 400038 China
| | - Ziwen Wang
- Institute of Rocket Force MedicineState Key Laboratory of TraumaBurns and Combined InjuryThird Military Medical University Chongqing 400038 China
| | - Lei Long
- Institute of Rocket Force MedicineState Key Laboratory of TraumaBurns and Combined InjuryThird Military Medical University Chongqing 400038 China
| | - Yawei Wang
- Institute of Rocket Force MedicineState Key Laboratory of TraumaBurns and Combined InjuryThird Military Medical University Chongqing 400038 China
| | - Fengying Liao
- Institute of Rocket Force MedicineState Key Laboratory of TraumaBurns and Combined InjuryThird Military Medical University Chongqing 400038 China
| | - Long Chen
- Institute of Rocket Force MedicineState Key Laboratory of TraumaBurns and Combined InjuryThird Military Medical University Chongqing 400038 China
| | - Chi Zhang
- Institute of Rocket Force MedicineState Key Laboratory of TraumaBurns and Combined InjuryThird Military Medical University Chongqing 400038 China
| | - Jintao He
- Institute of Rocket Force MedicineState Key Laboratory of TraumaBurns and Combined InjuryThird Military Medical University Chongqing 400038 China
| | - Yinghui Huang
- Institute of Rocket Force MedicineState Key Laboratory of TraumaBurns and Combined InjuryThird Military Medical University Chongqing 400038 China
| | - Zujuan Liu
- Institute of Rocket Force MedicineState Key Laboratory of TraumaBurns and Combined InjuryThird Military Medical University Chongqing 400038 China
| | - Yibo Gan
- Institute of Rocket Force MedicineState Key Laboratory of TraumaBurns and Combined InjuryThird Military Medical University Chongqing 400038 China
| | - Zelin Chen
- Institute of Rocket Force MedicineState Key Laboratory of TraumaBurns and Combined InjuryThird Military Medical University Chongqing 400038 China
| | - Yang Wang
- Institute of Rocket Force MedicineState Key Laboratory of TraumaBurns and Combined InjuryThird Military Medical University Chongqing 400038 China
| | - Yunsheng Liu
- Institute of Rocket Force MedicineState Key Laboratory of TraumaBurns and Combined InjuryThird Military Medical University Chongqing 400038 China
| | - Yu Wang
- Institute of Rocket Force MedicineState Key Laboratory of TraumaBurns and Combined InjuryThird Military Medical University Chongqing 400038 China
| | - Chunmeng Shi
- Institute of Rocket Force MedicineState Key Laboratory of TraumaBurns and Combined InjuryThird Military Medical University Chongqing 400038 China
| |
Collapse
|
68
|
Gomez M, Germain D. Cross talk between SOD1 and the mitochondrial UPR in cancer and neurodegeneration. Mol Cell Neurosci 2019; 98:12-18. [PMID: 31028834 DOI: 10.1016/j.mcn.2019.04.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 04/23/2019] [Indexed: 01/23/2023] Open
Abstract
The mitochondrial unfolded protein response (UPRmt) is rapidly gaining attention. While the CHOP (ATF4/5) axis of the UPRmt was the first to be described, other axes have subsequently been reported. Validation of this complex pathway in C. elegans has been extensively studied. However, validation of the UPRmt in mouse models of disease known to implicate mitochondrial reprogramming or dysfunction, such as cancer and neurodegeneration, respectively, is only beginning to emerge. This review summarizes recent findings and highlights the major role of the superoxide dismutase SOD1 in the communication between the mitochondria and the nucleus in these settings. While SOD1 has mostly been studied in the context of familial amyotrophic lateral sclerosis (fALS), recent studies suggest that SOD1 may be a potentially important mediator of the UPRmt and converge to emphasize an increasingly vital role of SOD1 as a therapeutic target in cancer.
Collapse
Affiliation(s)
- Maria Gomez
- Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, Department of Medicine, Division of Hematology/Oncology, New York, 10029, NY, USA
| | - Doris Germain
- Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, Department of Medicine, Division of Hematology/Oncology, New York, 10029, NY, USA.
| |
Collapse
|
69
|
Gourzones C, Bellanger C, Lamure S, Gadacha OK, De Paco EG, Vincent L, Cartron G, Klein B, Moreaux J. Antioxidant Defenses Confer Resistance to High Dose Melphalan in Multiple Myeloma Cells. Cancers (Basel) 2019; 11:cancers11040439. [PMID: 30925767 PMCID: PMC6521290 DOI: 10.3390/cancers11040439] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 03/15/2019] [Accepted: 03/20/2019] [Indexed: 12/25/2022] Open
Abstract
Background: Multiple myeloma (MM) is the second most common hematological cancer after lymphoma. It is characterized by the accumulation of clonal malignant plasma cells within the bone marrow. The development of drug resistance remains a major problem for effective treatment of MM. Understand the mechanisms underlying drug resistance in MM is a focal point to improve MM treatment. Methods: In the current study, we analyzed further the role of redox imbalance induction in melphalan-induced toxicity both in human myeloma cell lines (HMCLs) and primary myeloma cells from patients. Results: We developed an in-vitro model of short-term resistance to high-dose melphalan and identified that pretreatment with physiological concentration of GSH protects HMCLs from melphalan-induced cell cycle arrest and cytotoxicity. We validated these results using primary MM cells from patients co-cultured with their bone marrow microenvironment. GSH did not affect the ability of melphalan to induce DNA damages in MM cells. Interestingly, melphalan induced reactive oxygen species, a significant decrease in GSH concentration, protein and lipd oxydation together with NRF2 (NF-E2-related factor 2) pathway activation. Conclusions: Our data demonstrate that antioxidant defenses confers resistance to high dose melphalan in MM cells, supporting that redox status in MM cells could be determinant for patients’ response to melphalan.
Collapse
Affiliation(s)
- Claire Gourzones
- IGH, CNRS, University of Montpellier, 34000 Montpellier, France.
| | - Céline Bellanger
- IGH, CNRS, University of Montpellier, 34000 Montpellier, France.
| | - Sylvain Lamure
- Department of Clinical Hematology, CHU Montpellier, 34395 Montpellier, France.
| | | | | | - Laure Vincent
- Department of Clinical Hematology, CHU Montpellier, 34395 Montpellier, France.
| | - Guillaume Cartron
- Department of Clinical Hematology, CHU Montpellier, 34395 Montpellier, France.
- Univ Montpellier, UFR de Médecine, 34000 Montpellier, France.
- Univ Montpellier, UMR CNRS 5235, 34000 Montpellier, France.
| | - Bernard Klein
- IGH, CNRS, University of Montpellier, 34000 Montpellier, France.
- Univ Montpellier, UFR de Médecine, 34000 Montpellier, France.
- Department of Biological Hematology, CHU Montpellier, 34295 Montpellier, France.
| | - Jérôme Moreaux
- IGH, CNRS, University of Montpellier, 34000 Montpellier, France.
- Univ Montpellier, UFR de Médecine, 34000 Montpellier, France.
- Department of Biological Hematology, CHU Montpellier, 34295 Montpellier, France.
| |
Collapse
|
70
|
ATP7A delivers copper to the lysyl oxidase family of enzymes and promotes tumorigenesis and metastasis. Proc Natl Acad Sci U S A 2019; 116:6836-6841. [PMID: 30890638 DOI: 10.1073/pnas.1817473116] [Citation(s) in RCA: 146] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Lysyl oxidase (LOX) and LOX-like (LOXL) proteins are copper-dependent metalloenzymes with well-documented roles in tumor metastasis and fibrotic diseases. The mechanism by which copper is delivered to these enzymes is poorly understood. In this study, we demonstrate that the copper transporter ATP7A is necessary for the activity of LOX and LOXL enzymes. Silencing of ATP7A inhibited LOX activity in the 4T1 mammary carcinoma cell line, resulting in a loss of LOX-dependent mechanisms of metastasis, including the phosphorylation of focal adhesion kinase and myeloid cell recruitment to the lungs, in an orthotopic mouse model of breast cancer. ATP7A silencing was also found to attenuate LOX activity and metastasis of Lewis lung carcinoma cells in mice. Meta-analysis of breast cancer patients found that high ATP7A expression was significantly correlated with reduced survival. Taken together, these results identify ATP7A as a therapeutic target for blocking LOX- and LOXL-dependent malignancies.
Collapse
|
71
|
Li X, Chen Y, Zhao J, Shi J, Wang M, Qiu S, Hu Y, Xu Y, Cui Y, Liu C, Liu C. The Specific Inhibition of SOD1 Selectively Promotes Apoptosis of Cancer Cells via Regulation of the ROS Signaling Network. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:9706792. [PMID: 30911355 PMCID: PMC6398008 DOI: 10.1155/2019/9706792] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 11/21/2018] [Accepted: 12/02/2018] [Indexed: 12/17/2022]
Abstract
Multiple signaling pathways including ERK, PI3K-Akt, and NF-κB, which are essential for onset and development of cancer, can be activated by intracellularly sustained high levels of H2O2 provided by elevated activity and expression of copper/zinc superoxide dismutase (SOD1) that catalyzes the dismutation of O2 •- into H2O2. Here, tests performed by the utilization of our designed specific SOD1 inhibitor LD100 on cancer and normal cells reveal that the signaling pathways and their crosstalk to support cancer cell growth are repressed, but the signaling pathways to promote cancer cell cycle arrest and apoptosis are stimulated by specific SOD1 inhibition-mediated ROS changes. These regulated pathways constitute an ROS signaling network that determines the fate of cancer cells. This ROS signaling network is also regulated in SOD1 knockdown cells. These findings might facilitate disclosure of action mechanisms by copper-chelating anticancer agents and design of SOD1-targeting and ROS signaling pathway-interfering anticancer small molecules.
Collapse
Affiliation(s)
- Xiang Li
- Key Laboratory of Pesticide and Chemical Biology, Ministry of Education, School of Chemistry, Central China Normal University, Wuhan, 430079 Hubei, China
| | - Yuanyuan Chen
- Key Laboratory of Pesticide and Chemical Biology, Ministry of Education, School of Chemistry, Central China Normal University, Wuhan, 430079 Hubei, China
| | - Jidong Zhao
- Key Laboratory of Pesticide and Chemical Biology, Ministry of Education, School of Chemistry, Central China Normal University, Wuhan, 430079 Hubei, China
| | - Jiayuan Shi
- Key Laboratory of Pesticide and Chemical Biology, Ministry of Education, School of Chemistry, Central China Normal University, Wuhan, 430079 Hubei, China
| | - Mingfang Wang
- Key Laboratory of Pesticide and Chemical Biology, Ministry of Education, School of Chemistry, Central China Normal University, Wuhan, 430079 Hubei, China
| | - Shuang Qiu
- Key Laboratory of Pesticide and Chemical Biology, Ministry of Education, School of Chemistry, Central China Normal University, Wuhan, 430079 Hubei, China
| | - Yinghui Hu
- Key Laboratory of Pesticide and Chemical Biology, Ministry of Education, School of Chemistry, Central China Normal University, Wuhan, 430079 Hubei, China
| | - Yulin Xu
- Key Laboratory of Pesticide and Chemical Biology, Ministry of Education, School of Chemistry, Central China Normal University, Wuhan, 430079 Hubei, China
| | - Yanfang Cui
- Key Laboratory of Pesticide and Chemical Biology, Ministry of Education, School of Chemistry, Central China Normal University, Wuhan, 430079 Hubei, China
| | - Chunrong Liu
- Key Laboratory of Pesticide and Chemical Biology, Ministry of Education, School of Chemistry, Central China Normal University, Wuhan, 430079 Hubei, China
| | - Changlin Liu
- Key Laboratory of Pesticide and Chemical Biology, Ministry of Education, School of Chemistry, Central China Normal University, Wuhan, 430079 Hubei, China
| |
Collapse
|
72
|
Parascandolo A, Laukkanen MO. Carcinogenesis and Reactive Oxygen Species Signaling: Interaction of the NADPH Oxidase NOX1-5 and Superoxide Dismutase 1-3 Signal Transduction Pathways. Antioxid Redox Signal 2019; 30:443-486. [PMID: 29478325 PMCID: PMC6393772 DOI: 10.1089/ars.2017.7268] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 02/21/2018] [Accepted: 02/22/2018] [Indexed: 02/06/2023]
Abstract
SIGNIFICANCE Reduction/oxidation (redox) balance could be defined as an even distribution of reduction and oxidation complementary processes and their reaction end products. There is a consensus that aberrant levels of reactive oxygen species (ROS), commonly observed in cancer, stimulate primary cell immortalization and progression of carcinogenesis. However, the mechanism how different ROS regulate redox balance is not completely understood. Recent Advances: In the current review, we have summarized the main signaling cascades inducing NADPH oxidase NOX1-5 and superoxide dismutase (SOD) 1-3 expression and their connection to cell proliferation, immortalization, transformation, and CD34+ cell differentiation in thyroid, colon, lung, breast, and hematological cancers. CRITICAL ISSUES Interestingly, many of the signaling pathways activating redox enzymes or mediating the effect of ROS are common, such as pathways initiated from G protein-coupled receptors and tyrosine kinase receptors involving protein kinase A, phospholipase C, calcium, and small GTPase signaling molecules. FUTURE DIRECTIONS The clarification of interaction of signal transduction pathways could explain how cells regulate redox balance and may even provide means to inhibit the accumulation of harmful levels of ROS in human pathologies.
Collapse
|
73
|
Kepinska M, Kizek R, Milnerowicz H. Metallothionein and Superoxide Dismutase-Antioxidative Protein Status in Fullerene-Doxorubicin Delivery to MCF-7 Human Breast Cancer Cells. Int J Mol Sci 2018; 19:ijms19103253. [PMID: 30347787 PMCID: PMC6214080 DOI: 10.3390/ijms19103253] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 10/16/2018] [Accepted: 10/16/2018] [Indexed: 01/12/2023] Open
Abstract
Doxorubicin (DOX) is one of the most frequently used anticancer drugs in breast cancer treatment. However, clinical applications of DOX are restricted, largely due to the fact that its action disturbs the pro/antioxidant balance in both cancerous and non-cancerous cells. The aim of this study was to investigate the influence of fullerene (C60) in cell treatment by DOX on the proliferation of human breast cancer cells (MCF-7), concentration of metallothionein (MT) and superoxide dismutase (SOD), and SOD activity in these cells. The use of C60 in complexes with DOX causes a change in the level of cell proliferation of about 5% more than when caused by DOX alone (from 60–65% to 70%). The use of C60 as a DOX nanotransporter reduced the MT level increase induced by DOX. C60 alone caused an increase of SOD1 concentration. On the other hand, it led to a decrease of SOD activity. C60 in complex with DOX caused a decrease of the DOX-induced SOD activity level. Exposure of MCF-7 cells to DOX-C60 complexes results in a decrease in viable cells and may become a new therapeutic approach to breast cancer. The effects of C60 in complexes with DOX on MCF-7 cells included a decreased enzymatic (SOD activity) and nonenzymatic (MT) antioxidant status, thus indicating their prooxidant role in MCF-7 cells.
Collapse
Affiliation(s)
- Marta Kepinska
- Department of Biomedical and Environmental Analyses, Faculty of Pharmacy with Division of Laboratory Medicine, Wroclaw Medical University, Borowska 211, 50-556 Wroclaw, Poland.
| | - Rene Kizek
- Department of Biomedical and Environmental Analyses, Faculty of Pharmacy with Division of Laboratory Medicine, Wroclaw Medical University, Borowska 211, 50-556 Wroclaw, Poland.
- Department of Human Pharmacology and Toxicology, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences Brno, Palackeho nam. 1949, 612 42 Brno, Czech Republic.
| | - Halina Milnerowicz
- Department of Biomedical and Environmental Analyses, Faculty of Pharmacy with Division of Laboratory Medicine, Wroclaw Medical University, Borowska 211, 50-556 Wroclaw, Poland.
| |
Collapse
|
74
|
Chang YC, Fong Y, Tsai EM, Chang YG, Chou HL, Wu CY, Teng YN, Liu TC, Yuan SS, Chiu CC. Exogenous C₈-Ceramide Induces Apoptosis by Overproduction of ROS and the Switch of Superoxide Dismutases SOD1 to SOD2 in Human Lung Cancer Cells. Int J Mol Sci 2018; 19:ijms19103010. [PMID: 30279365 PMCID: PMC6213533 DOI: 10.3390/ijms19103010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 09/25/2018] [Accepted: 09/27/2018] [Indexed: 02/07/2023] Open
Abstract
Ceramides, abundant sphingolipids on the cell membrane, can act as signaling molecules to regulate cellular functions including cell viability. Exogenous ceramide has been shown to exert potent anti-proliferative effects against cancer cells, but little is known about how it affects reactive oxygen species (ROS) in lung cancer cells. In this study, we investigated the effect of N-octanoyl-D-erythro-sphingosine (C₈-ceramide) on human non-small-cell lung cancer H1299 cells. Flow cytometry-based assays indicated that C₈-ceramide increased the level of endogenous ROS in H1299 cells. Interestingly, the ratio of superoxide dismutases (SODs) SOD1 and SOD2 seem to be regulated by C₈-ceramide treatment. Furthermore, the accumulation of cell cycle G1 phase and apoptotic populations in C₈-ceramide-treated H1299 cells was observed. The results of the Western blot showed that C₈-ceramide causes a dramatically increased protein level of cyclin D1, a critical regulator of cell cycle G1/S transition. These results suggest that C₈-ceramide acts as a potent chemotherapeutic agent and may increase the endogenous ROS level by regulating the switch of SOD1 and SOD2, causing the anti-proliferation, and consequently triggering the apoptosis of NSCLC H1299 cells. Accordingly, our works may give a promising strategy for lung cancer treatment in the future.
Collapse
Affiliation(s)
- Yuli C Chang
- Department of Laboratory Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Yao Fong
- Chest Surgery, Chi-Mei Medical Center, Yung Kang City, Tainan 901, Taiwan.
| | - Eing-Mei Tsai
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
| | - Ya-Gin Chang
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Han Lin Chou
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Chang-Yi Wu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan;.
| | - Yen-Ni Teng
- Department of Biological Sciences and Technology, National University of Tainan, Tainan 700, Taiwan.
| | - Ta-Chih Liu
- Department of Laboratory Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
| | - Shyng-Shiou Yuan
- Translational Research Center, Cancer Center, Department of Medical Research, Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Chien-Chih Chiu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan;.
- Translational Research Center, Cancer Center, Department of Medical Research, Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Research Center for Environment Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| |
Collapse
|
75
|
Role of Oxidative and Nitro-Oxidative Damage in Silver Nanoparticles Cytotoxic Effect against Human Pancreatic Ductal Adenocarcinoma Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:8251961. [PMID: 30186549 PMCID: PMC6116403 DOI: 10.1155/2018/8251961] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Revised: 06/26/2018] [Accepted: 07/05/2018] [Indexed: 01/04/2023]
Abstract
Pancreatic ductal adenocarcinoma is one of the most aggressive human malignancies, where the 5-year survival rate is less than 4% worldwide. Successful treatment of pancreatic cancer is a challenge for today's oncology. Several studies showed that increased levels of oxidative stress may cause cancer cells damage and death. Therefore, we hypothesized that oxidative as well as nitro-oxidative stress is one of the mechanisms inducing pancreatic cancer programmed cell death. We decided to use silver nanoparticles (AgNPs) (2.6 and 18 nm) as a key factor triggering the reactive oxygen species (ROS) and reactive nitrogen species (RNS) in pancreatic ductal adenocarcinoma cells (PANC-1). Previously, we have found that AgNPs induced PANC-1 cells death. Furthermore, it is known that AgNPs may induce an accumulation of ROS and alteration of antioxidant systems in different type of tumors, and they are indicated as promising agents for cancer therapy. Then, the aim of our study was to evaluate the implication of oxidative and nitro-oxidative stress in this cytotoxic effect of AgNPs against PANC-1 cells. We determined AgNP-induced increase of ROS level in PANC-1 cells and pancreatic noncancer cell (hTERT-HPNE) for comparison purposes. We found that the increase was lower in noncancer cells. Reduction of mitochondrial membrane potential and changes in the cell cycle were also observed. Additionally, we determined the increase in RNS level: nitric oxide (NO) and nitric dioxide (NO2) in PANC-1 cells, together with increase in family of nitric oxide synthases (iNOS, eNOS, and nNOS) at protein and mRNA level. Disturbance of antioxidant enzymes: superoxide dismutase (SOD1, SOD2, and SOD3), glutathione peroxidase (GPX-4) and catalase (CAT) were proved at protein and mRNA level. Moreover, we showed cells ultrastructural changes, characteristic for oxidative damage. Summarizing, oxidative and nitro-oxidative stress and mitochondrial disruption are implicated in AgNPs-mediated death in human pancreatic ductal adenocarcinoma cells.
Collapse
|
76
|
Li S, Fu L, Tian T, Deng L, Li H, Xia W, Gong Q. Disrupting SOD1 activity inhibits cell growth and enhances lipid accumulation in nasopharyngeal carcinoma. Cell Commun Signal 2018; 16:28. [PMID: 29891006 PMCID: PMC5996554 DOI: 10.1186/s12964-018-0240-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 05/28/2018] [Indexed: 12/20/2022] Open
Abstract
Background SOD1 is an abundant enzyme that has been studied as a regulator of the antioxidant defence system, and this enzyme is well known for catalyzing the dismutation of superoxide into hydrogen peroxide. However the SOD1 in the progress of NPC and underlying mechanisms remain unclear. Methods In NPC tissue samples, SOD1 protein levels were measured by Western blot and immunohistochemical (IHC) staining. mRNA levels and SOD1 activity were monitored by qRT-PCR and SOD activity kit, respectively. Kaplan-Meier survival analysis was performed to explore the relationship between SOD1 expression and prognosis of NPC. The biological effects of SOD1 were investigated both in vitro by CCK-8, clonogenicity and apoptosis assays and in vivo by a xenograft mice model. Western blotting, ROS assay and triglyceride assays were applied to investigate the underlying molecular mechanism of pro-survival role of SOD1 in NPC. Results We observed a significant upregulation of SOD1 in NPC tissue and high SOD1 expression is a predictor of poor prognosis and is correlated with poor outcome. We confirmed the pro-survival role of SOD1 both in vitro and in vivo. We demonstrated that these mechanisms of SOD1 partly exist to maintain low levels of the superoxide anion and to avoid the accumulation of lipid droplets via enhanced CPT1A-mediated fatty acid oxidation. Conclusions The results of this study indicate that SOD1 is a potential prognostic biomarker and a promising target for NPC therapy. Electronic supplementary material The online version of this article (10.1186/s12964-018-0240-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shuai Li
- Department of Biochemistry and Molecular Biology, GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 510182, People's Republic of China
| | - Lanyan Fu
- Department of Biochemistry and Molecular Biology, GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 510182, People's Republic of China
| | - Tian Tian
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Liwen Deng
- Department of Biochemistry and Molecular Biology, GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 510182, People's Republic of China
| | - Huangbin Li
- Department of Biochemistry and Molecular Biology, GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 510182, People's Republic of China
| | - Weixiong Xia
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China.
| | - Qing Gong
- Department of Biochemistry and Molecular Biology, GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 510182, People's Republic of China.
| |
Collapse
|
77
|
Pedro NF, Biselli JM, Maniglia JV, Santi-Neto DD, Pavarino ÉC, Goloni-Bertollo EM, Biselli-Chicote PM. Candidate Biomarkers for Oral Squamous Cell Carcinoma: Differential Expression of Oxidative Stress-Related Genes. Asian Pac J Cancer Prev 2018; 19:1343-1349. [PMID: 29802697 PMCID: PMC6031819 DOI: 10.22034/apjcp.2018.19.5.1343] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Background: Alteration in the biotransformation of exogenous compounds can result in production of reactive oxygen species (ROS), which can predispose cells to malignant transformation in the head and neck. This study aimed to evaluate the expression of genes involved in antioxidant metabolism in the oral squamous cell carcinoma (OSCC). Methods: The expression of eighty-four genes was evaluated in OSCC and non-tumor tissues by quantitative real-time polymerase chain reaction using the TaqMan Gene Expression Array. The biological mechanisms related to the differentially expressed genes were investigated using Gene – NCBI, KEGG, UNIPROT and REACTOME databases. Results: Twenty-one genes encoding enzymes involved in antioxidant metabolism were differentially expressed in the OSCC case. Four genes (ATOX1, PRDX4, PRNP, and SOD2) were up-regulated, and seventeen (ALOX12, CAT, CSDE1, DHCR24, DUOX1, DUOX2, EPHX2, GLRX2, GPX3, GSR, GSTZ1, MGST3, PRDX1, OXR1, OXSR1, SOD1, and SOD3) were down-regulated. We identified 14 possible novel biomarkers for OSCC. The differentially expressed genes appeared related to important biological processes involved in carcinogenesis, such as inflammation, angiogenesis, apoptosis, genomic instability, invasion, survival, and cell proliferation. Conclusions: Our study identified novel biomarkers which might warrant further investigation regarding OSCC pathogenesis since the altered expression in the genes can modulate biological processes related to oxidative stress and predispose cells to malignant transformation in the oral cavity.
Collapse
Affiliation(s)
- Nayara Fernandes Pedro
- Genetics and Molecular Biology Research Unit (UPGEM), São José do Rio Preto Medical School (FAMERP), Avenida Brigadeiro Faria Lima, 5416, 15090-000, São Pedro, São José do Rio Preto, São Paulo, Brazil.
| | | | | | | | | | | | | |
Collapse
|
78
|
Hernández-Ramírez LC, Morgan RM, Barry S, D’Acquisto F, Prodromou C, Korbonits M. Multi-chaperone function modulation and association with cytoskeletal proteins are key features of the function of AIP in the pituitary gland. Oncotarget 2018; 9:9177-9198. [PMID: 29507682 PMCID: PMC5823669 DOI: 10.18632/oncotarget.24183] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 01/01/2018] [Indexed: 11/25/2022] Open
Abstract
Despite the well-recognized role of loss-of-function mutations of the aryl hydrocarbon receptor interacting protein gene (AIP) predisposing to pituitary adenomas, the pituitary-specific function of this tumor suppressor remains an enigma. To determine the repertoire of interacting partners for the AIP protein in somatotroph cells, wild-type and variant AIP proteins were used for pull-down/quantitative mass spectrometry experiments against lysates of rat somatotropinoma-derived cells; relevant findings were validated by co-immunoprecipitation and co-localization. Global gene expression was studied in AIP mutation positive and negative pituitary adenomas via RNA microarrays. Direct interaction with AIP was confirmed for three known and six novel partner proteins. Novel interactions with HSPA5 and HSPA9, together with known interactions with HSP90AA1, HSP90AB1 and HSPA8, indicate that the function/stability of multiple chaperone client proteins could be perturbed by a deficient AIP co-chaperone function. Interactions with TUBB, TUBB2A, NME1 and SOD1 were also identified. The AIP variants p.R304* and p.R304Q showed impaired interactions with HSPA8, HSP90AB1, NME1 and SOD1; p.R304* also displayed reduced binding to TUBB and TUBB2A, and AIP-mutated tumors showed reduced TUBB2A expression. Our findings suggest that cytoskeletal organization, cell motility/adhesion, as well as oxidative stress responses, are functions that are likely to be involved in the tumor suppressor activity of AIP.
Collapse
Affiliation(s)
- Laura C. Hernández-Ramírez
- Centre for Endocrinology, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, UK
- Present address: Section on Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD 20892-1862, USA
| | - Rhodri M.L. Morgan
- Genome Damage and Stability Centre, University of Sussex, Brighton, Falmer, BN1 9RQ, UK
- Present address: Protein Crystallography Facility, Centre for Structural Biology, Flowers Building, Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK
| | - Sayka Barry
- Centre for Endocrinology, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Fulvio D’Acquisto
- Centre for Microvascular Research, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, UK
| | | | - Márta Korbonits
- Centre for Endocrinology, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, UK
| |
Collapse
|
79
|
Zhang L, Er JC, Jiang H, Li X, Luo Z, Ramezani T, Feng Y, Tang MK, Chang YT, Vendrell M. A highly selective fluorogenic probe for the detection and in vivo imaging of Cu/Zn superoxide dismutase. Chem Commun (Camb) 2018; 52:9093-6. [PMID: 26940443 PMCID: PMC4943070 DOI: 10.1039/c6cc00095a] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Fine-tuning the BODIPY chemical structure to develop a highly selective fluorophore for Cu/Zn SOD.
Copper/zinc superoxide dismutase (Cu/Zn SOD) is an essential enzyme that protects tissue from oxidative damage. Herein we report the first fluorogenic probe (SODO) for the detection and in vivo imaging of Cu/Zn SOD. SODO represents a unique chemical probe for translational imaging studies of Cu/Zn SOD in inflammatory disorders.
Collapse
Affiliation(s)
- Liyun Zhang
- Institute of Technical Biology and Agriculture Engineering, Key Laboratory of Ion Beam Bioengineering, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, P. R. China.
| | - Jun Cheng Er
- Department of Chemistry, National University of Singapore, 3 Science Drive 2, 117543, Singapore and Graduate School for Integrative Sciences and Engineering, National University of Singapore, Centre for Life Sciences, #05-01, 28 Medical Drive, 117456 Singapore
| | - Hao Jiang
- School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, P. R. China
| | - Xin Li
- Institute of Technical Biology and Agriculture Engineering, Key Laboratory of Ion Beam Bioengineering, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, P. R. China.
| | - Zhaofeng Luo
- School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, P. R. China
| | - Thomas Ramezani
- MRC Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, EH16 4TJ Edinburgh, UK.
| | - Yi Feng
- MRC Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, EH16 4TJ Edinburgh, UK.
| | - Mui Kee Tang
- Department of Chemistry, National University of Singapore, 3 Science Drive 2, 117543, Singapore
| | - Young-Tae Chang
- Department of Chemistry, National University of Singapore, 3 Science Drive 2, 117543, Singapore
| | - Marc Vendrell
- MRC Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, EH16 4TJ Edinburgh, UK.
| |
Collapse
|
80
|
Martino T, Kudrolli TA, Kumar B, Salviano I, Mencalha AL, Coelho MGP, Justo G, Costa PRR, Sabino KCC, Lupold SE. The orally active pterocarpanquinone LQB-118 exhibits cytotoxicity in prostate cancer cell and tumor models through cellular redox stress. Prostate 2018; 78:140-151. [PMID: 29105806 PMCID: PMC5726914 DOI: 10.1002/pros.23455] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 10/24/2017] [Indexed: 12/17/2022]
Abstract
BACKGROUND The targeted induction of reactive oxygen species (ROS) is a developing mechanism for cancer therapy. LQB-118 is a pterocarpanquinone and ROS-inducing agent with proven antineoplastic activity. Here, LQB-118 efficacy and mechanism of activity, were examined in Prostate Cancer (PCa) cell and tumor models. METHODS PC3, LNCaP, and LAPC4 PCa cells were applied. Dicoumarol treatment was used to inhibit quinone reductase activity. N-acetylcysteine (NAC) was applied as a ROS scavenger. ROS production was quantified by H2 DCFDA flow cytometry. LQB-118 treated cells were evaluated for changes in lipid peroxidation, viability, and apoptosis. Treatment-induced gene expression was measured by RT-qPCR and Western Blot. SOD1 knockdown was achieved with siRNA or miRNA mimic transfection. MicroRNA specificity was determined by 3'UTR reporter assay. Oral LQB-118 treatment (10 mg/kg/day) efficacy was determined in athymic male nude mice bearing subcutaneous PC3 xenograft tumors. RESULTS LQB-118 treatment triggered PCa cell death and apoptosis. Therapeutic activity was at least partially dependent upon quinone reduction and ROS generation. LQB-118 treatment caused an increase in cellular ROS and lipid peroxidation. Treated cells exhibited elevated levels of NQO1, Nrf2, and SOD1. The miRNAs miR-206, miR-1, and miR-101 targeted and reduced SOD1 expression. The knockdown of SOD1, by siRNA or miRNA, enhanced LQB-118 cytotoxicity. Orally administered LQB-118 treatment significantly reduced the growth of established PCa xenograft tumors. CONCLUSION LQB-118 is a developing and orally active pterocarpanquinone agent that effectively kills PCa cells through quinone reduction and ROS generation. The inhibition SOD1 expression enhances LQB-118 activity, presumably by impairing the cellular antioxidant response.
Collapse
Affiliation(s)
- T Martino
- LIA-BPPN, Department of Biochemistry, State University of Rio de Janeiro, Rio de Janeiro, Brazil
- The James Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - TA Kudrolli
- The James Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - B Kumar
- The James Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - I Salviano
- Laboratory of Cancer Biology, Department of Biophysics and Biometry, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - AL Mencalha
- Laboratory of Cancer Biology, Department of Biophysics and Biometry, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - MGP Coelho
- LIA-BPPN, Department of Biochemistry, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - G Justo
- LIA-BPPN, Department of Biochemistry, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - PRR Costa
- IPPN, Laboratory of Chemistry Bioorganic, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - KCC Sabino
- LIA-BPPN, Department of Biochemistry, State University of Rio de Janeiro, Rio de Janeiro, Brazil
- Correspondence to: Dr. Katia Sabino, Department of Biochemistry, State University of Rio de Janeiro, Boulevard 28 de Setembro 87 fundos, 4°. Andar, Vila Isabel, Rio de Janeiro, Brazil. ; or Dr. Shawn Lupold, 600 N Wolfe St, Park 209, Baltimore, MD 21287 Phone: 410-502-4822, FAX: 410-502-7711.
| | - SE Lupold
- The James Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Correspondence to: Dr. Katia Sabino, Department of Biochemistry, State University of Rio de Janeiro, Boulevard 28 de Setembro 87 fundos, 4°. Andar, Vila Isabel, Rio de Janeiro, Brazil. ; or Dr. Shawn Lupold, 600 N Wolfe St, Park 209, Baltimore, MD 21287 Phone: 410-502-4822, FAX: 410-502-7711.
| |
Collapse
|
81
|
Younus H. Therapeutic potentials of superoxide dismutase. Int J Health Sci (Qassim) 2018; 12:88-93. [PMID: 29896077 PMCID: PMC5969776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Superoxide dismutases (SODs) constitute a very important antioxidant defense against oxidative stress in the body. The enzyme acts as a good therapeutic agent against reactive oxygen species-mediated diseases. The present review describes the therapeutic effects of SOD in various physiological and pathological conditions such as cancer, inflammatory diseases, cystic fibrosis, ischemia, aging, rheumatoid arthritis, neurodegenerative diseases, and diabetes. However, the enzyme has certain limitations in clinical applications. Therefore, SOD conjugates and mimetics have been developed to increase its therapeutic efficiency. Here, an overview is provided of some in vivo therapeutic effects observed with SOD.
Collapse
Affiliation(s)
- H. Younus
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, Uttar Pradesh, India,Address for correspondence: H. Younus, Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh - 202002, Uttar Pradesh, India. Tel.: +91 571 2720 388. Fax: +91 571 272 1776. E-mail:
| |
Collapse
|
82
|
Privat M, Rudewicz J, Sonnier N, Tamisier C, Ponelle-Chachuat F, Bignon YJ. Antioxydation And Cell Migration Genes Are Identified as Potential Therapeutic Targets in Basal-Like and BRCA1 Mutated Breast Cancer Cell Lines. Int J Med Sci 2018; 15:46-58. [PMID: 29333087 PMCID: PMC5765739 DOI: 10.7150/ijms.20508] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 10/11/2017] [Indexed: 12/26/2022] Open
Abstract
Basal-like breast cancers are among the most aggressive cancers and effective targeted therapies are still missing. In order to identify new therapeutic targets, we performed Methyl-Seq and RNA-Seq of 10 breast cancer cell lines with different phenotypes. We confirmed that breast cancer subtypes cluster the RNA-Seq data but not the Methyl-Seq data. Basal-like tumor hypermethylated phenotype was not confirmed in our study but RNA-Seq analysis allowed to identify 77 genes significantly overexpressed in basal-like breast cancer cell lines. Among them, 48 were overexpressed in triple negative breast cancers of TCGA data. Some molecular functions were overrepresented in this candidate gene list. Genes involved in antioxydation, such as SOD1, MGST3 and PRDX or cadherin-binding genes, such as PFN1, ITGB1 and ANXA1, could thus be considered as basal like breast cancer biomarkers. We then sought if these genes were linked to BRCA1, since this gene is often inactivated in basal-like breast cancers. Nine genes were identified overexpressed in both basal-like breast cancer cells and BRCA1 mutated cells. Amongst them, at least 3 genes code for proteins implicated in epithelial cell migration and epithelial to mesenchymal transition (VIM, ITGB1 and RhoA). Our study provided several potential therapeutic targets for triple negative and BRCA1 mutated breast cancers. It seems that migration and mesenchymal properties acquisition of basal-like breast cancer cells is a key functional pathway in these tumors with a high metastatic potential.
Collapse
Affiliation(s)
- Maud Privat
- Université Clermont Auvergne, Centre Jean Perrin, INSERM, U1240 Imagerie Moléculaire et Stratégies Théranostiques, F-63000 Clermont Ferrand, France
- Département d'Oncogénétique, Centre Jean Perrin, F-63000 Clermont Ferrand, France
| | - Justine Rudewicz
- Département d'Oncogénétique, Centre Jean Perrin, F-63000 Clermont Ferrand, France
| | - Nicolas Sonnier
- Université Clermont Auvergne, Centre Jean Perrin, INSERM, U1240 Imagerie Moléculaire et Stratégies Théranostiques, F-63000 Clermont Ferrand, France
- Département d'Oncogénétique, Centre Jean Perrin, F-63000 Clermont Ferrand, France
- Biological Resources Center BB-0033-00075, Centre Jean Perrin, F-63000 Clermont Ferrand, France
| | - Christelle Tamisier
- Département d'Oncogénétique, Centre Jean Perrin, F-63000 Clermont Ferrand, France
| | - Flora Ponelle-Chachuat
- Université Clermont Auvergne, Centre Jean Perrin, INSERM, U1240 Imagerie Moléculaire et Stratégies Théranostiques, F-63000 Clermont Ferrand, France
- Département d'Oncogénétique, Centre Jean Perrin, F-63000 Clermont Ferrand, France
| | - Yves-Jean Bignon
- Université Clermont Auvergne, Centre Jean Perrin, INSERM, U1240 Imagerie Moléculaire et Stratégies Théranostiques, F-63000 Clermont Ferrand, France
- Département d'Oncogénétique, Centre Jean Perrin, F-63000 Clermont Ferrand, France
- Biological Resources Center BB-0033-00075, Centre Jean Perrin, F-63000 Clermont Ferrand, France
| |
Collapse
|
83
|
Fu Q, Wu Y. RCAN1 in the inverse association between Alzheimer's disease and cancer. Oncotarget 2017; 9:54-66. [PMID: 29416595 PMCID: PMC5787488 DOI: 10.18632/oncotarget.23094] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 11/17/2017] [Indexed: 01/05/2023] Open
Abstract
The inverse association between Alzheimer’s disease (AD) and cancer has been reported in several population-based studies although both of them are age-related disorders. However, molecular mechanisms of the inverse association remain elusive. Increased expression of regulator of calcineurin 1 (RCAN1) promotes the pathogenesis of AD, while it suppresses cancer growth and progression in many types of cancer. Moreover, aberrant RCAN1 expression is detected in both AD and various types of cancer. It suggests that RCAN1 may play a key role in the inverse association between AD and cancer. In this article, we aim to review the role of RCAN1 in the inverse association and discuss underlying mechanisms, providing an insight into developing a novel approach to treat AD and cancer.
Collapse
Affiliation(s)
- Qiang Fu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Yili Wu
- Department of Psychiatry, Jining Medical University, Jining, Shandong, China.,Shandong Key Laboratory of Behavioral Medicine, Jining, Shandong, China.,Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong Province, Jining, Shandong, China
| |
Collapse
|
84
|
Impact of genotypic and phenotypic differences in sarcoma models on the outcome of photochemical internalization (PCI) of bleomycin. Photodiagnosis Photodyn Ther 2017; 20:35-47. [DOI: 10.1016/j.pdpdt.2017.08.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 08/11/2017] [Accepted: 08/15/2017] [Indexed: 02/03/2023]
|
85
|
Griess B, Tom E, Domann F, Teoh-Fitzgerald M. Extracellular superoxide dismutase and its role in cancer. Free Radic Biol Med 2017; 112:464-479. [PMID: 28842347 PMCID: PMC5685559 DOI: 10.1016/j.freeradbiomed.2017.08.013] [Citation(s) in RCA: 121] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 08/16/2017] [Accepted: 08/18/2017] [Indexed: 12/19/2022]
Abstract
Reactive oxygen species (ROS) are increasingly recognized as critical determinants of cellular signaling and a strict balance of ROS levels must be maintained to ensure proper cellular function and survival. Notably, ROS is increased in cancer cells. The superoxide dismutase family plays an essential physiological role in mitigating deleterious effects of ROS. Due to the compartmentalization of ROS signaling, EcSOD, the only superoxide dismutase in the extracellular space, has unique characteristics and functions in cellular signal transduction. In comparison to the other two intracellular SODs, EcSOD is a relatively new comer in terms of its tumor suppressive role in cancer and the mechanisms involved are less well understood. Nevertheless, the degree of differential expression of this extracellular antioxidant in cancer versus normal cells/tissues is more pronounced and prevalent than the other SODs. A significant association of low EcSOD expression with reduced cancer patient survival further suggests that loss of extracellular redox regulation promotes a conducive microenvironment that favors cancer progression. The vast array of mechanisms reported in mediating deregulation of EcSOD expression, function, and cellular distribution also supports that loss of this extracellular antioxidant provides a selective advantage to cancer cells. Moreover, overexpression of EcSOD inhibits tumor growth and metastasis, indicating a role as a tumor suppressor. This review focuses on the current understanding of the mechanisms of deregulation and tumor suppressive function of EcSOD in cancer.
Collapse
Affiliation(s)
- Brandon Griess
- Department of Biochemistry and Molecular Biology, Buffett Cancer Center, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Eric Tom
- Department of Biochemistry and Molecular Biology, Buffett Cancer Center, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Frederick Domann
- Free Radical and Radiation Biology Program, Radiation Oncology, University of Iowa, Iowa, IA 52242, United States
| | - Melissa Teoh-Fitzgerald
- Department of Biochemistry and Molecular Biology, Buffett Cancer Center, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, United States.
| |
Collapse
|
86
|
On the Origin of Superoxide Dismutase: An Evolutionary Perspective of Superoxide-Mediated Redox Signaling. Antioxidants (Basel) 2017; 6:antiox6040082. [PMID: 29084153 PMCID: PMC5745492 DOI: 10.3390/antiox6040082] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 10/23/2017] [Accepted: 10/27/2017] [Indexed: 12/15/2022] Open
Abstract
The field of free radical biology originated with the discovery of superoxide dismutase (SOD) in 1969. Over the last 5 decades, a plethora of research has been performed in species ranging from bacteria to mammals that has elucidated the molecular reaction, subcellular location, and specific isoforms of SOD. However, while humans have only begun to study this class of enzymes over the past 50 years, it has been estimated that these enzymes have existed for billions of years, and may be some of the original enzymes found in primitive life. As life evolved over this expanse of time, these enzymes have taken on new and different functional roles potentially in contrast to how they were originally derived. Herein, examination of the evolutionary history of these enzymes provides both an explanation and further inquiries into the modern-day role of SOD in physiology and disease.
Collapse
|
87
|
Thapa B, Bahadur KC R, Uludağ H. Novel targets for sensitizing breast cancer cells to TRAIL-induced apoptosis with siRNA delivery. Int J Cancer 2017; 142:597-606. [DOI: 10.1002/ijc.31079] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Revised: 09/09/2017] [Accepted: 09/19/2017] [Indexed: 12/18/2022]
Affiliation(s)
- Bindu Thapa
- Faculty of Pharmacy and Pharmaceutical Sciences; University of Alberta; Edmonton AB Canada
| | - Remant Bahadur KC
- Department of Chemical and Material Engineering, Faculty of Engineering; University of Alberta; Edmonton AB Canada
| | - Hasan Uludağ
- Faculty of Pharmacy and Pharmaceutical Sciences; University of Alberta; Edmonton AB Canada
- Department of Chemical and Material Engineering, Faculty of Engineering; University of Alberta; Edmonton AB Canada
- Department of Biomedical Engineering, Faculty of Medicine and Dentistry; University of Alberta; Edmonton AB Canada
| |
Collapse
|
88
|
Acylation of Superoxide Dismutase 1 (SOD1) at K122 Governs SOD1-Mediated Inhibition of Mitochondrial Respiration. Mol Cell Biol 2017; 37:MCB.00354-17. [PMID: 28739857 DOI: 10.1128/mcb.00354-17] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 07/11/2017] [Indexed: 12/24/2022] Open
Abstract
In this study, we employed proteomics to identify mechanisms of posttranslational regulation on cell survival signaling proteins. We focused on Cu-Zn superoxide dismutase (SOD1), which protects cells from oxidative stress. We found that acylation of K122 on SOD1, while not impacting SOD1 catalytic activity, suppressed the ability of SOD1 to inhibit mitochondrial metabolism at respiratory complex I. We found that deacylase depletion increased K122 acylation on SOD1, which blocked the suppression of respiration in a K122-dependent manner. In addition, we found that acyl-mimicking mutations at K122 decreased SOD1 accumulation in mitochondria, initially hinting that SOD1 may inhibit respiration directly within the intermembrane space (IMS). However, surprisingly, we found that forcing the K122 acyl mutants into the mitochondria with an IMS-targeting tag did not recover their ability to suppress respiration. Moreover, we found that suppressing or boosting respiration levels toggled SOD1 in or out of the mitochondria, respectively. These findings place SOD1-mediated inhibition of respiration upstream of its mitochondrial localization. Lastly, deletion-rescue experiments show that a respiration-defective mutant of SOD1 is also impaired in its ability to rescue cells from toxicity caused by SOD1 deletion. Together, these data suggest a previously unknown interplay between SOD1 acylation, metabolic regulation, and SOD1-mediated cell survival.
Collapse
|
89
|
Kenny TC, Germain D. From discovery of the CHOP axis and targeting ClpP to the identification of additional axes of the UPRmt driven by the estrogen receptor and SIRT3. J Bioenerg Biomembr 2017; 49:297-305. [PMID: 28799020 DOI: 10.1007/s10863-017-9722-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 05/11/2017] [Indexed: 12/18/2022]
Abstract
The mitochondrial UPR (UPRmt) is rapidly gaining attention. While most studies on the UPRmt have focused on its role in aging, emerging studies suggest an important role of the UPRmt in cancer. Further, several of the players of the UPRmt in mammalian cells have well reported roles in the maintenance of the organelle. The goal of this review is to emphasize aspects of the UPRmt that have been overlooked in the current literature, describe the role of specific players of the UPRmt in the biology of the mitochondria and highlight the intriguing possibility that targeting the UPRmt in cancer may be already within reach.
Collapse
Affiliation(s)
- Timothy C Kenny
- Department of Medicine, Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY, 10029, USA
| | - Doris Germain
- Department of Medicine, Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY, 10029, USA.
| |
Collapse
|
90
|
Zhang J, Sun QL, Luan ZD, Lian C, Sun L. Comparative transcriptome analysis of Rimicaris sp. reveals novel molecular features associated with survival in deep-sea hydrothermal vent. Sci Rep 2017; 7:2000. [PMID: 28515421 PMCID: PMC5435735 DOI: 10.1038/s41598-017-02073-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 04/05/2017] [Indexed: 11/09/2022] Open
Abstract
Shrimp of the family Alvinocarididae are the predominant megafauna of deep-sea hydrothermal vents. However, genome information on this family is currently unavailable. In the present study, by employing Illumina sequencing, we performed the first de novo transcriptome analysis of the gills of the shrimp Rimicaris sp. from the hydrothermal vent in Desmos, Manus Basin. The analysis was conducted in a comparative manner with the shrimp taken directly from the vent (GR samples) and the shrimp that had been maintained for ten days under normal laboratory condition (mGR samples). Among the 128,938 unigenes identified, a large number of differentially expressed genes (DEGs) between the GR and mGR samples were detected, including 2365 and 1607 genes significantly upregulated and downregulated, respectively, in GR. The DEGs covered diverse functional categories. Most of the DEGs associated with immunity were downregulated in GR, while most of the DEGs associated with sulfur metabolism and detoxification were upregulated in GR. These results provide the first comprehensive transcriptomic resource for hydrothermal vent Rimicaris and revealed varied categories of genes likely involved in deep-sea survival.
Collapse
Affiliation(s)
- Jian Zhang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China.,Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.,Deep Sea Research Center, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China
| | - Qing-Lei Sun
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China.,Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Zhen-Dong Luan
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.,Key Laboratory of Marine Geology and Environment, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China.,Deep Sea Research Center, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China
| | - Chao Lian
- Key Laboratory of Marine Geology and Environment, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China
| | - Li Sun
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China. .,Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.
| |
Collapse
|
91
|
Qin B, Shu Y, Xiao L, Lu T, Lin Y, Yang H, Lu Z. MicroRNA-150 targets ELK1 and modulates the apoptosis induced by ox-LDL in endothelial cells. Mol Cell Biochem 2017; 429:45-58. [PMID: 28110404 DOI: 10.1007/s11010-016-2935-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2016] [Accepted: 12/23/2016] [Indexed: 12/23/2022]
Abstract
Atherosclerosis, a chronic inflammatory disease, is the major cause of life-threatening complications such as myocardial infarction and stroke. Endothelial cells (ECs) apoptosis plays a vital role in the initiation and progression of atherosclerosis. Although a subset of microRNAs (miRNAs) have been identified as critical regulators of atherosclerosis, studies on their participation in endothelial apoptosis in atherosclerosis have been limited. In the current study, we show that miRNA-150 (miR-150) expression was substantially up-regulated during the oxidized low-density lipoprotein (ox-LDL)-induced apoptosis in human umbilical cord vein endothelial cells (HUVECs). Forced expression of miR-150 enhanced apoptosis in ECs, whereas inhibition of miR-150 could partly alleviate apoptotic cell death mediated by ox-LDL. Further analysis identified ELK1 as a direct target of miR-150, and ELK1 knockdown abolished the anti-apoptotic effect of miR-150 inhibitor. These findings reveal a novel role of miR-150 in endothelial apoptosis and indicate a therapeutic potential of miR-150 for endothelial dysfunction and atherosclerosis.
Collapse
Affiliation(s)
- Bing Qin
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, No. 600 Tianhe Road, Guangzhou, 510630, Guangdong, People's Republic of China
| | - Yaqing Shu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, No. 600 Tianhe Road, Guangzhou, 510630, Guangdong, People's Republic of China
| | - Li Xiao
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, No. 600 Tianhe Road, Guangzhou, 510630, Guangdong, People's Republic of China
| | - Tingting Lu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, No. 600 Tianhe Road, Guangzhou, 510630, Guangdong, People's Republic of China
| | - Yinyao Lin
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, No. 600 Tianhe Road, Guangzhou, 510630, Guangdong, People's Republic of China
| | - Huan Yang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Zhengqi Lu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, No. 600 Tianhe Road, Guangzhou, 510630, Guangdong, People's Republic of China.
| |
Collapse
|
92
|
de Carvalho MDC, De Mesquita JF, Eleutherio ECA. In Vivo Characterization of I91T Sod2 Polymorphism of Saccharomyces cerevisiae. J Cell Biochem 2017; 118:1078-1086. [DOI: 10.1002/jcb.25720] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 08/30/2016] [Indexed: 11/09/2022]
Affiliation(s)
| | - Joelma Freire De Mesquita
- Department of Genetics and Molecular Biology; Federal University of the State of Rio de Janeiro (UNIRIO); Rio de Janeiro 22290-240 Brazil
| | | |
Collapse
|
93
|
Wills QF, Mellado-Gomez E, Nolan R, Warner D, Sharma E, Broxholme J, Wright B, Lockstone H, James W, Lynch M, Gonzales M, West J, Leyrat A, Padilla-Parra S, Filippi S, Holmes C, Moore MD, Bowden R. The nature and nurture of cell heterogeneity: accounting for macrophage gene-environment interactions with single-cell RNA-Seq. BMC Genomics 2017; 18:53. [PMID: 28061811 PMCID: PMC5219790 DOI: 10.1186/s12864-016-3445-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Accepted: 12/20/2016] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Single-cell RNA-Seq can be a valuable and unbiased tool to dissect cellular heterogeneity, despite the transcriptome's limitations in describing higher functional phenotypes and protein events. Perhaps the most important shortfall with transcriptomic 'snapshots' of cell populations is that they risk being descriptive, only cataloging heterogeneity at one point in time, and without microenvironmental context. Studying the genetic ('nature') and environmental ('nurture') modifiers of heterogeneity, and how cell population dynamics unfold over time in response to these modifiers is key when studying highly plastic cells such as macrophages. RESULTS We introduce the programmable Polaris™ microfluidic lab-on-chip for single-cell sequencing, which performs live-cell imaging while controlling for the culture microenvironment of each cell. Using gene-edited macrophages we demonstrate how previously unappreciated knockout effects of SAMHD1, such as an altered oxidative stress response, have a large paracrine signaling component. Furthermore, we demonstrate single-cell pathway enrichments for cell cycle arrest and APOBEC3G degradation, both associated with the oxidative stress response and altered proteostasis. Interestingly, SAMHD1 and APOBEC3G are both HIV-1 inhibitors ('restriction factors'), with no known co-regulation. CONCLUSION As single-cell methods continue to mature, so will the ability to move beyond simple 'snapshots' of cell populations towards studying the determinants of population dynamics. By combining single-cell culture, live-cell imaging, and single-cell sequencing, we have demonstrated the ability to study cell phenotypes and microenvironmental influences. It's these microenvironmental components - ignored by standard single-cell workflows - that likely determine how macrophages, for example, react to inflammation and form treatment resistant HIV reservoirs.
Collapse
Affiliation(s)
- Quin F Wills
- Wellcome Trust Centre for Human Genetics (WTCHG), University of Oxford, Oxford, OX3 7BN, UK.
- Weatherall Institute of Molecular Medicine (WIMM), University of Oxford, Oxford, OX3 9DS, UK.
| | - Esther Mellado-Gomez
- Wellcome Trust Centre for Human Genetics (WTCHG), University of Oxford, Oxford, OX3 7BN, UK
| | - Rory Nolan
- Wellcome Trust Centre for Human Genetics (WTCHG), University of Oxford, Oxford, OX3 7BN, UK
- Division of Structural Biology, University of Oxford, Oxford, OX3 7BN, UK
| | - Damien Warner
- Sir William Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, UK
| | - Eshita Sharma
- Wellcome Trust Centre for Human Genetics (WTCHG), University of Oxford, Oxford, OX3 7BN, UK
| | - John Broxholme
- Wellcome Trust Centre for Human Genetics (WTCHG), University of Oxford, Oxford, OX3 7BN, UK
| | - Benjamin Wright
- Wellcome Trust Centre for Human Genetics (WTCHG), University of Oxford, Oxford, OX3 7BN, UK
| | - Helen Lockstone
- Wellcome Trust Centre for Human Genetics (WTCHG), University of Oxford, Oxford, OX3 7BN, UK
| | - William James
- Sir William Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, UK
| | - Mark Lynch
- Fluidigm Corporation, 7000 Shoreline Ct Ste 100, South San Francisco, CA, 94080-7603, USA
| | - Michael Gonzales
- Fluidigm Corporation, 7000 Shoreline Ct Ste 100, South San Francisco, CA, 94080-7603, USA
| | - Jay West
- Fluidigm Corporation, 7000 Shoreline Ct Ste 100, South San Francisco, CA, 94080-7603, USA
| | - Anne Leyrat
- Fluidigm Corporation, 7000 Shoreline Ct Ste 100, South San Francisco, CA, 94080-7603, USA
| | - Sergi Padilla-Parra
- Wellcome Trust Centre for Human Genetics (WTCHG), University of Oxford, Oxford, OX3 7BN, UK
- Division of Structural Biology, University of Oxford, Oxford, OX3 7BN, UK
| | - Sarah Filippi
- Department of Statistics, University of Oxford, Oxford, OX3 3LB, UK
| | - Chris Holmes
- Wellcome Trust Centre for Human Genetics (WTCHG), University of Oxford, Oxford, OX3 7BN, UK
- Department of Statistics, University of Oxford, Oxford, OX3 3LB, UK
| | - Michael D Moore
- Sir William Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, UK.
| | - Rory Bowden
- Wellcome Trust Centre for Human Genetics (WTCHG), University of Oxford, Oxford, OX3 7BN, UK.
| |
Collapse
|
94
|
Guiraut C, Cauchon N, Lepage M, Sébire G. Perinatal Arterial Ischemic Stroke Is Associated to Materno-Fetal Immune Activation and Intracranial Arteritis. Int J Mol Sci 2016; 17:ijms17121980. [PMID: 27898024 PMCID: PMC5187780 DOI: 10.3390/ijms17121980] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 11/14/2016] [Accepted: 11/21/2016] [Indexed: 12/22/2022] Open
Abstract
The medium-size intra-cranial arteries arising from the carotid bifurcation are prone to perinatal arterial ischemic strokes (PAIS). PAIS’ physiopathology needs to be better understood to develop preventive and therapeutic interventions that are currently missing. We hypothesized that materno-fetal inflammation leads to a vasculitis affecting selectively the carotidian tree and promoting a focal thrombosis and subsequent stroke. Dams were injected with saline or lipopolysaccharide (LPS) from Escherichia coli. A prothrombotic stress was applied on LPS-exposed vs. saline (S)-exposed middle cerebral arteries (MCA). Immunolabeling detected the inflammatory markers of interest. In S-exposed newborn pups, a constitutive higher density of macrophages combined to higher expressions of tumor necrosis factor-α (TNF-α), and interleukin 1β (IL-1β) was observed within the wall of intra- vs. extra-cranial cervicocephalic arteries. LPS-induced maternal and placental inflammatory responses mediated by IL-1β, TNF-α and monocyte chemotactic protein 1 (MCP-1) were associated with: (i) increased density of pro-inflammatory macrophages (M1 phenotype); and (ii) pro-inflammatory orientation of the IL-1 system (IL-1β/IL-1 receptor antagonist (IL-1Ra) ratio) within the wall of LPS-, vs. S-exposed, intra-cranial arteries susceptible to PAIS. LPS plus photothrombosis, but not sole photothrombosis, triggered ischemic strokes and subsequent motor impairments. Based on these preclinical results, the combination of pro-thrombotic stress and selective intra-cranial arteritis arising from end gestational maternal immune activation seem to play a role in the pathophysiology of human PAIS.
Collapse
Affiliation(s)
- Clémence Guiraut
- Département de Pédiatrie, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada.
| | - Nicole Cauchon
- Département de Médecine Nucléaire et Radiobiologie, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada.
| | - Martin Lepage
- Département de Médecine Nucléaire et Radiobiologie, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada.
| | - Guillaume Sébire
- Département de Pédiatrie, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada.
- Child Neurology Division, Department of Pediatrics, McGill University, Montréal, QC H4A 3J1, Canada.
| |
Collapse
|
95
|
Antioxidant defense of Nrf2vspro-inflammatory system of NF-κB during the amoebic liver infection in hamster. Parasitology 2016; 144:384-393. [DOI: 10.1017/s0031182016001967] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
SUMMARYEntamoeba histolyticais the causative agent of amoebic liver abscess (ALA), which course with an uncontrolled inflammation and nitro-oxidative stresses, although it is well known that amoeba has an effective defence mechanisms against this toxic environment, the underlying molecular factors responsible for progression of tissue damage remain largely unknown. The purpose of the present study was to determine during the acute stage of ALA in hamsters, the involvement of nuclear factor (erythroid-derived 2)-like 2 (Nrf2) and nuclear factor-kappa B (NF-κB), which are activated in response to oxidative stress. From 12 h post-infection the ALA was visible, haematoxylin-eosin and Masson's trichrome stains were consistent with these observations, and alanine aminotransferase, alkaline phosphatase and γ-glutamyl transpeptidase serum activities were increased too. At 48 h after infection, liver glycogen content was significantly reduced. Western blot analyses showed that 4-Hydroxy-2-nonenal peaked at 12 h, while glycogen synthase kinase-3β, cleaved caspase-3, pNF-κB, interleukin-1β and tumour necrosis factor-α were overexpressed from 12 to 48 h post-infection. Otherwise, Nrf2 and superoxide dismutase-1, decreased at 48 h and catalase declined at 36 and 48 h. Furthermore, heme oxygenase-1 was increased at 12 and 24 h and decreased to normal levels at 36 and 48 h. These findings suggest for the first time that the host antioxidant system of Nrf2 is influenced during ALA.
Collapse
|
96
|
Hambright HG, Ghosh R. Autophagy: In the cROSshairs of cancer. Biochem Pharmacol 2016; 126:13-22. [PMID: 27789215 DOI: 10.1016/j.bcp.2016.10.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 10/21/2016] [Indexed: 12/18/2022]
Abstract
Two prominent features of tumors that contribute to oncogenic survival signaling are redox disruption, or oxidative stress phenotype, and high autophagy signaling, making both phenomena ideal therapeutic targets. However, the relationship between redox disruption and autophagy signaling is not well characterized and the clinical impact of reactive oxygen species (ROS)-generating chemotherapeutics on autophagy merits immediate attention as autophagy largely contributes to chemotherapeutic resistance. In this commentary we focus on melanoma, using it as an example to provide clarity to current literature regarding the roles of autophagy and redox signaling which can be applicable to initiation and maintenance of most tumor types. Further, we address the crosstalk between ROS and autophagy signaling during pharmacological intervention and cell fate decisions. We attempt to elucidate the role of autophagy in regulating cell fate following treatment with ROS-generating agents in preclinical and clinical settings and discuss the emerging role of autophagy in cell fate decisions and as a cell death mechanism. We also address technical aspects of redox and autophagy evaluation in experimental design and data interpretation. Lastly, we present a provocative view of the clinical relevance, emerging challenges in dual targeting of redox and autophagy pathways for therapy, and the future directions to be addressed in order to advance both basic and translational aspects of this field.
Collapse
Affiliation(s)
- Heather Graham Hambright
- Department of Urology, University of Texas Health Science Center at San Antonio, South Texas Research Facility Campus, 8403 Floyd Curl Drive, San Antonio, TX 78229, USA; Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, South Texas Research Facility Campus, 8403 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Rita Ghosh
- Department of Urology, University of Texas Health Science Center at San Antonio, South Texas Research Facility Campus, 8403 Floyd Curl Drive, San Antonio, TX 78229, USA; Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, South Texas Research Facility Campus, 8403 Floyd Curl Drive, San Antonio, TX 78229, USA; Department of Pharmacology, University of Texas Health Science Center at San Antonio, South Texas Research Facility Campus, 8403 Floyd Curl Drive, San Antonio, TX 78229, USA; Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, South Texas Research Facility Campus, 8403 Floyd Curl Drive, San Antonio, TX 78229, USA.
| |
Collapse
|
97
|
Redox homeostasis of breast cancer lineages contributes to differential cell death response to exogenous hydrogen peroxide. Life Sci 2016; 158:7-13. [DOI: 10.1016/j.lfs.2016.06.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 05/31/2016] [Accepted: 06/16/2016] [Indexed: 02/07/2023]
|
98
|
Weigand A, Boos AM, Tasbihi K, Beier JP, Dalton PD, Schrauder M, Horch RE, Beckmann MW, Strissel PL, Strick R. Selective isolation and characterization of primary cells from normal breast and tumors reveal plasticity of adipose derived stem cells. Breast Cancer Res 2016; 18:32. [PMID: 26968831 PMCID: PMC4788819 DOI: 10.1186/s13058-016-0688-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Accepted: 02/19/2016] [Indexed: 02/08/2023] Open
Abstract
Background There is a need to establish more cell lines from breast tumors in contrast to immortalized cell lines from metastatic effusions in order to represent the primary tumor and not principally metastatic biology of breast cancer. This investigation describes the simultaneous isolation, characterization, growth and function of primary mammary epithelial cells (MEC), mesenchymal cells (MES) and adipose derived stem cells (ADSC) from four normal breasts, one inflammatory and one triple-negative ductal breast tumors. Methods A total of 17 cell lines were established and gene expression was analyzed for MEC and MES (n = 42) and ADSC (n = 48) and MUC1, pan-KRT, CD90 and GATA-3 by immunofluorescence. DNA fingerprinting to track cell line identity was performed between original primary tissues and isolates. Functional studies included ADSC differentiation, tumor MES and MEC invasion co-cultured with ADSC-conditioned media (CM) and MES adhesion and growth on 3D-printed scaffolds. Results Comparative analysis showed higher gene expression of EPCAM, CD49f, CDH1 and KRTs for normal MEC lines; MES lines e.g. Vimentin, CD10, ACTA2 and MMP9; and ADSC lines e.g. CD105, CD90, CDH2 and CDH11. Compared to the mean of all four normal breast cell lines, both breast tumor cell lines demonstrated significantly lower ADSC marker gene expression, but higher expression of mesenchymal and invasion gene markers like SNAI1 and MMP2. When compared with four normal ADSC differentiated lineages, both tumor ADSC showed impaired osteogenic and chondrogenic but enhanced adipogenic differentiation and endothelial-like structures, possibly due to high PDGFRB and CD34. Addressing a functional role for overproduction of adipocytes, we initiated 3D-invasion studies including different cell types from the same patient. CM from ADSC differentiating into adipocytes induced tumor MEC 3D-invasion via EMT and amoeboid phenotypes. Normal MES breast cells adhered and proliferated on 3D-printed scaffolds containing 20 fibers, but not on 2.5D-printed scaffolds with single fiber layers, important for tissue engineering. Conclusion Expression analyses confirmed successful simultaneous cell isolations of three different phenotypes from normal and tumor primary breast tissues. Our cell culture studies support that breast-tumor environment differentially regulates tumor ADSC plasticity as well as cell invasion and demonstrates applications for regenerative medicine. Electronic supplementary material The online version of this article (doi:10.1186/s13058-016-0688-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Annika Weigand
- Department of Plastic and Hand Surgery and Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Krankenhausstr. 12, Erlangen, D-91054, Germany.
| | - Anja M Boos
- Department of Plastic and Hand Surgery and Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Krankenhausstr. 12, Erlangen, D-91054, Germany
| | - Kereshmeh Tasbihi
- Department of Plastic and Hand Surgery and Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Krankenhausstr. 12, Erlangen, D-91054, Germany
| | - Justus P Beier
- Department of Plastic and Hand Surgery and Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Krankenhausstr. 12, Erlangen, D-91054, Germany
| | - Paul D Dalton
- Department of Functional Materials in Medicine and Dentistry, University of Würzburg, Pleicherwall 2, Würzburg, Germany
| | - Michael Schrauder
- Department of Obstetrics and Gynecology, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Raymund E Horch
- Department of Plastic and Hand Surgery and Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Krankenhausstr. 12, Erlangen, D-91054, Germany
| | - Matthias W Beckmann
- Department of Obstetrics and Gynecology, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Pamela L Strissel
- Department of Obstetrics and Gynecology, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Reiner Strick
- Department of Obstetrics and Gynecology, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
99
|
Flores-Pérez A, Marchat LA, Sánchez LL, Romero-Zamora D, Arechaga-Ocampo E, Ramírez-Torres N, Chávez JD, Carlos-Reyes Á, Astudillo-de la Vega H, Ruiz-García E, González-Pérez A, López-Camarillo C. Differential proteomic analysis reveals that EGCG inhibits HDGF and activates apoptosis to increase the sensitivity of non-small cells lung cancer to chemotherapy. Proteomics Clin Appl 2016; 10:172-182. [PMID: 26175166 DOI: 10.1002/prca.201500008] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 06/11/2015] [Accepted: 07/09/2015] [Indexed: 12/29/2022]
Abstract
PURPOSE To search for regulated proteins in response to green tea (-)-epigallocatechin-3-gallate (EGCG) in A549 lung cancer cells. EXPERIMENTAL DESIGN 2DE and ESI/multistage MS (ESI-MS/MS) were performed to identify modulated proteins in A549 cells treated with EGCG. Cell migration was evaluated by transwell assays. RNA interference was used to silence the hepatoma-derived growth factor (HDGF). Caspase-3, caspase-9, and HDGF were immunodetected by Western blot assays. Flow cytometry was used for detection of mitochondrial membrane potential and apoptosis. RESULTS We found that HDGF expression was threefold suppressed by EGCG treatment. Downregulation of HDGF by EGCG was confirmed using anti-HDGF antibodies in three lung cancer cell lines. EGCG treatment and HDGF abrogation by RNA interference resulted in a decreased migration of A549 cells. In addition, EGCG induced a marked synergistic effect with cisplatin in cell death. Consistently, an enhanced cytotoxicity in HDGF-silenced cells was also found. Cell death was associated to increased apoptosis, disruption of the mitochondrial membrane potential, and activation of caspase-3 and caspase-9. CONCLUSION AND CLINICAL RELEVANCE Our data suggest for the first time that abrogation of HDGF by EGCG enhances cisplatin-induced apoptosis and sensitize A549 cells to chemotherapy. Therefore, we propose that decreasing the HDGF levels by using EGCG may represent a novel strategy in lung cancer therapy.
Collapse
Affiliation(s)
- Ali Flores-Pérez
- Genomics Sciences Program, Autonomous University of Mexico City, Mexico City, Mexico
| | - Laurence A Marchat
- Molecular Biomedicine Program, National School of Medicine and Homeopathy, National Polytechnic Institute, Mexico
- Biotechnology Program, National School of Medicine and Homeopathy, National Polytechnic Institute, Mexico
| | - Lidia López Sánchez
- Genomics Sciences Program, Autonomous University of Mexico City, Mexico City, Mexico
| | - Diana Romero-Zamora
- Genomics Sciences Program, Autonomous University of Mexico City, Mexico City, Mexico
| | | | - Nayeli Ramírez-Torres
- Genomics Sciences Program, Autonomous University of Mexico City, Mexico City, Mexico
| | - José Díaz Chávez
- Oncogenomics Laboratory, National Institute of Cancerology, Mexico
| | | | - Horacio Astudillo-de la Vega
- Laboratory of Translational Cancer Research and Cellular Therapy, Oncology Hospital, Medical Center Siglo XXI, Mexico
| | - Erika Ruiz-García
- Translational Medicine Laboratory, National Institute of Cancerology, Mexico
| | | | - César López-Camarillo
- Genomics Sciences Program, Autonomous University of Mexico City, Mexico City, Mexico
| |
Collapse
|
100
|
Amara S, Ivy MT, Myles EL, Tiriveedhi V. Sodium channel γENaC mediates IL-17 synergized high salt induced inflammatory stress in breast cancer cells. Cell Immunol 2015; 302:1-10. [PMID: 26723502 DOI: 10.1016/j.cellimm.2015.12.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 12/15/2015] [Accepted: 12/18/2015] [Indexed: 01/03/2023]
Abstract
Chronic inflammation is known to play a critical role in the development of cancer. Recent evidence suggests that high salt in the tissue microenvironment induces chronic inflammatory milieu. In this report, using three breast cancer-related cell lines, we determined the molecular basis of the potential synergistic inflammatory effect of sodium chloride (NaCl) with interleukin-17 (IL-17). Combined treatment of high NaCl (0.15M) with sub-effective IL-17 (0.1 nM) induced enhanced growth in breast cancer cells along with activation of reactive nitrogen and oxygen (RNS/ROS) species known to promote cancer. Similar effect was not observed with equi-molar mannitol. This enhanced of ROS/RNS activity correlates with upregulation of γENaC an inflammatory sodium channel. The similar culture conditions have also induced expression of pro-inflammatory cytokines such as IL-6, TNFα etc. Taken together, these data suggest that high NaCl in the cellular microenvironment induces a γENaC mediated chronic inflammatory response with a potential pro-carcinogenic effect.
Collapse
Affiliation(s)
- Suneetha Amara
- Department of Medicine, Mercy Hospital, St Louis, MO, USA
| | - Michael T Ivy
- Department of Biological Sciences, Tennessee State University, Nashville, TN, USA
| | - Elbert L Myles
- Department of Biological Sciences, Tennessee State University, Nashville, TN, USA
| | | |
Collapse
|