51
|
Gasparetto M, Pei S, Minhajuddin M, Stevens B, Smith CA, Seligman P. Low ferroportin expression in AML is correlated with good risk cytogenetics, improved outcomes and increased sensitivity to chemotherapy. Leuk Res 2019; 80:1-10. [PMID: 30852438 DOI: 10.1016/j.leukres.2019.02.011] [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: 09/29/2018] [Revised: 02/25/2019] [Accepted: 02/27/2019] [Indexed: 12/31/2022]
Abstract
Iron metabolism is altered in a variety of cancers; however, little is known about the role of iron metabolism in the biology and response to therapy of acute myeloid leukemia (AML). Here we show that SLC40A1, the gene encoding the iron exporter ferroportin (FPN), is variably expressed among primary AMLs and that low levels are associated with good prognosis and improved outcomes. In particular, core binding factor (CBF) AMLs, which are associated with good outcomes with chemotherapy, consistently have low level of SLC40A1 expression. AML cell lines that expressed relatively low levels of FPN endogenously, or were engineered via gene knockdown, had an increased sensitivity to chemotherapy relative to controls expressing high levels of FPN. Primary FPNlow AML bulk cells also had increased sensitivity to Ara-C treatment, iron treatment and the combination of Ara-C and iron relative to FPNhigh cells. FPNlow leukemic stem cells (LSCs) had decreased viability following addition of iron alone and in combination with Ara-C treatment relative to FPNhigh LSCs. Together these observations suggest a model where FPN mediated iron metabolism may play a role in chemosensitivity and outcome to therapy in AML.
Collapse
Affiliation(s)
- Maura Gasparetto
- Division of Hematology, University of Colorado Medical Center, Aurora, CO, USA.
| | - Shanshan Pei
- Division of Hematology, University of Colorado Medical Center, Aurora, CO, USA
| | | | - Brett Stevens
- Division of Hematology, University of Colorado Medical Center, Aurora, CO, USA
| | - Clayton A Smith
- Division of Hematology, University of Colorado Medical Center, Aurora, CO, USA
| | - Paul Seligman
- Division of Hematology, University of Colorado Medical Center, Aurora, CO, USA
| |
Collapse
|
52
|
Pinton L, Masetto E, Vettore M, Solito S, Magri S, D'Andolfi M, Del Bianco P, Lollo G, Benoit JP, Okada H, Diaz A, Della Puppa A, Mandruzzato S. The immune suppressive microenvironment of human gliomas depends on the accumulation of bone marrow-derived macrophages in the center of the lesion. J Immunother Cancer 2019; 7:58. [PMID: 30813960 PMCID: PMC6391795 DOI: 10.1186/s40425-019-0536-x] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Accepted: 02/13/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Systemic and local immune suppression plays a significant role in glioma progression. Glioma microenvironment contains both brain-resident microglial cells (MG) and bone marrow-derived macrophages (BMDM), but the study of their functional and immune regulatory activity has been hampered until now by the lack of markers allowing a proper identification and isolation to collect pure populations. METHODS Myeloid and lymphoid infiltrate were characterized in grade II, III and IV gliomas by multicolor flow cytometry, along with the composition of the cell subsets of circulating myeloid cells. Macrophages were sorted and tested for their immunosuppressive ability. Moreover, following preoperative administration of 5-aminolevulinic acid to patients, distinct areas of tumor lesion were surgically removed and analyzed, based on protoporphyrin IX fluorescence emission. RESULTS The immune microenvironment of grade II to grade IV gliomas contains a large proportion of myeloid cells and a small proportion of lymphocytes expressing markers of dysfunctional activity. BMDM and resident MG cells were characterized through a combination of markers, thus permitting their geographical identification in the lesions, their sorting and subsequent analysis of the functional characteristics. The infiltration by BMDM reached the highest percentages in grade IV gliomas, and it increased from the periphery to the center of the lesion, where it exerted a strong immunosuppression that was, instead, absent in the marginal area. By contrast, MG showed little or no suppression. Functional differences, such as iron metabolism and phagocytosis, characterized resident versus blood-derived macrophages. Significant alterations in circulating monocytes were present in grade IV patients, correlating with accumulation of tumor macrophages. CONCLUSIONS Grade IV gliomas have an alteration in both circulating and tumor-associated myeloid cells and, differently from grade II and III gliomas, show a significant presence of blood-derived, immune suppressive macrophages. BMDM and MG have different functional properties.
Collapse
Affiliation(s)
- Laura Pinton
- Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
| | - Elena Masetto
- Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
| | - Marina Vettore
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Via Gattamelata, 64 35128, Padova, Italy
| | - Samantha Solito
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Via Gattamelata, 64 35128, Padova, Italy
| | - Sara Magri
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Via Gattamelata, 64 35128, Padova, Italy
| | - Marta D'Andolfi
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Via Gattamelata, 64 35128, Padova, Italy
| | | | - Giovanna Lollo
- LUNAM Universite - Micro et Nanomedecines Biomimetiques, F-49933, Angers, France
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, LAGEP UMR 5007, F-69100, VILLEURBANNE, Lyon, France
| | - Jean-Pierre Benoit
- INSERM U1066/CNRS 6021 University of ANGERS, cedex 9, 49933, Angers, France
| | - Hideho Okada
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Aaron Diaz
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | | | - Susanna Mandruzzato
- Veneto Institute of Oncology IOV - IRCCS, Padova, Italy.
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Via Gattamelata, 64 35128, Padova, Italy.
| |
Collapse
|
53
|
Gan B, Jiang X, Mi Z, Zhang C, Bai R, Shuai Q, Xie Y. Straightforward synthesis of amino-3-hydroxypyridin-4-one iron chelators via BBr 3-mediated tandem reduction of azides and deprotection of methyl ether. SYNTHETIC COMMUN 2019. [DOI: 10.1080/00397911.2018.1542002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Bing Gan
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology , Hangzhou , People’s Republic of China
| | - Xiaoying Jiang
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology , Hangzhou , People’s Republic of China
| | - Zhisheng Mi
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology , Hangzhou , People’s Republic of China
| | - Changjung Zhang
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology , Hangzhou , People’s Republic of China
| | - Renren Bai
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology , Hangzhou , People’s Republic of China
| | - Qi Shuai
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology , Hangzhou , People’s Republic of China
| | - Yuanyuan Xie
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology , Hangzhou , People’s Republic of China
| |
Collapse
|
54
|
Schoenfeld JD, Alexander MS, Waldron TJ, Sibenaller ZA, Spitz DR, Buettner GR, Allen BG, Cullen JJ. Pharmacological Ascorbate as a Means of Sensitizing Cancer Cells to Radio-Chemotherapy While Protecting Normal Tissue. Semin Radiat Oncol 2019; 29:25-32. [PMID: 30573181 PMCID: PMC6310038 DOI: 10.1016/j.semradonc.2018.10.006] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Chemoradiation has remained the standard of care treatment for many of the most aggressive cancers. However, despite effective toxicity to cancer cells, current chemoradiation regimens are limited in efficacy due to significant normal cell toxicity. Thus, efforts have been made to identify agents demonstrating selective toxicity, whereby treatments simultaneously sensitize cancer cells to protect normal cells from chemoradiation. Pharmacological ascorbate (intravenous infusions of vitamin C resulting in plasma ascorbate concentrations ≥20 mM; P-AscH-) has demonstrated selective toxicity in a variety of preclinical tumor models and is currently being assessed as an adjuvant to standard-of-care therapies in several early phase clinical trials. This review summarizes the most current preclinical and clinical data available demonstrating the multidimensional role of P-AscH- in cancer therapy including: selective toxicity to cancer cells via a hydrogen peroxide (H2O2)-mediated mechanism; action as a sensitizing agent of cancer cells to chemoradiation; a protectant of normal tissues exposed to chemoradiation; and its safety and tolerability in clinical trials.
Collapse
Affiliation(s)
- Joshua D Schoenfeld
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Iowa City, IA
| | - Matthew S Alexander
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Iowa City, IA; Department of Surgery, Iowa City, IA
| | - Timothy J Waldron
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Iowa City, IA; University of Iowa Carver College of Medicine, Iowa City, IA; The Holden Comprehensive Cancer Center, Iowa City, IA
| | - Zita A Sibenaller
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Iowa City, IA
| | - Douglas R Spitz
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Iowa City, IA; University of Iowa Carver College of Medicine, Iowa City, IA; The Holden Comprehensive Cancer Center, Iowa City, IA
| | - Garry R Buettner
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Iowa City, IA; University of Iowa Carver College of Medicine, Iowa City, IA; The Holden Comprehensive Cancer Center, Iowa City, IA
| | - Bryan G Allen
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Iowa City, IA; University of Iowa Carver College of Medicine, Iowa City, IA; The Holden Comprehensive Cancer Center, Iowa City, IA
| | - Joseph J Cullen
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Iowa City, IA; Department of Surgery, Iowa City, IA; University of Iowa Carver College of Medicine, Iowa City, IA; The Holden Comprehensive Cancer Center, Iowa City, IA; Veterans Affairs Medical Center, Iowa City, IA.
| |
Collapse
|
55
|
Gaur K, Vázquez-Salgado A, Duran-Camacho G, Dominguez-Martinez I, Benjamín-Rivera J, Fernández-Vega L, Carmona Sarabia L, Cruz García A, Pérez-Deliz F, Méndez Román J, Vega-Cartagena M, Loza-Rosas S, Rodriguez Acevedo X, Tinoco A. Iron and Copper Intracellular Chelation as an Anticancer Drug Strategy. INORGANICS 2018. [DOI: https://doi.org/10.3390/inorganics6040126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
A very promising direction in the development of anticancer drugs is inhibiting the molecular pathways that keep cancer cells alive and able to metastasize. Copper and iron are two essential metals that play significant roles in the rapid proliferation of cancer cells and several chelators have been studied to suppress the bioavailability of these metals in the cells. This review discusses the major contributions that Cu and Fe play in the progression and spreading of cancer and evaluates select Cu and Fe chelators that demonstrate great promise as anticancer drugs. Efforts to improve the cellular delivery, efficacy, and tumor responsiveness of these chelators are also presented including a transmetallation strategy for dual targeting of Cu and Fe. To elucidate the effectiveness and specificity of Cu and Fe chelators for treating cancer, analytical tools are described for measuring Cu and Fe levels and for tracking the metals in cells, tissue, and the body.
Collapse
|
56
|
Gaur K, Vázquez-Salgado AM, Duran-Camacho G, Dominguez-Martinez I, Benjamín-Rivera JA, Fernández-Vega L, Sarabia LC, García AC, Pérez-Deliz F, Méndez Román JA, Vega-Cartagena M, Loza-Rosas SA, Acevedo XR, Tinoco AD. Iron and Copper Intracellular Chelation as an Anticancer Drug Strategy. INORGANICS 2018; 6:126. [PMID: 33912613 PMCID: PMC8078164 DOI: 10.3390/inorganics6040126] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
A very promising direction in the development of anticancer drugs is inhibiting the molecular pathways that keep cancer cells alive and able to metastasize. Copper and iron are two essential metals that play significant roles in the rapid proliferation of cancer cells and several chelators have been studied to suppress the bioavailability of these metals in the cells. This review discusses the major contributions that Cu and Fe play in the progression and spreading of cancer and evaluates select Cu and Fe chelators that demonstrate great promise as anticancer drugs. Efforts to improve the cellular delivery, efficacy, and tumor responsiveness of these chelators are also presented including a transmetallation strategy for dual targeting of Cu and Fe. To elucidate the effectiveness and specificity of Cu and Fe chelators for treating cancer, analytical tools are described for measuring Cu and Fe levels and for tracking the metals in cells, tissue, and the body.
Collapse
Affiliation(s)
- Kavita Gaur
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA
| | | | - Geraldo Duran-Camacho
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA
| | | | - Josué A Benjamín-Rivera
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA
| | - Lauren Fernández-Vega
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA
| | - Lesly Carmona Sarabia
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA
| | - Angelys Cruz García
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA
| | - Felipe Pérez-Deliz
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA
| | - José A Méndez Román
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA
| | - Melissa Vega-Cartagena
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA
| | - Sergio A Loza-Rosas
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA
| | | | - Arthur D Tinoco
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA
| |
Collapse
|
57
|
Vela D. Iron Metabolism in Prostate Cancer; From Basic Science to New Therapeutic Strategies. Front Oncol 2018; 8:547. [PMID: 30538952 PMCID: PMC6277552 DOI: 10.3389/fonc.2018.00547] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Accepted: 11/05/2018] [Indexed: 01/09/2023] Open
Abstract
An increasing amount of research has recently strengthened the case for the existence of iron dysmetabolism in prostate cancer. It is characterized with a wide array of differential expression of iron-related proteins compared to normal cells. These proteins control iron entry, cellular iron distribution but also iron exit from prostate cells. Iron dysmetabolism is not an exclusive feature of prostate cancer cells, but it is observed in other cells of the tumor microenvironment. Disrupting the machinery that secures iron for prostate cancer cells can retard tumor growth and its invasive potential. This review unveils the current understanding of the ways that prostate cancer cells secure iron in the tumor milieu and how can we exploit this knowledge for therapeutic purposes.
Collapse
Affiliation(s)
- Driton Vela
- Department of Physiology, University of Prishtina, Prishtina, Kosovo
| |
Collapse
|
58
|
Pfeifhofer-Obermair C, Tymoszuk P, Petzer V, Weiss G, Nairz M. Iron in the Tumor Microenvironment-Connecting the Dots. Front Oncol 2018; 8:549. [PMID: 30534534 PMCID: PMC6275298 DOI: 10.3389/fonc.2018.00549] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 11/06/2018] [Indexed: 12/18/2022] Open
Abstract
Iron metabolism and tumor biology are intimately linked. Iron facilitates the production of oxygen radicals, which may either result in iron-induced cell death, ferroptosis, or contribute to mutagenicity and malignant transformation. Once transformed, malignant cells require high amounts of iron for proliferation. In addition, iron has multiple regulatory effects on the immune system, thus affecting tumor surveillance by immune cells. For these reasons, inconsiderate iron supplementation in cancer patients has the potential of worsening disease course and outcome. On the other hand, chronic immune activation in the setting of malignancy alters systemic iron homeostasis and directs iron fluxes into myeloid cells. While this response aims at withdrawing iron from tumor cells, it may impair the effector functions of tumor-associated macrophages and will result in iron-restricted erythropoiesis and the development of anemia, subsequently. This review summarizes our current knowledge of the interconnections of iron homeostasis with cancer biology, discusses current clinical controversies in the treatment of anemia of cancer and focuses on the potential roles of iron in the solid tumor microenvironment, also speculating on yet unknown molecular mechanisms.
Collapse
Affiliation(s)
- Christa Pfeifhofer-Obermair
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Innsbruck, Austria
| | - Piotr Tymoszuk
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Innsbruck, Austria
| | - Verena Petzer
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Innsbruck, Austria
| | - Günter Weiss
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Innsbruck, Austria.,Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, Innsbruck, Austria
| | - Manfred Nairz
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
59
|
Algso MAS, Kivrak A. New strategy for the synthesis of 3-ethynyl-2-(thiophen-2-yl)benzo[b]thiophene derivatives. CHEMICAL PAPERS 2018. [DOI: 10.1007/s11696-018-0640-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
60
|
Yang J, Li W, Luo L, Jiang M, Zhu C, Qin B, Yin H, Yuan X, Yin X, Zhang J, Luo Z, Du Y, You J. Hypoxic tumor therapy by hemoglobin-mediated drug delivery and reversal of hypoxia-induced chemoresistance. Biomaterials 2018; 182:145-156. [DOI: 10.1016/j.biomaterials.2018.08.004] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 08/01/2018] [Accepted: 08/02/2018] [Indexed: 02/06/2023]
|
61
|
Links Between Iron and Lipids: Implications in Some Major Human Diseases. Pharmaceuticals (Basel) 2018; 11:ph11040113. [PMID: 30360386 PMCID: PMC6315991 DOI: 10.3390/ph11040113] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 10/18/2018] [Accepted: 10/19/2018] [Indexed: 12/30/2022] Open
Abstract
Maintenance of iron homeostasis is critical to cellular health as both its excess and insufficiency are detrimental. Likewise, lipids, which are essential components of cellular membranes and signaling mediators, must also be tightly regulated to hinder disease progression. Recent research, using a myriad of model organisms, as well as data from clinical studies, has revealed links between these two metabolic pathways, but the mechanisms behind these interactions and the role these have in the progression of human diseases remains unclear. In this review, we summarize literature describing cross-talk between iron and lipid pathways, including alterations in cholesterol, sphingolipid, and lipid droplet metabolism in response to changes in iron levels. We discuss human diseases correlating with both iron and lipid alterations, including neurodegenerative disorders, and the available evidence regarding the potential mechanisms underlying how iron may promote disease pathogenesis. Finally, we review research regarding iron reduction techniques and their therapeutic potential in treating patients with these debilitating conditions. We propose that iron-mediated alterations in lipid metabolic pathways are involved in the progression of these diseases, but further research is direly needed to elucidate the mechanisms involved.
Collapse
|
62
|
Brzóska K, Bartłomiejczyk T, Sochanowicz B, Cymerman M, Grudny J, Kołakowski J, Kruszewski M, Śliwiński P, Roszkowski-Śliż K, Kapka-Skrzypczak L. Carcinogenesis-related changes in iron metabolism in chronic obstructive pulmonary disease subjects with lung cancer. Oncol Lett 2018; 16:6831-6837. [PMID: 30405827 DOI: 10.3892/ol.2018.9459] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 08/10/2018] [Indexed: 12/17/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is often accompanied by lung cancer. In our previous work, it was observed that matrix metalloproteinase-3 and haptoglobin (HP) polymorphisms were potential markers of enhanced susceptibility to lung cancer development among male COPD subjects. Here, results are reported on blood serum levels of several proteins involved in iron metabolism, inflammation and the oxidative stress response compared between the same groups of subjects. The blood serum levels of tumor necrosis factor α (TNFα), transferrin, hepcidin, ferritin, soluble transferrin receptor and 8-oxo-2'-deoxyguanosine were compared, as well as total iron-binding capacity (TIBC) and ceruloplasmin ferroxidase activity in two groups of subjects: Male COPD patients (54 subjects) and male COPD patients diagnosed with lung cancer (53 subjects). Statistically significant differences were identified between the two groups in transferrin and TNFα levels, as well as in TIBC; all three parameters were lower in the group consisting of COPD patients diagnosed with lung cancer (P<0.01). It was also revealed that HP genotype 1/2 was concomitant with low transferrin blood level in subjects with COPD; this apparent dependence was absent in the COPD + cancer subjects. The results indicate a role of iron metabolism in the susceptibility to lung cancer in COPD-affected subjects. They also emphasize the importance of individual capacity for an effective response to oxidative stress during the pathogenic process as HP is a plasma protein that binds free hemoglobin and its polymorphism results in proteins with altered hemoglobin-binding capacity and different antioxidant and iron-recycling functions.
Collapse
Affiliation(s)
- Kamil Brzóska
- Institute of Nuclear Chemistry and Technology, Centre for Radiobiology and Biological Dosimetry, 03-195 Warsaw, Poland
| | - Teresa Bartłomiejczyk
- Institute of Nuclear Chemistry and Technology, Centre for Radiobiology and Biological Dosimetry, 03-195 Warsaw, Poland
| | - Barbara Sochanowicz
- Institute of Nuclear Chemistry and Technology, Centre for Radiobiology and Biological Dosimetry, 03-195 Warsaw, Poland
| | - Magdalena Cymerman
- Institute of Nuclear Chemistry and Technology, Centre for Radiobiology and Biological Dosimetry, 03-195 Warsaw, Poland
| | - Jacek Grudny
- Institute of Tuberculosis and Lung Diseases, Third Department of Lung Diseases, 01-138 Warsaw, Poland
| | - Jacek Kołakowski
- Institute of Tuberculosis and Lung Diseases, Department of Diagnosis and Treatment of Respiratory Insufficiency, 01-138 Warsaw, Poland
| | - Marcin Kruszewski
- Institute of Nuclear Chemistry and Technology, Centre for Radiobiology and Biological Dosimetry, 03-195 Warsaw, Poland.,Institute of Rural Health, Department of Molecular Biology and Translational Research, 20-090 Lublin, Poland.,University of Information Technology and Management, Faculty of Medicine, Department of Medical Biology and Translational Research, 35-225 Rzeszów, Poland
| | - Paweł Śliwiński
- Institute of Tuberculosis and Lung Diseases, Department of Diagnosis and Treatment of Respiratory Insufficiency, 01-138 Warsaw, Poland
| | - Kazimierz Roszkowski-Śliż
- Institute of Tuberculosis and Lung Diseases, Third Department of Lung Diseases, 01-138 Warsaw, Poland
| | - Lucyna Kapka-Skrzypczak
- Institute of Rural Health, Department of Molecular Biology and Translational Research, 20-090 Lublin, Poland.,University of Information Technology and Management, Faculty of Medicine, Department of Medical Biology and Translational Research, 35-225 Rzeszów, Poland
| |
Collapse
|
63
|
Algso MAS, Kivrak A, Konus M, Yilmaz C, Kurt-Kizildoğan A. Synthesis and biological evaluation of novel benzothiophene derivatives. J CHEM SCI 2018. [DOI: 10.1007/s12039-018-1523-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
64
|
Puig S, Ramos-Alonso L, Romero AM, Martínez-Pastor MT. The elemental role of iron in DNA synthesis and repair. Metallomics 2018; 9:1483-1500. [PMID: 28879348 DOI: 10.1039/c7mt00116a] [Citation(s) in RCA: 217] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Iron is an essential redox element that functions as a cofactor in many metabolic pathways. Critical enzymes in DNA metabolism, including multiple DNA repair enzymes (helicases, nucleases, glycosylases, demethylases) and ribonucleotide reductase, use iron as an indispensable cofactor to function. Recent striking results have revealed that the catalytic subunit of DNA polymerases also contains conserved cysteine-rich motifs that bind iron-sulfur (Fe/S) clusters that are essential for the formation of stable and active complexes. In line with this, mitochondrial and cytoplasmic defects in Fe/S cluster biogenesis and insertion into the nuclear iron-requiring enzymes involved in DNA synthesis and repair lead to DNA damage and genome instability. Recent studies have shown that yeast cells possess multi-layered mechanisms that regulate the ribonucleotide reductase function in response to fluctuations in iron bioavailability to maintain optimal deoxyribonucleotide concentrations. Finally, a fascinating DNA charge transport model indicates how the redox active Fe/S centers present in DNA repair machinery components are critical for detecting and repairing DNA mismatches along the genome by long-range charge transfers through double-stranded DNA. These unexpected connections between iron and DNA replication and repair have to be considered to properly understand cancer, aging and other DNA-related diseases.
Collapse
Affiliation(s)
- Sergi Puig
- Departamento de Biotecnología, Instituto de Agroquímica y Tecnología de Alimentos (IATA), Consejo Superior de Investigaciones Científicas (CSIC), Ave. Agustín Escardino 7, 46980, Paterna, Valencia, Spain.
| | | | | | | |
Collapse
|
65
|
Malarz K, Mrozek-Wilczkiewicz A, Serda M, Rejmund M, Polanski J, Musiol R. The role of oxidative stress in activity of anticancer thiosemicarbazones. Oncotarget 2018; 9:17689-17710. [PMID: 29707141 PMCID: PMC5915149 DOI: 10.18632/oncotarget.24844] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 02/28/2018] [Indexed: 01/21/2023] Open
Abstract
Thiosemicarbazones are chelators of transition metals such as iron or copper whose anticancer potency is intensively investigated. Although two compounds from this class have entered clinical trials, their precise mechanism of action is still unknown. Recent studies have suggested the mobilization of the iron ions from a cell, as well as the inhibition of ribonucleotide reductase, and the formation of reactive oxygen species. The complexity and vague nature of this mechanism not only impedes a more rational design of novel compounds, but also the further development of those that are highly active that are already in the preclinical phase. In the current work, a series of highly active thiosemicarbazones was studied for their antiproliferative activity in vitro. Our experiments indicate that these complexes have ionophoric properties and redox activity. They appeared to be very effective generating reactive oxygen species and deregulating the antioxidative potential of a cell. Moreover, the genes that are responsible for antioxidant capacity were considerably deregulated, which led to the induction of apoptosis and cell cycle arrest. On the other hand, good intercalating properties of the studied compounds may explain their ability to cleave DNA strands and to also poison related enzymes through the formation of reactive oxygen species. These findings may help to explain the particularly high selectivity that they have over normal cells, which generally have a stronger redox equilibrium.
Collapse
Affiliation(s)
- Katarzyna Malarz
- Institute of Chemistry, University of Silesia in Katowice, Katowice, Poland
- Silesian Center for Education and Interdisciplinary Research, University of Silesia in Katowice, Chorzów, Poland
| | - Anna Mrozek-Wilczkiewicz
- Silesian Center for Education and Interdisciplinary Research, University of Silesia in Katowice, Chorzów, Poland
- A. Chełkowski Institute of Physics, University of Silesia in Katowice, Katowice, Poland
| | - Maciej Serda
- Institute of Chemistry, University of Silesia in Katowice, Katowice, Poland
| | - Marta Rejmund
- Institute of Chemistry, University of Silesia in Katowice, Katowice, Poland
| | - Jaroslaw Polanski
- Institute of Chemistry, University of Silesia in Katowice, Katowice, Poland
| | - Robert Musiol
- Institute of Chemistry, University of Silesia in Katowice, Katowice, Poland
| |
Collapse
|
66
|
Chitambar CR, Al-Gizawiy MM, Alhajala HS, Pechman KR, Wereley JP, Wujek R, Clark PA, Kuo JS, Antholine WE, Schmainda KM. Gallium Maltolate Disrupts Tumor Iron Metabolism and Retards the Growth of Glioblastoma by Inhibiting Mitochondrial Function and Ribonucleotide Reductase. Mol Cancer Ther 2018; 17:1240-1250. [PMID: 29592883 DOI: 10.1158/1535-7163.mct-17-1009] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 02/15/2018] [Accepted: 03/23/2018] [Indexed: 11/16/2022]
Abstract
Gallium, a metal with antineoplastic activity, binds transferrin (Tf) and enters tumor cells via Tf receptor1 (TfR1); it disrupts iron homeostasis leading to cell death. We hypothesized that TfR1 on brain microvascular endothelial cells (BMEC) would facilitate Tf-Ga transport into the brain enabling it to target TfR-bearing glioblastoma. We show that U-87 MG and D54 glioblastoma cell lines and multiple glioblastoma stem cell (GSC) lines express TfRs, and that their growth is inhibited by gallium maltolate (GaM) in vitro After 24 hours of incubation with GaM, cells displayed a loss of mitochondrial reserve capacity followed by a dose-dependent decrease in oxygen consumption and a decrease in the activity of the iron-dependent M2 subunit of ribonucleotide reductase (RRM2). IHC staining of rat and human tumor-bearing brains showed that glioblastoma, but not normal glial cells, expressed TfR1 and RRM2, and that glioblastoma expressed greater levels of H- and L-ferritin than normal brain. In an orthotopic U-87 MG glioblastoma xenograft rat model, GaM retarded the growth of brain tumors relative to untreated control (P = 0.0159) and reduced tumor mitotic figures (P = 0.045). Tumors in GaM-treated animals displayed an upregulation of TfR1 expression relative to control animals, thus indicating that gallium produced tumor iron deprivation. GaM also inhibited iron uptake and upregulated TfR1 expression in U-87 MG and D54 cells in vitro We conclude that GaM enters the brain via TfR1 on BMECs and targets iron metabolism in glioblastoma in vivo, thus inhibiting tumor growth. Further development of novel gallium compounds for brain tumor treatment is warranted. Mol Cancer Ther; 17(6); 1240-50. ©2018 AACR.
Collapse
Affiliation(s)
| | - Mona M Al-Gizawiy
- Department of Radiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Hisham S Alhajala
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Kimberly R Pechman
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Janine P Wereley
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Robert Wujek
- Department of Radiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Paul A Clark
- Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - John S Kuo
- Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - William E Antholine
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Kathleen M Schmainda
- Department of Radiology, Medical College of Wisconsin, Milwaukee, Wisconsin.,Department of Biophysics, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
67
|
Wei L, Wang Y, Lin L, Zhang L, Shi Y, Xiang P, Cao S, Shen M, Yang P. Identification of potential serum biomarkers of acute paraquat poisoning in humans using an iTRAQ quantitative proteomic. RSC Adv 2018; 8:10598-10609. [PMID: 35540476 PMCID: PMC9078879 DOI: 10.1039/c7ra12956d] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 01/28/2019] [Accepted: 03/07/2018] [Indexed: 12/18/2022] Open
Abstract
Paraquat (PQ) poisoning has high mortality rates in many countries. Due to it readily being absorbed through the gastrointestinal tract and rapidly excreted in the urine, few biomarkers possess satisfactory specificity and sensitivity in diagnostic and forensic practices. To investigate serum biomarkers in patients with PQ poisoning, pooled sera was analyzed using a proteomic approach based on iTRAQ coupled LC-MS/MS. Of the 413 proteins identified with high confidence, 81 were found to be differentially expressed (1.5-fold change) in the sera of patients with PQ poisoning. The differential expression pattern of 4 of these proteins was validated by enzyme-linked immunosorbent assay (ELISA) in clinical samples. A sera sample from a PQ poisoning patient has shown relatively increased abundance of S100A8 and S100A9. The overexpression of S100A8 and S100A9 was further validated in the lung tissue of PQ-treated rat associated with lung damage. Meanwhile, we identified another two down-expressed proteins, transferrin receptor protein 1 (TfR1) and serum amyloid P-component (SAP), which may be also practicable in human clinical samples as PQ poisoning serum biomarkers. Furthermore, receiver operating characteristic curve analysis confirmed that the expression levels of S100 alarmins, TfR1 and SAP in patient serum could provide a discriminatory diagnostic test for predicting PQ poisoning in patients. Therefore, our results suggest that increased serum levels of S100 alarmins and decreased serum levels of TfR1 and SAP may constitute potential biomarkers for the prediction of PQ poisoning in humans, and might be novel therapeutic targets in PQ poisoning.
Collapse
Affiliation(s)
- Liming Wei
- Institutes of Biomedical Sciences & Department of Chemistry, Fudan University Shanghai China
- Shanghai Songjiang District Central Hospital Shanghai China
| | - Yi Wang
- Institutes of Biomedical Sciences & Department of Chemistry, Fudan University Shanghai China
| | - Ling Lin
- Institutes of Biomedical Sciences & Department of Chemistry, Fudan University Shanghai China
| | - Lei Zhang
- Institutes of Biomedical Sciences & Department of Chemistry, Fudan University Shanghai China
| | - Yan Shi
- Shanghai Key Laboratory of Forensic Medicine, Shanghai Forensic Service Platform, Institute of Forensic Science, Ministry of Justice Shanghai China
| | - Ping Xiang
- Shanghai Key Laboratory of Forensic Medicine, Shanghai Forensic Service Platform, Institute of Forensic Science, Ministry of Justice Shanghai China
| | - Shujun Cao
- Shanghai Songjiang District Central Hospital Shanghai China
| | - Min Shen
- Shanghai Key Laboratory of Forensic Medicine, Shanghai Forensic Service Platform, Institute of Forensic Science, Ministry of Justice Shanghai China
| | - Pengyuan Yang
- Institutes of Biomedical Sciences & Department of Chemistry, Fudan University Shanghai China
| |
Collapse
|
68
|
Akam EA, Utterback RD, Marcero JR, Dailey HA, Tomat E. Disulfide-masked iron prochelators: Effects on cell death, proliferation, and hemoglobin production. J Inorg Biochem 2018; 180:186-193. [PMID: 29324291 PMCID: PMC5956897 DOI: 10.1016/j.jinorgbio.2017.12.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Revised: 12/20/2017] [Accepted: 12/24/2017] [Indexed: 10/18/2022]
Abstract
The iron metabolism of malignant cells, which is altered to ensure higher acquisition and utilization, motivates the investigation of iron chelation strategies in cancer treatment. In a prochelation approach aimed at increasing intracellular specificity, disulfide reduction/activation switches are incorporated on iron-binding scaffolds resulting in intracellularly activated scavengers. Herein, this strategy is applied to several tridentate donor sets including thiosemicarbazones, aroylhydrazones and semicarbazones. The novel prochelator systems are antiproliferative in breast adenocarcinoma cell lines (MCF-7 and metastatic MDA-MB-231) and do not result in the intracellular generation of oxidative stress. Consistent with iron deprivation, the tested prochelators lead to cell-cycle arrest at the G1/S interface and induction of apoptosis. Notably, although hemoglobin-synthesizing blood cells have the highest iron need in the human body, no significant impact on hemoglobin production was observed in the MEL (murine erythroleukemia) model of differentiating erythroid cells. This study provides new information on the intracellular effects of disulfide-based prochelators and indicates aroylhydrazone (AH1-S)2 as a promising prototype of a new class of antiproliferative prochelator systems.
Collapse
Affiliation(s)
- E A Akam
- Department of Chemistry and Biochemistry, The University of Arizona, United States
| | - R D Utterback
- Department of Chemistry and Biochemistry, The University of Arizona, United States
| | - J R Marcero
- Department of Microbiology and Department of Biochemistry and Molecular Biology, University of Georgia, United States
| | - H A Dailey
- Department of Microbiology and Department of Biochemistry and Molecular Biology, University of Georgia, United States
| | - E Tomat
- Department of Chemistry and Biochemistry, The University of Arizona, United States.
| |
Collapse
|
69
|
Hajipour Verdom B, Abdolmaleki P, Behmanesh M. The Static Magnetic Field Remotely Boosts the Efficiency of Doxorubicin through Modulating ROS Behaviors. Sci Rep 2018; 8:990. [PMID: 29343746 PMCID: PMC5772617 DOI: 10.1038/s41598-018-19247-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 12/18/2017] [Indexed: 01/27/2023] Open
Abstract
Exposure to magnetic field (MF) can affect cellular metabolism remotely. Cardio-toxic effects of Doxorubicin (DOXO) have limited clinical uses at high dose. MF due to its effect on reactive oxygen species (ROS) lifetime, may provide a suitable choice to boost the efficacy of this drug at low dose. Here, we investigated the potential effects of homogenous static magnetic field (SMF) on DOXO-induced toxicity and proliferation rate of cancer cells. The results indicated that SMF similar to DOXO decreased the cell viability as well as the proliferation rate of MCF-7 and HFF cells. Moreover, combination of 10 mT SMF and 0.1 µM DOXO decreased the viability and proliferation rate of cancer and normal cells in a synergetic manner. In spite of high a GSH level in cancer cell, SMF boosts the generation and lifetime of ROS at low dose of DOXO, and overcame to GSH mediated drug resistance. The results also confirmed that SMF exposure decreased 50% iron content of cells, which is attributed to iron homeostasis. In conclusion, these findings suggest that SMF can decrease required dose of chemotherapy drugs such as DOXO and thereby decrease their side effect.
Collapse
Affiliation(s)
- Behnam Hajipour Verdom
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University (TMU), Tehran, Iran
| | - Parviz Abdolmaleki
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University (TMU), Tehran, Iran.
| | - Mehrdad Behmanesh
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University (TMU), Tehran, Iran
| |
Collapse
|
70
|
Kaushal M, Lobana TS, Nim L, Kaur J, Bala R, Hundal G, Arora DS, Garcia-Santos I, Duff CE, Jasinski JP. Synthesis, structures, antimicrobial activity and biosafety evaluation of pyridine-2-formaldehyde-N-susbtituted-thiosemicarbazonates of copper(ii). NEW J CHEM 2018. [DOI: 10.1039/c8nj03619e] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Copper(ii) complexes with pyridine based thiosemicarbazones have shown high antimicrobial potential against different microbial strains, and were found to be biosafe with several complexes displaying high cell viability (90–98%).
Collapse
Affiliation(s)
- Mani Kaushal
- Department of Chemistry
- Guru Nanak Dev University
- Amritsar-143 005
- India
| | - Tarlok S. Lobana
- Department of Chemistry
- Guru Nanak Dev University
- Amritsar-143 005
- India
| | - Lovedeep Nim
- Department of Microbiology
- Guru Nanak Dev University
- Amritsar-143 005
- India
| | - Jaskamal Kaur
- Department of Chemistry
- Guru Nanak Dev University
- Amritsar-143 005
- India
| | - Ritu Bala
- Department of Chemistry
- Guru Nanak Dev University
- Amritsar-143 005
- India
| | - Geeta Hundal
- Department of Chemistry
- Guru Nanak Dev University
- Amritsar-143 005
- India
| | - Daljit S. Arora
- Department of Microbiology
- Guru Nanak Dev University
- Amritsar-143 005
- India
| | - Isabel Garcia-Santos
- Departamento de Quimica Inorganica
- Facultad de Farmacia
- Universidad de Santiago
- 15782-Santiago
- Spain
| | | | | |
Collapse
|
71
|
Yu Q, Zhang B, Li J, Du T, Yi X, Li M, Chen W, Alvarez PJJ. Graphene oxide significantly inhibits cell growth at sublethal concentrations by causing extracellular iron deficiency. Nanotoxicology 2017; 11:1102-1114. [DOI: 10.1080/17435390.2017.1398357] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Qilin Yu
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, College of Life Science, Nankai University, Tianjin, China
| | - Bing Zhang
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, College of Life Science, Nankai University, Tianjin, China
| | - Jianrong Li
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, College of Life Science, Nankai University, Tianjin, China
| | - Tingting Du
- College of Environmental Science and Engineering, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, Nankai University, Tianjin, China
| | - Xiao Yi
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, College of Life Science, Nankai University, Tianjin, China
| | - Mingchun Li
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, College of Life Science, Nankai University, Tianjin, China
| | - Wei Chen
- College of Environmental Science and Engineering, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, Nankai University, Tianjin, China
| | - Pedro J. J. Alvarez
- Department of Civil and Environmental Engineering, Rice University, Houston, TX, USA
| |
Collapse
|
72
|
Santiago T, DeVaux RS, Kurzatkowska K, Espinal R, Herschkowitz JI, Hepel M. Surface-enhanced Raman scattering investigation of targeted delivery and controlled release of gemcitabine. Int J Nanomedicine 2017; 12:7763-7776. [PMID: 29123391 PMCID: PMC5661449 DOI: 10.2147/ijn.s149306] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Advanced and metastatic cancer forms are extremely difficult to treat and require high doses of chemotherapeutics, inadvertently affecting also healthy cells. As a result, the observed survival rates are very low. For instance, gemcitabine (GEM), one of the most effective chemotherapeutic drugs used for the treatment of breast and pancreatic cancers, sees only a 20% efficacy in penetrating cancer tissue, resulting in <5% survival rate in pancreatic cancer. Here, we present a method for delivering the drug that offers mitigation of side effects, as well as a targeted delivery and controlled release of the drug, improving its overall efficacy. By modifying the surface of gold nanoparticles (AuNPs) with covalently bonded thiol linkers, we have immobilized GEM on the nanoparticle (NP) through a pH-sensitive amide bond. This bond prevents the drug from being metabolized or acting on tissue at physiological pH 7.4, but breaks, releasing the drug at acidic pH, characteristic of cancer cells. Further functionalization of the NP with folic acid and/or transferrin (TF) offers a targeted delivery, as cancer cells overexpress folate and TF receptors, which can mediate the endocytosis of the NP carrying the drug. Thus, through the modification of AuNPs, we have been able to produce a nanocarrier containing GEM and folate/TF ligands, which is capable of targeted controlled-release delivery of the drug, reducing the side effects of the drug and increasing its efficacy. Here, we demonstrate the pH-dependent GEM release, using an ultrasensitive surface-enhanced Raman scattering spectroscopy to monitor the GEM loading onto the nanocarrier and follow its stimulated release. Further in vitro studies with model triple-negative breast cancer cell line MDA-MB-231 have corroborated the utility of the proposed nanocarrier method allowing the administration of high drug doses to targeted cancer cells.
Collapse
Affiliation(s)
- Ty Santiago
- Department of Chemistry, State University of New York at Potsdam, Potsdam
| | - Rebecca Sinnott DeVaux
- Department of Biomedical Sciences, Cancer Research Center, University at Albany, State University of New York, Rensselaer, NY, USA
| | | | - Ricardo Espinal
- Department of Chemistry, State University of New York at Potsdam, Potsdam
| | - Jason I Herschkowitz
- Department of Biomedical Sciences, Cancer Research Center, University at Albany, State University of New York, Rensselaer, NY, USA
| | - Maria Hepel
- Department of Chemistry, State University of New York at Potsdam, Potsdam
| |
Collapse
|
73
|
Li J, Xing X, Sun B, Zhao Y, Wu Z. Metallofullerenol Inhibits Cellular Iron Uptake by Inducing Transferrin Tetramerization. Chem Asian J 2017; 12:2646-2651. [PMID: 28815927 DOI: 10.1002/asia.201700910] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 08/16/2017] [Indexed: 12/24/2022]
Abstract
Herein, A549 tumor cell proliferation was confirmed to be positively dependent on the concentration of Fe3+ or transferrin (Tf). Gd@C82 (OH)22 or C60 (OH)22 effectively inhibited the iron uptake and the subsequent proliferation of A549 cells. The conformational changes of Tf mixed with FeCl3 , GdCl3 , C60 (OH)22 or Gd@C82 (OH)22 were obtained by SAXS. The results demonstrate that Tf homodimers can be decomposed into monomers in the presence of FeCl3 , GdCl3 or C60 (OH)22 , but associated into tetramers in the presence of Gd@C82 (OH)22 . The larger change of SAXS shapes between Tf+C60 (OH)22 and Tf+FeCl3 implies that C60 (OH)22 is bound to Tf, blocking the iron-binding site. The larger deviation of the SAXS shape from a possible crystal structure of Tf tetramer implies that Gd@C82 (OH)22 is bound to the Tf tetramer, thus disturbing iron transport. This study well explains the inhibition mechanism of Gd@C82 (OH)22 and C60 (OH)22 on the iron uptake and the proliferation of A549 tumor cells and highlights the specific interactions of a nanomedicine with the target biomolecules in cancer therapy.
Collapse
Affiliation(s)
- Jinxia Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
| | - Xueqing Xing
- Beijing Synchrotron Radiation Facility, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
| | - Baoyun Sun
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
| | - Yuliang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Zhonghua Wu
- Beijing Synchrotron Radiation Facility, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
74
|
G9a regulates breast cancer growth by modulating iron homeostasis through the repression of ferroxidase hephaestin. Nat Commun 2017; 8:274. [PMID: 28819251 PMCID: PMC5561105 DOI: 10.1038/s41467-017-00350-9] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 06/23/2017] [Indexed: 01/16/2023] Open
Abstract
G9a, a H3K9 methyltransferase, shows elevated expression in many types of human cancers, particularly breast cancer. However, the tumorigenic mechanism of G9a is still far from clear. Here we report that G9a exerts its oncogenic function in breast cancer by repressing hephaestin and destruction cellular iron homeostasis. In the case of pharmacological inhibition or short hairpin RNA interference-mediated suppression of G9a, the expression and activity of hephaestin increases, leading to the observed decrease of intracellular labile iron content and the disturbance of breast cancer cell growth in vitro and in vivo. We also provide evidence that G9a interacts with HDAC1 and YY1 to form a multi-molecular complex that contributes to hephaestin silencing. Furthermore, high G9a expression and low hephaestin expression correlate with poor survival of breast cancer are investigated. All these suggest a G9a-dependent epigenetic program in the control of iron homeostasis and tumor growth in breast cancer. G9a is a histone methyltransferase highly expressed in several cancers including breast cancer. Here the authors propose a mechanism through which G9a promotes breast cancer by regulating iron metabolism through the repression of ferroxidase hephaestin.
Collapse
|
75
|
Kazan HH, Urfali-Mamatoglu C, Gunduz U. Iron metabolism and drug resistance in cancer. Biometals 2017; 30:629-641. [DOI: 10.1007/s10534-017-0037-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 07/26/2017] [Indexed: 01/17/2023]
|
76
|
Petrović Peroković V, Car Ž, Opačak-Bernardi T, Martin-Kleiner I, Kralj M, Tomić S. In vitro antiproliferative study of novel adamantyl pyridin-4-ones. Mol Divers 2017; 21:881-891. [PMID: 28695468 DOI: 10.1007/s11030-017-9763-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 06/30/2017] [Indexed: 12/23/2022]
Abstract
The preparation of several N-aryl-substituted (phenyl, p-methylphenyl, p-methoxyphenyl, p-nitrophenyl, p-aminophenyl, p-hydroxyphenyl) 3-hydroxy-2-methylpyridin-4-ones as well as their adamantyl derivatives is described, and their in vitro antitumor properties were investigated. The compounds were synthesized in good yields using efficient synthetic routes and methods. Prepared derivatives were evaluated in an antiproliferative in vitro study on 4 cancer cell lines, namely HCT 116 (colon carcinoma), H 460 (lung carcinoma), MCF-7 (breast carcinoma) and K562 (chronic myelogenous leukemia). All tested compounds showed antiproliferative activity ranging from moderate to strong on all inspected cell lines with 4 adamantane containing derivatives being active and selective at low micromolar IC[Formula: see text] concentrations on HCT 116, H 460 and MCF-7. LDH cytotoxicity assay revealed that cytotoxic effects occur after 48 h of exposure. It was shown that there was no change in caspase activity in the treated cells, but there were changes in the cell cycle. All treated samples showed reduced number of cells in the S phase with increased G0/G1 (4b, 5a, 5b) and G2/M (4a) phase.
Collapse
Affiliation(s)
- V Petrović Peroković
- Department of Chemistry, Faculty of Science, University of Zagreb, Horvatovac 102a, 10000, Zagreb, Croatia.
| | - Ž Car
- Department of Chemistry, Faculty of Science, University of Zagreb, Horvatovac 102a, 10000, Zagreb, Croatia
| | - T Opačak-Bernardi
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, Cara Hadrijana 10/E, 31000, Osijek, Croatia
| | - I Martin-Kleiner
- Laboratory of Experimental Therapy, Division of Molecular Medicine, Ruđer Bošković Institute, Bijenička 54, 10000, Zagreb, Croatia
| | - M Kralj
- Laboratory of Experimental Therapy, Division of Molecular Medicine, Ruđer Bošković Institute, Bijenička 54, 10000, Zagreb, Croatia
| | - S Tomić
- Department of Chemistry, Faculty of Science, University of Zagreb, Horvatovac 102a, 10000, Zagreb, Croatia
| |
Collapse
|
77
|
Wang Q, Franz KJ. The hydrolytic susceptibility of prochelator BSIH in aqueous solutions. Bioorg Med Chem Lett 2017; 27:4165-4170. [PMID: 28734582 DOI: 10.1016/j.bmcl.2017.07.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 07/06/2017] [Indexed: 01/17/2023]
Abstract
The prochelator BSIH ((E)-N'-(2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzylidene)isonicotinohydrazide) contains a boronate group that prevents metal coordination until reaction with peroxide releases the iron chelator SIH ((E)-N'-(2-hydroxybenzylidene)isonicotinohydrazide). BSIH exists in aqueous buffer and cell culture media in equilibrium with its hydrolysis products isoniazid and (2-formylphenyl)boronic acid (FBA). The relative concentrations of these species limit the yield of intact SIH available for targeted iron chelation. While the hydrolysis fragments are nontoxic to retinal pigment epithelial cells, these results suggest that modifications to BSIH that improve its hydrolytic stability yet maintain its low inherent cytotoxicity are desirable for creating more efficient prochelators for protection against cellular oxidative damage.
Collapse
Affiliation(s)
- Qin Wang
- Duke University, Department of Chemistry, 124 Science Dr., Durham, NC 27708, USA
| | - Katherine J Franz
- Duke University, Department of Chemistry, 124 Science Dr., Durham, NC 27708, USA.
| |
Collapse
|
78
|
Design and drug-like properties of new 5-methoxysalicylaldehyde based hydrazones with anti-breast cancer activity. J Appl Biomed 2017. [DOI: 10.1016/j.jab.2017.04.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
|
79
|
Abstract
Lipocalin 2 (Lcn2), an innate immune protein, has emerged as a critical iron regulatory protein during physiological and inflammatory conditions. As a bacteriostatic factor, Lcn2 obstructs the siderophore iron-acquiring strategy of bacteria and thus inhibits bacterial growth. As part of host nutritional immunity, Lcn2 facilitates systemic, cellular, and mucosal hypoferremia during inflammation, in addition to stabilizing the siderophore-bound labile iron pool. In this review, we summarize recent advances in understanding the interaction between Lcn2 and iron, and its effects in various inflammatory diseases. Lcn2 exerts mostly a protective role in infectious and inflammatory bowel diseases, whereas both beneficial and detrimental functions have been documented in neurodegenerative diseases, metabolic syndrome, renal disorders, skin disorders, and cancer. Further animal and clinical studies are necessary to unveil the multifaceted roles of Lcn2 in iron dysregulation during inflammation and to explore its therapeutic potential for treating inflammatory diseases.
Collapse
Affiliation(s)
- Xia Xiao
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, Pennsylvania 16802;
| | - Beng San Yeoh
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, Pennsylvania 16802;
| | - Matam Vijay-Kumar
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, Pennsylvania 16802; .,Department of Medicine, The Pennsylvania State University Medical Center, Hershey, Pennsylvania 17033
| |
Collapse
|
80
|
Kostos L, Chai KL, Tam CS, Bazargan A. The positive effect of deferasirox on erythropoiesis in a patient with a dual diagnosis of myelofibrosis and diffuse large B-cell lymphoma. Leuk Lymphoma 2017; 58:2720-2723. [DOI: 10.1080/10428194.2017.1281414] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Louise Kostos
- Department of Haematology, St Vincent’s Hospital, Melbourne, Australia
| | - Khai Li Chai
- Department of Haematology, St Vincent’s Hospital, Melbourne, Australia
| | - Constantine S. Tam
- Department of Haematology, St Vincent’s Hospital, Melbourne, Australia
- Department of Haematology, Peter MacCallum Cancer Centre, Melbourne, Australia
- University of Melbourne, Parkville, Victoria, Australia
| | - Ali Bazargan
- Department of Haematology, St Vincent’s Hospital, Melbourne, Australia
| |
Collapse
|
81
|
Lipocalin-2 and iron trafficking in the tumor microenvironment. Pharmacol Res 2017; 120:146-156. [PMID: 28342790 DOI: 10.1016/j.phrs.2017.03.018] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 03/16/2017] [Accepted: 03/17/2017] [Indexed: 02/07/2023]
Abstract
Iron is an essential element for virtually all organisms. It facilitates cell proliferation and growth but also contributes to major hallmarks of cancer such as tumor initiation, growth, and metastasis. Often, iron handling of tumor cells is disturbed, with altered iron acquisition, efflux, and storage. Targeting perturbed iron metabolic pathways might open opportunities towards novel approaches in cancer treatment. It is becoming clear that cells of the tumor microenvironment such as macrophages contribute to tumor progression. Since macrophages evolved a multitude of mechanisms to sequester, transport, store, and release iron it can be speculated that tumor cells educate them to supply iron to support tumor growth. Recent evidence supports the existence of transferrin-independent iron transport mechanisms in the tumor microenvironment, which points to local iron transport proteins such as lipocalin-2 and/or low molecular weight iron-trafficking substances such as siderophores. We hypothesize that tumor cells educate immune cells, i.e. macrophages in their neighborhood to make them delivering iron for the benefit of cancer progression. In particular, we pay attention to recent developments, pointing to lipocalin-2 and siderophores as alternative iron transport molecules in the tumor microenvironment.
Collapse
|
82
|
Rychtarcikova Z, Lettlova S, Tomkova V, Korenkova V, Langerova L, Simonova E, Zjablovskaja P, Alberich-Jorda M, Neuzil J, Truksa J. Tumor-initiating cells of breast and prostate origin show alterations in the expression of genes related to iron metabolism. Oncotarget 2017; 8:6376-6398. [PMID: 28031527 PMCID: PMC5351639 DOI: 10.18632/oncotarget.14093] [Citation(s) in RCA: 70] [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: 01/12/2016] [Accepted: 11/30/2016] [Indexed: 12/22/2022] Open
Abstract
The importance of iron in the growth and progression of tumors has been widely documented. In this report, we show that tumor-initiating cells (TICs), represented by spheres derived from the MCF7 cell line, exhibit higher intracellular labile iron pool, mitochondrial iron accumulation and are more susceptible to iron chelation. TICs also show activation of the IRP/IRE system, leading to higher iron uptake and decrease in iron storage, suggesting that level of properly assembled cytosolic iron-sulfur clusters (FeS) is reduced. This finding is confirmed by lower enzymatic activity of aconitase and FeS cluster biogenesis enzymes, as well as lower levels of reduced glutathione, implying reduced FeS clusters synthesis/utilization in TICs. Importantly, we have identified specific gene signature related to iron metabolism consisting of genes regulating iron uptake, mitochondrial FeS cluster biogenesis and hypoxic response (ABCB10, ACO1, CYBRD1, EPAS1, GLRX5, HEPH, HFE, IREB2, QSOX1 and TFRC). Principal component analysis based on this signature is able to distinguish TICs from cancer cells in vitro and also Leukemia-initiating cells (LICs) from non-LICs in the mouse model of acute promyelocytic leukemia (APL). Majority of the described changes were also recapitulated in an alternative model represented by MCF7 cells resistant to tamoxifen (TAMR) that exhibit features of TICs. Our findings point to the critical importance of redox balance and iron metabolism-related genes and proteins in the context of cancer and TICs that could be potentially used for cancer diagnostics or therapy.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacology
- Biological Transport
- Breast Neoplasms/drug therapy
- Breast Neoplasms/enzymology
- Breast Neoplasms/genetics
- Breast Neoplasms/pathology
- Dose-Response Relationship, Drug
- Drug Resistance, Neoplasm
- Female
- Gene Expression Profiling
- Gene Expression Regulation, Enzymologic
- Gene Expression Regulation, Neoplastic
- Humans
- Iron/metabolism
- Iron Chelating Agents/pharmacology
- Leukemia, Promyelocytic, Acute/enzymology
- Leukemia, Promyelocytic, Acute/genetics
- MCF-7 Cells
- Male
- Mice, Transgenic
- Mitochondria/enzymology
- Neoplastic Stem Cells/drug effects
- Neoplastic Stem Cells/enzymology
- Neoplastic Stem Cells/pathology
- Phenotype
- Principal Component Analysis
- Prostatic Neoplasms/drug therapy
- Prostatic Neoplasms/enzymology
- Prostatic Neoplasms/genetics
- Prostatic Neoplasms/pathology
- Spheroids, Cellular
- Tamoxifen/pharmacology
- Transcriptome
Collapse
Affiliation(s)
- Zuzana Rychtarcikova
- Institute of Biotechnology, Czech Academy of Sciences, Prague, Czech Republic
- Charles University in Prague, Faculty of Pharmacy in Hradec Kralove, Hradec Kralove, Czech Republic
| | - Sandra Lettlova
- Institute of Biotechnology, Czech Academy of Sciences, Prague, Czech Republic
- Charles University in Prague, Faculty of Sciences, Prague, Czech Republic
| | - Veronika Tomkova
- Institute of Biotechnology, Czech Academy of Sciences, Prague, Czech Republic
- Charles University in Prague, Faculty of Sciences, Prague, Czech Republic
| | - Vlasta Korenkova
- Institute of Biotechnology, Czech Academy of Sciences, Prague, Czech Republic
| | - Lucie Langerova
- Institute of Biotechnology, Czech Academy of Sciences, Prague, Czech Republic
| | - Ekaterina Simonova
- Institute of Biotechnology, Czech Academy of Sciences, Prague, Czech Republic
| | - Polina Zjablovskaja
- Institute of Molecular Genetics, Czech Academy of Sciences, Prague, Czech Republic
| | | | - Jiri Neuzil
- Institute of Biotechnology, Czech Academy of Sciences, Prague, Czech Republic
- School of Medical Science, Menzies Health Institute Queensland, Southport, Queensland, Australia
| | - Jaroslav Truksa
- Institute of Biotechnology, Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
83
|
Understanding the role of the kynurenine pathway in human breast cancer immunobiology. Oncotarget 2016; 7:6506-20. [PMID: 26646699 PMCID: PMC4872729 DOI: 10.18632/oncotarget.6467] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 11/25/2015] [Indexed: 02/06/2023] Open
Abstract
Breast cancer (BrCa) is the leading cause of cancer related death in women. While current diagnostic modalities provide opportunities for early medical intervention, significant proportions of breast tumours escape treatment and metastasize. Gaining increasing recognition as a factor in tumour metastasis is the local immuno-surveillance environment. Following identification of the role played by the enzyme indoleamine dioxygenase 1 (IDO1) in mediating maternal foetal tolerance, the kynurenine pathway (KP) of tryptophan metabolism has emerged as a key metabolic pathway contributing to immune escape. In inflammatory conditions activation of the KP leads to the production of several immune-modulating metabolites including kynurenine, kynurenic acid, 3-hydroxykynurenine, anthranilic acid, 3-hydroxyanthranilic acid, picolinic acid and quinolinic acid. KP over-activation was first described in BrCa patients in the early 1960s. More evidence has since emerged to suggest that the IDO1 is elevated in advanced BrCa patients and is associated with poor prognosis. Further, IDO1 positive breast tumours have a positive correlation with the density of immune suppressive Foxp3+ T regulatory cells and lymph node metastasis. The analysis of clinical microarray data in invasive BrCa compared to normal tissue showed, using two microarray databank (cBioportal and TCGA), that 86.3% and 91.4% BrCa patients have altered KP enzyme expression respectively. Collectively, these data highlight the key roles played by KP activation in BrCa, particularly in basal BrCa subtypes where expression of most KP enzymes was altered. Accordingly, the use of KP enzyme inhibitors in addition to standard chemotherapy regimens may present a viable therapeutic approach.
Collapse
|
84
|
Intracellular Iron Chelation Modulates the Macrophage Iron Phenotype with Consequences on Tumor Progression. PLoS One 2016; 11:e0166164. [PMID: 27806101 PMCID: PMC5091876 DOI: 10.1371/journal.pone.0166164] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 10/23/2016] [Indexed: 12/23/2022] Open
Abstract
A growing body of evidence suggests that macrophage polarization dictates the expression of iron-regulated genes. Polarization towards iron sequestration depletes the microenvironment, whereby extracellular pathogen growth is limited and inflammation is fostered. In contrast, iron release contributes to cell proliferation, which is important for tissue regeneration. Moreover, macrophages constitute a major component of the infiltrates in most solid tumors. Considering the pivotal role of macrophages for iron homeostasis and their presence in association with poor clinical prognosis in tumors, we approached the possibility to target macrophages with intracellular iron chelators. Analyzing the expression of iron-regulated genes at mRNA and protein level in primary human macrophages, we found that the iron-release phenotype is a characteristic of polarized macrophages that, in turn, stimulate tumor cell growth and progression. The application of the intracellular iron chelator (TC3-S)2 shifted the macrophage phenotype from iron release towards sequestration, as determined by the iron-gene profile and atomic absorption spectroscopy (AAS). Moreover, whereas the addition of macrophage supernatants to tumor cells induced tumor growth and metastatic behavior, the supernatant of chelator-treated macrophages reversed this effect. Iron chelators demonstrated potent anti-neoplastic properties in a number of cancers, both in cell culture and in clinical trials. Our results suggest that iron chelation could affect not only cancer cells but also the tumor microenvironment by altering the iron-release phenotype of tumor-associated macrophages (TAMs). The study of iron chelators in conjunction with the effect of TAMs on tumor growth could lead to an improved understanding of the role of iron in cancer biology and to novel therapeutic avenues for iron chelation approaches.
Collapse
|
85
|
|
86
|
Todokoro T, Fukuda K, Matsumura K, Irie M, Hata Y. Production of the natural iron chelator deferriferrichrysin from Aspergillus oryzae and evaluation as a novel food-grade antioxidant. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2016; 96:2998-3006. [PMID: 26399367 DOI: 10.1002/jsfa.7469] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 08/27/2015] [Accepted: 09/18/2015] [Indexed: 06/05/2023]
Abstract
BACKGROUND Deferriferrichrysin (Dfcy) is a siderophore found in foods fermented by Aspergillus oryzae and is a promising candidate for an antioxidant food additive because of its high binding constant toward iron. However, the Dfcy concentration is typically low in foods and cultures. RESULTS We optimised culture conditions to improve Dfcy production to 2800 mg L(-1) from 22.5 mg L(-1) under typical conditions. Then, we evaluated the potential of Dfcy as a food additive by measuring its safety, stability, and antioxidant activity. Dfcy was sufficiently stable that over 90% remained after pasteurisation at 63 °C for 30 min at pH 3-11, or after sterilisation at 120 °C for 4 min at pH 4-6. Dfcy showed high antioxidant activity in an oil-in-water model, where inhibition of lipid oxidation was measured by peroxide value (PV) and thiobarbituric acid reactive substances (TBARS) assays. Dfcy decreased PV and TBARS by 83% and 75%, respectively. Antioxidant activity of Dfcy was equal to or higher than that of the synthetic chelator EDTA. CONCLUSION Our study provides the first practical method for production of Dfcy. Dfcy can be a novel food-grade antioxidant and the first natural alternative to the synthesised iron chelator EDTA. © 2015 Society of Chemical Industry.
Collapse
MESH Headings
- Animals
- Antioxidants/adverse effects
- Antioxidants/chemistry
- Antioxidants/economics
- Antioxidants/isolation & purification
- Aspergillus oryzae/chemistry
- Aspergillus oryzae/growth & development
- Aspergillus oryzae/metabolism
- Fermentation
- Food Preservatives/adverse effects
- Food Preservatives/chemistry
- Food Preservatives/economics
- Food Preservatives/isolation & purification
- Food-Processing Industry/economics
- Fungal Proteins/metabolism
- Hot Temperature/adverse effects
- Industrial Waste/analysis
- Industrial Waste/economics
- Iron Chelating Agents/adverse effects
- Iron Chelating Agents/chemistry
- Iron Chelating Agents/economics
- Iron Chelating Agents/isolation & purification
- Japan
- Models, Chemical
- Mutagenicity Tests
- Oryza/chemistry
- Peptide Hydrolases/metabolism
- Peptides, Cyclic/adverse effects
- Peptides, Cyclic/chemistry
- Peptides, Cyclic/economics
- Peptides, Cyclic/isolation & purification
- Plant Proteins, Dietary/chemistry
- Plant Proteins, Dietary/economics
- Plant Proteins, Dietary/isolation & purification
- Plant Proteins, Dietary/metabolism
- Protein Hydrolysates/chemistry
- Protein Hydrolysates/economics
- Protein Hydrolysates/isolation & purification
- Protein Hydrolysates/metabolism
- Saccharomyces cerevisiae/growth & development
- Saccharomyces cerevisiae/metabolism
- Seeds/chemistry
- Toxicity Tests, Acute
- Wine/analysis
- Wine/microbiology
Collapse
Affiliation(s)
- Takehiko Todokoro
- Research Institute, Gekkeikan Sake Co., Ltd, 101 Shimotoba-koyanagi-cho, Fushimi-ku, Kyoto, 612-8385, Japan
| | - Katsuharu Fukuda
- Research Institute, Gekkeikan Sake Co., Ltd, 101 Shimotoba-koyanagi-cho, Fushimi-ku, Kyoto, 612-8385, Japan
| | - Kengo Matsumura
- Research Institute, Gekkeikan Sake Co., Ltd, 101 Shimotoba-koyanagi-cho, Fushimi-ku, Kyoto, 612-8385, Japan
| | - Motoko Irie
- Research Institute, Gekkeikan Sake Co., Ltd, 101 Shimotoba-koyanagi-cho, Fushimi-ku, Kyoto, 612-8385, Japan
| | - Yoji Hata
- Research Institute, Gekkeikan Sake Co., Ltd, 101 Shimotoba-koyanagi-cho, Fushimi-ku, Kyoto, 612-8385, Japan
| |
Collapse
|
87
|
Zubair H, Azim S, Khan HY, Ullah MF, Wu D, Singh AP, Hadi SM, Ahmad A. Mobilization of Intracellular Copper by Gossypol and Apogossypolone Leads to Reactive Oxygen Species-Mediated Cell Death: Putative Anticancer Mechanism. Int J Mol Sci 2016; 17:ijms17060973. [PMID: 27331811 PMCID: PMC4926505 DOI: 10.3390/ijms17060973] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 06/07/2016] [Accepted: 06/14/2016] [Indexed: 12/25/2022] Open
Abstract
There is compelling evidence that serum, tissue and intracellular levels of copper are elevated in all types of cancer. Copper has been suggested as an important co-factor for angiogenesis. It is also a major metal ion present inside the nucleus, bound to DNA bases, particularly guanine. We have earlier proposed that the interaction of phenolic-antioxidants with intracellular copper leads to the generation of reactive oxygen species (ROS) that ultimately serve as DNA cleaving agents. To further validate our hypothesis we show here that the antioxidant gossypol and its semi-synthetic derivative apogossypolone induce copper-mediated apoptosis in breast MDA-MB-231, prostate PC3 and pancreatic BxPC-3 cancer cells, through the generation of ROS. MCF10A breast epithelial cells refractory to the cytotoxic property of these compounds become sensitized to treatment against gossypol, as well as apogossypolone, when pre-incubated with copper. Our present results confirm our earlier findings and strengthen our hypothesis that plant-derived antioxidants mobilize intracellular copper instigating ROS-mediated cellular DNA breakage. As cancer cells exist under significant oxidative stress, this increase in ROS-stress to cytotoxic levels could be a successful anticancer approach.
Collapse
Affiliation(s)
- Haseeb Zubair
- Department of Biochemistry, Faculty of Life Sciences, AMU, Aligarh 202002, India.
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA.
| | - Shafquat Azim
- Department of Biochemistry, Faculty of Life Sciences, AMU, Aligarh 202002, India.
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA.
| | - Husain Yar Khan
- Department of Biochemistry, Faculty of Life Sciences, AMU, Aligarh 202002, India.
- UoN Chair of Oman's Medicinal Plants and Marine Natural Products, University of Nizwa, Birkat Al Mauz, PO Box 33, 616 Nizwa, Oman.
| | - Mohammad Fahad Ullah
- Department of Biochemistry, Faculty of Life Sciences, AMU, Aligarh 202002, India.
- Prince Fahad Research Chair, Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, University of Tabuk, P.O. Box 741, Tabuk-71491, Saudi Arabia.
| | - Daocheng Wu
- Key Laboratory of Biomedical Information Engineering of Education Ministry, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710048, China.
| | - Ajay Pratap Singh
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA.
| | - Sheikh Mumtaz Hadi
- Department of Biochemistry, Faculty of Life Sciences, AMU, Aligarh 202002, India.
| | - Aamir Ahmad
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA.
| |
Collapse
|
88
|
Substituted N-aminothioglycolurils containing thiosemicarbazone moiety and their cytotoxic activity in vitro. Mol Divers 2016; 20:837-846. [PMID: 27216444 DOI: 10.1007/s11030-016-9671-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 04/24/2016] [Indexed: 01/22/2023]
Abstract
A library of hybrid molecules bearing thioglycoluril and (hetero)aromatic aldehyde thiosemicarbazone moieties was synthesized via a tandem hydrazone formation-ring contraction reaction of 5,7-dialkyl-3-thioxoperhydroimidazo[4,5-e]-1,2,4-triazin-6-ones with (hetero)aromatic aldehydes. All synthesized compounds were tested for their cytotoxic activity against rhabdomyosarcoma, A549, and MS human cancer cell lines by MTT-assay. Among the derivatives, (E)-4-benzylideneamino-1,3-dimethyl-5-thioxohexahydroimidazo[4,5-d]imidazol-2(1H)-one 1f was found to have the most marked antiproliferative activity toward the tested cell lines (1f: IC[Formula: see text] 23.7, and 6.4 [Formula: see text]M, respectively). The IC[Formula: see text] value of thioglycoluril 1f against normal human embryonic kidney cells HEK293 was 72.5 [Formula: see text]M, which appeared to be 3-11-fold higher than IC[Formula: see text] values of 1f against human cancer cells.
Collapse
|
89
|
Roztocki K, Senkovska I, Kaskel S, Matoga D. Carboxylate-Hydrazone Mixed-Linker Metal-Organic Frameworks: Synthesis, Structure, and Selective Gas Adsorption. Eur J Inorg Chem 2016. [DOI: 10.1002/ejic.201600134] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Kornel Roztocki
- Faculty of Chemistry; Jagiellonian University; Ingardena 3 30-060 Kraków Poland
| | - Irena Senkovska
- Department of Inorganic Chemistry; Technische Universität Dresden; Bergstrasse 66 01062 Dresden Germany
| | - Stefan Kaskel
- Department of Inorganic Chemistry; Technische Universität Dresden; Bergstrasse 66 01062 Dresden Germany
| | - Dariusz Matoga
- Faculty of Chemistry; Jagiellonian University; Ingardena 3 30-060 Kraków Poland
| |
Collapse
|
90
|
Jansová H, Bureš J, Macháček M, Hašková P, Jirkovská A, Roh J, Wang Q, Franz KJ, Kovaříková P, Šimůnek T. Characterization of cytoprotective and toxic properties of iron chelator SIH, prochelator BSIH and their degradation products. Toxicology 2016; 350-352:15-24. [PMID: 27046792 DOI: 10.1016/j.tox.2016.03.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 03/24/2016] [Accepted: 03/31/2016] [Indexed: 02/01/2023]
Abstract
Free cellular iron catalyzes the formation of toxic hydroxyl radicals and therefore chelation of iron could be a promising therapeutic approach in pathological states associated with oxidative stress. Salicylaldehyde isonicotinoyl hydrazone (SIH) is a strong intracellular iron chelator with well documented potential to protect against oxidative damage both in vitro and in vivo. Due to the short biological half-life of SIH and risk of toxicity due to iron depletion, boronate prochelator BSIH has been designed. BSIH cannot bind iron until it is activated by certain reactive oxygen species to active chelator SIH. The aim of this study was to examine the toxicity and cytoprotective potential of BSIH, SIH, and their decomposition products against hydrogen peroxide-induced injury of H9c2 cardiomyoblast cells. Using HPLC, we observed that salicylaldehyde was the main decomposition products of SIH and BSIH, although a small amount of salicylic acid was also detected. In the case of BSIH, the concentration of formed salicylaldehyde consistently exceeded that of SIH. Isoniazid and salicylic acid were not toxic nor did they provide any antioxidant protective effect in H9c2 cells. In contrast, salicylaldehyde was able to chelate intracellular iron and significantly preserve cellular viability and mitochondrial inner membrane potential induced by hydrogen peroxide. However it was consistently less effective than SIH. The inherent toxicities of salicylaldehyde and SIH were similar. Hence, although SIH - the active chelating agent formed following the BSIH activation - undergoes rapid hydrolysis, its principal decomposition product salicylaldehyde accounts markedly for both cytoprotective and toxic properties.
Collapse
Affiliation(s)
- Hana Jansová
- Faculty of Pharmacy, Charles University in Prague, 500 05 Hradec Králové, Czech Republic
| | - Jan Bureš
- Faculty of Pharmacy, Charles University in Prague, 500 05 Hradec Králové, Czech Republic
| | - Miloslav Macháček
- Faculty of Pharmacy, Charles University in Prague, 500 05 Hradec Králové, Czech Republic
| | - Pavlína Hašková
- Faculty of Pharmacy, Charles University in Prague, 500 05 Hradec Králové, Czech Republic
| | - Anna Jirkovská
- Faculty of Pharmacy, Charles University in Prague, 500 05 Hradec Králové, Czech Republic
| | - Jaroslav Roh
- Faculty of Pharmacy, Charles University in Prague, 500 05 Hradec Králové, Czech Republic
| | - Qin Wang
- Department of Chemistry, Duke University, Durham, NC 22708, USA
| | | | - Petra Kovaříková
- Faculty of Pharmacy, Charles University in Prague, 500 05 Hradec Králové, Czech Republic
| | - Tomáš Šimůnek
- Faculty of Pharmacy, Charles University in Prague, 500 05 Hradec Králové, Czech Republic.
| |
Collapse
|
91
|
Sanvisens N, Romero AM, Zhang C, Wu X, An X, Huang M, Puig S. Yeast Dun1 Kinase Regulates Ribonucleotide Reductase Small Subunit Localization in Response to Iron Deficiency. J Biol Chem 2016; 291:9807-17. [PMID: 26970775 DOI: 10.1074/jbc.m116.720862] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Indexed: 12/25/2022] Open
Abstract
Ribonucleotide reductase (RNR) is an essential iron-dependent enzyme that catalyzes deoxyribonucleotide synthesis in eukaryotes. Living organisms have developed multiple strategies to tightly modulate RNR function to avoid inadequate or unbalanced deoxyribonucleotide pools that cause DNA damage and genome instability. Yeast cells activate RNR in response to genotoxic stress and iron deficiency by facilitating redistribution of its small heterodimeric subunit Rnr2-Rnr4 from the nucleus to the cytoplasm, where it forms an active holoenzyme with large Rnr1 subunit. Dif1 protein inhibits RNR by promoting nuclear import of Rnr2-Rnr4. Upon DNA damage, Dif1 phosphorylation by the Dun1 checkpoint kinase and its subsequent degradation enhances RNR function. In this report, we demonstrate that Dun1 kinase triggers Rnr2-Rnr4 redistribution to the cytoplasm in response to iron deficiency. We show that Rnr2-Rnr4 relocalization by low iron requires Dun1 kinase activity and phosphorylation site Thr-380 in the Dun1 activation loop, but not the Dun1 forkhead-associated domain. By using different Dif1 mutant proteins, we uncover that Dun1 phosphorylates Dif1 Ser-104 and Thr-105 residues upon iron scarcity. We observe that the Dif1 phosphorylation pattern differs depending on the stimuli, which suggests different Dun1 activating pathways. Importantly, the Dif1-S104A/T105A mutant exhibits defects in nucleus-to-cytoplasm redistribution of Rnr2-Rnr4 by iron limitation. Taken together, these results reveal that, in response to iron starvation, Dun1 kinase phosphorylates Dif1 to stimulate Rnr2-Rnr4 relocalization to the cytoplasm and promote RNR function.
Collapse
Affiliation(s)
- Nerea Sanvisens
- From the Departamento de Biotecnología, Instituto de Agroquímica y Tecnología de Alimentos (IATA), Consejo Superior de Investigaciones Científicas (CSIC), Paterna, Valencia 46980, Spain and
| | - Antonia M Romero
- From the Departamento de Biotecnología, Instituto de Agroquímica y Tecnología de Alimentos (IATA), Consejo Superior de Investigaciones Científicas (CSIC), Paterna, Valencia 46980, Spain and
| | - Caiguo Zhang
- the Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Xiaorong Wu
- the Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Xiuxiang An
- the Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Mingxia Huang
- the Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Sergi Puig
- From the Departamento de Biotecnología, Instituto de Agroquímica y Tecnología de Alimentos (IATA), Consejo Superior de Investigaciones Científicas (CSIC), Paterna, Valencia 46980, Spain and
| |
Collapse
|
92
|
Marques O, Porto G, Rêma A, Faria F, Cruz Paula A, Gomez-Lazaro M, Silva P, Martins da Silva B, Lopes C. Local iron homeostasis in the breast ductal carcinoma microenvironment. BMC Cancer 2016; 16:187. [PMID: 26944411 PMCID: PMC4779214 DOI: 10.1186/s12885-016-2228-y] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 02/29/2016] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND While the deregulation of iron homeostasis in breast epithelial cells is acknowledged, iron-related alterations in stromal inflammatory cells from the tumor microenvironment have not been explored. METHODS Immunohistochemistry for hepcidin, ferroportin 1 (FPN1), transferrin receptor 1 (TFR1) and ferritin (FT) was performed in primary breast tissues and axillary lymph nodes in order to dissect the iron-profiles of epithelial cells, lymphocytes and macrophages. Furthermore, breast carcinoma core biopsies frozen in optimum cutting temperature (OCT) compound were subjected to imaging flow cytometry to confirm FPN1 expression in the cell types previously evaluated and determine its cellular localization. RESULTS We confirm previous results by showing that breast cancer epithelial cells present an 'iron-utilization phenotype' with an increased expression of hepcidin and TFR1, and decreased expression of FT. On the other hand, lymphocytes and macrophages infiltrating primary tumors and from metastized lymph nodes display an 'iron-donor' phenotype, with increased expression of FPN1 and FT, concomitant with an activation profile reflected by a higher expression of TFR1 and hepcidin. A higher percentage of breast carcinomas, compared to control mastectomy samples, present iron accumulation in stromal inflammatory cells, suggesting that these cells may constitute an effective tissue iron reservoir. Additionally, not only the deregulated expression of iron-related proteins in epithelial cells, but also on lymphocytes and macrophages, are associated with clinicopathological markers of breast cancer poor prognosis, such as negative hormone receptor status and tumor size. CONCLUSIONS The present results reinforce the importance of analyzing the tumor microenvironment in breast cancer, extending the contribution of immune cells to local iron homeostasis in the tumor microenvironment context.
Collapse
Affiliation(s)
- Oriana Marques
- Laboratory of Immunogenetics - Autoimmunity and Neurosciences, Unit for Multidisciplinary Biomedical Research (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Rua Jorge Viterbo Ferreira 228,Edif 2 Piso 4, P-4050313, Porto, Portugal. .,Pathology and Molecular Immunology Department, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal. .,Basic and Clinical Research on Iron Biology, Instituto de Biologia Molecular e Celular (IBMC), University of Porto, Porto, Portugal. .,Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal.
| | - Graça Porto
- Pathology and Molecular Immunology Department, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal. .,Hematology Service, Hospital de Santo António, Centro Hospitalar do Porto, Porto, Portugal.
| | - Alexandra Rêma
- Pathology and Molecular Immunology Department, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal.
| | - Fátima Faria
- Pathology and Molecular Immunology Department, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal.
| | - Arnaud Cruz Paula
- Pathology and Molecular Immunology Department, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal. .,Department of Pathology, Portuguese Oncology Institute (IPO), Porto, Portugal.
| | - Maria Gomez-Lazaro
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal. .,Instituto Nacional de Engenharia Biomédica (INEB), University of Porto, Porto, Portugal.
| | - Paula Silva
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal. .,Faculty of Medicine of University of Porto (FMUP), Porto, Portugal. .,Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal.
| | - Berta Martins da Silva
- Laboratory of Immunogenetics - Autoimmunity and Neurosciences, Unit for Multidisciplinary Biomedical Research (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Rua Jorge Viterbo Ferreira 228,Edif 2 Piso 4, P-4050313, Porto, Portugal. .,Pathology and Molecular Immunology Department, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal.
| | - Carlos Lopes
- Pathology and Molecular Immunology Department, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal. .,Department of Pathology, Portuguese Oncology Institute (IPO), Porto, Portugal.
| |
Collapse
|
93
|
Gautam R, Akam EA, Astashkin AV, Loughrey JJ, Tomat E. Sirtuin inhibitor sirtinol is an intracellular iron chelator. Chem Commun (Camb) 2016; 51:5104-7. [PMID: 25715179 DOI: 10.1039/c5cc00829h] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Sirtinol is a known inhibitor of sirtuin proteins, a family of deacetylases involved in the pathophysiology of aging. Spectroscopic and structural data reveal that this compound is also an iron chelator forming high-spin ferric species in vitro and in cultured leukemia cells. Interactions with the highly regulated iron pool therefore contribute to its overall intracellular agenda.
Collapse
Affiliation(s)
- R Gautam
- University of Arizona, Department of Chemistry and Biochemistry, 1306 E. University Blvd., Tucson AZ 85721, USA.
| | | | | | | | | |
Collapse
|
94
|
Jeong SM, Hwang S, Seong RH. Transferrin receptor regulates pancreatic cancer growth by modulating mitochondrial respiration and ROS generation. Biochem Biophys Res Commun 2016; 471:373-9. [PMID: 26869514 DOI: 10.1016/j.bbrc.2016.02.023] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 02/06/2016] [Indexed: 12/13/2022]
Abstract
The transferrin receptor (TfR1) is upregulated in malignant cells and its expression is associated with cancer progression. Because of its pre-eminent role in cell proliferation, TfR1 has been an important target for the development of cancer therapy. Although TfR1 is highly expressed in pancreatic cancers, what it carries out in these refractory cancers remains poorly understood. Here we report that TfR1 supports mitochondrial respiration and ROS production in human pancreatic ductal adenocarcinoma (PDAC) cells, which is required for their tumorigenic growth. Elevated TfR1 expression in PDAC cells contributes to oxidative phosphorylation, which allows for the generation of ROS. Importantly, mitochondrial-derived ROS are essential for PDAC growth. However, exogenous iron supplement cannot rescue the defects caused by TfR1 knockdown. Moreover, we found that TfR1 expression determines PDAC cells sensitivity to oxidative stress. Together, our findings reveal that TfR1 can contribute to the mitochondrial respiration and ROS production, which have essential roles in growth and survival of pancreatic cancer.
Collapse
Affiliation(s)
- Seung Min Jeong
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea; Institute for Aging and Metabolic Diseases, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea.
| | - Sunsook Hwang
- School of Biological Sciences and Institute of Molecular Biology and Genetics, Seoul National University, Seoul 151-742, South Korea
| | - Rho Hyun Seong
- School of Biological Sciences and Institute of Molecular Biology and Genetics, Seoul National University, Seoul 151-742, South Korea
| |
Collapse
|
95
|
Ultra high pressure homogenization of almond milk: Physico-chemical and physiological effects. Food Chem 2016; 192:82-9. [DOI: 10.1016/j.foodchem.2015.06.063] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 05/06/2015] [Accepted: 06/20/2015] [Indexed: 01/19/2023]
|
96
|
Dumoga S, Dey N, Kaur A, Singh S, Mishra AK, Kakkar D. Novel biotin-functionalized lipidic nanocarriers for encapsulating BpT and Bp4eT iron chelators: evaluation of potential anti-tumour efficacy by in vitro, in vivo and pharmacokinetic studies in A549 mice models. RSC Adv 2016. [DOI: 10.1039/c6ra03079c] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
This work proposes a novel strategy for delivery of iron chelators to the tumour cells which is exemplified in A549 mice models by using lipidic nanocarriers and introducing biotin based targeting.
Collapse
Affiliation(s)
- Shweta Dumoga
- Institute of Nuclear Medicine and Allied Sciences
- Timarpur
- Delhi-110054
- Department of Chemistry
- University of Delhi
| | - Namit Dey
- Institute of Nuclear Medicine and Allied Sciences
- Timarpur
- Delhi-110054
| | - Anivind Kaur
- Institute of Nuclear Medicine and Allied Sciences
- Timarpur
- Delhi-110054
| | | | - Anil K. Mishra
- Institute of Nuclear Medicine and Allied Sciences
- Timarpur
- Delhi-110054
| | - Dipti Kakkar
- Institute of Nuclear Medicine and Allied Sciences
- Timarpur
- Delhi-110054
| |
Collapse
|
97
|
Wani WA, Baig U, Shreaz S, Shiekh RA, Iqbal PF, Jameel E, Ahmad A, Mohd-Setapar SH, Mushtaque M, Ting Hun L. Recent advances in iron complexes as potential anticancer agents. NEW J CHEM 2016. [DOI: 10.1039/c5nj01449b] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The iron complexes discussed in this review highlight their promising future as anticancer agents.
Collapse
Affiliation(s)
- Waseem A. Wani
- Institute of Bioproduct Development
- Universiti Teknologi Malaysia
- 81310 UTM Skudai
- Malaysia
| | - Umair Baig
- Center of Excellence for Scientific Research Collaboration with MIT
- King Fahd University of Petroleum and Minerals
- Dhahran 31261
- Saudi Arabia
| | - Sheikh Shreaz
- Oral Microbiology Laboratory
- Department of Bioclinical Sciences
- Faculty of Dentistry
- Health Sciences Center
- Kuwait University
| | - Rayees Ahmad Shiekh
- Department of Chemistry
- Faculty of Science
- Taibah University
- Al Madinah Al Munawarrah
- Saudi Arabia
| | | | - Ehtesham Jameel
- Department of Chemistry
- B. R. Ambedkar Bihar University
- Muzaffarpur
- India
| | - Akil Ahmad
- Center of Lipids Engineering and Applied Research
- Ibnu Sina Institute for Industrial and Scientific Research
- Universiti Teknologi Malaysia
- 81310 UTM Skudai
- Malaysia
| | - Siti Hamidah Mohd-Setapar
- Center of Lipids Engineering and Applied Research
- Ibnu Sina Institute for Industrial and Scientific Research
- Universiti Teknologi Malaysia
- 81310 UTM Skudai
- Malaysia
| | - Md. Mushtaque
- Department of Physical and Molecular Sciences (Chemistry)
- Al-Falah University
- Faridabad
- India
| | - Lee Ting Hun
- Institute of Bioproduct Development
- Universiti Teknologi Malaysia
- 81310 UTM Skudai
- Malaysia
| |
Collapse
|
98
|
Corcé V, Gouin SG, Renaud S, Gaboriau F, Deniaud D. Recent advances in cancer treatment by iron chelators. Bioorg Med Chem Lett 2015; 26:251-256. [PMID: 26684852 DOI: 10.1016/j.bmcl.2015.11.094] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 11/12/2015] [Accepted: 11/25/2015] [Indexed: 01/19/2023]
Abstract
The development of new therapeutic alternatives for cancers is a major public health priority. Among the more promising approaches, the iron depletion strategy based on metal chelation in the tumoral environment has been particularly studied in recent decades. After a short description of the importance of iron for cancer cell proliferation, we will review the different iron chelators developed as potential chemotherapeutics. Finally, the recent efforts to vectorize the chelating agents specifically in the microtumoral environment will be discussed in detail.
Collapse
Affiliation(s)
- Vincent Corcé
- LUNAM Université, CEISAM, Chimie Et Interdisciplinarité, Synthèse, Analyse, Modélisation, UMR CNRS 6230, UFR des Sciences et des Techniques, 2, rue de la Houssinière, BP 92208, 44322 Nantes Cedex 3, France
| | - Sébastien G Gouin
- LUNAM Université, CEISAM, Chimie Et Interdisciplinarité, Synthèse, Analyse, Modélisation, UMR CNRS 6230, UFR des Sciences et des Techniques, 2, rue de la Houssinière, BP 92208, 44322 Nantes Cedex 3, France
| | - Stéphanie Renaud
- INSERM, UMR 991, CHRU Pontchaillou, 35033 Rennes, France; Université de Rennes 1, 35043 Rennes, France
| | - François Gaboriau
- INSERM, UMR 991, CHRU Pontchaillou, 35033 Rennes, France; Université de Rennes 1, 35043 Rennes, France
| | - David Deniaud
- LUNAM Université, CEISAM, Chimie Et Interdisciplinarité, Synthèse, Analyse, Modélisation, UMR CNRS 6230, UFR des Sciences et des Techniques, 2, rue de la Houssinière, BP 92208, 44322 Nantes Cedex 3, France
| |
Collapse
|
99
|
Hafizi M, Hajarizadeh A, Atashi A, Kalanaky S, Fakharzadeh S, Masoumi Z, Nazaran MH, Soleimani M. Nanochelating based nanocomplex, GFc7, improves quality and quantity of human mesenchymal stem cells during in vitro expansion. Stem Cell Res Ther 2015; 6:226. [PMID: 26597909 PMCID: PMC4657224 DOI: 10.1186/s13287-015-0216-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2015] [Revised: 04/30/2015] [Accepted: 10/28/2015] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION Human mesenchymal stem cells (hMSCs) have been approved for therapeutic applications. Despite the advances in this field, in vitro approaches are still required to improve the essential indices that would pave the way to a bright horizon for an efficient transplantation in the future. Nanotechnology could help to improve these approaches. Studies signified the important role of iron in stem cell metabolism and efficiency of copper chelation application for stem cell expansion METHODS For the first time, based on novel Nanochelating technology, we design an iron containing copper chelator nano complex, GFc7 and examined on hMSCs during in vitro expansion. In this study, the hMSCs were isolated, characterized and expanded in vitro in two media (with or without GFc7). Then proliferation, cell viability, cell cycle analysis, surface markers, HLADR, pluripotency genes expression, homing and antioxidative defense at genes and protein expression were investigated. Also we analyzed the spontaneous differentiation and examined osteogenic and lipogenic differentiation. RESULTS GFc7 affected the expression of key genes, improving both the stemness and fitness of the cells in a precise and balanced manner. We observed significant increases in cell proliferation, enhanced expression of pluripotency genes and homing markers, improved antioxidative defense, repression of genes involved in spontaneous differentiation and exposing the hMSCs to differentiation medium indicated that pretreatment with GFc7 increased the quality and rate of differentiation. CONCLUSIONS Thus, GFc7 appears to be a potential new supplement for cell culture medium for increasing the efficiency of transplantation.
Collapse
Affiliation(s)
- Maryam Hafizi
- Stem Cell Technology Research Center, Tehran, Iran.
- Department of Research and Development, Sodour Ahrar Shargh Company, Tehran, Iran.
| | | | - Amir Atashi
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Somayeh Kalanaky
- Department of Research and Development, Sodour Ahrar Shargh Company, Tehran, Iran.
| | - Saideh Fakharzadeh
- Department of Research and Development, Sodour Ahrar Shargh Company, Tehran, Iran.
| | | | | | - Masoud Soleimani
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
100
|
Pelivan K, Miklos W, van Schoonhoven S, Koellensperger G, Gille L, Berger W, Heffeter P, Kowol CR, Keppler BK. Differences in protein binding and excretion of Triapine and its Fe(III) complex. J Inorg Biochem 2015; 160:61-9. [PMID: 26507768 DOI: 10.1016/j.jinorgbio.2015.10.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 09/02/2015] [Accepted: 10/05/2015] [Indexed: 11/26/2022]
Abstract
Triapine has been investigated as anticancer drug in multiple clinical phase I/II trials. Although promising anti-leukemic activity was observed, Triapine was ineffective against solid tumors. The reasons are currently widely unknown. The biological activity of Triapine is strongly connected to its iron complex (Fe-Triapine) which is pharmacologically not investigated. Here, novel analytical tools for Triapine and Fe-Triapine were developed and applied for cell extracts and body fluids of treated mice. Triapine and its iron complex showed a completely different behavior: for Triapine, low protein binding was observed in contrast to fast protein adduct formation of Fe-Triapine. Notably, both drugs were rapidly cleared from the body (serum half-life time <1h). Remarkably, in contrast to Triapine, where (in accordance to clinical data) basically no renal excretion was found, the iron complex was effectively excreted via urine. Moreover, no Fe-Triapine was detected in serum or cytosolic extracts after Triapine treatment. Taken together, our study will help to further understand the biological behavior of Triapine and its Fe-complex and allow the development of novel thiosemicarbazones with pronounced activity against solid tumor types.
Collapse
Affiliation(s)
- Karla Pelivan
- Institute of Inorganic Chemistry, University of Vienna, Waehringer Strasse 42, 1090 Vienna, Austria
| | - Walter Miklos
- Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria
| | - Sushilla van Schoonhoven
- Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria
| | - Gunda Koellensperger
- Institute of Analytical Chemistry, University of Vienna, Waehringer Strasse 38, 1090 Vienna, Austria
| | - Lars Gille
- Institute of Pharmacology and Toxicology, Department of Biomedical Sciences, University of Veterinary Medicine Vienna, Veterinaerplatz 1, 1210 Vienna, Austria
| | - Walter Berger
- Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria; Research Platform "Translational Cancer Therapy Research", University of Vienna and Medical University of Vienna, Vienna, Austria
| | - Petra Heffeter
- Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria; Research Platform "Translational Cancer Therapy Research", University of Vienna and Medical University of Vienna, Vienna, Austria.
| | - Christian R Kowol
- Institute of Inorganic Chemistry, University of Vienna, Waehringer Strasse 42, 1090 Vienna, Austria; Research Platform "Translational Cancer Therapy Research", University of Vienna and Medical University of Vienna, Vienna, Austria.
| | - Bernhard K Keppler
- Institute of Inorganic Chemistry, University of Vienna, Waehringer Strasse 42, 1090 Vienna, Austria; Research Platform "Translational Cancer Therapy Research", University of Vienna and Medical University of Vienna, Vienna, Austria
| |
Collapse
|