101
|
Gasparetto M, Pei S, Minhajuddin M, Khan N, Pollyea DA, Myers JR, Ashton JM, Becker MW, Vasiliou V, Humphries KR, Jordan CT, Smith CA. Targeted therapy for a subset of acute myeloid leukemias that lack expression of aldehyde dehydrogenase 1A1. Haematologica 2017; 102:1054-1065. [PMID: 28280079 PMCID: PMC5451337 DOI: 10.3324/haematol.2016.159053] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 03/08/2017] [Indexed: 12/20/2022] Open
Abstract
Aldehyde dehydrogenase 1A1 (ALDH1A1) activity is high in hematopoietic stem cells and functions in part to protect stem cells from reactive aldehydes and other toxic compounds. In contrast, we found that approximately 25% of all acute myeloid leukemias expressed low or undetectable levels of ALDH1A1 and that this ALDH1A1− subset of leukemias correlates with good prognosis cytogenetics. ALDH1A1− cell lines as well as primary leukemia cells were found to be sensitive to treatment with compounds that directly and indirectly generate toxic ALDH substrates including 4-hydroxynonenal and the clinically relevant compounds arsenic trioxide and 4-hydroperoxycyclophosphamide. In contrast, normal hematopoietic stem cells were relatively resistant to these compounds. Using a murine xenotransplant model to emulate a clinical treatment strategy, established ALDH1A1− leukemias were also sensitive to in vivo treatment with cyclophosphamide combined with arsenic trioxide. These results demonstrate that targeting ALDH1A1− leukemic cells with toxic ALDH1A1 substrates such as arsenic and cyclophosphamide may be a novel targeted therapeutic strategy for this subset of acute myeloid leukemias.
Collapse
Affiliation(s)
| | - Shanshan Pei
- Division of Hematology, University of Colorado, Aurora, CO, USA
| | | | - Nabilah Khan
- Division of Hematology, University of Colorado, Aurora, CO, USA
| | | | - Jason R Myers
- Genomics Research Center, University of Rochester, NY, USA
| | - John M Ashton
- Genomics Research Center, University of Rochester, NY, USA
| | - Michael W Becker
- Department of Medicine, University of Rochester Medical Center, NY, USA
| | - Vasilis Vasiliou
- Department of Environmental Health Sciences, Yale University, New Haven, CT, USA
| | - Keith R Humphries
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, BC, Canada
| | - Craig T Jordan
- Division of Hematology, University of Colorado, Aurora, CO, USA
| | - Clayton A Smith
- Division of Hematology, University of Colorado, Aurora, CO, USA
| |
Collapse
|
102
|
Inhibition of IRE1α-driven pro-survival pathways is a promising therapeutic application in acute myeloid leukemia. Oncotarget 2017; 7:18736-49. [PMID: 26934650 PMCID: PMC4951325 DOI: 10.18632/oncotarget.7702] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 01/29/2016] [Indexed: 01/07/2023] Open
Abstract
Survival of cancer cells relies on the unfolded protein response (UPR) to resist stress triggered by the accumulation of misfolded proteins within the endoplasmic reticulum (ER). The IRE1α-XBP1 pathway, a key branch of the UPR, is activated in many cancers. Here, we show that the expression of both mature and spliced forms of XBP1 (XBP1s) is up-regulated in acute myeloid leukemia (AML) cell lines and AML patient samples. IRE1α RNase inhibitors [MKC-3946, 2-hydroxy-1-naphthaldehyde (HNA), STF-083010 and toyocamycin] blocked XBP1 mRNA splicing and exhibited cytotoxicity against AML cells. IRE1α inhibition induced caspase-dependent apoptosis and G1 cell cycle arrest at least partially by regulation of Bcl-2 family proteins, G1 phase controlling proteins (p21cip1, p27kip1 and cyclin D1), as well as chaperone proteins. Xbp1 deleted murine bone marrow cells were resistant to growth inhibition by IRE1α inhibitors. Combination of HNA with either bortezomib or AS2O3 was synergistic in AML cytotoxicity associated with induction of p-JNK and reduction of p-PI3K and p-MAPK. Inhibition of IRE1α RNase activity increased expression of many miRs in AML cells including miR-34a. Inhibition of miR-34a conferred cellular resistance to HNA. Our results strongly suggest that targeting IRE1α driven pro-survival pathways represent an exciting therapeutic approach for the treatment of AML.
Collapse
|
103
|
Peluso I, Palmery M, Vitalone A. Green Tea and Bone Marrow Transplantation: From Antioxidant Activity to Enzymatic and Multidrug-resistance Modulation. Crit Rev Food Sci Nutr 2017; 56:2251-60. [PMID: 26047551 DOI: 10.1080/10408398.2013.826175] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Epigallocatechin-3-gallate (EGCG), the main flavonoid of green tea (GT), could play an active role in the prevention of oxidative-stress-related diseases, such as hematologic malignancies. Some effects of EGCG are not imputable to antioxidant activity, but involve modulation of antioxidant enzymes and uric acid (UA) levels. The latter is the major factor responsible of the plasma non-enzymatic antioxidant capacity (NEAC). However, hyperuricemia is a frequent clinical feature caused by tumor lysis syndrome or cyclosporine side effects, both before and after bone marrow transplantation (BMT). Besides this, food-drug interactions could be associated with GT consumption and could have clinical implications. The molecular mechanisms involved in the redox and drug metabolizing/transporting pathways were discussed, with particular reference to the potential role of GT and EGCG in BMT. Moreover, on reviewing data on NEAC, isoprostanes, uric acid, and various enzymes from human studies on GT, its extract, or EGCG, an increase in NEAC, without effect on isoprostanes, and contrasting results on UA and enzymes were observed. Currently, few and contrasting available evidences suggest caution for GT consumption in BMT patients and more studies are needed to better understand the potential impact of EGCG on oxidative stress and metabolizing/transporting systems.
Collapse
Affiliation(s)
- Ilaria Peluso
- a Department of Physiology and Pharmacology "V. Erspamer" , "Sapienza" University of Rome , Rome , Italy
| | - Maura Palmery
- a Department of Physiology and Pharmacology "V. Erspamer" , "Sapienza" University of Rome , Rome , Italy
| | - Annabella Vitalone
- a Department of Physiology and Pharmacology "V. Erspamer" , "Sapienza" University of Rome , Rome , Italy
| |
Collapse
|
104
|
Rebechi MT, Pratz KW. Genomic instability is a principle pathologic feature of FLT3 ITD kinase activity in acute myeloid leukemia leading to clonal evolution and disease progression. Leuk Lymphoma 2017; 58:1-11. [PMID: 28278729 DOI: 10.1080/10428194.2017.1283031] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Acute Myeloid Leukemia with FLT3 ITD mutations are associated with a poor prognosis characterized by a higher relapse rate, shorter relapse free survival, and decreased likelihood of response to therapy at relapse. FLT3 ITD signaling drives cell proliferation and survival. FLT3 ITD AML disease progression is associated with cytogenetic evolution and acquired tyrosine kinase inhibitor (TKI) resistance suggesting a potential role of genomic instability. There is growing evidence demonstrating a relationship between FLT3 signaling and increased DNA damage, specifically through increased reactive oxygen species (ROS) resulting in double-strand breaks (DSB), as well as impaired DNA repair, involving deficiencies in the non-homologous end joining (NHEJ), alternative non-homologous end joining (ALT NHEJ) and homologous recombination (HR) pathways. The role of genomic instability in the pathogenesis of FLT3 ITD AML warrants further examination as it offers potential therapeutic targets.
Collapse
Affiliation(s)
- Melanie T Rebechi
- a Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University , Baltimore , MD , USA
| | - Keith W Pratz
- a Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University , Baltimore , MD , USA
| |
Collapse
|
105
|
Dwivedi P, Greis KD. Granulocyte colony-stimulating factor receptor signaling in severe congenital neutropenia, chronic neutrophilic leukemia, and related malignancies. Exp Hematol 2017; 46:9-20. [PMID: 27789332 PMCID: PMC5241233 DOI: 10.1016/j.exphem.2016.10.008] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 10/15/2016] [Accepted: 10/17/2016] [Indexed: 01/17/2023]
Abstract
Granulocyte colony-stimulating factor is a hematopoietic cytokine that stimulates neutrophil production and hematopoietic stem cell mobilization by initiating the dimerization of homodimeric granulocyte colony-stimulating factor receptor. Different mutations of CSF3R have been linked to a unique spectrum of myeloid disorders and related malignancies. Myeloid disorders caused by the CSF3R mutations include severe congenital neutropenia, chronic neutrophilic leukemia, and atypical chronic myeloid leukemia. In this review, we provide an analysis of granulocyte colony-stimulating factor receptor, various mutations, and their roles in the severe congenital neutropenia, chronic neutrophilic leukemia, and malignant transformation, as well as the clinical implications and some perspective on approaches that could expand our knowledge with respect to the normal signaling mechanisms and those associated with mutations in the receptor.
Collapse
MESH Headings
- Animals
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Congenital Bone Marrow Failure Syndromes
- Genetic Predisposition to Disease
- Humans
- Janus Kinases/metabolism
- Leukemia, Myeloid, Acute/etiology
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Neutrophilic, Chronic/etiology
- Leukemia, Neutrophilic, Chronic/metabolism
- MAP Kinase Signaling System
- Mutation
- Neutropenia/congenital
- Neutropenia/etiology
- Neutropenia/metabolism
- Phosphatidylinositol 3-Kinases/metabolism
- Protein Binding
- Protein Interaction Domains and Motifs/genetics
- Proto-Oncogene Proteins c-akt/metabolism
- Receptors, Granulocyte Colony-Stimulating Factor/chemistry
- Receptors, Granulocyte Colony-Stimulating Factor/genetics
- Receptors, Granulocyte Colony-Stimulating Factor/metabolism
- STAT Transcription Factors/metabolism
- Signal Transduction
Collapse
Affiliation(s)
- Pankaj Dwivedi
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Kenneth D Greis
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH.
| |
Collapse
|
106
|
Subcellular localization of the FLT3-ITD oncogene plays a significant role in the production of NOX- and p22phox-derived reactive oxygen species in acute myeloid leukemia. Leuk Res 2017; 52:34-42. [DOI: 10.1016/j.leukres.2016.11.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Revised: 11/09/2016] [Accepted: 11/10/2016] [Indexed: 12/26/2022]
|
107
|
Zhao M, Zhu P, Fujino M, Zhuang J, Guo H, Sheikh I, Zhao L, Li XK. Oxidative Stress in Hypoxic-Ischemic Encephalopathy: Molecular Mechanisms and Therapeutic Strategies. Int J Mol Sci 2016; 17:ijms17122078. [PMID: 27973415 PMCID: PMC5187878 DOI: 10.3390/ijms17122078] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 12/02/2016] [Accepted: 12/06/2016] [Indexed: 12/14/2022] Open
Abstract
Hypoxic-ischemic encephalopathy (HIE) is one of the leading causes of morbidity and mortality in neonates. Because of high concentrations of sensitive immature cells, metal-catalyzed free radicals, non-saturated fatty acids, and low concentrations of antioxidant enzymes, the brain requires high levels of oxygen supply and is, thus, extremely sensitive to hypoxia. Strong evidence indicates that oxidative stress plays an important role in pathogenesis and progression. Following hypoxia and ischemia, reactive oxygen species (ROS) production rapidly increases and overwhelms antioxidant defenses. A large excess of ROS will directly modify or degenerate cellular macromolecules, such as membranes, proteins, lipids, and DNA, and lead to a cascading inflammatory response, and protease secretion. These derivatives are involved in a complex interplay of multiple pathways (e.g., inflammation, apoptosis, autophagy, and necrosis) which finally lead to brain injury. In this review, we highlight the molecular mechanism for oxidative stress in HIE, summarize current research on therapeutic strategies utilized in combating oxidative stress, and try to explore novel potential clinical approaches.
Collapse
Affiliation(s)
- Mingyi Zhao
- Department of Pediatrics, the Third Xiangya Hospital, Central South University, Changsha 410006, China.
| | - Ping Zhu
- Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510100, China.
| | - Masayuki Fujino
- National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-8535, Japan.
- National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan.
| | - Jian Zhuang
- Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510100, China.
| | - Huiming Guo
- Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510100, China.
| | - IdrisAhmed Sheikh
- Department of Pediatrics, the Third Xiangya Hospital, Central South University, Changsha 410006, China.
| | - Lingling Zhao
- Department of Pediatrics, the Third Xiangya Hospital, Central South University, Changsha 410006, China.
| | - Xiao-Kang Li
- Department of Pediatrics, the Third Xiangya Hospital, Central South University, Changsha 410006, China.
- National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-8535, Japan.
| |
Collapse
|
108
|
NOX-driven ROS formation in cell transformation of FLT3-ITD-positive AML. Exp Hematol 2016; 44:1113-1122. [DOI: 10.1016/j.exphem.2016.08.008] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 08/28/2016] [Indexed: 12/22/2022]
|
109
|
Carter-Cooper BA, Fletcher S, Ferraris D, Choi EY, Kronfli D, Dash S, Truong P, Sausville EA, Lapidus RG, Emadi A. Synthesis, characterization and antineoplastic activity of bis-aziridinyl dimeric naphthoquinone - A novel class of compounds with potent activity against acute myeloid leukemia cells. Bioorg Med Chem Lett 2016; 27:6-10. [PMID: 27890379 DOI: 10.1016/j.bmcl.2016.11.045] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 11/15/2016] [Accepted: 11/17/2016] [Indexed: 01/05/2023]
Abstract
The synthesis, characterization and antileukemic activity of rationally designed amino dimeric naphthoquinone (BiQ) possessing aziridine as alkylating moiety is described. Bis-aziridinyl BiQ decreased proliferation of acute myeloid leukemia (AML) cell lines and primary cells from patients, and exhibited potent (nanomolar) inhibition of colony formation and overall cell survival in AML cells. Effective production of reactive oxygen species (ROS) and double stranded DNA breaks (DSB) induced by bis-aziridinyl BiQ is reported. Bis-dimethylamine BiQ, as the isostere of bis-aziridinyl BiQ but without the alkylating moiety did not show as potent anti-AML activity. Systemic administration of bis-aziridinyl BiQ was well tolerated in NSG mice.
Collapse
Affiliation(s)
- Brandon A Carter-Cooper
- University of Maryland School of Medicine, Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States
| | - Steven Fletcher
- Department of Pharmaceutical Sciences, University of Maryland, School of Pharmacy, Baltimore, MD, United States
| | - Dana Ferraris
- Department of Chemistry, McDaniel College, Westminster, MD, United States
| | - Eun Yong Choi
- University of Maryland School of Medicine, Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States
| | - Dahlia Kronfli
- University of Maryland School of Medicine, Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States
| | - Smaraki Dash
- University of Maryland School of Medicine, Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States
| | - Phuc Truong
- Department of Chemistry, McDaniel College, Westminster, MD, United States
| | - Edward A Sausville
- University of Maryland School of Medicine, Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States
| | - Rena G Lapidus
- University of Maryland School of Medicine, Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States
| | - Ashkan Emadi
- University of Maryland School of Medicine, Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States.
| |
Collapse
|
110
|
Identification of H7 as a novel peroxiredoxin I inhibitor to induce differentiation of leukemia cells. Oncotarget 2016; 7:3873-83. [PMID: 26716647 PMCID: PMC4826176 DOI: 10.18632/oncotarget.6763] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 11/28/2015] [Indexed: 01/02/2023] Open
Abstract
Identifying novel targets to enhance leukemia-cell differentiation is an urgent requirment. We have recently proposed that inhibiting the antioxidant enzyme peroxiredoxin I (Prdx I) may induce leukemia-cell differentiation. However, this concept remains to be confirmed. In this work, we identified H7 as a novel Prdx I inhibitor through virtual screening, in vitro activity assay, and surface plasmon resonance assay. Cellular thermal shift assay showed that H7 directly bound to Prdx I but not to Prdxs II–V in cells. H7 treatment also increased reactive oxygen species (ROS) level and cell differentiation in leukemia cells, as reflected by the upregulation of the cell surface differentiation marker CD11b/CD14 and the morphological maturation of cells. The differentiation-induction effect of H7 was further observed in some non-acute promyelocytic leukemia (APL) and primary leukemia cells apart from APL NB4 cells. Moreover, the ROS scavenger N-acetyl cysteine significantly reversed the H7-induced cell differentiation. We demonstrated as well that H7-induced cell differentiation was associated with the activation of the ROS-Erk1/2-C/EBPβ axis. Finally, we showed H7 treatment induced cell differentiation in an APL mouse model. All of these data confirmed that Prdx I was novel target for inducing leukemia-cell differentiation and that H7 was a novel lead compound for optimizing Prdx I inhibition.
Collapse
|
111
|
Shahrabi S, Khosravi A, Shahjahani M, Rahim F, Saki N. Genetics and Epigenetics of Myelodysplastic Syndromes and Response to Drug Therapy: New Insights. Oncol Rev 2016; 10:311. [PMID: 28058097 PMCID: PMC5178845 DOI: 10.4081/oncol.2016.311] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Accepted: 12/06/2016] [Indexed: 12/12/2022] Open
Abstract
Myelodysplastic syndromes (MDS) are a heterogeneous group of hematologic neoplasms ocurring mostly in the elderly. The clinical outcome of MDS patients is still poor despite progress in treatment approaches. About 90% of patients harbor at least one somatic mutation. This review aimed to assess the potential of molecular abnormalities in understanding pathogenesis, prognosis, diagnosis and in guiding choice of proper therapy in MDS patients. Papers related to this topic from 2000 to 2016 in PubMed and Scopus databases were searched and studied. The most common molecular abnormalities were TET2, ASXL1 as well as molecules involved in spliceosome machinery (U2AF1, SRSF2 and SF3B1). Patients with defects in TET2 molecule show better response to treatment with azacitidine. IDH and DNMT3A mutations are associated with a good response to decitabine therapy. In addition, patients with del5q subtype harboring TP53 mutation do not show a good response to lenalidomide therapy. In general, the results of this study show that molecular abnormalities can be associated with the occurrence of a specific morphological phenotype in patients. Therefore, considering the morphology of patients, different gene profiling methods can be selected to choice the most appropriate therapeutic measure in these patients in addition to faster and more cost-effective diagnosis of molecular abnormalities.
Collapse
Affiliation(s)
- Saeid Shahrabi
- Department of Biochemistry and Hematology, Semnan University of Medical Sciences, Semnan
| | - Abbas Khosravi
- Health Research Institute, Thalassemia and Hemoglobinopathy Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz
| | - Mohammad Shahjahani
- Colestan Hospital Clinical Research Development Unit. Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Fakher Rahim
- Health Research Institute, Thalassemia and Hemoglobinopathy Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz
| | - Najmaldin Saki
- Health Research Institute, Thalassemia and Hemoglobinopathy Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz
- Colestan Hospital Clinical Research Development Unit. Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
112
|
Khan N, Hills RK, Knapper S, Steadman L, Qureshi U, Rector JL, Bradbury C, Russell NH, Vyas P, Burnett AK, Grimwade D, Hole PS, Freeman SD. Normal Hematopoietic Progenitor Subsets Have Distinct Reactive Oxygen Species, BCL2 and Cell-Cycle Profiles That Are Decoupled from Maturation in Acute Myeloid Leukemia. PLoS One 2016; 11:e0163291. [PMID: 27669008 PMCID: PMC5036879 DOI: 10.1371/journal.pone.0163291] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 09/05/2016] [Indexed: 02/07/2023] Open
Abstract
In acute myeloid leukemia (AML) quiescence and low oxidative state, linked to BCL2 mitochondrial regulation, endow leukemic stem cells (LSC) with treatment-resistance. LSC in CD34+ and more mature CD34- AML have heterogeneous immunophenotypes overlapping with normal stem/progenitor cells (SPC) but may be differentiated by functional markers. We therefore investigated the oxidative/reactive oxygen species (ROS) profile, its relationship with cell-cycle/BCL2 for normal SPC, and whether altered in AML and myelodysplasia (MDS). In control BM (n = 24), ROS levels were highest in granulocyte-macrophage progenitors (GMP) and CD34- myeloid precursors but megakaryocyte-erythroid progenitors had equivalent levels to CD34+CD38low immature-SPC although they were ki67high. BCL2 upregulation was specific to GMPs. This profile was also observed for CD34+SPC in MDS-without-excess-blasts (MDS-noEB, n = 12). Erythroid CD34- precursors were, however, abnormally ROS-high in MDS-noEB, potentially linking oxidative stress to cell loss. In pre-treatment AML (n = 93) and MDS-with-excess-blasts (MDS-RAEB) (n = 14), immunophenotypic mature-SPC had similar ROS levels to co-existing immature-SPC. However ROS levels varied between AMLs; Flt3ITD+/NPM1wild-type CD34+SPC had higher ROS than NPM1mutated CD34+ or CD34- SPC. An aberrant ki67lowBCL2high immunophenotype was observed in CD34+AML (most prominent in Flt3ITD AMLs) but also in CD34- AMLs and MDS-RAEB, suggesting a shared redox/pro-survival adaptation. Some patients had BCL2 overexpression in CD34+ ROS-high as well as ROS-low fractions which may be indicative of poor early response to standard chemotherapy. Thus normal SPC subsets have distinct ROS, cell-cycle, BCL2 profiles that in AML /MDS-RAEB are decoupled from maturation. The combined profile of these functional properties in AML subpopulations may be relevant to differential treatment resistance.
Collapse
Affiliation(s)
- Naeem Khan
- Department of Clinical Immunology, Institute of Immunology and Immunotherapy, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Robert K. Hills
- Department of Haematology, Cardiff University School of Medicine, University Hospital Wales, Cardiff, United Kingdom
| | - Steve Knapper
- Department of Haematology, Cardiff University School of Medicine, University Hospital Wales, Cardiff, United Kingdom
| | - Lora Steadman
- Department of Clinical Immunology, Institute of Immunology and Immunotherapy, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Ushna Qureshi
- Department of Clinical Immunology, Institute of Immunology and Immunotherapy, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Jerrald L. Rector
- Department of Clinical Immunology, Institute of Immunology and Immunotherapy, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Charlotte Bradbury
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - Nigel H. Russell
- Centre for Clinical Haematology, Nottingham University Hospital NHS Trust, Nottingham, United Kingdom
| | - Paresh Vyas
- Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Alan K. Burnett
- Department of Haematology, Cardiff University School of Medicine, University Hospital Wales, Cardiff, United Kingdom
| | - David Grimwade
- Department of Medical and Molecular Genetics, King’s College London School of Medicine, Guy’s & St. Thomas’ NHS Foundation Trust, London, United Kingdom
| | - Paul S. Hole
- Department of Haematology, Cardiff University School of Medicine, University Hospital Wales, Cardiff, United Kingdom
| | - Sylvie D. Freeman
- Department of Clinical Immunology, Institute of Immunology and Immunotherapy, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| |
Collapse
|
113
|
AbdulSalam SF, Thowfeik FS, Merino EJ. Excessive Reactive Oxygen Species and Exotic DNA Lesions as an Exploitable Liability. Biochemistry 2016; 55:5341-52. [PMID: 27582430 DOI: 10.1021/acs.biochem.6b00703] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Although the terms "excessive reactive oxygen species (ROS)" and "oxidative stress" are widely used, the implications of oxidative stress are often misunderstood. ROS are not a single species but a variety of compounds, each with unique biochemical properties and abilities to react with biomolecules. ROS cause activation of growth signals through thiol oxidation and may lead to DNA damage at elevated levels. In this review, we first discuss a conceptual framework for the interplay of ROS and antioxidants. This review then describes ROS signaling using FLT3-mediated growth signaling as an example. We then focus on ROS-mediated DNA damage. High concentrations of ROS result in various DNA lesions, including 8-oxo-7,8-dihydro-guanine, oxazolone, DNA-protein cross-links, and hydantoins, that have unique biological impacts. Here we delve into the biochemistry of nine well-characterized DNA lesions. Within each lesion, the types of repair mechanisms, the mutations induced, and their effects on transcription and replication are discussed. Finally, this review will discuss biochemically inspired implications for cancer therapy. Several teams have put forward designs to harness the excessive ROS and the burdened DNA repair systems of tumor cells for treating cancer. We discuss inhibition of the antioxidant system, the targeting of DNA repair, and ROS-activated prodrugs.
Collapse
Affiliation(s)
- Safnas F AbdulSalam
- Department of Chemistry, University of Cincinnati , 404 Crosley Tower, Cincinnati, Ohio 45221-0172, United States
| | - Fathima Shazna Thowfeik
- Department of Chemistry, University of Cincinnati , 404 Crosley Tower, Cincinnati, Ohio 45221-0172, United States
| | - Edward J Merino
- Department of Chemistry, University of Cincinnati , 404 Crosley Tower, Cincinnati, Ohio 45221-0172, United States
| |
Collapse
|
114
|
Fiskus W, Coothankandaswamy V, Chen J, Ma H, Ha K, Saenz DT, Krieger SS, Mill CP, Sun B, Huang P, Mumm JS, Melnick AM, Bhalla KN. SIRT2 Deacetylates and Inhibits the Peroxidase Activity of Peroxiredoxin-1 to Sensitize Breast Cancer Cells to Oxidant Stress-Inducing Agents. Cancer Res 2016; 76:5467-78. [PMID: 27503926 DOI: 10.1158/0008-5472.can-16-0126] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 06/21/2016] [Indexed: 12/21/2022]
Abstract
SIRT2 is a protein deacetylase with tumor suppressor activity in breast and liver tumors where it is mutated; however, the critical substrates mediating its antitumor activity are not fully defined. Here we demonstrate that SIRT2 binds, deacetylates, and inhibits the peroxidase activity of the antioxidant protein peroxiredoxin (Prdx-1) in breast cancer cells. Ectopic overexpression of SIRT2, but not its catalytically dead mutant, increased intracellular levels of reactive oxygen species (ROS) induced by hydrogen peroxide, which led to increased levels of an overoxidized and multimeric form of Prdx-1 with activity as a molecular chaperone. Elevated levels of SIRT2 sensitized breast cancer cells to intracellular DNA damage and cell death induced by oxidative stress, as associated with increased levels of nuclear FOXO3A and the proapoptotic BIM protein. In addition, elevated levels of SIRT2 sensitized breast cancer cells to arsenic trioxide, an approved therapeutic agent, along with other intracellular ROS-inducing agents. Conversely, antisense RNA-mediated attenuation of SIRT2 reversed ROS-induced toxicity as demonstrated in a zebrafish embryo model system. Collectively, our findings suggest that the tumor suppressor activity of SIRT2 requires its ability to restrict the antioxidant activity of Prdx-1, thereby sensitizing breast cancer cells to ROS-induced DNA damage and cell cytotoxicity. Cancer Res; 76(18); 5467-78. ©2016 AACR.
Collapse
Affiliation(s)
- Warren Fiskus
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | | | - Hongwei Ma
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Kyungsoo Ha
- Department of Molecular Physiology, Baylor College of Medicine, Houston, Texas
| | - Dyana T Saenz
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Stephanie S Krieger
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Christopher P Mill
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Baohua Sun
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Peng Huang
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston Texas
| | - Jeffrey S Mumm
- Wilmer Eye Institute and the McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Ari M Melnick
- Division of Hematology and Medical Oncology, Weill Cornell Medical College, New York, New York
| | - Kapil N Bhalla
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
115
|
Singh AK, Awasthi D, Dubey M, Nagarkoti S, Kumar A, Chandra T, Barthwal MK, Tripathi AK, Dikshit M. High oxidative stress adversely affects NFκB mediated induction of inducible nitric oxide synthase in human neutrophils: Implications in chronic myeloid leukemia. Nitric Oxide 2016; 58:28-41. [PMID: 27264783 DOI: 10.1016/j.niox.2016.06.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 05/31/2016] [Accepted: 06/01/2016] [Indexed: 02/07/2023]
Abstract
Increasing evidence support bimodal action of nitric oxide (NO) both as a promoter and as an impeder of oxygen free radicals in neutrophils (PMNs), however impact of high oxidative stress on NO generation is less explored. In the present study, we comprehensively investigated the effect of high oxidative stress on inducible nitric oxide synthase (iNOS) expression and NO generation in human PMNs. Our findings suggest that PMA or diamide induced oxidative stress in PMNs from healthy volunteers, and high endogenous ROS in PMNs of chronic myeloid leukemia (CML) patients attenuate basal as well as LPS/cytokines induced NO generation and iNOS expression in human PMNs. Mechanistically, we found that under high oxidative stress condition, S-glutathionylation of NFκB (p50 and p65 subunits) severely limits iNOS expression due to its reduced binding to iNOS promoter, which was reversed in presence of DTT. Furthermore, by using pharmacological inhibitors, scavengers and molecular approaches, we identified that enhanced ROS generation via NOX2 and mitochondria, reduced Grx1/2 expression and GSH level associated with NFκB S-glutathionylation in PMNs from CML patients. Altogether data obtained suggest that oxidative status act as an important regulator of NO generation/iNOS expression, and under enhanced oxidative stress condition, NOX2-mtROS-NFκB S-glutathionylation is a feed forward loop, which attenuate NO generation and iNOS expression in human PMNs.
Collapse
Affiliation(s)
| | - Deepika Awasthi
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Megha Dubey
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Sheela Nagarkoti
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Ashutosh Kumar
- Department of Pathology, King George's Medical University, Lucknow, India
| | - Tulika Chandra
- Department of Transfusion Medicine, King George's Medical University, Lucknow, India
| | | | - Anil Kumar Tripathi
- Department of Clinical Haematology & Medical Oncology, King George's Medical University, Lucknow, India
| | - Madhu Dikshit
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, India.
| |
Collapse
|
116
|
Oxidative stress and hypoxia in normal and leukemic stem cells. Exp Hematol 2016; 44:540-60. [PMID: 27179622 DOI: 10.1016/j.exphem.2016.04.012] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 04/06/2016] [Accepted: 04/09/2016] [Indexed: 12/20/2022]
Abstract
The main hematopoietic stem cell (HSC) functions, self-renewal and differentiation, are finely regulated by both intrinsic mechanisms such as transcriptional and epigenetic regulators and extrinsic signals originating in the bone marrow microenvironment (HSC niche) or in the body (humoral mediators). The interaction between regulatory signals and cellular metabolism is an emerging area. Several metabolic pathways function differently in HSCs compared with progenitors and differentiated cells. Hypoxia, acting through hypoxia-inducing factors, has emerged as a key regulator of stem cell biology and acts by maintaining HSC quiescence and a condition of metabolic dormancy based on anaerobic glycolytic energetic metabolism, with consequent low production reactive oxygen species (ROS) and high antioxidant defense. Hematopoietic cell differentiation is accompanied by changes in oxidative metabolism (decrease of anaerobic glycolysis and increase of oxidative phosphorylation) and increased levels of ROS. Leukemic stem cells, defined as the cells that initiate and maintain the leukemic process, show peculiar metabolic properties in that they are more dependent on oxidative respiration than on glycolysis and are more sensitive to oxidative stress than normal HSCs. Several mitochondrial abnormalities have been described in acute myeloid leukemia (AML) cells, explaining the shift to aerobic glycolysis observed in these cells and offering the unique opportunity for therapeutic metabolic targeting. Finally, frequent mutations of the mitochondrial isocitrate dehydrogenase-2 (IDH2) enzyme are observed in AML cells, in which the mutated enzyme acts as an oncogenic driver and can be targeted using specific inhibitors under clinical evaluation with promising results.
Collapse
|
117
|
Jia X, Gu Z, Chen W, Jiao J. Tigecycline targets nonsmall cell lung cancer through inhibition of mitochondrial function. Fundam Clin Pharmacol 2016; 30:297-306. [PMID: 27009695 DOI: 10.1111/fcp.12199] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 02/28/2016] [Accepted: 03/18/2016] [Indexed: 12/18/2022]
Affiliation(s)
- Xuefeng Jia
- Department of Oncology; The First People's Hospital of Jining; No.6 Jiankang road Jining Shandong Province China
| | - Zhenfang Gu
- Department of Oncology; Affiliated Hospital of Jining Medical University; No.79 Guhuai road Jining Shandong Province China
| | - Wenming Chen
- Department of Oncology; The First People's Hospital of Jining; No.6 Jiankang road Jining Shandong Province China
| | - Junbo Jiao
- Department of Oncology; The First People's Hospital of Jining; No.6 Jiankang road Jining Shandong Province China
| |
Collapse
|
118
|
Zhang J, Ahn KS, Kim C, Shanmugam MK, Siveen KS, Arfuso F, Samym RP, Deivasigamanim A, Lim LHK, Wang L, Goh BC, Kumar AP, Hui KM, Sethi G. Nimbolide-Induced Oxidative Stress Abrogates STAT3 Signaling Cascade and Inhibits Tumor Growth in Transgenic Adenocarcinoma of Mouse Prostate Model. Antioxid Redox Signal 2016; 24:575-89. [PMID: 26649526 DOI: 10.1089/ars.2015.6418] [Citation(s) in RCA: 145] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
AIMS Prostate cancer (PCa) is one of the most commonly diagnosed cancers worldwide. Currently available therapies for metastatic PCa are only marginally effective, hence novel treatment modalities are urgently required. Considerable evidence(s) suggest that deregulated activation of oncogenic transcription factor, signal transducer and activator of transcription 3 (STAT3) plays a pivotal role in the development and progression of PCa. Thus, agents that can abrogate STAT3 activation could form the basis of novel therapy for PCa patients. In the present study, we analyzed whether the potential anticancer effects of nimbolide (NL), a limonoid triterpene derived from Azadirachta indica, against PCa cell lines and transgenic adenocarcinoma of mouse prostate (TRAMP) model are mediated through the negative regulation of STAT3 pathway. RESULTS Data from the in vitro studies indicated that NL could significantly inhibit cell viability, induce apoptosis, and suppress cellular invasion and migration. Interestingly, NL also abrogated STAT3 activation and this effect was found to be mediated via an increased production of reactive oxygen species (ROS) due to GSH/GSSG imbalance. Oral administration of NL significantly suppressed the tumor growth and metastasis in TRAMP mouse model without exhibiting any significant adverse effects. INNOVATION The present study demonstrates the critical role of GSH/GSSG imbalance-mediated ROS production contributing to the STAT3 inhibitory and tumor-suppressive effect of NL in PCa. CONCLUSION Overall, our findings indicate that NL exhibits significant anticancer effects in PCa that may be primarily mediated through the ROS-regulated inhibition of STAT3 signaling cascade.
Collapse
Affiliation(s)
- Jingwen Zhang
- 1 Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore
| | - Kwang Seok Ahn
- 2 College of Korean Medicine, Kyung Hee University , Seoul, Republic of Korea
| | - Chulwon Kim
- 2 College of Korean Medicine, Kyung Hee University , Seoul, Republic of Korea
| | - Muthu K Shanmugam
- 1 Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore
| | - Kodappully Sivaraman Siveen
- 1 Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore
| | - Frank Arfuso
- 3 School of Biomedical Sciences, CHIRI Biosciences Research Precinct, Curtin University , Perth, Australia
| | - Ramar Perumal Samym
- 4 Department of Anatomy, Venom and Toxin Research Programme, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore .,5 Department of Microbiology Infectious Diseases Programme, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore .,6 Department of Physiology, NUS Immunology Programme, Centre for Life Sciences, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore
| | - Amudha Deivasigamanim
- 7 Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research , National Cancer Centre, Singapore, Singapore
| | - Lina Hsiu Kim Lim
- 6 Department of Physiology, NUS Immunology Programme, Centre for Life Sciences, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore
| | - Lingzhi Wang
- 1 Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore .,8 Centre for Translational Medicine (CeTM), Cancer Science Institute, National University of Singapore , Singapore, Singapore
| | - Boon Cher Goh
- 1 Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore .,8 Centre for Translational Medicine (CeTM), Cancer Science Institute, National University of Singapore , Singapore, Singapore
| | - Alan Prem Kumar
- 1 Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore .,8 Centre for Translational Medicine (CeTM), Cancer Science Institute, National University of Singapore , Singapore, Singapore .,9 School of Biomedical Sciences, Curtin University , Perth, Australia .,10 Department of Biological Sciences, University of North Texas , Denton, Texas
| | - Kam Man Hui
- 7 Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research , National Cancer Centre, Singapore, Singapore
| | - Gautam Sethi
- 1 Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore .,9 School of Biomedical Sciences, Curtin University , Perth, Australia
| |
Collapse
|
119
|
Gonçalves AC, Alves R, Baldeiras I, Cortesão E, Carda JP, Branco CC, Oliveiros B, Loureiro L, Pereira A, Nascimento Costa JM, Sarmento-Ribeiro AB, Mota-Vieira L. Genetic variants involved in oxidative stress, base excision repair, DNA methylation, and folate metabolism pathways influence myeloid neoplasias susceptibility and prognosis. Mol Carcinog 2016; 56:130-148. [PMID: 26950655 DOI: 10.1002/mc.22478] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Revised: 01/22/2016] [Accepted: 02/17/2016] [Indexed: 12/27/2022]
Abstract
Myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML) share common features: elevated oxidative stress, DNA repair deficiency, and aberrant DNA methylation. We performed a hospital-based case-control study to evaluate the association in variants of genes involved in oxidative stress, folate metabolism, DNA repair, and DNA methylation with susceptibility and prognosis of these malignancies. To that end, 16 SNPs (one per gene: CAT, CYBA, DNMT1, DNMT3A, DNMT3B, GPX1, KEAP1, MPO, MTRR, NEIL1, NFE2F2, OGG1, SLC19A1, SOD1, SOD2, and XRCC1) were genotyped in 191 patients (101 MDS and 90 AML) and 261 controls. We also measured oxidative stress (reactive oxygen species/total antioxidant status ratio), DNA damage (8-hydroxy-2'-deoxyguanosine), and DNA methylation (5-methylcytosine) in 50 subjects (40 MDS and 10 controls). Results showed that five genes (GPX1, NEIL1, NFE2L2, OGG1, and SOD2) were associated with MDS, two (DNMT3B and SLC19A1) with AML, and two (CYBA and DNMT1) with both diseases. We observed a correlation of CYBA TT, GPX1 TT, and SOD2 CC genotypes with increased oxidative stress levels, as well as NEIL1 TT and OGG1 GG genotypes with higher DNA damage. The 5-methylcytosine levels were negatively associated with DNMT1 CC, DNMT3A CC, and MTRR AA genotypes, and positively with DNMT3B CC genotype. Furthermore, DNMT3A, MTRR, NEIL1, and OGG1 variants modulated AML transformation in MDS patients. Additionally, DNMT3A, OGG1, GPX1, and KEAP1 variants influenced survival of MDS and AML patients. Altogether, data suggest that genetic variability influence predisposition and prognosis of MDS and AML patients, as well AML transformation rate in MDS patients. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Ana Cristina Gonçalves
- Laboratory of Oncobiology and Hematology (LOH) and University Clinic of Hematology, Faculty of Medicine, University of Coimbra-FMUC, Coimbra, Portugal.,Center of Investigation in Environment, Genetics and Oncobiology (CIMAGO), FMUC, Coimbra, Portugal.,Center for Neuroscience and Cell Biology and Institute for Biomedical Imaging and Life Sciences (CNC.IBILI), Coimbra, Portugal
| | - Raquel Alves
- Laboratory of Oncobiology and Hematology (LOH) and University Clinic of Hematology, Faculty of Medicine, University of Coimbra-FMUC, Coimbra, Portugal.,Center of Investigation in Environment, Genetics and Oncobiology (CIMAGO), FMUC, Coimbra, Portugal.,Center for Neuroscience and Cell Biology and Institute for Biomedical Imaging and Life Sciences (CNC.IBILI), Coimbra, Portugal
| | - Inês Baldeiras
- Center for Neuroscience and Cell Biology and Institute for Biomedical Imaging and Life Sciences (CNC.IBILI), Coimbra, Portugal.,Department of Neurology, Laboratory of Neurochemistry, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.,Faculty of Medicine, University of Coimbra-FMUC, Coimbra, Portugal
| | - Emília Cortesão
- Laboratory of Oncobiology and Hematology (LOH) and University Clinic of Hematology, Faculty of Medicine, University of Coimbra-FMUC, Coimbra, Portugal.,Center of Investigation in Environment, Genetics and Oncobiology (CIMAGO), FMUC, Coimbra, Portugal.,Clinical Hematology Department, Centro Hospitalar e Universitário de Coimbra, EPE (CHUC, EPE), Coimbra, Portugal
| | - José Pedro Carda
- Laboratory of Oncobiology and Hematology (LOH) and University Clinic of Hematology, Faculty of Medicine, University of Coimbra-FMUC, Coimbra, Portugal.,Clinical Hematology Department, Centro Hospitalar e Universitário de Coimbra, EPE (CHUC, EPE), Coimbra, Portugal
| | - Claudia C Branco
- Molecular Genetics and Pathology Unit, Hospital of Divino Espírito Santo of Ponta Delgada, EPE, Ponta Delgada, São Miguel Island, Azores, Portugal.,Azores Genetics Research Group, Instituto Gulbenkian de Ciência, Oeiras, Portugal.,Faculty of Sciences, BioISI-Biosystems and Integrative Sciences Institute, University of Lisboa, Lisbon, Portugal
| | - Bárbara Oliveiros
- Laboratory for Biostatistics and Medical Informatics, FMUC, Coimbra, Portugal
| | - Luísa Loureiro
- Department of Medicine, Hospital Distrital da Figueira da Foz, EPE (HDFF, EPE), Figueira da Foz, Portugal
| | - Amélia Pereira
- Center of Investigation in Environment, Genetics and Oncobiology (CIMAGO), FMUC, Coimbra, Portugal.,Department of Medicine, Hospital Distrital da Figueira da Foz, EPE (HDFF, EPE), Figueira da Foz, Portugal
| | - José Manuel Nascimento Costa
- Department of Oncology, Centro Hospitalar e Universitário de Coimbra, EPE (CHUC, EPE), Coimbra, Portugal.,Faculty of Medicine, University Clinic of Oncology, University of Coimbra-FMUC, Coimbra, Portugal
| | - Ana Bela Sarmento-Ribeiro
- Laboratory of Oncobiology and Hematology (LOH) and University Clinic of Hematology, Faculty of Medicine, University of Coimbra-FMUC, Coimbra, Portugal.,Center of Investigation in Environment, Genetics and Oncobiology (CIMAGO), FMUC, Coimbra, Portugal.,Center for Neuroscience and Cell Biology and Institute for Biomedical Imaging and Life Sciences (CNC.IBILI), Coimbra, Portugal.,Clinical Hematology Department, Centro Hospitalar e Universitário de Coimbra, EPE (CHUC, EPE), Coimbra, Portugal
| | - Luisa Mota-Vieira
- Molecular Genetics and Pathology Unit, Hospital of Divino Espírito Santo of Ponta Delgada, EPE, Ponta Delgada, São Miguel Island, Azores, Portugal.,Azores Genetics Research Group, Instituto Gulbenkian de Ciência, Oeiras, Portugal.,Faculty of Sciences, BioISI-Biosystems and Integrative Sciences Institute, University of Lisboa, Lisbon, Portugal
| |
Collapse
|
120
|
Fairfield H, Falank C, Avery L, Reagan MR. Multiple myeloma in the marrow: pathogenesis and treatments. Ann N Y Acad Sci 2016; 1364:32-51. [PMID: 27002787 PMCID: PMC4806534 DOI: 10.1111/nyas.13038] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Multiple myeloma (MM) is a B cell malignancy resulting in osteolytic lesions and fractures. In the disease state, bone healing is limited owing to increased osteoclastic and decreased osteoblastic activity, as well as an MM-induced forward-feedback cycle where bone-embedded growth factors further enhance tumor progression as bone is resorbed. Recent work on somatic mutation in MM tumors has provided insight into cytogenetic changes associated with this disease; the initiating driver mutations causing MM are diverse because of the complexity and multitude of mutations inherent in MM tumor cells. This manuscript provides an overview of MM pathogenesis by summarizing cytogenic changes related to oncogenes and tumor suppressors associated with MM, reviewing risk factors, and describing the disease progression from monoclonal gammopathy of undetermined significance to overt MM. It also highlights the importance of the bone marrow microenvironment (BMM) in the establishment and progression of MM, as well as associated MM-induced bone disease, and the relationship of the bone marrow to current and future therapeutics. This review highlights why understanding the basic biology of the healthy and diseased BMM is crucial in the quest for better treatments and work toward a cure for genetically diverse diseases such as MM.
Collapse
Affiliation(s)
| | | | | | - Michaela R Reagan
- Maine Medical Center Research Institute, Scarborough, Maine
- University of Maine, Orono, Maine
| |
Collapse
|
121
|
Xiao M, Zhang L, Zhou Y, Rajoria P, Wang C. Pyrvinium selectively induces apoptosis of lymphoma cells through impairing mitochondrial functions and JAK2/STAT5. Biochem Biophys Res Commun 2016; 469:716-22. [DOI: 10.1016/j.bbrc.2015.12.059] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 12/15/2015] [Indexed: 12/28/2022]
|
122
|
Bănescu C, Iancu M, Trifa AP, Cândea M, Benedek Lazar E, Moldovan VG, Duicu C, Tripon F, Crauciuc A, Dobreanu M. From Six Gene Polymorphisms of the Antioxidant System, Only GPX Pro198Leu and GSTP1 Ile105Val Modulate the Risk of Acute Myeloid Leukemia. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2016:2536705. [PMID: 26823947 PMCID: PMC4707325 DOI: 10.1155/2016/2536705] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 10/25/2015] [Indexed: 02/05/2023]
Abstract
Oxidative stress might contribute to the occurrence of cancers, including the hematological ones. Various genetic polymorphisms were shown to increase the quantity of reactive oxygen species, a phenomenon that is able to induce mutations and thus promote cancers. The purpose of the study was to evaluate the association between CAT C262T, GPX1 Pro198Leu, MnSOD Ala16Val, GSTM1, GSTT1, and GSTP1 Ile105Val gene polymorphisms and acute myeloid leukemia risk, in a case-control study comprising 102 patients and 303 controls. No association was observed between AML and variant genotypes of CAT, MnSOD, GSTM1, and GSTT1 polymorphisms. Our data revealed a statistically significant difference regarding the frequencies of GPX1 Pro198Leu and GSTP1 Ile105Val variant genotypes between AML patients and controls (p < 0.001). Our results showed no association in the distribution of any of the CAT C262T, GPX1 Pro198Leu, GSTM1, GSTT1, and GSTP1 polymorphisms regarding age, gender, FAB subtype, cytogenetic risk groups, FLT3 and DNMT3 gene mutations, and overall survival. Our data suggests that the presence of variant allele and genotype of GPX1 Pro198Leu and GSTP1 Ile105Val gene polymorphisms may modulate the risk of developing AML.
Collapse
Affiliation(s)
- Claudia Bănescu
- Department of Medical Genetics, University of Medicine and Pharmacy Targu Mures, 38 Gh Marinescu Street, 540139 Targu Mures, Romania
| | - Mihaela Iancu
- Department of Medical Informatics and Biostatistics, “Iuliu Hatieganu” University of Medicine and Pharmacy, Cluj-Napoca, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania
| | - Adrian P. Trifa
- Department of Medical Genetics, “Iuliu Hatieganu” University of Medicine and Pharmacy, Cluj-Napoca, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania
| | - Marcela Cândea
- Hematology Clinic 1, University of Medicine and Pharmacy Targu Mures, 38 Gh Marinescu Street, 540139 Targu Mures, Romania
| | - Erzsebet Benedek Lazar
- Hematology Clinic 2, University of Medicine and Pharmacy Targu Mures, 38 Gh Marinescu Street, 540139 Targu Mures, Romania
| | - Valeriu G. Moldovan
- Department of Medical Genetics, University of Medicine and Pharmacy Targu Mures, 38 Gh Marinescu Street, 540139 Targu Mures, Romania
| | - Carmen Duicu
- Pediatric Clinic, University of Medicine and Pharmacy Targu Mures, 38 Gh Marinescu Street, 540139 Targu Mures, Romania
| | - Florin Tripon
- Department of Medical Genetics, University of Medicine and Pharmacy Targu Mures, 38 Gh Marinescu Street, 540139 Targu Mures, Romania
| | - Andrei Crauciuc
- Department of Medical Genetics, University of Medicine and Pharmacy Targu Mures, 38 Gh Marinescu Street, 540139 Targu Mures, Romania
| | - Minodora Dobreanu
- Department of Laboratory Medicine, University of Medicine and Pharmacy Targu Mures, 38 Gh Marinescu Street, 540139 Targu Mures, Romania
| |
Collapse
|
123
|
7-formyl-10-methylisoellipticine, a novel ellipticine derivative, induces mitochondrial reactive oxygen species (ROS) and shows anti-leukaemic activity in mice. Invest New Drugs 2015; 34:15-23. [DOI: 10.1007/s10637-015-0302-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 11/01/2015] [Indexed: 01/05/2023]
|
124
|
Differentiation and apoptosis induction by lovastatin and γ-tocotrienol in HL-60 cells via Ras/ERK/NF-κB and Ras/Akt/NF-κB signaling dependent down-regulation of glyoxalase 1 and HMG-CoA reductase. Cell Signal 2015. [DOI: 10.1016/j.cellsig.2015.07.014] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
125
|
Bazi A, Keramati MR, Gholamin M. Role of Oxidative Stress in Modulating Unfolded Protein Response Activity in Chronic Myeloid Leukemia Cell Line. IRANIAN BIOMEDICAL JOURNAL 2015; 20:63-7. [PMID: 26432458 PMCID: PMC4689283 DOI: 10.7508/ibj.2016.01.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Background: Recently, it has been revealed that tyrosine kinase inhibitors (TKIs) act through inducing both oxidative and endoplasmic reticulum (ER) stress in chronic myeloid leukemia cells. However, ER stress signaling triggers both apoptotic and survival processes within cells. Nevertheless, mechanisms by which TKIs avoid the pro-survival effects are not clear. The aim of this study was to evaluate the potential role of oxidative stress in activity of unfolded protein response (UPR) survival pathway within K562 cell line. Methods: The expression of UPR survival target genes, Xbp1, and Grp94 (glucose requiring protein 94) was studied in single and combined exposure to oxidative and ER stress in K562 cell line by quantitative and qualitative PCR. Results: The expression of UPR-related survival gene Grp94 was hampered by exposing to oxidative stress in cell induced with ER stress. Conclusion: Interaction of oxidative and ER stress may role as a mediator influencing UPR signaling activity.
Collapse
Affiliation(s)
- Ali Bazi
- Cancer Molecular Pathology Research Center, Imam Reza Hospital, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Faculty of Allied Medical Sciences, Zabol University of Medical Sciences, Zabol, Iran
| | - Mohammad Reza Keramati
- Cancer Molecular Pathology Research Center, Imam Reza Hospital, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehran Gholamin
- Division of Human Genetics, Immunology Research Center, Avicenna Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
126
|
Chen YL, Kan WM. Down-regulation of superoxide dismutase 1 by PMA is involved in cell fate determination and mediated via protein kinase D2 in myeloid leukemia cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:2662-75. [PMID: 26241492 DOI: 10.1016/j.bbamcr.2015.07.025] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Revised: 07/10/2015] [Accepted: 07/31/2015] [Indexed: 11/26/2022]
Abstract
Myeloid leukemia cells maintain a high intracellular ROS level and use redox signals for survival. The metabolism of ROS also affects cell fate, including cell death and differentiation. Superoxide dismutases (SODs) are major antioxidant enzymes that have high levels of expression in myeloid leukemia cells. However, the role of SODs in the regulation of myeloid leukemia cells' biological function is still unclear. To investigate the function of SODs in myeloid leukemia cell death and differentiation, we used myeloid leukemia cell lines K562, MEG-01, TF-1, and HEL cells for this study. We found that PMA-induced megakaryocytic differentiation in myeloid leukemia cells is accompanied by cell death and SOD1 down-regulation, while SOD2 expression is not affected. The role of SOD1 is verified when ATN-224, a SOD1 specific inhibitor, inhibits cell proliferation and promotes cell death in myeloid leukemia cells without PMA treatment. Moreover, inhibition or silencing of SODs further increases cell death and decreases polyploidization induced by PMA while they were partially reversed by SOD1 overexpression. Thus, SOD1 expression is required for myeloid leukemia cell fate determination. In addition, the knockdown of PKD2 reduces cell death and promotes polyploidization induced by PMA. PMA/PKD2-mediated necrosis via PARP cleavage involves both SOD1-dependent and -independent pathways. Finally, ATN-224 enhanced the inhibition of cell proliferation by Ara-C. Taken together, the results demonstrate that SOD1 regulates cell death and differentiation in myeloid leukemia cells. ATN-224 may be beneficial for myeloid leukemia therapy.
Collapse
Affiliation(s)
- Yu-Lin Chen
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | - Wai-Ming Kan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan; Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan.
| |
Collapse
|
127
|
Mehde AA, Yusof F, Mehdi WA, Zainulabdeen JA. CD26: A Prognostic Marker of Acute Lymphoblastic Leukemia in Children in the Post Remission Induction Phase. Asian Pac J Cancer Prev 2015; 16:5059-62. [DOI: 10.7314/apjcp.2015.16.12.5059] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
128
|
Zu B, Shi Y, Xu M, You G, Huang Z, Gao M, Feng W. ARE/SUZ12 dual specifically-regulated adenoviral TK/GCV system for CML blast crisis cells. J Exp Clin Cancer Res 2015; 34:56. [PMID: 26017281 PMCID: PMC4456766 DOI: 10.1186/s13046-015-0139-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2014] [Accepted: 02/17/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Treatment of blast phase chronic myeloid leukemia (BP-CML) remains a challenge, and the median survival is less than 6 months. Because effective treatments are lacking, we studied tight targeting of blast crisis CML cells using adenoviral (Ad) vectors expressing a HSV-TK system under dual control of a specific SUZ12 promoter and an antioxidant response element (ARE). METHODS A potential SUZ12 promoter fragment was designed with bioinformatics databases and identified with a luciferase assay. Next, we cloned the ARE element of the NQO1 gene and developed Ad vectors expressing TK kinase or luciferase under the dual control of a specific SUZ12 promoter and an ARE element. An in vitro transfection assay with Ad-ARE/SUZ12-Luc was used to determine promoter activity of ARE/SUZ12 regulatory element in blast crisis CML cells. After incubating human BP-CML-derived cells with Ad-ARE/SUZ12-TK and ganciclovir, Western blot, CCK8, Immunofluorescent assays and Annexin V assays were conducted to assess the efficacy of an ARE/SUZ12 dual-specific TK/GCV system for BP-CML cell lines. RESULTS Here, luciferase data confirmed significantly higher and specificer promoter activity of the ARE/SUZ12 composite component in CML blast crisis-derived cell lines (K562, KCL22, and K562/G01) compared to HepG2 cells, and Ad-AS-TK/GCV system could exhibit enhanced apoptotic effects and decreased cell viability for BP-CML cell lines. Additionally, Ad-AS-TK/GCV system altered expression of cycle-related and apoptosis-related proteins in BP-CML cell lines. CONCLUSIONS Thus, ARE/SUZ12 dual targeting TK/GCV system was effective in killing BP-CML cells. Moreover, efficacy and specificity of CML cell eradication were enhanced by synergistic effects of ARE/SUZ12 dual-specific regulation. We conclude that suicide gene-targeted therapy might hold promise for BP-CML treatment.
Collapse
Affiliation(s)
- Bailing Zu
- Department of Clinical Hematology, Key Laboratory of Laboratory Medical Diagnostics of Ministry of Education, Chongqing Medical University, No.1, Yixueyuan Road, Chongqing, 400016, People's Republic of China.
| | - Yi Shi
- Department of Clinical Laboratory, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China.
| | - Min Xu
- Department of Clinical Hematology, Key Laboratory of Laboratory Medical Diagnostics of Ministry of Education, Chongqing Medical University, No.1, Yixueyuan Road, Chongqing, 400016, People's Republic of China.
| | - Guoling You
- Department of Clinical Laboratory, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Zhenglan Huang
- Department of Clinical Hematology, Key Laboratory of Laboratory Medical Diagnostics of Ministry of Education, Chongqing Medical University, No.1, Yixueyuan Road, Chongqing, 400016, People's Republic of China.
| | - Miao Gao
- Department of Clinical Hematology, Key Laboratory of Laboratory Medical Diagnostics of Ministry of Education, Chongqing Medical University, No.1, Yixueyuan Road, Chongqing, 400016, People's Republic of China.
| | - Wenli Feng
- Department of Clinical Hematology, Key Laboratory of Laboratory Medical Diagnostics of Ministry of Education, Chongqing Medical University, No.1, Yixueyuan Road, Chongqing, 400016, People's Republic of China.
| |
Collapse
|
129
|
Gonçalves AC, Cortesão E, Oliveiros B, Alves V, Espadana AI, Rito L, Magalhães E, Lobão MJ, Pereira A, Nascimento Costa JM, Mota-Vieira L, Sarmento-Ribeiro AB. Oxidative stress and mitochondrial dysfunction play a role in myelodysplastic syndrome development, diagnosis, and prognosis: A pilot study. Free Radic Res 2015; 49:1081-94. [PMID: 25968944 DOI: 10.3109/10715762.2015.1035268] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The imbalance between reactive oxygen species (ROS) production and their elimination by antioxidants leads to oxidative stress. Depending on their concentration, ROS can trigger apoptosis or stimulate cell proliferation. We hypothesized that oxidative stress and mitochondrial dysfunction may participate not only in apoptosis detected in some myelodysplastic syndrome (MDS) patients, but also in increasing proliferation in other patients. We investigated the involvement of oxidative stress and mitochondrial dysfunction in MDS pathogenesis, as well as assessed their diagnostic and prognostic values. Intracellular peroxides, superoxide, superoxide/peroxides ratio, reduced glutathione (GSH), and mitochondrial membrane potential (Δψ(mit)) levels were analyzed in bone marrow cells from 27 MDS patients and 12 controls, by flow cytometry. We observed that all bone marrow cell types from MDS patients had increased intracellular peroxide levels and decreased GSH content, compared with control cells. Moreover, oxidative stress levels were MDS subtype- and risk group-dependent. Low-risk patients had the highest ROS levels, which can be related with their high apoptosis; and intermediate-2-risk patients had high Δψ(mit) that may be associated with their proliferative potential. GSH levels were negatively correlated with transfusion dependency, and peroxide levels were positively correlated with serum ferritin level. GSH content proved to be an accurate parameter to discriminate patients from controls. Finally, patients with high ROS or low GSH levels, as well as high superoxide/peroxides ratio had lower overall survival. Our results suggest that oxidative stress and mitochondrial dysfunction are involved in MDS development, and that oxidative stress parameters may constitute novel diagnosis and/or prognosis biomarkers for MDS.
Collapse
Affiliation(s)
- A C Gonçalves
- Laboratory of Oncobiology and Hematology, FMUC - Faculty of Medicine, University of Coimbra , Coimbra , Portugal
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
130
|
Gonçalves AC, Cortesão E, Oliveiros B, Alves V, Espadana AI, Rito L, Magalhães E, Pereira S, Pereira A, Costa JMN, Mota-Vieira L, Sarmento-Ribeiro AB. Oxidative stress levels are correlated with P15 and P16 gene promoter methylation in myelodysplastic syndrome patients. Clin Exp Med 2015; 16:333-43. [PMID: 25982567 DOI: 10.1007/s10238-015-0357-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 05/03/2015] [Indexed: 11/24/2022]
Abstract
Oxidative stress and abnormal DNA methylation have been implicated in some types of cancer, namely in myelodysplastic syndromes (MDS). Since both mechanisms are observed in MDS patients, we analyzed the correlation of intracellular levels of peroxides, superoxide anion, and glutathione (GSH), as well as ratios of peroxides/GSH and superoxide/GSH, with the methylation status of P15 and P16 gene promoters in bone marrow leukocytes from MDS patients. Compared to controls, these patients had lower GSH content, higher peroxide levels, peroxides/GSH and superoxide/GSH ratios, as well as higher methylation frequency of P15 and P16 gene promoters. Moreover, patients with methylated P15 gene had higher oxidative stress levels than patients without methylation (peroxides: 460 ± 42 MIF vs 229 ± 25 MIF, p = 0.001; superoxide: 383 ± 48 MIF vs 243 ± 17 MIF, p = 0.022; peroxides/GSH: 2.50 ± 0.08 vs 1.04 ± 0.34, p < 0.001; superoxide/GSH: 1.76 ± 0.21 vs 1.31 ± 0.10, p = 0.007). Patients with methylated P16 and at least one methylated gene had higher peroxide levels as well as peroxides/GSH ratio than patients without methylation. Interestingly, oxidative stress levels allow the discrimination of patients without methylation from ones with methylated P15, methylated P16, or at least one methylated (P15 or P16) promoter. Taken together, these findings support the hypothesis that oxidative stress is correlated with P15 and P16 hypermethylation.
Collapse
Affiliation(s)
- Ana Cristina Gonçalves
- Applied Molecular Biology, University Clinic of Hematology, Center of Investigation in Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra (FMUC), Azinhaga de Santa Comba - Celas, 3000-548, Coimbra, Portugal.,Center for Neuroscience and Cell Biology (CNC), Coimbra, Portugal
| | - Emília Cortesão
- Applied Molecular Biology, University Clinic of Hematology, Center of Investigation in Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra (FMUC), Azinhaga de Santa Comba - Celas, 3000-548, Coimbra, Portugal.,Clinical Hematology Department, Centro Hospitalar e Universitário de Coimbra (CHUC), Coimbra, Portugal
| | - Barbara Oliveiros
- Laboratory for Biostatistics and Medical Informatics, Faculty of Medicine, University of Coimbra (FMUC), Coimbra, Portugal
| | - Vera Alves
- Immunology, Faculty of Medicine, University of Coimbra (FMUC), Coimbra, Portugal
| | - Ana Isabel Espadana
- Clinical Hematology Department, Centro Hospitalar e Universitário de Coimbra (CHUC), Coimbra, Portugal
| | - Luís Rito
- Applied Molecular Biology, University Clinic of Hematology, Center of Investigation in Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra (FMUC), Azinhaga de Santa Comba - Celas, 3000-548, Coimbra, Portugal.,Clinical Hematology Department, Centro Hospitalar e Universitário de Coimbra (CHUC), Coimbra, Portugal
| | - Emília Magalhães
- Clinical Hematology Department, Centro Hospitalar e Universitário de Coimbra (CHUC), Coimbra, Portugal
| | - Sónia Pereira
- Medicine Department, Hospital Distrital da Figueira da Foz, Figueira da Foz, Portugal
| | - Amélia Pereira
- Medicine Department, Hospital Distrital da Figueira da Foz, Figueira da Foz, Portugal
| | - José Manuel Nascimento Costa
- Oncology Department, Centro Hospital e Universitário de Coimbra (CHUC), Coimbra, Portugal.,University Clinic of Oncology, Faculty of Medicine, University of Coimbra (FMUC), Coimbra, Portugal
| | - Luisa Mota-Vieira
- Molecular Genetics and Pathology Unit, Hospital of Divino Espírito Santo of Ponta Delgada EPE, São Miguel Island, Azores, Portugal.,Azores Genetics Research Group, Instituto Gulbenkian de Ciência, Oeiras, Portugal.,Faculty of Sciences, Biosystems & Integrative Sciences Institute (BioISI), University of Lisboa, Lisbon, Portugal
| | - Ana Bela Sarmento-Ribeiro
- Applied Molecular Biology, University Clinic of Hematology, Center of Investigation in Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra (FMUC), Azinhaga de Santa Comba - Celas, 3000-548, Coimbra, Portugal. .,Center for Neuroscience and Cell Biology (CNC), Coimbra, Portugal. .,Clinical Hematology Department, Centro Hospitalar e Universitário de Coimbra (CHUC), Coimbra, Portugal.
| |
Collapse
|
131
|
Traumatic brain injury and NADPH oxidase: a deep relationship. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:370312. [PMID: 25918580 PMCID: PMC4397034 DOI: 10.1155/2015/370312] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 03/18/2015] [Indexed: 12/21/2022]
Abstract
Traumatic brain injury (TBI) represents one of the major causes of mortality and disability in the world.
TBI is characterized by primary damage resulting from the mechanical forces applied to the head as a direct result of the trauma and by the subsequent secondary injury due to a complex cascade of biochemical events that eventually lead to neuronal cell death. Oxidative stress plays a pivotal role in the genesis of the delayed harmful effects contributing to permanent damage. NADPH oxidases (Nox), ubiquitary membrane multisubunit enzymes whose unique function is the production of reactive oxygen species (ROS), have been shown to be a major source of ROS in the brain and to be involved in several neurological diseases. Emerging evidence demonstrates that Nox is upregulated after TBI, suggesting Nox critical role in the onset and development of this pathology.
In this review, we summarize the current evidence about the role of Nox enzymes in the pathophysiology of TBI.
Collapse
|
132
|
Abstract
The mechanism by which reactive oxygen species (ROS) are produced by tumour cells remained incompletely understood until the discovery over the last 15 years of the family of NADPH oxidases (NOXs 1–5 and dual oxidases DUOX1/2) which are structural homologues of gp91phox, the major membrane-bound component of the respiratory burst oxidase of leucocytes. Knowledge of the roles of the NOX isoforms in cancer is rapidly expanding. Recent evidence suggests that both NOX1 and DUOX2 species produce ROS in the gastrointestinal tract as a result of chronic inflammatory stress; cytokine induction (by interferon-γ, tumour necrosis factor α, and interleukins IL-4 and IL-13) of NOX1 and DUOX2 may contribute to the development of colorectal and pancreatic carcinomas in patients with inflammatory bowel disease and chronic pancreatitis, respectively. NOX4 expression is increased in pre-malignant fibrotic states which may lead to carcinomas of the lung and liver. NOX5 is highly expressed in malignant melanomas, prostate cancer and Barrett's oesophagus-associated adenocarcinomas, and in the last it is related to chronic gastro-oesophageal reflux and inflammation. Over-expression of functional NOX proteins in many tissues helps to explain tissue injury and DNA damage from ROS that accompany pre-malignant conditions, as well as elucidating the potential mechanisms of NOX-related damage that contribute to both the initiation and the progression of a wide range of solid and haematopoietic malignancies.
Collapse
|
133
|
Wang W, Zhang Y, Lu W, Liu K. Mitochondrial reactive oxygen species regulate adipocyte differentiation of mesenchymal stem cells in hematopoietic stress induced by arabinosylcytosine. PLoS One 2015; 10:e0120629. [PMID: 25768922 PMCID: PMC4359087 DOI: 10.1371/journal.pone.0120629] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 01/28/2015] [Indexed: 01/28/2023] Open
Abstract
Objective The increase in adipocytes induced by chemotherapeutic drugs may play a negative role in hematopoietic recovery. However, the mechanism underlying adipocyte differentiation of mesenchymal stem cells (MSCs) in hematopoietic stress is still unknown. Hence, the involvement of reactive oxygen species (ROS) in adipocyte differentiation under hematopoietic stress was investigated in vitro and in vivo. Methods The roles of cellular ROS in adipogenesis were investigated in vivo through an adipocyte hyperplasia marrow model under hematopoietic stress induced by arabinosylcytosine (Ara-C) and in vitro via adipocyte differentiation of human MSCs. ROS levels were detected using the CM-H2DCFDA probe and Mito-SOX dye. Adipogenesis was evaluated by histopathology and oil red O staining, whereas detection of mRNA levels of antioxidant enzymes and adipogenesis markers was performed using quantitative real-time polymerase chain reaction analysis. Results ROS were found to play an important role in regulating adipocyte differentiation of MSCs by activating peroxisome proliferator-activated receptor gamma (PPARγ,) while the antioxidant N-acetyl-L-cysteine acts through ROS to inhibit adipocyte differentiation. The elevated ROS levels induced by Ara-C were caused by both over-generation of mitochondrial ROS and reduction of antioxidant enzymes (Cu/Zn Superoxide dismutase and catalase). Our findings suggest that a mitochondrial-targeted antioxidant could diminish adipocyte differentiation.
Collapse
Affiliation(s)
- Weimin Wang
- Department of Hematology, Peking University People's Hospital, Beijing, China
- Institute of Hematology, Peking University, Beijing, China
| | - Yao Zhang
- Department of Hematology, Peking University People's Hospital, Beijing, China
- Institute of Hematology, Peking University, Beijing, China
| | - Wenyi Lu
- Department of Hematology, Peking University People's Hospital, Beijing, China
- Institute of Hematology, Peking University, Beijing, China
| | - Kaiyan Liu
- Department of Hematology, Peking University People's Hospital, Beijing, China
- Institute of Hematology, Peking University, Beijing, China
- * E-mail:
| |
Collapse
|
134
|
Stanicka J, Russell EG, Woolley JF, Cotter TG. NADPH oxidase-generated hydrogen peroxide induces DNA damage in mutant FLT3-expressing leukemia cells. J Biol Chem 2015; 290:9348-61. [PMID: 25697362 DOI: 10.1074/jbc.m113.510495] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Indexed: 11/06/2022] Open
Abstract
Internal tandem duplication of the FMS-like tyrosine kinase (FLT3-ITD) receptor is present in 20% of acute myeloid leukemia (AML) patients and it has been associated with an aggressive AML phenotype. FLT3-ITD expressing cell lines have been shown to generate increased levels of reactive oxygen species (ROS) and DNA double strand breaks (DSBs). However, the molecular basis of how FLT3-ITD-driven ROS leads to the aggressive form of AML is not clearly understood. Our group has previously reported that inhibition of FLT3-ITD signaling results in post-translational down-regulation of p22(phox), a small membrane-bound subunit of the NADPH oxidase (NOX) complex. Here we demonstrated that 32D cells, a myeloblast-like cell line transfected with FLT3-ITD, have a higher protein level of p22(phox) and p22(phox)-interacting NOX isoforms than 32D cells transfected with the wild type FLT3 receptor (FLT3-WT). The inhibition of NOX proteins, p22(phox), and NOX protein knockdowns caused a reduction in ROS, as measured with a hydrogen peroxide (H2O2)-specific dye, peroxy orange 1 (PO1), and nuclear H2O2, as measured with nuclear peroxy emerald 1 (NucPE1). These reductions in the level of H2O2 following the NOX knockdowns were accompanied by a decrease in the number of DNA DSBs. We showed that 32D cells that express FLT3-ITD have a higher level of both oxidized DNA and DNA DSBs than their wild type counterparts. We also observed that NOX4 and p22(phox) localize to the nuclear membrane in MV4-11 cells expressing FLT3-ITD. Taken together these data indicate that NOX and p22(phox) mediate the ROS production from FLT3-ITD that signal to the nucleus causing genomic instability.
Collapse
Affiliation(s)
- Joanna Stanicka
- From the Tumour Biology Laboratory, School of Biochemistry and Cell Biology, Bioscience Research Institute, University College Cork, Cork, Ireland
| | - Eileen G Russell
- From the Tumour Biology Laboratory, School of Biochemistry and Cell Biology, Bioscience Research Institute, University College Cork, Cork, Ireland
| | - John F Woolley
- From the Tumour Biology Laboratory, School of Biochemistry and Cell Biology, Bioscience Research Institute, University College Cork, Cork, Ireland
| | - Thomas G Cotter
- From the Tumour Biology Laboratory, School of Biochemistry and Cell Biology, Bioscience Research Institute, University College Cork, Cork, Ireland
| |
Collapse
|
135
|
Ali T, Saeed K, Abdullah M, Murtaza I. Antihematotoxic Role of Bunium persicum Seed Differential Extracts in Animal Model: Reactive Oxygen Species Might Be a Contributor. Osong Public Health Res Perspect 2015; 5:358-63. [PMID: 25562045 PMCID: PMC4281623 DOI: 10.1016/j.phrp.2014.11.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Revised: 09/15/2014] [Accepted: 10/01/2014] [Indexed: 11/29/2022] Open
Abstract
Objectives Humans have been using plants as natural medicines since prehistoric times. Bunium persicum is a rich source of oils with different biological activities such as antioxidative and antimicrobial activities. The aim of this study is to evaluate the antihematotoxic and antioxidative effects of the differential extracts of B. persicum against leukemic blood induced hematotoxicity in an animal model. Methods This study was performed on animals, which were divided into several groups: normal control, disease control, and groups that were administered with differential extracts of plants. We measured the concentration of free radical [reactive oxygen species (ROS)] and hematological parameters as blast cells from the tibia and femur in different groups. Results The ROS level and blast cells count were higher in disease control groups than in groups treated with varying amounts of B. persicum extract and the normal group. Moreover, there was an imbalance in hematological parameters in untreated and treated groups with a correlation between free radicals and plant extract administration. Conclusion These findings may indicate a possible link between free radicals and hematotoxicity and blast cells, while depicting a potential therapeutic role for B. persicum against ROS-induced hematotoxicity.
Collapse
|
136
|
Park S, Park JA, Kim YE, Song S, Kwon HJ, Lee Y. Suberoylanilide hydroxamic acid induces ROS-mediated cleavage of HSP90 in leukemia cells. Cell Stress Chaperones 2015; 20:149-57. [PMID: 25119188 PMCID: PMC4255254 DOI: 10.1007/s12192-014-0533-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 07/03/2014] [Accepted: 07/29/2014] [Indexed: 12/20/2022] Open
Abstract
Heat shock protein 90 (HSP90) is a molecular chaperone that supports stability of client proteins. We found that HSP90 was cleaved to 55 kDa protein after treatment with histone deacetylase (HDAC) inhibitors including suberoylanilide hydroxamic acid (SAHA) in several leukemia cell lines. We further analyzed molecular changes induced by SAHA in K562 cells. The SAHA-induced cleavage of HSP90 was blocked by a pan-caspase inhibitor, z-VAD-fmk, implying that the process is dependent on caspase activity. However, the experiments using antagonistic and agonistic Fas antibodies revealed that the cleavage of HSP90 was not dependent on Fas signaling. SAHA induced generation of reactive oxygen species (ROS), and the cleavage of HSP90 was blocked by a ROS scavenger N-acetylcystein (NAC). We also confirmed that hydrogen peroxide (H2O2) induced cleavage of HSP90 in a similar manner. Caspase 2, 3, 4, 6, 8, and 10 were activated by treatment with SAHA, and the activities were reduced by the pretreatment of NAC. Treatment of the cells with caspase 10 inhihitor, but not other inhibitors of caspases activated by SAHA, prevented cleavage of HSP90 by SAHA. SAHA-induced ROS generation and HSP90 cleavage were dependent on newly synthesized unknown proteins. Taken together, our results suggest that the cleavage of HSP90 by SAHA is mediated by ROS generation and caspase 10 activation. HSP90 cleavage may provide an additional mechanism involved in anti-cancer effects of HDAC inhibitors.
Collapse
Affiliation(s)
- Sangkyu Park
- />Department of Biochemistry, College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk 361-763 Republic of Korea
| | - Jeong-A Park
- />Department of Biochemistry, College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk 361-763 Republic of Korea
| | - Young-Eun Kim
- />Department of Biochemistry, College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk 361-763 Republic of Korea
- />Biotechnology Research Institute, Chungbuk National University, Cheongju, Chungbuk 361-763 Republic of Korea
| | - Sukgil Song
- />College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk 361-763 Republic of Korea
| | - Hyung-Joo Kwon
- />Center for Medical Science Research, Hallym University, Chuncheon, 200-720 Republic of Korea
- />Department of Microbiology, College of Medicine, Hallym University, Chuncheon, 200-720 Republic of Korea
| | - Younghee Lee
- />Department of Biochemistry, College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk 361-763 Republic of Korea
- />Biotechnology Research Institute, Chungbuk National University, Cheongju, Chungbuk 361-763 Republic of Korea
- />Department of Biochemistry, College of Natural Sciences, Chungbuk National University, 52 Naesudong-Ro, Heungduk-Gu, Cheongju, Chungbuk 361-763 Republic of Korea
| |
Collapse
|
137
|
Muhammad F, Qi W, Wang A, Gu J, Du J, Zhu G. Using oxidant susceptibility of thiol stabilized nanoparticles to develop an inflammation triggered drug release system. J Mater Chem B 2015; 3:1597-1604. [DOI: 10.1039/c4tb01709a] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Ultrasmall thiol passivated ZnS NPs are prepared using a newly developed synthetic protocol. Exposure to hydroxyl radicals results in oxidation of the thiol groups, thus destabilizing the ZnS nanolids to open drug encompassing pores for attaining an inflammation responsive drug delivery system.
Collapse
Affiliation(s)
- Faheem Muhammad
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry
- College of Chemistry
- Jilin University
- Changchun
- China
| | - Wenxiu Qi
- College of Life Science
- Jilin University
- Changchun
- China
| | - Aifei Wang
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry
- College of Chemistry
- Jilin University
- Changchun
- China
| | - Jingkai Gu
- College of Life Science
- Jilin University
- Changchun
- China
| | - Jianshi Du
- China Japan Union Hospital
- Jilin University
- Changchun
- China
| | - Guangshan Zhu
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry
- College of Chemistry
- Jilin University
- Changchun
- China
| |
Collapse
|
138
|
Rasool M, Farooq S, Malik A, Shaukat A, Manan A, Asif M, Sani S, Qazi MH, Kamal MA, Iqbal Z, Hussain A. Assessment of circulating biochemical markers and antioxidative status in acute lymphoblastic leukemia (ALL) and acute myeloid leukemia (AML) patients. Saudi J Biol Sci 2015; 22:106-111. [PMID: 25561892 PMCID: PMC4281600 DOI: 10.1016/j.sjbs.2014.09.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Revised: 08/31/2014] [Accepted: 09/01/2014] [Indexed: 01/14/2023] Open
Abstract
Various circulating biochemical markers are indicators of pathological state in leukemia and its subtypes. Increased oxidative stress and decreased antioxidant factors portray clear image associated with malignancies during subtypes of leukemia. In this research work we investigated the inter-relationship among the subtypes of leukemia with circulating biochemical markers and oxidative stress in the Pakistani population. This research work was conducted on a total number of 70 subjects in which 20 were control participants and 50 were suffering from leukemia and divided into two subtypes (ALL and AML). Various circulating biomarkers were investigated including hematological, hepatic and renal profiles as well as oxidative stress markers, electrolytes and vitamins C and E. Results show that vitamin E was found to be decreased in diseased sub-types (P < 0.05). Malondialdehyde (MDA) levels were very high in disease sub-types (ALL-B = 8.69 ± 1.59; ALL-T = 8.78 ± 0.97; AML = 8.50 ± 1.29) compared to controls (1.22 ± 0.10; P < 0.05) while the levels of antioxidants [superoxide dismutase (SOD), glutathione peroxidase (GPx), reduced glutathione (GSH), catalase (CAT)], platelets, as well as electrolytes (Ca and Mg) were reduced in patients suffering from leukemia (sub-types). Enhanced levels of oxidative stress (MDA) and decreased levels of enzymatic and non-enzymatic antioxidants reflect the pathological state and impaired cell control in patients suffering from leukemia (subtypes) and show a strong correlation with oxidative stress, indicating that patients' biological systems are under oxidative stress.
Collapse
Affiliation(s)
- Mahmood Rasool
- Center of Excellence in Genomic Medicine Research (CEGMR), King Abdulaziz University, Jeddah, Saudi Arabia
| | - Shahzad Farooq
- Institute of Molecular Biology and Biotechnology (IMBB), The University of Lahore, Lahore, Pakistan
| | - Arif Malik
- Institute of Molecular Biology and Biotechnology (IMBB), The University of Lahore, Lahore, Pakistan
| | - Aysha Shaukat
- Institute of Molecular Biology and Biotechnology (IMBB), The University of Lahore, Lahore, Pakistan
| | - Abdul Manan
- Institute of Molecular Biology and Biotechnology (IMBB), The University of Lahore, Lahore, Pakistan
| | - Muhammad Asif
- Department of Biotechnology and Informatics, BUITEMS, Quetta, Pakistan
| | - Suleman Sani
- Institute of Molecular Biology and Biotechnology (IMBB), The University of Lahore, Lahore, Pakistan
| | - Mahmood Husain Qazi
- Centre for Research in Molecular Medicine (CRiMM), The University of Lahore, Lahore, Pakistan
| | - Mohammad Amjad Kamal
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Zafar Iqbal
- College of Applied Medical Sciences, King Saud Bin Abdulaziz University of Health Sciences, National Guards Health Affairs, Riyadh, Saudi Arabia
| | - Abrar Hussain
- Department of Biotechnology and Informatics, BUITEMS, Quetta, Pakistan
| |
Collapse
|
139
|
Muhammad F, Wang A, Miao L, Wang P, Li Q, Liu J, Du J, Zhu G. Synthesis of oxidant prone nanosilver to develop H2O2 responsive drug delivery system. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2014; 31:514-21. [PMID: 25486873 DOI: 10.1021/la503922j] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Our immune system uses toxicity of hydrogen peroxide to kill off bacterial invaders. In this contribution, we intended to integrate ROS producing capability of immune system with oxidant-sensitive nature of antibacterial silver nanoparticles (Ag NPs) to develop an oxidant drug delivery system. Prior to execute this strategy, we have developed an efficient one-pot synthetic protocol to produce ultrasmall (5 nm), water-stable, and oxidant-prone Ag NPs. Notably, the yield of as-synthesized Ag NPs is 10-fold higher than standard citrate reduction route. The resulting therapeutically active and well-dispersed Ag NPs are used as nanolids to cap the drug loaded nanochannels of porous silica. Upon exposing to H2O2, dissolution-accompanied aggregation of Ag nanolids unleashes the encapsulated therapeutic entities from channels of nanocarrier. Combination of antibacterial and anti-inflammatory drugs in single nanocarriers can potentially augment the effectiveness of various therapies.
Collapse
Affiliation(s)
- Faheem Muhammad
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, College of Chemistry, ‡China Japan Union Hospital, and §State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University , Changchun 130012, China
| | | | | | | | | | | | | | | |
Collapse
|
140
|
Udensi UK, Tchounwou PB. Dual effect of oxidative stress on leukemia cancer induction and treatment. J Exp Clin Cancer Res 2014; 33:106. [PMID: 25519934 PMCID: PMC4320640 DOI: 10.1186/s13046-014-0106-5] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 12/01/2014] [Indexed: 02/07/2023] Open
Abstract
Oxidative stress (OS) has been characterized by an imbalance between the production of reactive oxygen species (ROS) and a biological system's ability to repair oxidative damage or to neutralize the reactive intermediates including peroxides and free radicals. High ROS production has been associated with significant decrease in antioxidant defense mechanisms leading to protein, lipid and DNA damage and subsequent disruption of cellular functions. In humans, OS has been reported to play a role in the pathogenesis of neurodegenerative diseases such as Alzheimer's disease, Huntington's disease, Lou Gehrig's disease, multiple sclerosis and Parkinson's disease, as well as atherosclerosis, autism, cancer, heart failure, and myocardial infarction. Although OS has been linked to the etiology and development of chronic diseases, many chemotherapeutic drugs have been shown to exert their biologic activity through induction of OS in affected cells. This review highlights the controversial role of OS in the development and progression of leukemia cancer and the therapeutic application of increased OS and antioxidant approaches to the treatment of leukemia patients.
Collapse
Affiliation(s)
- Udensi K Udensi
- NIH/NIMHD RCMI Center for Environmental Health, College of Science, Engineering and Technology, Jackson State University, Jackson, MS, 39217, USA.
| | - Paul B Tchounwou
- NIH/NIMHD RCMI Center for Environmental Health, College of Science, Engineering and Technology, Jackson State University, Jackson, MS, 39217, USA.
| |
Collapse
|
141
|
Esposito MT, So CWE. DNA damage accumulation and repair defects in acute myeloid leukemia: implications for pathogenesis, disease progression, and chemotherapy resistance. Chromosoma 2014; 123:545-61. [PMID: 25112726 DOI: 10.1007/s00412-014-0482-9] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Revised: 07/23/2014] [Accepted: 07/24/2014] [Indexed: 12/15/2022]
Abstract
DNA damage repair mechanisms are vital to maintain genomic integrity. Mutations in genes involved in the DNA damage response (DDR) can increase the risk of developing cancer. In recent years, a variety of polymorphisms in DDR genes have been associated with increased risk of developing acute myeloid leukemia (AML) or of disease relapse. Moreover, a growing body of literature has indicated that epigenetic silencing of DDR genes could contribute to the leukemogenic process. In addition, a variety of AML oncogenes have been shown to induce replication and oxidative stress leading to accumulation of DNA damage, which affects the balance between proliferation and differentiation. Conversely, upregulation of DDR genes can provide AML cells with escape mechanisms to the DDR anticancer barrier and induce chemotherapy resistance. The current review summarizes the DDR pathways in the context of AML and describes how aberrant DNA damage response can affect AML pathogenesis, disease progression, and resistance to standard chemotherapy, and how defects in DDR pathways may provide a new avenue for personalized therapeutic strategies in AML.
Collapse
Affiliation(s)
- Maria Teresa Esposito
- Leukemia and Stem Cell Biology Group, Department of Hematological Medicine, King's College London, Denmark Hill campus, SE5 9NU, London, UK
| | | |
Collapse
|
142
|
Ye ZW, Zhang J, Townsend DM, Tew KD. Oxidative stress, redox regulation and diseases of cellular differentiation. Biochim Biophys Acta Gen Subj 2014; 1850:1607-21. [PMID: 25445706 DOI: 10.1016/j.bbagen.2014.11.010] [Citation(s) in RCA: 170] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Revised: 10/31/2014] [Accepted: 11/10/2014] [Indexed: 12/18/2022]
Abstract
BACKGROUND Within cells, there is a narrow concentration threshold that governs whether reactive oxygen species (ROS) induce toxicity or act as second messengers. SCOPE OF REVIEW We discuss current understanding of how ROS arise, facilitate cell signaling, cause toxicities and disease related to abnormal cell differentiation and those (primarily) sulfur based pathways that provide nucleophilicity to offset these effects. PRIMARY CONCLUSIONS Cellular redox homeostasis mediates a plethora of cellular pathways that determine life and death events. For example, ROS intersect with GSH based enzyme pathways to influence cell differentiation, a process integral to normal hematopoiesis, but also affecting a number of diverse cell differentiation related human diseases. Recent attempts to manage such pathologies have focused on intervening in some of these pathways, with the consequence that differentiation therapy targeting redox homeostasis has provided a platform for drug discovery and development. GENERAL SIGNIFICANCE The balance between electrophilic oxidative stress and protective biomolecular nucleophiles predisposes the evolution of modern life forms. Imbalances of the two can produce aberrant redox homeostasis with resultant pathologies. Understanding the pathways involved provides opportunities to consider interventional strategies. This article is part of a Special Issue entitled Redox regulation of differentiation and de-differentiation.
Collapse
Affiliation(s)
- Zhi-Wei Ye
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, 70 President St., DD410, Charleston, SC 29425, USA
| | - Jie Zhang
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, 70 President St., DD410, Charleston, SC 29425, USA
| | - Danyelle M Townsend
- Department of Pharmaceutical and Biomedical Sciences, Medical University of South Carolina, 274 Calhoun Street MSC 141, Charleston, SC 29425-1410, USA
| | - Kenneth D Tew
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, 70 President St., DD410, Charleston, SC 29425, USA.
| |
Collapse
|
143
|
Hasselbalch HC, Thomassen M, Hasselbalch Riley C, Kjær L, Stauffer Larsen T, Jensen MK, Bjerrum OW, Kruse TA, Skov V. Whole blood transcriptional profiling reveals deregulation of oxidative and antioxidative defence genes in myelofibrosis and related neoplasms. Potential implications of downregulation of Nrf2 for genomic instability and disease progression. PLoS One 2014; 9:e112786. [PMID: 25397683 PMCID: PMC4232509 DOI: 10.1371/journal.pone.0112786] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 10/15/2014] [Indexed: 01/16/2023] Open
Abstract
The Philadelphia-negative chronic myeloproliferative neoplasms - essential thrombocythemia (ET), polycythemia vera (PV), and myelofibrosis (MF) (MPNs) - have recently been shown to be associated with chronic inflammation, oxidative stress and accumulation of reactive oxygen species (ROS). Using whole blood transcriptional profiling, we report that several oxidative stress and anti-oxidative stress genes are significantly deregulated in MPNs. Among the twenty most up- and downregulated genes, ATOX1, DEFB122, GPX8, PRDX2, PRDX6, PTGS1, and SEPP1 were progressively upregulated from ET over PV to PMF, whereas AKR1B1, CYBA, SIRT2, TTN, and UCP2 were progressively downregulated in ET, PV and PMF (all FDR <0.05). The gene Nrf2, encoding the transcription factor nuclear factor erythroid 2-related factor 2 (NFE2L2 or Nrf2) was significantly downregulated in all MPNs. Nrf2 has a key role in the regulation of the oxidative stress response and modulates both migration and retention of hematopoietic stem cells (HSCs) in their niche. The patogenetic importance of Nrf2 depletion in the context of expansion of the hematopoietic progenitor pool in MPNs is discussed with particular focus upon the implications of concomitant downregulation of Nrf2 and CXCR4 for stem cell mobilization.
Collapse
Affiliation(s)
- Hans Carl Hasselbalch
- Department of Hematology, Roskilde Hospital, University of Copenhagen, Roskilde, Denmark
- * E-mail:
| | - Mads Thomassen
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
| | | | - Lasse Kjær
- Department of Hematology, Roskilde Hospital, University of Copenhagen, Roskilde, Denmark
| | | | - Morten K. Jensen
- Department of Hematology, Roskilde Hospital, University of Copenhagen, Roskilde, Denmark
| | - Ole Weis Bjerrum
- Department of Hematology L, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Torben A. Kruse
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
| | - Vibe Skov
- Department of Hematology, Roskilde Hospital, University of Copenhagen, Roskilde, Denmark
| |
Collapse
|
144
|
Muhammad F, Wang A, Qi W, Zhang S, Zhu G. Intracellular antioxidants dissolve man-made antioxidant nanoparticles: using redox vulnerability of nanoceria to develop a responsive drug delivery system. ACS APPLIED MATERIALS & INTERFACES 2014; 6:19424-19433. [PMID: 25312332 DOI: 10.1021/am5055367] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Regeneratable antioxidant property of nanoceria has widely been explored to minimize the deleterious influences of reactive oxygen species. Limited information is, however, available regarding the biological interactions and subsequent fate of nanoceria in body fluids. This study demonstrates a surprising dissolution of stable and ultrasmall (4 nm) cerium oxide nanoparticles (CeO2 NPs) in response to biologically prevalent antioxidant molecules (glutathione, vitamin C). Such a redox sensitive behavior of CeO2 NPs is subsequently exploited to design a redox responsive drug delivery system for transporting anticancer drug (camptothecin). Upon exposing the CeO2 capped and drug loaded nanoconstruct to vitamin c or glutathione, dissolution-accompanied aggregation of CeO2 nanolids unleashes the drug molecules from porous silica to achieve a significant anticancer activity. Besides stimuli responsive drug delivery, immobilization of nanoceria onto the surface of mesoporous silica also facilitates us to gain a basic insight into the biotransformation of CeO2 in physiological mediums.
Collapse
Affiliation(s)
- Faheem Muhammad
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, College of Chemistry, ‡College of Life Science, Jilin University , Changchun 130012, P. R. China
| | | | | | | | | |
Collapse
|
145
|
Truitt L, Hutchinson C, DeCoteau JF, Geyer CR. Chaetocin antileukemia activity against chronic myelogenous leukemia cells is potentiated by bone marrow stromal factors and overcomes innate imatinib resistance. Oncogenesis 2014; 3:e122. [PMID: 25329721 PMCID: PMC4216903 DOI: 10.1038/oncsis.2014.37] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Revised: 09/02/2014] [Accepted: 09/09/2014] [Indexed: 02/07/2023] Open
Abstract
Chronic myelogenous leukemia (CML) is maintained by a minor population of leukemic stem cells (LSCs) that exhibit innate resistance to tyrosine kinase inhibitors (TKIs) targeting BCR-ABL. Innate resistance can be induced by secreted bone marrow stromal cytokines and growth factors (BMSFs) that protect CML-LSCs from TKIs, resulting in minimal residual disease. Developing strategies to eradicate innate TKI resistance in LSCs is critical for preventing disease relapse. Cancer cells balance reactive oxygen species (ROS) at higher than normal levels, promoting their proliferation and survival, but also making them susceptible to damage by ROS-generating agents. Bcr-Abl increases cellular ROS levels, which can be reduced with TKI inhibitors, whereas, BMSFs increase ROS levels. We hypothesized that BMSF-mediated increases in ROS would trigger ROS damage in TKI-treated CML-LSCs when exposed to chaetocin, a mycotoxin that imposes oxidative stress by inhibiting thioredoxin reductase-1. Here, we showed that chaetocin suppressed viability and colony formation, and induced apoptosis of the murine hematopoietic cell line TonB210 with and without Bcr-Abl expression, and these effects were potentiated by BMSFs. In contrast, imatinib activities in Bcr-Abl-positive TonB210 cells were inhibited by BMSFs. Further, BMSFs did not inhibit imatinib activities when TonB210 cells expressing Bcr-Abl were cotreated with chaetocin. Chaetocin showed similar activities against LSC-enriched CML cell populations isolated from a murine transplant model of CML blast crisis that were phenotypically negative for lineage markers and positive for Sca-1 and c-Kit (CML-LSK). BMSFs and chaetocin increased ROS in CML-LSK cells and addition of BMSFs and chaetocin resulted in higher levels compared with chaetocin or BMSF treatment alone. Pretreatment of CML-LSKs with the antioxidant N-acetylcysteine blocked chaetocin cytotoxicity, even in the presence of BMSFs, demonstrating the importance ROS for chaetocin activities. Chaetocin effects on self-renewal of CML-LSKs were assessed by transplanting CML-LSKs into secondary recipients following ex vivo exposure to chaetocin, in the presence or absence of BMSFs. Disease latency in mice transplanted with CML-LSKs following chaetocin treatment more than doubled compared with untreated CML-LSKs or BMSFs-treated CML-LSKs. Mice transplanted with CML-LSKs following chaetocin treatment in the presence of BMSFs had significantly extended survival time compared with mice transplanted with CML-LSKs treated with chaetocin alone. Our findings indicate that chaetocin activity against CML-LSKs is significantly enhanced in the presence of BMSFs and suggest that chaetocin may be effective as a codrug to complement TKIs in CML treatment by disrupting the innate resistance of CML-LSKs through an ROS dependent mechanism.
Collapse
Affiliation(s)
- L Truitt
- Cancer Stem Cell Research Group, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Department of Pathology and Laboratory Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - C Hutchinson
- Cancer Stem Cell Research Group, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Department of Biochemistry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - J F DeCoteau
- Cancer Stem Cell Research Group, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Department of Pathology and Laboratory Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - C R Geyer
- Cancer Stem Cell Research Group, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Department of Pathology and Laboratory Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Department of Biochemistry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
146
|
Ludin A, Gur-Cohen S, Golan K, Kaufmann KB, Itkin T, Medaglia C, Lu XJ, Ledergor G, Kollet O, Lapidot T. Reactive oxygen species regulate hematopoietic stem cell self-renewal, migration and development, as well as their bone marrow microenvironment. Antioxid Redox Signal 2014; 21:1605-19. [PMID: 24762207 PMCID: PMC4175025 DOI: 10.1089/ars.2014.5941] [Citation(s) in RCA: 245] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
SIGNIFICANCE Blood forming, hematopoietic stem cells (HSCs) mostly reside in the bone marrow in a quiescent, nonmotile state via adhesion interactions with stromal cells and macrophages. Quiescent, proliferating, and differentiating stem cells have different metabolism, and accordingly different amounts of intracellular reactive oxygen species (ROS). Importantly, ROS is not just a byproduct of metabolism, but also plays a role in stem cell state and function. RECENT ADVANCES ROS levels are dynamic and reversibly dictate enhanced cycling and myeloid bias in ROS(high) short-term repopulating stem cells, and ROS(low) quiescent long-term repopulating stem cells. Low levels of ROS, regulated by intrinsic factors such as cell respiration or nicotinamide adenine dinucleotide phosphate-oxidase (NADPH oxidase) activity, or extrinsic factors such as stem cell factor or prostaglandin E2 are required for maintaining stem cell self-renewal. High ROS levels, due to stress and inflammation, induce stem cell differentiation and enhanced motility. CRITICAL ISSUES Stem cells need to be protected from high ROS levels to avoid stem cell exhaustion, insufficient host immunity, and leukemic transformation that may occur during chronic inflammation. However, continuous low ROS production will lead to lack of stem cell function and opportunistic infections. Ultimately, balanced ROS levels are crucial for maintaining the small stem cell pool and host immunity, both in homeostasis and during stress situations. FUTURE DIRECTIONS Deciphering the signaling pathway of ROS in HSC will provide a better understanding of ROS roles in switching HSC from quiescence to activation and vice versa, and will also shed light on the possible roles of ROS in leukemia initiation and development.
Collapse
Affiliation(s)
- Aya Ludin
- 1 Department of Immunology, Weizmann Institute of Science , Rehovot, Israel
| | | | | | | | | | | | | | | | | | | |
Collapse
|
147
|
Karmali R, Dalovisio A, Borgia JA, Venugopal P, Kim BW, Grant-Szymanski K, Hari P, Lazarus H. All in the family: Clueing into the link between metabolic syndrome and hematologic malignancies. Blood Rev 2014; 29:71-80. [PMID: 25433571 DOI: 10.1016/j.blre.2014.09.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Revised: 09/11/2014] [Accepted: 09/18/2014] [Indexed: 10/24/2022]
Abstract
Metabolic syndrome constitutes a constellation of findings including central obesity, insulin resistance/type 2 diabetes mellitus (DM), dyslipidemia and hypertension. Metabolic syndrome affects 1 in 4 adults in the United States and is rapidly rising in prevalence, largely driven by the dramatic rise in obesity and insulin resistance/DM. Being central to the development of metabolic syndrome and its other related diseases, much focus has been placed on identifying the mitogenic effects of obesity and insulin resistance/DM as mechanistic clues of the link between metabolic syndrome and cancer. Pertinent mechanisms identified include altered lipid signaling, adipokine and inflammatory cytokine effects, and activation of PI3K/Akt/mTOR and RAS/RAF/MAPK/ERK pathways via dysregulated insulin/insulin-like growth factor-1 (IGF-1) signaling. Through variable activation of these multiple pathways, obesity and insulin resistance/DM pre-dispose to hematologic malignancies, imposing the aggressive and chemo-resistant phenotypes typically seen in cancer patients with underlying metabolic syndrome. Growing understanding of these pathways has identified druggable cancer targets, rationalizing the development and testing of agents like PI3K inhibitor idelalisib, mTOR inhibitors everolimus and temsirolimus, and IGF-1 receptor inhibitor linsitinib. It has also led to exploration of obesity and diabetes-directed therapies including statins and oral hypoglycemic for the management of metabolic syndrome-related hematologic neoplasms.
Collapse
Affiliation(s)
- Reem Karmali
- Division of Hematology, Oncology and Cell Therapy, Rush University Medical Center, 1725 W Harrison Street, Suite 809, Chicago, IL 60612, United States.
| | - Andrew Dalovisio
- Division of Hematology, Oncology and Cell Therapy, Rush University Medical Center, 1725 W Harrison Street, Suite 809, Chicago, IL 60612, United States.
| | - Jeffrey A Borgia
- Department of Pathology, Rush Proteomics and Biomarker Development Core, Rush University Medical Center, 1735 W Harrison Street, Suite 554, Chicago, IL 60612, United States; Department of Biochemistry, Rush Proteomics and Biomarker Development Core, Rush University Medical Center, 1735 W Harrison Street, Suite 554, Chicago, IL 60612, United States.
| | - Parameswaran Venugopal
- Division of Hematology, Oncology and Cell Therapy, Rush University Medical Center, 1725 W Harrison Street, Suite 809, Chicago, IL 60612, United States.
| | - Brian W Kim
- Division of Endocrinology, Rush University Medical Center, Cohn Building Room 226, 1735 W Harrison Street, Chicago, IL 60612, United States.
| | - Kelly Grant-Szymanski
- Division of Hematology, Oncology and Cell Therapy, Rush University Medical Center, 1725 W Harrison Street, Suite 809, Chicago, IL 60612, United States.
| | - Parameswaran Hari
- Medical College of Wisconsin, 9200 W. Wisconsin Ave., Clinical Cancer Center, Milwaukee, WI 53226, United States.
| | - Hillard Lazarus
- Case Western Reserve University, Novel Cell Therapy, 11100 Euclid Ave., Wearn Bldg. Room 341, Cleveland, OH 44106-5089, United States.
| |
Collapse
|
148
|
Gao H, Wu X, Simon L, Fossett N. Antioxidants maintain E-cadherin levels to limit Drosophila prohemocyte differentiation. PLoS One 2014; 9:e107768. [PMID: 25226030 PMCID: PMC4167200 DOI: 10.1371/journal.pone.0107768] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Accepted: 08/12/2014] [Indexed: 01/01/2023] Open
Abstract
Mitochondrial reactive oxygen species (ROS) regulate a variety of biological processes by networking with signal transduction pathways to maintain homeostasis and support adaptation to stress. In this capacity, ROS have been shown to promote the differentiation of progenitor cells, including mammalian embryonic and hematopoietic stem cells and Drosophila hematopoietic progenitors (prohemocytes). However, many questions remain about how ROS alter the regulatory machinery to promote progenitor differentiation. Here, we provide evidence for the hypothesis that ROS reduce E-cadherin levels to promote Drosophila prohemocyte differentiation. Specifically, we show that knockdown of the antioxidants, Superoxide dismutatase 2 and Catalase reduce E-cadherin protein levels prior to the loss of Odd-skipped-expressing prohemocytes. Additionally, over-expression of E-cadherin limits prohemocyte differentiation resulting from paraquat-induced oxidative stress. Furthermore, two established targets of ROS, Enhancer of Polycomb and FOS, control the level of E-cadherin protein expression. Finally, we show that knockdown of either Superoxide dismutatase 2 or Catalase leads to an increase in the E-cadherin repressor, Serpent. As a result, antioxidants and targets of ROS can control E-cadherin protein levels, and over-expression of E-cadherin can ameliorate the prohemocyte response to oxidative stress. Collectively, these data strongly suggest that ROS promote differentiation by reducing E-cadherin levels. In mammalian systems, ROS promote embryonic stem cell differentiation, whereas E-cadherin blocks differentiation. However, it is not known if elevated ROS reduce E-cadherin to promote embryonic stem cell differentiation. Thus, our findings may have identified an important mechanism by which ROS promote stem/progenitor cell differentiation.
Collapse
Affiliation(s)
- Hongjuan Gao
- Center for Vascular and Inflammatory Diseases and the Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Xiaorong Wu
- Center for Vascular and Inflammatory Diseases and the Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - LaTonya Simon
- Department of Chemical and Biochemical Engineering, University of Maryland Baltimore County, Baltimore, MD, United States of America
| | - Nancy Fossett
- Center for Vascular and Inflammatory Diseases and the Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, United States of America
- * E-mail:
| |
Collapse
|
149
|
Hypoxia inhibits JAK2V617F activation via suppression of SHP-2 function in myeloproliferative neoplasm cells. Exp Hematol 2014; 42:783-92.e1. [DOI: 10.1016/j.exphem.2014.05.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 04/27/2014] [Accepted: 05/14/2014] [Indexed: 11/22/2022]
|
150
|
Zhang H, Fang H, Wang K. Reactive oxygen species in eradicating acute myeloid leukemic stem cells. Stem Cell Investig 2014; 1:13. [PMID: 27358859 DOI: 10.3978/j.issn.2306-9759.2014.04.03] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 04/20/2014] [Indexed: 12/13/2022]
Abstract
Leukemic stem cells (LSCs) have been proven to drive leukemia initiation, progression and relapse, and are increasingly being used as a critical target for therapeutic intervention. As an essential feature in LSCs, reactive oxygen species (ROS) homeostasis has been extensively exploited in the past decade for targeting LSCs in acute myeloid leukemia (AML). Most, if not all, agents that show therapeutic benefits are able to alter redox status by inducing ROS, which confers selectivity in eradicating AML stem cells but sparing normal counterparts. In this review, we provide the comprehensive update of ROS-generating agents in the context of their impacts on our understanding of the pathogenesis of AML and its therapy. We anticipate that further characterizing these ROS agents will help us combat against AML in the coming era of LSC-targeting strategy.
Collapse
Affiliation(s)
- Hui Zhang
- 1 State Key Laboratory of Medical Genomics and Shanghai Institute of Hematology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China ; 2 Pediatric department, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Hai Fang
- 1 State Key Laboratory of Medical Genomics and Shanghai Institute of Hematology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China ; 2 Pediatric department, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Kankan Wang
- 1 State Key Laboratory of Medical Genomics and Shanghai Institute of Hematology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China ; 2 Pediatric department, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| |
Collapse
|