51
|
Lima K, Lopes LR, Machado-Neto JA. Exploring redox vulnerabilities in JAK2 V617F-positive cellular models. Hematol Transfus Cell Ther 2020; 43:430-436. [PMID: 32962959 PMCID: PMC8573030 DOI: 10.1016/j.htct.2020.08.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 07/09/2020] [Accepted: 08/15/2020] [Indexed: 11/30/2022] Open
Abstract
Background In Philadelphia chromosome-negative myeloproliferative neoplasm (MPN) models, reactive oxygen species (ROS) are elevated and have been implicated in genomic instability, JAK2/STAT signaling amplification, and disease progression. Although the potential effects of ROS on the MPN phenotype, the effects of ruxolitinib treatment on ROS regulation have been poorly explored. Herein, we have reported the impact of ruxolitinib on redox signaling transcriptional network, and the effects of diphenyleneiodonium (DPI), a pan NOX inhibitor, in JAK2V617F-driven cellular models. Method Redox signaling-related genes were investigated in SET2 cells upon ruxolitinib treatment by RNA-seq (GEO accession GSE69827). SET2 and HEL cells, which represent JAK2V617F-positive MPN cellular models with distinct sensitivity to apoptosis induced by ruxolitinib, were used. Cell viability was evaluated by MTT, apoptosis by annexin V/PI and flow cytometry, and cell signaling by quantitative PCR and Western blot. Main results Ruxolitinib impacted on a network composed of redox signaling-related genes, and DUOX1 and DUOX2 were identified as potential modulators of ruxolitinib response. In SET2 and HEL cells, DPI reduced cell viability and, at low doses, it significantly potentiated ruxolitinib-induced apoptosis. In the molecular scenario, DPI inhibited STAT3, STAT5 and S6 ribosomal protein phosphorylation and induced PARP1 cleavage in JAK2V617F-positive cells. DPI combined with ruxolitinib increased PARP1 cleavage in SET2 cells and potentiated ruxolitinib-reduced STAT3, STAT5 and S6 ribosomal protein in HEL cells. Conclusion Our study reveals a potential adaptation mechanism for resistance against ruxolitinib by transcriptionally reprogramming redox signaling in JAK2V617F cells and exposes redox vulnerabilities with therapeutic value in MPN cellular models.
Collapse
Affiliation(s)
- Keli Lima
- Biomedical Sciences Institute, Universidade de São Paulo, São Paulo, SP, Brazil
| | | | | |
Collapse
|
52
|
Cellular and Molecular Mechanisms of Environmental Pollutants on Hematopoiesis. Int J Mol Sci 2020; 21:ijms21196996. [PMID: 32977499 PMCID: PMC7583016 DOI: 10.3390/ijms21196996] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/20/2020] [Accepted: 09/21/2020] [Indexed: 02/07/2023] Open
Abstract
Hematopoiesis is a complex and intricate process that aims to replenish blood components in a constant fashion. It is orchestrated mostly by hematopoietic progenitor cells (hematopoietic stem cells (HSCs)) that are capable of self-renewal and differentiation. These cells can originate other cell subtypes that are responsible for maintaining vital functions, mediate innate and adaptive immune responses, provide tissues with oxygen, and control coagulation. Hematopoiesis in adults takes place in the bone marrow, which is endowed with an extensive vasculature conferring an intense flow of cells. A myriad of cell subtypes can be found in the bone marrow at different levels of activation, being also under constant action of an extensive amount of diverse chemical mediators and enzymatic systems. Bone marrow platelets, mature erythrocytes and leukocytes are delivered into the bloodstream readily available to meet body demands. Leukocytes circulate and reach different tissues, returning or not returning to the bloodstream. Senescent leukocytes, specially granulocytes, return to the bone marrow to be phagocytized by macrophages, restarting granulopoiesis. The constant high production and delivery of cells into the bloodstream, alongside the fact that blood cells can also circulate between tissues, makes the hematopoietic system a prime target for toxic agents to act upon, making the understanding of the bone marrow microenvironment vital for both toxicological sciences and risk assessment. Environmental and occupational pollutants, therapeutic molecules, drugs of abuse, and even nutritional status can directly affect progenitor cells at their differentiation and maturation stages, altering behavior and function of blood compounds and resulting in impaired immune responses, anemias, leukemias, and blood coagulation disturbances. This review aims to describe the most recently investigated molecular and cellular toxicity mechanisms of current major environmental pollutants on hematopoiesis in the bone marrow.
Collapse
|
53
|
Bratek E, Ziembowicz A, Salinska E. N-Acetylaspartylglutamate (NAAG) Pretreatment Reduces Hypoxic-Ischemic Brain Damage and Oxidative Stress in Neonatal Rats. Antioxidants (Basel) 2020; 9:antiox9090877. [PMID: 32957477 PMCID: PMC7555246 DOI: 10.3390/antiox9090877] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/08/2020] [Accepted: 09/14/2020] [Indexed: 12/15/2022] Open
Abstract
N-acetylaspartylglutamate (NAAG), the most abundant peptide transmitter in the mammalian nervous system, activates mGluR3 at presynaptic sites, inhibiting the release of glutamate, and acts on mGluR3 on astrocytes, stimulating the release of neuroprotective growth factors (TGF-β). NAAG can also affect N-methyl-d-aspartate (NMDA) receptors in both synaptic and extrasynaptic regions. NAAG reduces neurodegeneration in a neonatal rat model of hypoxia-ischemia (HI), although the exact mechanism is not fully recognized. In the present study, the effect of NAAG application 24 or 1 h before experimental birth asphyxia on oxidative stress markers and the potential mechanisms of neuroprotection on 7-day old rats was investigated. The intraperitoneal application of NAAG at either time point before HI significantly reduced the weight deficit of the ischemic brain hemisphere, radical oxygen species (ROS) content and activity of antioxidant enzymes, and increased the concentration of reduced glutathione (GSH). No additional increase in the TGF-β concentration was observed after NAAG application. The fast metabolism of NAAG and the decrease in TGF-β concentration that resulted from NAAG pretreatment, performed up to 24 h before HI, excluded the involvement mGluR3 in neuroprotection. The observed effect may be explained by the activation of NMDA receptors induced by NAAG pretreatment 24 h before HI. Inhibition of the NAAG effect by memantine supports this conclusion. NAAG preconditioning 1 h before HI results in a mixture of mGluR3 and NMDA receptor activation. Preconditioning with NAAG induces the antioxidative defense system triggered by mild excitotoxicity in neurons. Moreover, this response to NAAG pretreatment is consistent with the commonly accepted mechanism of preconditioning. However, this theory requires further investigation.
Collapse
|
54
|
Eastman CL, D'Ambrosio R, Ganesh T. Modulating neuroinflammation and oxidative stress to prevent epilepsy and improve outcomes after traumatic brain injury. Neuropharmacology 2020; 172:107907. [PMID: 31837825 PMCID: PMC7274911 DOI: 10.1016/j.neuropharm.2019.107907] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 11/26/2019] [Accepted: 12/05/2019] [Indexed: 12/14/2022]
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability in young adults worldwide. TBI survival is associated with persistent neuropsychiatric and neurological impairments, including posttraumatic epilepsy (PTE). To date, no pharmaceutical treatment has been found to prevent PTE or ameliorate neurological/neuropsychiatric deficits after TBI. Brain trauma results in immediate mechanical damage to brain cells and blood vessels that may never be fully restored given the limited regenerative capacity of brain tissue. This primary insult unleashes cascades of events, prominently including neuroinflammation and massive oxidative stress that evolve over time, expanding the brain injury, but also clearing cellular debris and establishing homeostasis in the region of damage. Accumulating evidence suggests that oxidative stress and neuroinflammatory sequelae of TBI contribute to posttraumatic epileptogenesis. This review will focus on possible roles of reactive oxygen species (ROS), their interactions with neuroinflammation in posttraumatic epileptogenesis, and emerging therapeutic strategies after TBI. We propose that inhibitors of the professional ROS-generating enzymes, the NADPH oxygenases and myeloperoxidase alone, or combined with selective inhibition of cyclooxygenase mediated signaling may have promise for the treatment or prevention of PTE and other sequelae of TBI. This article is part of the special issue entitled 'New Epilepsy Therapies for the 21st Century - From Antiseizure Drugs to Prevention, Modification and Cure of Epilepsy'.
Collapse
Affiliation(s)
- Clifford L Eastman
- Department of Neurological Surgery, 325 Ninth Ave., Seattle, WA, 98104, USA.
| | - Raimondo D'Ambrosio
- Department of Neurological Surgery, 325 Ninth Ave., Seattle, WA, 98104, USA; Regional Epilepsy Center, University of Washington, 325 Ninth Ave., Seattle, WA, 98104, USA
| | - Thota Ganesh
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 1510 Clifton Rd, Atlanta, GA, 30322, Georgia.
| |
Collapse
|
55
|
El Dor M, Dakik H, Polomski M, Haudebourg E, Brachet M, Gouilleux F, Prié G, Zibara K, Mazurier F. VAS3947 Induces UPR-Mediated Apoptosis through Cysteine Thiol Alkylation in AML Cell Lines. Int J Mol Sci 2020; 21:ijms21155470. [PMID: 32751795 PMCID: PMC7432790 DOI: 10.3390/ijms21155470] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 07/25/2020] [Accepted: 07/28/2020] [Indexed: 12/16/2022] Open
Abstract
Nicotinamide adenine dinucleotide phosphate (NADPH) oxidases (NOX) involvement has been established in the oncogenic cell signaling of acute myeloid leukemia (AML) cells and in the crosstalk with their niche. We have shown an expression of NOX subunits in AML cell lines while NOX activity is lacking in the absence of exogenous stimulation. Here, we used AML cell lines as models to investigate the specificity of VAS3947, a current NOX inhibitor. Results demonstrated that VAS3947 induces apoptosis in AML cells independently of its anti-NOX activity. High-performance liquid chromatography (HPLC) and mass spectrometry analyses revealed that VAS3947 thiol alkylates cysteine residues of glutathione (GSH), while also interacting with proteins. Remarkably, VAS3947 decreased detectable GSH in the MV-4-11 cell line, thereby suggesting possible oxidative stress induction. However, a decrease in both cytoplasmic and mitochondrial reactive oxygen species (ROS) levels was observed by flow cytometry without disturbance of mitochondrial mass and membrane potential. Thus, assuming the consequences of VAS3947 treatment on protein structure, we examined its impact on endoplasmic reticulum (ER) stress. An acute unfolded protein response (UPR) was triggered shortly after VAS3947 exposure, through the activation of inositol-requiring enzyme 1α (IRE1α) and PKR-like endoplasmic reticulum kinase (PERK) pathways. Overall, VAS3947 induces apoptosis independently of anti-NOX activity, via UPR activation, mainly due to aggregation and misfolding of proteins.
Collapse
Affiliation(s)
- Maya El Dor
- EA 7501 GICC, University of Tours, CNRS ERL7001 LNOx, Bâtiment Dutrochet, 10 boulevard Tonnellé, BP3223, CEDEX 1, 37032 Tours, France; (M.E.D.); (H.D.); (M.B.); (F.G.)
- PRASE, Beirut, Lebanon;
| | - Hassan Dakik
- EA 7501 GICC, University of Tours, CNRS ERL7001 LNOx, Bâtiment Dutrochet, 10 boulevard Tonnellé, BP3223, CEDEX 1, 37032 Tours, France; (M.E.D.); (H.D.); (M.B.); (F.G.)
| | - Marion Polomski
- EA 7501 GICC, University of Tours, IMT, 31 Avenue Monge, 37200 Tours, France; (M.P.); (E.H.); (G.P.)
| | - Eloi Haudebourg
- EA 7501 GICC, University of Tours, IMT, 31 Avenue Monge, 37200 Tours, France; (M.P.); (E.H.); (G.P.)
| | - Marie Brachet
- EA 7501 GICC, University of Tours, CNRS ERL7001 LNOx, Bâtiment Dutrochet, 10 boulevard Tonnellé, BP3223, CEDEX 1, 37032 Tours, France; (M.E.D.); (H.D.); (M.B.); (F.G.)
| | - Fabrice Gouilleux
- EA 7501 GICC, University of Tours, CNRS ERL7001 LNOx, Bâtiment Dutrochet, 10 boulevard Tonnellé, BP3223, CEDEX 1, 37032 Tours, France; (M.E.D.); (H.D.); (M.B.); (F.G.)
| | - Gildas Prié
- EA 7501 GICC, University of Tours, IMT, 31 Avenue Monge, 37200 Tours, France; (M.P.); (E.H.); (G.P.)
| | - Kazem Zibara
- PRASE, Beirut, Lebanon;
- Biology Department, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon
| | - Frédéric Mazurier
- EA 7501 GICC, University of Tours, CNRS ERL7001 LNOx, Bâtiment Dutrochet, 10 boulevard Tonnellé, BP3223, CEDEX 1, 37032 Tours, France; (M.E.D.); (H.D.); (M.B.); (F.G.)
- EA 7501 GICC, University of Tours, IMT, 31 Avenue Monge, 37200 Tours, France; (M.P.); (E.H.); (G.P.)
- Correspondence: ; Tel.: +33-2-47-36-60-75
| |
Collapse
|
56
|
Bovine Dialyzable Leukocyte Extract IMMUNEPOTENT-CRP Induces Selective ROS-Dependent Apoptosis in T-Acute Lymphoblastic Leukemia Cell Lines. JOURNAL OF ONCOLOGY 2020; 2020:1598503. [PMID: 32587616 PMCID: PMC7298273 DOI: 10.1155/2020/1598503] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 03/15/2020] [Accepted: 03/31/2020] [Indexed: 12/12/2022]
Abstract
Immunotherapies strengthen the immune system to fight multiple diseases such as infections, immunodeficiencies, and autoimmune diseases, and recently, they are being used as an adjuvant in cancer treatment. IMMUNEPOTENT-CRP (I-CRP) is an immunotherapy made of bovine dialyzable leukocyte extract (bDLE) that has chemoprotective and immunomodulatory effects in different cellular populations of the immune system and antitumor activity in different cancer cell lines. Our recent results suggest that the antineoplastic effect of I-CRP is due to the characteristics of cancer cells. To confirm, we evaluated whether the selectivity is due to cell lineage or characteristics of cancer cells, testing cytotoxicity in T-acute lymphoblastic leukemia cells and their cell death mechanism. Here, we assessed the effect of I-CRP on cell viability and cell death. To determine the mechanism of cell death, we tested cell cycle, mitochondrial and nuclear alterations, and caspases and reactive oxygen species (ROS) and their role in cell death mechanism. Our results show that I-CRP does not affect cell viability in noncancer cells and induces selective cytotoxicity in a dose-dependent manner in leukemic cell lines. I-CRP also induces mitochondrial damage through proapoptotic and antiapoptotic protein modulation (Bax and Bcl-2) and ROS production, nuclear alterations including DNA damage (γ-H2Ax), overexpression of p53, cell cycle arrest, and DNA degradation. I-CRP induced ROS-dependent apoptosis in leukemic cells. Overall, here, we show that I-CRP cytotoxicity is selective to leukemic cells, inducing ROS-dependent apoptosis. This research opens the door to further exploration of their role in the immune system and the cell death mechanism that could potentially work in conjunction with other therapies including hematological malignances.
Collapse
|
57
|
Transient receptor potential ion channel TRPM2 promotes AML proliferation and survival through modulation of mitochondrial function, ROS, and autophagy. Cell Death Dis 2020; 11:247. [PMID: 32312983 PMCID: PMC7170900 DOI: 10.1038/s41419-020-2454-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 04/02/2020] [Accepted: 04/06/2020] [Indexed: 01/01/2023]
Abstract
Transient receptor potential melastatin 2 (TRPM2) ion channel has an essential function in maintaining cell survival following oxidant injury. Here, we show that TRPM2 is highly expressed in acute myeloid leukemia (AML). The role of TRPM2 in AML was studied following depletion with CRISPR/Cas9 technology in U937 cells. In in vitro experiments and in xenografts, depletion of TRPM2 in AML inhibited leukemia proliferation, and doxorubicin sensitivity was increased. Mitochondrial function including oxygen consumption rate and ATP production was reduced, impairing cellular bioenergetics. Mitochondrial membrane potential and mitochondrial calcium uptake were significantly decreased in depleted cells. Mitochondrial reactive oxygen species (ROS) were significantly increased, and Nrf2 was decreased, reducing the antioxidant response. In TRPM2-depleted cells, ULK1, Atg7, and Atg5 protein levels were decreased, leading to autophagy inhibition. Consistently, ATF4 and CREB, two master transcription factors for autophagosome biogenesis, were reduced in TRPM2-depleted cells. In addition, Atg13 and FIP200, which are known to stabilize ULK1 protein, were decreased. Reconstitution with TRPM2 fully restored proliferation, viability, and autophagy; ATF4 and CREB fully restored proliferation and viability but only partially restored autophagy. TRPM2 expression reduced the elevated ROS found in depleted cells. These data show that TRPM2 has an important role in AML proliferation and survival through regulation of key transcription factors and target genes involved in mitochondrial function, bioenergetics, the antioxidant response, and autophagy. Targeting TRPM2 may represent a novel therapeutic approach to inhibit myeloid leukemia growth and enhance susceptibility to chemotherapeutic agents through multiple pathways.
Collapse
|
58
|
Robinson AJ, Hopkins GL, Rastogi N, Hodges M, Doyle M, Davies S, Hole PS, Omidvar N, Darley RL, Tonks A. Reactive Oxygen Species Drive Proliferation in Acute Myeloid Leukemia via the Glycolytic Regulator PFKFB3. Cancer Res 2020; 80:937-949. [PMID: 31862780 PMCID: PMC7611211 DOI: 10.1158/0008-5472.can-19-1920] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 11/15/2019] [Accepted: 12/17/2019] [Indexed: 02/07/2023]
Abstract
Acute myeloid leukemia (AML) is a heterogeneous clonal disorder with a poor clinical outcome. Previously, we showed that overproduction of reactive oxygen species (ROS), arising from constitutive activation of NOX2 oxidase, occurs in >60% of patients with AML and that ROS production promotes proliferation of AML cells. We show here that the process most significantly affected by ROS overproduction is glycolysis. Whole metabolome analysis of 20 human primary AML showed that blasts generating high levels of ROS have increased glucose uptake and correspondingly increased glucose metabolism. In support of this, exogenous ROS increased glucose consumption while inhibition of NOX2 oxidase decreased glucose consumption. Mechanistically, ROS promoted uncoupling protein 2 (UCP2) protein expression and phosphorylation of AMPK, upregulating the expression of a key regulatory glycolytic enzyme, 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase (PFKFB3). Overexpression of PFKFB3 promoted glucose uptake and cell proliferation, whereas downregulation of PFKFB3 strongly suppressed leukemia growth both in vitro and in vivo in the NSG model. These experiments provide direct evidence that oxidase-derived ROS promotes the growth of leukemia cells via the glycolytic regulator PFKFB3. Targeting PFKFB3 may therefore present a new mode of therapy for this disease with a poor outcome. SIGNIFICANCE: These findings show that ROS generated by NOX2 in AML cells promotes glycolysis by activating PFKFB3 and suggest PFKFB3 as a novel therapeutic target in AML.
Collapse
Affiliation(s)
- Andrew J Robinson
- Department of Haematology, Division of Cancer & Genetics, School of Medicine, Cardiff University, Wales, United Kingdom
| | - Goitseone L Hopkins
- Department of Haematology, Division of Cancer & Genetics, School of Medicine, Cardiff University, Wales, United Kingdom
| | - Namrata Rastogi
- Department of Haematology, Division of Cancer & Genetics, School of Medicine, Cardiff University, Wales, United Kingdom
| | - Marie Hodges
- Department of Haematology, Division of Cancer & Genetics, School of Medicine, Cardiff University, Wales, United Kingdom
- Cardiff Experimental and Cancer Medicine Centre (ECMC), School of Medicine, Cardiff University, Wales, United Kingdom
| | - Michelle Doyle
- Department of Haematology, Division of Cancer & Genetics, School of Medicine, Cardiff University, Wales, United Kingdom
- Cardiff Experimental and Cancer Medicine Centre (ECMC), School of Medicine, Cardiff University, Wales, United Kingdom
| | - Sara Davies
- Department of Haematology, Division of Cancer & Genetics, School of Medicine, Cardiff University, Wales, United Kingdom
| | - Paul S Hole
- Department of Haematology, Division of Cancer & Genetics, School of Medicine, Cardiff University, Wales, United Kingdom
| | - Nader Omidvar
- Department of Haematology, Division of Cancer & Genetics, School of Medicine, Cardiff University, Wales, United Kingdom
| | - Richard L Darley
- Department of Haematology, Division of Cancer & Genetics, School of Medicine, Cardiff University, Wales, United Kingdom
| | - Alex Tonks
- Department of Haematology, Division of Cancer & Genetics, School of Medicine, Cardiff University, Wales, United Kingdom.
| |
Collapse
|
59
|
Sun DL, Poddar S, Pan RD, Rosser EW, Abt ER, Van Valkenburgh J, Le TM, Lok V, Hernandez SP, Song J, Li J, Turlik A, Chen X, Cheng CA, Chen W, Mona CE, Stuparu AD, Vergnes L, Reue K, Damoiseaux R, Zink JI, Czernin J, Donahue TR, Houk KN, Jung ME, Radu CG. Isoquinoline thiosemicarbazone displays potent anticancer activity with in vivo efficacy against aggressive leukemias. RSC Med Chem 2020; 11:392-410. [PMID: 33479645 DOI: 10.1039/c9md00594c] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Accepted: 02/12/2020] [Indexed: 11/21/2022] Open
Abstract
A potent class of isoquinoline-based α-N-heterocyclic carboxaldehyde thiosemicarbazone (HCT) compounds has been rediscovered; based upon this scaffold, three series of antiproliferative agents were synthesized through iterative rounds of methylation and fluorination modifications, with anticancer activities being potentiated by physiologically relevant levels of copper. The lead compound, HCT-13, was highly potent against a panel of pancreatic, small cell lung carcinoma, prostate cancer, and leukemia models, with IC50 values in the low-to-mid nanomolar range. Density functional theory (DFT) calculations showed that fluorination at the 6-position of HCT-13 was beneficial for ligand-copper complex formation, stability, and ease of metal-center reduction. Through a chemical genomics screen, we identify DNA damage response/replication stress response (DDR/RSR) pathways, specifically those mediated by ataxia-telangiectasia and Rad3-related protein kinase (ATR), as potential compensatory mechanism(s) of action following HCT-13 treatment. We further show that the cytotoxicity of HCT-13 is copper-dependent, that it promotes mitochondrial electron transport chain (mtETC) dysfunction, induces production of reactive oxygen species (ROS), and selectively depletes guanosine nucleotide pools. Lastly, we identify metabolic hallmarks for therapeutic target stratification and demonstrate the in vivo efficacy of HCT-13 against aggressive models of acute leukemias in mice.
Collapse
Affiliation(s)
- Daniel L Sun
- Department of Molecular and Medical Pharmacology , University of California, Los Angeles , California 90095 , USA . .,Ahmanson Translational Imaging Division , University of California, Los Angeles , California 90095 , USA.,Department of Chemistry and Biochemistry , University of California, Los Angeles , California 90095 , USA .
| | - Soumya Poddar
- Department of Molecular and Medical Pharmacology , University of California, Los Angeles , California 90095 , USA . .,Ahmanson Translational Imaging Division , University of California, Los Angeles , California 90095 , USA
| | - Roy D Pan
- Department of Molecular and Medical Pharmacology , University of California, Los Angeles , California 90095 , USA . .,Ahmanson Translational Imaging Division , University of California, Los Angeles , California 90095 , USA.,Department of Chemistry and Biochemistry , University of California, Los Angeles , California 90095 , USA .
| | - Ethan W Rosser
- Department of Molecular and Medical Pharmacology , University of California, Los Angeles , California 90095 , USA . .,Ahmanson Translational Imaging Division , University of California, Los Angeles , California 90095 , USA.,Department of Chemistry and Biochemistry , University of California, Los Angeles , California 90095 , USA .
| | - Evan R Abt
- Department of Molecular and Medical Pharmacology , University of California, Los Angeles , California 90095 , USA . .,Ahmanson Translational Imaging Division , University of California, Los Angeles , California 90095 , USA
| | - Juno Van Valkenburgh
- Department of Molecular and Medical Pharmacology , University of California, Los Angeles , California 90095 , USA . .,Ahmanson Translational Imaging Division , University of California, Los Angeles , California 90095 , USA.,Department of Chemistry and Biochemistry , University of California, Los Angeles , California 90095 , USA .
| | - Thuc M Le
- Department of Molecular and Medical Pharmacology , University of California, Los Angeles , California 90095 , USA . .,Ahmanson Translational Imaging Division , University of California, Los Angeles , California 90095 , USA
| | - Vincent Lok
- Department of Molecular and Medical Pharmacology , University of California, Los Angeles , California 90095 , USA .
| | - Selena P Hernandez
- Department of Chemistry and Biochemistry , University of California, Los Angeles , California 90095 , USA .
| | - Janet Song
- Department of Molecular and Medical Pharmacology , University of California, Los Angeles , California 90095 , USA .
| | - Joanna Li
- Department of Molecular and Medical Pharmacology , University of California, Los Angeles , California 90095 , USA .
| | - Aneta Turlik
- Department of Chemistry and Biochemistry , University of California, Los Angeles , California 90095 , USA .
| | - Xiaohong Chen
- Department of Chemistry and Biochemistry , University of California, Los Angeles , California 90095 , USA .
| | - Chi-An Cheng
- Department of Chemistry and Biochemistry , University of California, Los Angeles , California 90095 , USA . .,Department of Bioengineering , University of California, Los Angeles , CA 90095 , USA
| | - Wei Chen
- Department of Chemistry and Biochemistry , University of California, Los Angeles , California 90095 , USA .
| | - Christine E Mona
- Department of Molecular and Medical Pharmacology , University of California, Los Angeles , California 90095 , USA . .,Ahmanson Translational Imaging Division , University of California, Los Angeles , California 90095 , USA
| | - Andreea D Stuparu
- Department of Molecular and Medical Pharmacology , University of California, Los Angeles , California 90095 , USA . .,Ahmanson Translational Imaging Division , University of California, Los Angeles , California 90095 , USA
| | - Laurent Vergnes
- Department of Human Genetics , David Geffen School of Medicine , University of California, Los Angeles , California 90095 , USA
| | - Karen Reue
- Department of Human Genetics , David Geffen School of Medicine , University of California, Los Angeles , California 90095 , USA.,Molecular Biology Institute , University of California, Los Angeles , California 90095 , USA
| | - Robert Damoiseaux
- UCLA Metabolomic Center , University of California, Los Angeles , Los Angeles , California 90095 , USA
| | - Jeffrey I Zink
- Department of Chemistry and Biochemistry , University of California, Los Angeles , California 90095 , USA .
| | - Johannes Czernin
- Department of Molecular and Medical Pharmacology , University of California, Los Angeles , California 90095 , USA . .,Ahmanson Translational Imaging Division , University of California, Los Angeles , California 90095 , USA
| | - Timothy R Donahue
- Department of Molecular and Medical Pharmacology , University of California, Los Angeles , California 90095 , USA . .,Ahmanson Translational Imaging Division , University of California, Los Angeles , California 90095 , USA.,Department of Surgery , University of California, Los Angeles , CA 90095 , USA
| | - Kendall N Houk
- Department of Chemistry and Biochemistry , University of California, Los Angeles , California 90095 , USA .
| | - Michael E Jung
- Department of Chemistry and Biochemistry , University of California, Los Angeles , California 90095 , USA .
| | - Caius G Radu
- Department of Molecular and Medical Pharmacology , University of California, Los Angeles , California 90095 , USA . .,Ahmanson Translational Imaging Division , University of California, Los Angeles , California 90095 , USA
| |
Collapse
|
60
|
Akiyama H, Umezawa Y, Watanabe D, Okada K, Ishida S, Nogami A, Miura O. Inhibition of USP9X Downregulates JAK2-V617F and Induces Apoptosis Synergistically with BH3 Mimetics Preferentially in Ruxolitinib-Persistent JAK2-V617F-Positive Leukemic Cells. Cancers (Basel) 2020; 12:cancers12020406. [PMID: 32050632 PMCID: PMC7072561 DOI: 10.3390/cancers12020406] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 01/29/2020] [Accepted: 02/06/2020] [Indexed: 01/17/2023] Open
Abstract
JAK2-V617F plays a key role in the pathogenesis of myeloproliferative neoplasm. However, its inhibitor ruxolitinib has shown limited clinical efficacies because of the ruxolitinib-persistent proliferation of JAK2-V617F-positive cells. We here demonstrate that the USP9X inhibitor WP1130 or EOAI3402143 (G9) inhibited proliferation and induced apoptosis more efficiently in cells dependent on JAK2-V617F than on cytokine-activated JAK2. WP1130 preferentially downregulated activated and autophosphorylated JAK2-V617F by enhancing its K63-linked polyubiquitination and inducing its aggresomal translocation to block downstream signaling. Furthermore, JAK2-V617F associated physically with USP9X in leukemic HEL cells. Induction of apoptosis by inhibition of USP9X was mediated through the intrinsic mitochondria-mediated pathway, synergistically enhanced by BH3 mimetics, prevented by overexpression of Bcl-xL, and required oxidative stress to activate stress-related MAP kinases p38 and JNK as well as DNA damage responses in HEL cells. Although autophosphorylated JAK2-V617F was resistant to WP1130 in the ruxolitinib-persistent HEL-R cells, these cells expressed Bcl-2 and Bcl-xL at lower levels and showed an increased sensitivity to WP1130 as well as BH3 mimetics as compared with ruxolitinib-naive HEL cells. Thus, USP9X represents a promising target along with anti-apoptotic Bcl-2 family members for novel therapeutic strategies against JAK2-V617F-positive myeloproliferative neoplasms, particularly under the ruxolitinib persistence conditions.
Collapse
Affiliation(s)
- Hiroki Akiyama
- Department of Hematology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8519, Japan; (H.A.); (Y.U.); (D.W.); (K.O.); (S.I.); (A.N.)
| | - Yoshihiro Umezawa
- Department of Hematology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8519, Japan; (H.A.); (Y.U.); (D.W.); (K.O.); (S.I.); (A.N.)
| | - Daisuke Watanabe
- Department of Hematology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8519, Japan; (H.A.); (Y.U.); (D.W.); (K.O.); (S.I.); (A.N.)
| | - Keigo Okada
- Department of Hematology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8519, Japan; (H.A.); (Y.U.); (D.W.); (K.O.); (S.I.); (A.N.)
| | - Shinya Ishida
- Department of Hematology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8519, Japan; (H.A.); (Y.U.); (D.W.); (K.O.); (S.I.); (A.N.)
| | - Ayako Nogami
- Department of Hematology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8519, Japan; (H.A.); (Y.U.); (D.W.); (K.O.); (S.I.); (A.N.)
- Department of Clinical Laboratory, Medical Hospital, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyoku, Tokyo 113-8519, Japan
| | - Osamu Miura
- Department of Hematology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8519, Japan; (H.A.); (Y.U.); (D.W.); (K.O.); (S.I.); (A.N.)
- Correspondence:
| |
Collapse
|
61
|
Inhibition of Xanthine Oxidoreductase Enhances the Potential of Tyrosine Kinase Inhibitors against Chronic Myeloid Leukemia. Antioxidants (Basel) 2020; 9:antiox9010074. [PMID: 31952182 PMCID: PMC7022995 DOI: 10.3390/antiox9010074] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 01/13/2020] [Accepted: 01/13/2020] [Indexed: 12/28/2022] Open
Abstract
Chronic myeloid leukemia (CML) is characterized by the expression of the oncogenic kinase BCR-ABL. Although tyrosine kinase inhibitors (TKIs) against BCR-ABL represent the standard therapeutic option for CML, resistances to TKIs can be a serious problem. Thus, the search for novel therapeutic approaches is still needed. CML cells show an increased ROS production, which is required for maintaining the BCR-ABL signaling cascade active. In line with that, reducing ROS levels could be an interesting therapeutic strategy for the clinical management of resistant CML. To analyze the therapeutic potential of xanthine oxidoreductase (XOR) in CML, we tested the effect of XOR inhibitor allopurinol. Here, we show for the first time the therapeutic potential of allopurinol against BCR-ABL-positive CML cells. Allopurinol reduces the proliferation and clonogenic ability of the CML model cell lines K562 and KCL22. More importantly, the combination of allopurinol with imatinib or nilotinib reduced cell proliferation in a synergistic manner. Moreover, the co-treatment arms hampered cell clonogenic capacity and induced cell death more strongly than each single-agent arm. The reduction of intracellular ROS levels and the attenuation of the BCR-ABL signaling cascade may explain these effects. Finally, the self-renewal potential of primary bone marrow cells from CML patients was also severely reduced especially by the combination of allopurinol with TKIs. In summary, here we show that XOR inhibition is an interesting therapeutic option for CML, which can enhance the effectiveness of the TKIs currently used in clinics.
Collapse
|
62
|
Al Ageeli E. Alterations of Mitochondria and Related Metabolic Pathways in Leukemia: A Narrative Review. SAUDI JOURNAL OF MEDICINE & MEDICAL SCIENCES 2019; 8:3-11. [PMID: 31929772 PMCID: PMC6945320 DOI: 10.4103/sjmms.sjmms_112_18] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 02/12/2019] [Accepted: 07/21/2019] [Indexed: 12/14/2022]
Abstract
Dysregulation of mitochondrial function often precedes malignant transformation of hematopoietic stem cells (HSCs). Mitochondria have a direct role in the maintenance of HSC functions. For example, D-2-hydroxyglutarate, generated due to the activity of mutated mitochondrial isocitrate dehydrogenase (IDH), has been implicated in the pathogenesis of leukemia. Furthermore, disturbances in the fatty acid breakdown and pyruvate oxidation are often seen in leukemic cells. These and other abnormalities expedite leukemogenesis and chemoresistance of leukemic cells. However, it needs to be elucidated whether these aberrations are the result or cause of leukemogenesis. Accordingly, for this review, a search was carried out in PubMed and Google Scholar databases until June 2019 to assess the relationship between metabolic pathways in altered mitochondria and leukemia development. In the present review, an overview of mitochondria-related mechanisms and their abnormalities in leukemia is presented, with mitochondrial pathways and factors, such as mitophagy, intermediary metabolism enzymes, oncometabolites and reactive oxygen species' generation, discussed as potential diagnostic and therapeutic targets in leukemia.
Collapse
Affiliation(s)
- Essam Al Ageeli
- Department of Medical Biochemistry (Medical Genetics), Faculty of Medicine, Jazan University, Jazan, Saudi Arabia
| |
Collapse
|
63
|
Rodríguez-García A, Morales ML, Garrido-García V, García-Baquero I, Leivas A, Carreño-Tarragona G, Sánchez R, Arenas A, Cedena T, Ayala RM, Bautista JM, Martínez-López J, Linares M. Protein Carbonylation in Patients with Myelodysplastic Syndrome: An Opportunity for Deferasirox Therapy. Antioxidants (Basel) 2019; 8:E508. [PMID: 31652983 PMCID: PMC6912333 DOI: 10.3390/antiox8110508] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 10/18/2019] [Accepted: 10/22/2019] [Indexed: 12/21/2022] Open
Abstract
Control of oxidative stress in the bone marrow (BM) is key for maintaining the interplay between self-renewal, proliferation, and differentiation of hematopoietic cells. Breakdown of this regulation can lead to diseases characterized by BM failure such as the myelodysplastic syndromes (MDS). To better understand the role of oxidative stress in MDS development, we compared protein carbonylation as an indicator of oxidative stress in the BM of patients with MDS and control subjects, and also patients with MDS under treatment with the iron chelator deferasirox (DFX). As expected, differences in the pattern of protein carbonylation were observed in BM samples between MDS patients and controls, with an increase in protein carbonylation in the former. Strikingly, patients under DFX treatment had lower levels of protein carbonylation in BM with respect to untreated patients. Proteomic analysis identified four proteins with high carbonylation levels in MDS BM cells. Finally, as oxidative stress-related signaling pathways can modulate the cell cycle through p53, we analyzed the expression of the p53 target gene p21 in BM cells, finding that it was significantly upregulated in patients with MDS and was significantly downregulated after DFX treatment. Overall, our results suggest that the fine-tuning of oxidative stress levels in the BM of patients with MDS might control malignant progression.
Collapse
Affiliation(s)
- Alba Rodríguez-García
- Department of Hematology, 16473 Hospital Universitario 12 de Octubre, Hematological Malignancies Clinical Research Unit H120-CNIO, 28041 Madrid, Spain.
| | - María Luz Morales
- Department of Hematology, 16473 Hospital Universitario 12 de Octubre, Hematological Malignancies Clinical Research Unit H120-CNIO, 28041 Madrid, Spain.
| | - Vanesa Garrido-García
- Department of Hematology, 16473 Hospital Universitario 12 de Octubre, Hematological Malignancies Clinical Research Unit H120-CNIO, 28041 Madrid, Spain.
| | - Irene García-Baquero
- Department of Hematology, 16473 Hospital Universitario 12 de Octubre, Hematological Malignancies Clinical Research Unit H120-CNIO, 28041 Madrid, Spain.
| | - Alejandra Leivas
- Department of Hematology, 16473 Hospital Universitario 12 de Octubre, Hematological Malignancies Clinical Research Unit H120-CNIO, 28041 Madrid, Spain.
| | - Gonzalo Carreño-Tarragona
- Department of Hematology, 16473 Hospital Universitario 12 de Octubre, Hematological Malignancies Clinical Research Unit H120-CNIO, 28041 Madrid, Spain.
| | - Ricardo Sánchez
- Department of Hematology, 16473 Hospital Universitario 12 de Octubre, Hematological Malignancies Clinical Research Unit H120-CNIO, 28041 Madrid, Spain.
| | - Alicia Arenas
- Department of Hematology, 16473 Hospital Universitario 12 de Octubre, Hematological Malignancies Clinical Research Unit H120-CNIO, 28041 Madrid, Spain.
| | - Teresa Cedena
- Department of Hematology, 16473 Hospital Universitario 12 de Octubre, Hematological Malignancies Clinical Research Unit H120-CNIO, 28041 Madrid, Spain.
| | - Rosa María Ayala
- Department of Hematology, 16473 Hospital Universitario 12 de Octubre, Hematological Malignancies Clinical Research Unit H120-CNIO, 28041 Madrid, Spain.
| | - José M Bautista
- Department of Biochemistry and Molecular Biology and Research Institute Hospital 12 de Octubre, Universidad Complutense de Madrid, Ciudad Universitaria, 28040 Madrid, Spain.
| | - Joaquín Martínez-López
- Department of Hematology, 16473 Hospital Universitario 12 de Octubre, Hematological Malignancies Clinical Research Unit H120-CNIO, 28041 Madrid, Spain.
- Department of Medicine, Universidad Complutense de Madrid, Ciudad Universitaria, 28040 Madrid, Spain.
| | - María Linares
- Department of Hematology, 16473 Hospital Universitario 12 de Octubre, Hematological Malignancies Clinical Research Unit H120-CNIO, 28041 Madrid, Spain.
- Department of Biochemistry and Molecular Biology and Research Institute Hospital 12 de Octubre, Universidad Complutense de Madrid, Ciudad Universitaria, 28040 Madrid, Spain.
| |
Collapse
|
64
|
Ammar M, Ben Mahmoud L, Medhaffar M, Ghozzi H, Sahnoun Z, Hakim A, Mseddi M, Elloumi M, Zeghal K. Relationship of oxidative stress in the resistance to imatinib in Tunisian patients with chronic myeloid leukemia: A retrospective study. J Clin Lab Anal 2019; 34:e23050. [PMID: 31617242 PMCID: PMC7031571 DOI: 10.1002/jcla.23050] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 08/23/2019] [Accepted: 09/13/2019] [Indexed: 01/08/2023] Open
Abstract
Background This work aimed to evaluate oxidative stress in chronic myeloid leukemia (CML) patients treated with tunisian (IM) vs controls and in CML patients with resistance to IM vs patients without resistance to IM. Methods The study included 40 CML patients and 34 controls. Of 40 patients with CML, 26 patients were developed in resistance to IM. The oxidant/antioxidant markers were evaluated by spectrophotometric methods for all used samples. Results For CML patients, increased malondialdehyde (MDA) and advanced oxidation protein products (AOPP) levels were found compared to controls (P < .001; P = .01). Higher catalase (CAT) activity (P = .048) and lower superoxide dismutase (SOD) and glutathione peroxidase (GPx) activities, reduced Glutathione (GSH) and vitamin C levels were found in CML patients (P < .001). The comparison between the resistant vs no‐resistant CML patients revealed higher MDA level (P = .02) and CAT and SOD activities in IM‐resistant patients (P = .04, P = .03). GPx activity was reduced (P = .04). Furthermore, increased mean ratio of MDA/GSH, MDA/GPx, and SOD/(GPx + CAT) was found in IM‐resistant patients as compared with no‐resistant (P = .01, P = .01, P = .035). The mean ratio of GPx/GSH in the IM‐resistant CML patients was lower than in IM no‐resistant one (P = .039). For IM‐resistant patients, we found negative correlation between MDA level and the ratio SOD/(CAT + GPx) (r = −0.46, P = .002); and positive correlation between SOD and (CAT + GPx) activities (r = 0.38, P = .06) and between GSH level and GPx activity (r = 0.53, P = .01). Conclusions Our results have shown a highly disturbed oxidative profile in IM‐resistant CML patients as compared to no‐resistant. The H2O2 has a key role in the resistance to IM treatment.
Collapse
Affiliation(s)
- Mariam Ammar
- Department of Pharmacology, Faculty of Medicine, University of Sfax, Sfax, Tunisia.,Faculty of Sciences of Sfax, University of Sfax, Sfax, Tunisia
| | - Lobna Ben Mahmoud
- Department of Pharmacology, Faculty of Medicine, University of Sfax, Sfax, Tunisia
| | - Moez Medhaffar
- Department of Hematology, Hedi Chaker Hospital, University of Sfax, Sfax, Tunisia
| | - Hanen Ghozzi
- Department of Pharmacology, Faculty of Medicine, University of Sfax, Sfax, Tunisia
| | - Zouheir Sahnoun
- Department of Pharmacology, Faculty of Medicine, University of Sfax, Sfax, Tunisia
| | - Ahmed Hakim
- Department of Pharmacology, Faculty of Medicine, University of Sfax, Sfax, Tunisia
| | - Malek Mseddi
- Department of Pharmacology, Faculty of Medicine, University of Sfax, Sfax, Tunisia
| | - Moez Elloumi
- Department of Hematology, Hedi Chaker Hospital, University of Sfax, Sfax, Tunisia
| | - Khaled Zeghal
- Department of Pharmacology, Faculty of Medicine, University of Sfax, Sfax, Tunisia
| |
Collapse
|
65
|
The Human Transient Receptor Potential Melastatin 2 Ion Channel Modulates ROS Through Nrf2. Sci Rep 2019; 9:14132. [PMID: 31575956 PMCID: PMC6773863 DOI: 10.1038/s41598-019-50661-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 09/16/2019] [Indexed: 12/20/2022] Open
Abstract
Transient receptor potential melastatin channel subfamily member 2 (TRPM2) has an essential role in protecting cell viability through modulation of oxidative stress. TRPM2 is highly expressed in cancer. When TRPM2 is inhibited, mitochondria are dysfunctional, ROS levels are increased, and cell viability is reduced. Here, the importance of NF-E2-related factor (Nrf2) in TRPM2-mediated suppression of oxidant stress was explored. In TRPM2 depleted cells, antioxidant cofactors glutathione, NADPH, and NADH were significantly reduced. Cytoplasmic and nuclear expression of Nrf2 and of IQGAP1, a modulator of Nrf2 stability regulated by intracellular calcium, were decreased. Antioxidant enzymes transcriptionally regulated by Nrf2 and involved in GSH, NADPH, and NADH generation were significantly lower including PRX1 and PRX3, GPX4, GSTP1, GCLC, and MTHFD2. The glutamine pathway leading to GSH production was suppressed, and ATP and GTP levels were impaired. Reconstitution with wild type TRPM2 or Nrf2, but not TRPM2 pore mutant E960D, rescued expression of enzymes downstream of Nrf2 and restored GSH and GTP. Cell viability, ROS, NADPH, NADH, and ATP levels were fully rescued by TRPM2 and partially by Nrf2. These data show that TRPM2 maintains cell survival following oxidative stress through modulation of antioxidant pathways and cofactors regulated by Nrf2.
Collapse
|
66
|
Hexokinase II inhibition by 3-bromopyruvate sensitizes myeloid leukemic cells K-562 to anti-leukemic drug, daunorubicin. Biosci Rep 2019; 39:BSR20190880. [PMID: 31506393 PMCID: PMC6757186 DOI: 10.1042/bsr20190880] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 07/19/2019] [Accepted: 08/12/2019] [Indexed: 12/31/2022] Open
Abstract
An increased metabolic flux towards Warburg phenotype promotes survival, proliferation and causes therapeutic resistance, in leukemic cells. Hexokinase-II (HK-II) is expressed predominantly in cancer cells, which promotes Warburg metabolic phenotype and protects the cancer cells from drug-induced apoptosis. The HK-II inhibitor 3- Bromopyruvate (3-BP) dissociates HK-II from mitochondrial complex, which leads to enhanced sensitization of leukemic cells to anti-leukemic drugs. In the present study, we analyzed the Warburg characteristics viz. HK-II expression, glucose uptake, endogenous reactive oxygen species (ROS) level of leukemic cell lines K-562 and THP-1 and then investigated if 3-BP can sensitize the leukemic cells K-562 to anti-leukemic drug Daunorubicin (DNR). We found that both K-562 and THP-1 cells have multi-fold high levels of HK-II, glucose uptake and endogenous ROS with respect to normal PBMCs. The combined treatment (CT) of 3-BP and DNR showed synergistic effect on the growth inhibition (GI) of K-562 and THP-1 cells. This growth inhibitory effect was attributed to 3-BP induced S-phase block and DNR induced G2/M block, resulted in reduced proliferation due to CT. Further, CT resulted in low HK-II level in mitochondrial fraction, high intracellular calcium and elevated apoptosis as compared with individual treatment of DNR and 3-BP. Moreover, CT caused enhanced DNA damage and hyperpolarized mitochondria, leading to cell death. Taken together, these results suggest that 3-BP synergises the anticancer effects of DNR in the chronic myeloid leukemic cell K-562, and may act as an effective adjuvant to anti-leukemic chemotherapy.
Collapse
|
67
|
Di Marcantonio D, Sykes SM. Flow Cytometric Analysis of Mitochondrial Reactive Oxygen Species in Murine Hematopoietic Stem and Progenitor Cells and MLL-AF9 Driven Leukemia. J Vis Exp 2019:10.3791/59593. [PMID: 31545325 PMCID: PMC7239511 DOI: 10.3791/59593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
We present a flow cytometric approach for analyzing mitochondrial ROS in various live bone marrow (BM)-derived stem and progenitor cell populations from healthy mice as well as mice with AML driven by MLL-AF9. Specifically, we describe a two-step cell staining process, whereby healthy or leukemia BM cells are first stained with a fluorogenic dye that detects mitochondrial superoxides, followed by staining with fluorochrome-linked monoclonal antibodies that are used to distinguish various healthy and malignant hematopoietic progenitor populations. We also provide a strategy for acquiring and analyzing the samples by flow cytometry. The entire protocol can be carried out in a timeframe as short as 3-4 h. We also highlight the key variables to consider as well as the advantages and limitations of monitoring ROS production in the mitochondrial compartment of live hematopoietic and leukemia stem and progenitor subpopulations using fluorogenic dyes by flow cytometry. Furthermore, we present data that mitochondrial ROS abundance varies among distinct healthy HSPC sub-populations and leukemia progenitors and discuss the possible applications of this technique in hematologic research.
Collapse
Affiliation(s)
| | - Stephen M Sykes
- Blood Cell Development and Function Program, Fox Chase Cancer Center;
| |
Collapse
|
68
|
Jin Y, Lv M, Tao Y, Xu S, He J, Zhang J, Zhao W. A water-soluble BODIPY-based fluorescent probe for rapid and selective detection of hypochlorous acid in living cells. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2019; 219:569-575. [PMID: 31085435 DOI: 10.1016/j.saa.2019.04.085] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 04/29/2019] [Accepted: 04/29/2019] [Indexed: 06/09/2023]
Abstract
We designed and synthesized 4,4-di-(4'-methylmercaptophenoxy)-8-(N-methylpyridinium-2-yl)-1,3,5,7-tetramethyl-4-bora-3a,4a-diaza-s-indacene (probe 1) as a water-soluble BODIPY derivative for rapid and selective detection of hypochlorous acid. The pyridinium-2-yl linked at the meso position of BODIPY core was used to maintain highly fluorescent nature and to increase water solubility. Methylmercaptophenoxy was selected as responsive site installed on the boron atom (to replace the fluorine atom) and induced the photoinduced electron-transfer (PeT) effect to quench the fluorescence of BODIPY. The probe exhibited 83.9 μg mL-1 solubility in PBS (10 mM, pH 7.4), and possessed very low fluorescence (Φf = 0.0013). Upon addition of HClO, the probe could display a distinct response in 1 min and generate marked fluorescence enhancement by 100-fold due to the oxidation of thioether into sulfoxide to terminate PeT process. A limit of detection of 53 nM was calculated for HClO in the linear response range from 0 μM to 10 μM, and the probe was successfully applied to image HClO in living cells.
Collapse
Affiliation(s)
- Yue Jin
- Key Laboratory for Special Functional Materials of Ministry of Education, School of Materials Science and Engineering, Henan University, Kaifeng 475004, PR China
| | - Minghuan Lv
- Key Laboratory for Special Functional Materials of Ministry of Education, School of Materials Science and Engineering, Henan University, Kaifeng 475004, PR China
| | - Yuanfang Tao
- Key Laboratory for Special Functional Materials of Ministry of Education, School of Materials Science and Engineering, Henan University, Kaifeng 475004, PR China
| | - Shuang Xu
- School of Pharmacy, Fudan University, Shanghai 201203, PR China
| | - Jinling He
- Key Laboratory for Special Functional Materials of Ministry of Education, School of Materials Science and Engineering, Henan University, Kaifeng 475004, PR China
| | - Jian Zhang
- Key Laboratory for Special Functional Materials of Ministry of Education, School of Materials Science and Engineering, Henan University, Kaifeng 475004, PR China.
| | - Weili Zhao
- Key Laboratory for Special Functional Materials of Ministry of Education, School of Materials Science and Engineering, Henan University, Kaifeng 475004, PR China; School of Pharmacy, Fudan University, Shanghai 201203, PR China.
| |
Collapse
|
69
|
Hosseini M, Rezvani HR, Aroua N, Bosc C, Farge T, Saland E, Guyonnet-Dupérat V, Zaghdoudi S, Jarrou L, Larrue C, Sabatier M, Mouchel PL, Gotanègre M, Piechaczyk M, Bossis G, Récher C, Sarry JE. Targeting Myeloperoxidase Disrupts Mitochondrial Redox Balance and Overcomes Cytarabine Resistance in Human Acute Myeloid Leukemia. Cancer Res 2019; 79:5191-5203. [PMID: 31358527 DOI: 10.1158/0008-5472.can-19-0515] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 05/29/2019] [Accepted: 07/19/2019] [Indexed: 11/16/2022]
Abstract
Chemotherapies alter cellular redox balance and reactive oxygen species (ROS) content. Recent studies have reported that chemoresistant cells have an increased oxidative state in hematologic malignancies. In this study, we demonstrated that chemoresistant acute myeloid leukemia (AML) cells had a lower level of mitochondrial and cytosolic ROS in response to cytarabine (AraC) and overexpressed myeloperoxidase (MPO), a heme protein that converts hydrogen peroxide to hypochlorous acid (HOCl), compared with sensitive AML cells. High MPO-expressing AML cells were less sensitive to AraC in vitro and in vivo. They also produced higher levels of HOCl and exhibited an increased rate of mitochondrial oxygen consumption when compared with low MPO-expressing AML cells. Targeting MPO expression or enzyme activity sensitized AML cells to AraC treatment by triggering oxidative damage and sustaining oxidative stress, particularly in high MPO-expressing AML cells. This sensitization stemmed from mitochondrial superoxide accumulation, which impaired oxidative phosphorylation and cellular energetic balance, driving apoptotic death and selective eradication of chemoresistant AML cells in vitro and in vivo. Altogether, this study uncovers a noncanonical function of MPO enzyme in maintaining redox balance and mitochondrial energetic metabolism, therefore affecting downstream pathways involved in AML chemoresistance. SIGNIFICANCE: These findings demonstrate the role of myeloperoxidase in the regulation of ROS levels and sensitivity of AML cells to cytarabine, an essential chemotherapeutic backbone in the therapy of AML.
Collapse
Affiliation(s)
- Mohsen Hosseini
- Centre de Recherches en Cancérologie de Toulouse, UMR1037, Inserm, Equipe Labellisée LIGUE 2018, Toulouse, France.,University of Toulouse, Toulouse, France
| | - Hamid Reza Rezvani
- INSERM U1035, Bordeaux, France.,Université de Bordeaux, Bordeaux, France
| | - Nesrine Aroua
- Centre de Recherches en Cancérologie de Toulouse, UMR1037, Inserm, Equipe Labellisée LIGUE 2018, Toulouse, France.,University of Toulouse, Toulouse, France
| | - Claudie Bosc
- Centre de Recherches en Cancérologie de Toulouse, UMR1037, Inserm, Equipe Labellisée LIGUE 2018, Toulouse, France.,University of Toulouse, Toulouse, France
| | - Thomas Farge
- Centre de Recherches en Cancérologie de Toulouse, UMR1037, Inserm, Equipe Labellisée LIGUE 2018, Toulouse, France.,University of Toulouse, Toulouse, France
| | - Estelle Saland
- Centre de Recherches en Cancérologie de Toulouse, UMR1037, Inserm, Equipe Labellisée LIGUE 2018, Toulouse, France.,University of Toulouse, Toulouse, France
| | | | - Sonia Zaghdoudi
- Centre de Recherches en Cancérologie de Toulouse, UMR1037, Inserm, Equipe Labellisée LIGUE 2018, Toulouse, France.,University of Toulouse, Toulouse, France
| | - Latifa Jarrou
- Centre de Recherches en Cancérologie de Toulouse, UMR1037, Inserm, Equipe Labellisée LIGUE 2018, Toulouse, France.,University of Toulouse, Toulouse, France
| | - Clément Larrue
- Centre de Recherches en Cancérologie de Toulouse, UMR1037, Inserm, Equipe Labellisée LIGUE 2018, Toulouse, France.,University of Toulouse, Toulouse, France
| | - Marie Sabatier
- Centre de Recherches en Cancérologie de Toulouse, UMR1037, Inserm, Equipe Labellisée LIGUE 2018, Toulouse, France.,University of Toulouse, Toulouse, France
| | - Pierre Luc Mouchel
- Centre de Recherches en Cancérologie de Toulouse, UMR1037, Inserm, Equipe Labellisée LIGUE 2018, Toulouse, France.,University of Toulouse, Toulouse, France.,Service d'Hématologie, Institut Universitaire du Cancer de Toulouse-Oncopole, CHU de Toulouse, Toulouse, France
| | - Mathilde Gotanègre
- Centre de Recherches en Cancérologie de Toulouse, UMR1037, Inserm, Equipe Labellisée LIGUE 2018, Toulouse, France.,University of Toulouse, Toulouse, France
| | - Marc Piechaczyk
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Equipe Labellisée LIGUE, Montpellier, France
| | - Guillaume Bossis
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Equipe Labellisée LIGUE, Montpellier, France
| | - Christian Récher
- Centre de Recherches en Cancérologie de Toulouse, UMR1037, Inserm, Equipe Labellisée LIGUE 2018, Toulouse, France.,University of Toulouse, Toulouse, France.,Service d'Hématologie, Institut Universitaire du Cancer de Toulouse-Oncopole, CHU de Toulouse, Toulouse, France
| | - Jean-Emmanuel Sarry
- Centre de Recherches en Cancérologie de Toulouse, UMR1037, Inserm, Equipe Labellisée LIGUE 2018, Toulouse, France. .,University of Toulouse, Toulouse, France
| |
Collapse
|
70
|
Akiyama H, Umezawa Y, Ishida S, Okada K, Nogami A, Miura O. Inhibition of USP9X induces apoptosis in FLT3-ITD-positive AML cells cooperatively by inhibiting the mutant kinase through aggresomal translocation and inducing oxidative stress. Cancer Lett 2019; 453:84-94. [DOI: 10.1016/j.canlet.2019.03.046] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 03/12/2019] [Accepted: 03/28/2019] [Indexed: 12/14/2022]
|
71
|
Overview of thioredoxin system and targeted therapies for acute leukemia. Mitochondrion 2019; 47:38-46. [PMID: 31029641 DOI: 10.1016/j.mito.2019.04.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 03/15/2019] [Accepted: 04/24/2019] [Indexed: 12/11/2022]
|
72
|
Riccio P, Sessa R, de Nicola S, Petruzziello F, Trombetti S, Menna G, Pepe G, Maddalena P, Izzo P, Grosso M. GATA-1 isoforms differently contribute to the production and compartmentation of reactive oxygen species in the myeloid leukemia cell line K562. J Cell Physiol 2019; 234:20829-20846. [PMID: 31049966 PMCID: PMC6767011 DOI: 10.1002/jcp.28688] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 03/12/2019] [Accepted: 03/19/2019] [Indexed: 12/11/2022]
Abstract
Maintenance of a balanced expression of the two isoforms of the transcription factor GATA‐1, the full‐length protein (GATA‐1FL) and a shorter isoform (GATA‐1
S), contributes to control hematopoiesis, whereas their dysregulation can alter the differentiation/proliferation potential of hematopoietic precursors thereby eventually leading to a variety of hematopoietic disorders. Although it is well established that these isoforms play opposite roles in these remarkable processes, most of the molecular pathways involved remain unknown. Here, we demonstrate that GATA‐1FL and GATA‐1S are able to differently influence intracellular redox states and reactive oxygen species (ROS) compartmentation in the erythroleukemic K562 cell line, thus shedding novel mechanistic insights into the processes of cell proliferation and apoptosis resistance in myeloid precursors. Furthermore, given the role played by ROS signaling as a strategy to escape apoptosis and evade cell‐mediated immunity in myeloid cells, this study highlights a mechanism through which aberrant expression of GATA‐1 isoforms could play a role in the leukemogenic process.
Collapse
Affiliation(s)
- Patrizia Riccio
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Raffaele Sessa
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Sergio de Nicola
- CNR-SPIN, National Research Council, Institute for Superconductors, Innovative Materials and Devices, Naples, Italy
| | - Fara Petruzziello
- Pediatric Hematology Unit, Santobono-Pausilipon Hospital, Naples, Italy
| | - Silvia Trombetti
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Giuseppe Menna
- Pediatric Hematology Unit, Santobono-Pausilipon Hospital, Naples, Italy
| | - Giampiero Pepe
- CNR-SPIN, National Research Council, Institute for Superconductors, Innovative Materials and Devices, Naples, Italy.,Department of Physics, University of Naples Federico II, Naples, Italy
| | | | - Paola Izzo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Michela Grosso
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| |
Collapse
|
73
|
Identification of ortho-naphthoquinones as anti-AML agents by highly efficient oxidation of phenols. Bioorg Chem 2019; 86:97-102. [DOI: 10.1016/j.bioorg.2019.01.025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 01/12/2019] [Accepted: 01/12/2019] [Indexed: 01/07/2023]
|
74
|
Abstract
The TRP ion channel TRPM2 has an essential function in cell survival and protects the viability of a number of cell types after oxidative stress. It is highly expressed in many cancers including breast, prostate, and pancreatic cancer, melanoma, leukemia, and neuroblastoma, suggesting it promotes cancer cell survival. TRPM2 is activated by production of ADP-ribose (ADPR) following oxidative stress, which binds to the C-terminus of TRPM2, resulting in channel opening. In a number of cancers including neuroblastoma, TRPM2 has been shown to preserve viability and mechanisms have been identified. Activation of TRPM2 results in expression of transcription factors and kinases important in cell proliferation and survival including HIF-1/2α, CREB, nuclear factor (erythroid-derived 2)-related factor-2 (Nrf2), and Pyk2, and Src phosphorylation. Together, HIF-1/2α and CREB regulate expression of genes encoding proteins with roles in mitochondrial function including members of the electron transport complex involved in ATP production. These contribute to lower mitochondrial ROS production while expression of antioxidants regulated by HIF-1/2α, FOXO3a, CREB, and Nrf2 is maintained. CREB is also important in control of expression of key proteins involved in autophagy. When TRPM2-mediated calcium influx is inhibited, mitochondria are dysfunctional, cellular bioenergetics are reduced, production of ROS is increased, and autophagy and DNA repair are impaired, decreasing tumor growth and increasing chemotherapy sensitivity. Inhibition of TRPM2 expression or function results in decreased tumor proliferation and/or viability in many malignancies including breast, gastric, pancreatic, prostate, head and neck cancers, melanoma, neuroblastoma, and T-cell and acute myelogenous leukemia. However, in a small number of malignancies, activation of TRPM2 rather than inhibition has been reported to reduce tumor cell survival. Here, TRPM2-mediated Ca2+ signaling and mechanisms of regulation of cancer cell growth and survival are reviewed and controversies discussed. Evidence suggests that targeting TRPM2 may be a novel therapeutic approach in many cancers.
Collapse
Affiliation(s)
- Barbara A Miller
- Departments of Pediatrics, and Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, P.O. Box 850, Hershey, PA 17033, USA.
| |
Collapse
|
75
|
Lenzi M, Cocchi V, Novaković A, Karaman M, Sakač M, Mandić A, Pojić M, Barbalace MC, Angeloni C, Hrelia P, Malaguti M, Hrelia S. Meripilus giganteus ethanolic extract exhibits pro-apoptotic and anti-proliferative effects in leukemic cell lines. Altern Ther Health Med 2018; 18:300. [PMID: 30419892 PMCID: PMC6233556 DOI: 10.1186/s12906-018-2366-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 10/30/2018] [Indexed: 01/19/2023]
Abstract
Background The interest towards botanicals and plant extracts has strongly risen due to their numerous biological effects and ability to counteract chronic diseases development. Among these effects, chemoprevention which represents the possibility to counteract the cancerogenetic process is one of the most studied. The extracts of mushroom Meripilus giganteus (MG) (Phylum of Basidiomycota) showed to exert antimicrobic, antioxidant and antiproliferative effects. Therefore, since its effect in leukemic cell lines has not been previously evaluated, we studied its potential chemopreventive effect in Jurkat and HL-60 cell lines. Methods MG ethanolic extract was characterized for its antioxidant activity and scavenging effect against different radical species. Moreover, its phenolic profile was evaluated by HPLC-MS-MS analyses. Flow cytometry (FCM) analyses of Jurkat and HL-60 cells treated with MG extract (0–750 μg/mL) for 24–72 h- allowed to evaluate its cytotoxicity, pro-apoptotic and anti-proliferative effect. To better characterize MG pro-apoptotic mechanism ROS intracellular level and the gene expression level of FAS, BAX and BCL2 were also evaluated. Moreover, to assess MG extract selectivity towards cancer cells, its cytotoxicity was also evaluated in human peripheral blood lymphocytes (PBL). Results MG extract induced apoptosis in Jurkat and HL-60 cells in a dose- and time- dependent manner by increasing BAX/BCL2 ratio, reducing ROS intracellular level and inducing FAS gene expression level. In fact, reduced ROS level is known to be related to the activation of apoptosis in leukemic cells by the involvement of death receptors. MG extract also induced cell-cycle arrest in HL-60 cells. Moreover, IC50 at 24 h treatment resulted 2 times higher in PBL than in leukemic cell lines. Conclusions Our data suggest that MG extract might be considered a promising and partially selective chemopreventive agent since it is able to modulate different mechanisms in transformed cells at concentrations lower than in non-transformed ones.
Collapse
|
76
|
Kim C, Lee SG, Yang WM, Arfuso F, Um JY, Kumar AP, Bian J, Sethi G, Ahn KS. Formononetin-induced oxidative stress abrogates the activation of STAT3/5 signaling axis and suppresses the tumor growth in multiple myeloma preclinical model. Cancer Lett 2018; 431:123-141. [DOI: 10.1016/j.canlet.2018.05.038] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Revised: 05/23/2018] [Accepted: 05/23/2018] [Indexed: 01/13/2023]
|
77
|
Prieto-Bermejo R, Romo-González M, Pérez-Fernández A, Ijurko C, Hernández-Hernández Á. Reactive oxygen species in haematopoiesis: leukaemic cells take a walk on the wild side. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:125. [PMID: 29940987 PMCID: PMC6019308 DOI: 10.1186/s13046-018-0797-0] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 06/15/2018] [Indexed: 02/08/2023]
Abstract
Oxidative stress is related to ageing and degenerative diseases, including cancer. However, a moderate amount of reactive oxygen species (ROS) is required for the regulation of cellular signalling and gene expression. A low level of ROS is important for maintaining quiescence and the differentiation potential of haematopoietic stem cells (HSCs), whereas the level of ROS increases during haematopoietic differentiation; thus, suggesting the importance of redox signalling in haematopoiesis. Here, we will analyse the importance of ROS for haematopoiesis and include evidence showing that cells from leukaemia patients live under oxidative stress. The potential sources of ROS will be described. Finally, the level of oxidative stress in leukaemic cells can also be harnessed for therapeutic purposes. In this regard, the reliance of front-line anti-leukaemia chemotherapeutics on increased levels of ROS for their mechanism of action, as well as the active search for novel compounds that modulate the redox state of leukaemic cells, will be analysed.
Collapse
Affiliation(s)
- Rodrigo Prieto-Bermejo
- Department of Biochemistry and Molecular Biology, University of Salamanca, Lab. 122, Edificio Departamental, Plaza Doctores de la Reina s/n, 37007, Salamanca, Spain.,IBSAL (Instituto de investigación Biomédica de Salamanca), Salamanca, Spain
| | - Marta Romo-González
- Department of Biochemistry and Molecular Biology, University of Salamanca, Lab. 122, Edificio Departamental, Plaza Doctores de la Reina s/n, 37007, Salamanca, Spain.,IBSAL (Instituto de investigación Biomédica de Salamanca), Salamanca, Spain
| | - Alejandro Pérez-Fernández
- Department of Biochemistry and Molecular Biology, University of Salamanca, Lab. 122, Edificio Departamental, Plaza Doctores de la Reina s/n, 37007, Salamanca, Spain.,IBSAL (Instituto de investigación Biomédica de Salamanca), Salamanca, Spain
| | - Carla Ijurko
- Department of Biochemistry and Molecular Biology, University of Salamanca, Lab. 122, Edificio Departamental, Plaza Doctores de la Reina s/n, 37007, Salamanca, Spain.,IBSAL (Instituto de investigación Biomédica de Salamanca), Salamanca, Spain
| | - Ángel Hernández-Hernández
- Department of Biochemistry and Molecular Biology, University of Salamanca, Lab. 122, Edificio Departamental, Plaza Doctores de la Reina s/n, 37007, Salamanca, Spain. .,IBSAL (Instituto de investigación Biomédica de Salamanca), Salamanca, Spain.
| |
Collapse
|
78
|
Zhang Y, Li L, Yu C, Senyuk V, Li F, Quigley JG, Zhu T, Qian Z. miR-9 upregulation leads to inhibition of erythropoiesis by repressing FoxO3. Sci Rep 2018; 8:6519. [PMID: 29695725 PMCID: PMC5916915 DOI: 10.1038/s41598-018-24628-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 04/04/2018] [Indexed: 12/20/2022] Open
Abstract
MicroRNAs (miRNAs) are emerging as critical regulators of normal and malignant hematopoiesis. In previous studies of acute myeloid leukemia miR-9 overexpression was commonly observed. Here, we show that ectopic expression of miR-9 in vitro and in vivo significantly blocks differentiation of erythroid progenitor cells with an increase in reactive oxygen species (ROS) production. Consistent with this observation, ROS scavenging enzymes, including superoxide dismutase (Sod2), Catalase (Cat), and glutathine peroxidase (Gpx1), are down-regulated by miR-9. In addition, miR-9 suppresses expression of the erythroid transcriptional regulator FoxO3, and its down-stream targets Btg1 and Cited 2 in erythroid progenitor cells, while expression of a constitutively active form of FoxO3 (FoxO3-3A) reverses miR-9-induced suppression of erythroid differentiation, and inhibits miR-9-induced ROS production. Thus, our findings indicate that aberrant expression of miR-9 blocks erythropoiesis by deregulating FoxO3-mediated pathways, which may contribute to the ineffective erythropoiesis observed in patients with hematological malignancies.
Collapse
Affiliation(s)
- Yunyuan Zhang
- Department of Clinical laboratory, The Affiliated Hospital of Qingdao University Medical College, Qingdao, 266003, China
- Department of Medicine and Cancer Research Center, University of Illinois Hospital and Health Sciences System, Chicago, IL, USA
| | - Liping Li
- Department of Medicine and Cancer Research Center, University of Illinois Hospital and Health Sciences System, Chicago, IL, USA
- Fudan University ZhongShan Hospital, Shanghai, China
| | - Chunjie Yu
- Department of Medicine and Cancer Research Center, University of Illinois Hospital and Health Sciences System, Chicago, IL, USA
| | - Vitalyi Senyuk
- Department of Medicine and Cancer Research Center, University of Illinois Hospital and Health Sciences System, Chicago, IL, USA
| | - Fuxing Li
- Department of Medicine and Cancer Research Center, University of Illinois Hospital and Health Sciences System, Chicago, IL, USA
- Department of Pediatrics, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - John G Quigley
- Department of Medicine and Cancer Research Center, University of Illinois Hospital and Health Sciences System, Chicago, IL, USA
| | - Tongyu Zhu
- Fudan University ZhongShan Hospital, Shanghai, China
| | - Zhijian Qian
- Department of Medicine and Cancer Research Center, University of Illinois Hospital and Health Sciences System, Chicago, IL, USA.
| |
Collapse
|
79
|
Samimi A, Kalantari H, Lorestani MZ, Shirzad R, Saki N. Oxidative stress in normal hematopoietic stem cells and leukemia. APMIS 2018; 126:284-294. [PMID: 29575200 DOI: 10.1111/apm.12822] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Accepted: 01/22/2018] [Indexed: 12/19/2022]
Abstract
Leukemia is developed following the abnormal proliferation of immature hematopoietic cells in the blood when hematopoietic stem cells lose the ability to turn into mature cells at different stages of maturation and differentiation. Leukemia initiating cells are specifically dependent upon the suppression of oxidative stress in the hypoglycemic bone marrow (BM) environment to be able to start their activities. Relevant literature was identified by a PubMed search (2000-2017) of English-language literature using the terms 'oxidative stress,' 'reactive oxygen species,' 'hematopoietic stem cell,' and 'leukemia.' The generation and degradation of free radicals is a main component of the metabolism in aerobic organisms. A certain level of ROS is required for proper cellular function, but values outside this range will result in oxidative stress (OS). Long-term overactivity of reactive oxygen species (ROS) has harmful effects on the function of cells and their vital macromolecules, including the transformation of proteins into autoantigens and increased degradation of protein/DNA, which eventually leads to the change in pathways involved in the development of cancer and several other disorders. According to the metabolic disorders of cancer, the relationship between OS changes, the viability of cancer cells, and their response to chemotherapeutic agents affecting this pathway are undeniable. Recently, studies have been conducted to determine the effect of herbal agents and cancer chemotherapy drugs on oxidative stress pathways. By emphasizing the role of oxidative stress on stem cells in the incidence of leukemia, this paper attempts to state and summarize this subject.
Collapse
Affiliation(s)
- Azin Samimi
- Department of Pharmacology and Toxicology, School of Pharmacy, Ahvaz Jundishpur University of Medical Sciences, Ahvaz, Iran
| | - Heybatullah Kalantari
- Department of Pharmacology and Toxicology, School of Pharmacy, Ahvaz Jundishpur University of Medical Sciences, Ahvaz, Iran
| | - Marzieh Zeinvand Lorestani
- Department of Pharmacology and Toxicology, School of Pharmacy, Ahvaz Jundishpur University of Medical Sciences, Ahvaz, Iran
| | - Reza Shirzad
- WHO-Collaborating Centre for Reference and Research on Rabies, Pasteur Institute of Iran, Tehran, Iran
| | - Najmaldin Saki
- Research Center of Thalassemia & Hemoglobinopathy, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
80
|
Ferroni P, Barbanti P, Della-Morte D, Palmirotta R, Jirillo E, Guadagni F. Redox Mechanisms in Migraine: Novel Therapeutics and Dietary Interventions. Antioxid Redox Signal 2018; 28:1144-1183. [PMID: 28990418 DOI: 10.1089/ars.2017.7260] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
SIGNIFICANCE Migraine represents the third most prevalent and the seventh most disabling human disorder. Approximately 30% of migraine patients experience transient, fully reversible, focal neurological symptoms (aura) preceding the attack. Recent Advances: Awareness of the hypothesis that migraine actually embodies a spectrum of illnesses-ranging from episodic to chronic forms-is progressively increasing and poses novel challenges for clarifying the underlying pathophysiological mechanisms of migraine as well as for the development of novel therapeutic interventions. Several theories have evolved to the current concept that a combination of genetic, epigenetic, and environmental factors may play a role in migraine pathogenesis, although their relative importance is still being debated. CRITICAL ISSUES One critical issue that deserves a particular attention is the role of oxidative stress in migraine. Indeed, potentially harmful oxidative events occur during the migraine attack and long-lasting or frequent migraine episodes may increase brain exposure to oxidative events that can lead to chronic transformation. Moreover, a wide variety of dietary, environmental, physiological, behavioral, and pharmacological migraine triggers may act through oxidative stress, with clear implications for migraine treatment and prophylaxis. Interestingly, almost all current prophylactic migraine agents exert antioxidant effects. FUTURE DIRECTIONS Increasing awareness of the role of oxidative stress and/or decreased antioxidant defenses in migraine pathogenesis and progression to a chronic condition lays the foundations for the design of novel prophylactic approaches, which, by reducing brain oxidative phenomena, could favorably modify the clinical course of migraine. Antioxid. Redox Signal. 28, 1144-1183.
Collapse
Affiliation(s)
- Patrizia Ferroni
- 1 Department of Human Sciences and Quality of Life Promotion, San Raffaele Roma Open University , Rome, Italy
- 2 IRCCS San Raffaele Pisana , Rome, Italy
| | - Piero Barbanti
- 3 Headache and Pain Unit, Department of Neurological, Motor and Sensorial Sciences, IRCCS San Raffaele Pisana , Rome, Italy
| | - David Della-Morte
- 1 Department of Human Sciences and Quality of Life Promotion, San Raffaele Roma Open University , Rome, Italy
- 2 IRCCS San Raffaele Pisana , Rome, Italy
- 4 Department of Systems Medicine, University of Rome "Tor Vergata ," Rome, Italy
| | - Raffaele Palmirotta
- 5 Department of Biomedical Sciences and Human Oncology, "A. Moro" University , Bari, Italy
| | - Emilio Jirillo
- 6 Department of Basic Medical Sciences, Neuroscience and Sensory Organs, "A. Moro" University , Bari, Italy
| | - Fiorella Guadagni
- 1 Department of Human Sciences and Quality of Life Promotion, San Raffaele Roma Open University , Rome, Italy
- 2 IRCCS San Raffaele Pisana , Rome, Italy
| |
Collapse
|
81
|
Oncogenic STAT5 signaling promotes oxidative stress in chronic myeloid leukemia cells by repressing antioxidant defenses. Oncotarget 2018; 8:41876-41889. [PMID: 27566554 PMCID: PMC5522035 DOI: 10.18632/oncotarget.11480] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 07/28/2016] [Indexed: 12/12/2022] Open
Abstract
STAT5 transcription factors are frequently activated in hematopoietic neoplasms and are targets of various tyrosine kinase oncogenes. Evidences for a crosstalk between STAT5 and reactive oxygen species (ROS) metabolism have recently emerged but mechanisms involved in STAT5-mediated regulation of ROS still remain elusive. We demonstrate that sustained activation of STAT5 induced by Bcr-Abl in chronic myeloid leukemia (CML) cells promotes ROS production by repressing expression of two antioxidant enzymes, catalase and glutaredoxin-1(Glrx1). Downregulation of catalase and Glrx1 expression was also observed in primary cells from CML patients. Catalase was shown not only to reduce ROS levels but also, to induce quiescence in Bcr-Abl-positive leukemia cells. Furthermore, reduction of STAT5 phosphorylation and upregulation of catalase and Glrx1 were also evidenced in leukemia cells co-cultured with bone marrow stromal cells to mimic a leukemic niche. This caused downregulation of ROS levels and enhancement of leukemic cell quiescence. These data support a role of persistent STAT5 signaling in the regulation of ROS production in myeloid leukemias and highlight the repression of antioxidant defenses as an important regulatory mechanism.
Collapse
|
82
|
Dutta D, Chong NS, Lim SH. Endogenous volatile organic compounds in acute myeloid leukemia: origins and potential clinical applications. J Breath Res 2018; 12:034002. [PMID: 29463782 DOI: 10.1088/1752-7163/aab108] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Not unlike many cancer types, acute myeloid leukemia (AML) exhibits many metabolic changes and reprogramming, causing changes in lipid metabolism. Some of the distinct molecular abnormalities associated with AML also modify the metabolic changes. Both processes result in changes in the production of endogenous volatile organic compounds (VOCs). The increasing availability of highly sensitive methods for detecting trace chemicals provides the opportunity to investigate the role of patient-specific VOC finger-prints as biomarkers for detecting early relapse or minimal residual disease in AML. Since VOC production is reliant on metabolic activities, when combined with currently available methods, VOC analysis may identify within a group of patients with flow cytometric or molecular evidence of residual disease those most at risk for disease relapse.
Collapse
Affiliation(s)
- Dibyendu Dutta
- Department of Professional Sciences, Middle Tennessee State University, Murfreesboro, Tennessee, United States of America
| | | | | |
Collapse
|
83
|
Xing P, Feng Y, Niu Y, Li Q, Zhang Z, Dong L, Wang C. A Water-Soluble, Two-Photon Probe for Imaging Endogenous Hypochlorous Acid in Live Tissue. Chemistry 2018; 24:5748-5753. [DOI: 10.1002/chem.201800249] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Panfei Xing
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences; University of Macau; Avenida da Universidade Macau SAR P. R. China
| | - Yanxian Feng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences; University of Macau; Avenida da Universidade Macau SAR P. R. China
| | - Yiming Niu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences; University of Macau; Avenida da Universidade Macau SAR P. R. China
| | - Qiu Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences; University of Macau; Avenida da Universidade Macau SAR P. R. China
| | - Zhe Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences; University of Macau; Avenida da Universidade Macau SAR P. R. China
| | - Lei Dong
- State Key Laboratory of Pharmaceutical Biotechnology; Nanjing University Institution; Nanjing 210093 P. R. China
| | - Chunming Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences; University of Macau; Avenida da Universidade Macau SAR P. R. China
| |
Collapse
|
84
|
Mahdavi M, Asghari S, Rahnamay M, Dehghan G, Feizi MAH, Balalaie S. Cytotoxicity, oxidative stress, and apoptosis in K562 leukemia cells induced by an active compound from pyrano-pyridine derivatives. Hum Exp Toxicol 2018; 37:1105-1116. [DOI: 10.1177/0960327118756719] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Recent studies have reported the potential of pyrano-pyridine compounds in inhibiting cell growth and apoptosis induction in cancer cells. Here, we investigated the effect of new pyrano-pyridine derivatives on proliferation, oxidative damages, and apoptosis in K562 leukemia cells. Among different tested compounds, we found 8-(4-chlorobenzylidene)-2-amino-4-(4-chlorophenyl)-5, 6, 7, 8-tetrahydro-6-phenethyl-4H-pyrano-[3,2-c]pyridine-3-carbonitrile (4-CP.P) as the most effective compound with IC50 value of 20 μM. Gel electrophoresis, fluorescence microscopy, and flow cytometry analyses indicated the apoptosis induction ability of 4-CP.P in K562 cells. Further analyses revealed that 4-CP.P induces significant increase in cellular reactive oxygen species production, lipid peroxidation, protein oxidation, and total thiol depletion. Interestingly, while 4-CP.P significantly increased the activity of superoxide dismutase, it reduced the catalase activity in a time-dependent manner. These data propose that 4-CP.P treatment causes free radicals accumulation that ultimately leads to oxidative stress condition and apoptosis induction. Therefore, we report the 4-CP.P as a novel, potent compound as a chemotherapeutic agent in cancer treatment.
Collapse
Affiliation(s)
- M Mahdavi
- Department of Biology, Faculty of Natural Science, University of Tabriz, Tabriz, Iran
| | - S Asghari
- Department of Biology, Faculty of Natural Science, University of Tabriz, Tabriz, Iran
| | - M Rahnamay
- Department of Biology, Faculty of Natural Science, University of Tabriz, Tabriz, Iran
| | - G Dehghan
- Department of Biology, Faculty of Natural Science, University of Tabriz, Tabriz, Iran
| | - MAH Feizi
- Department of Biology, Faculty of Natural Science, University of Tabriz, Tabriz, Iran
| | - S Balalaie
- Department of Chemistry, Faculty of Science, K. N. Toosi University of Tech, Tehran, Iran
| |
Collapse
|
85
|
Cuapio A, Post M, Cerny-Reiterer S, Gleixner KV, Stefanzl G, Basilio J, Herndlhofer S, Sperr WR, Brons NHC, Casanova E, Zimmer J, Valent P, Hofer E. Maintenance therapy with histamine plus IL-2 induces a striking expansion of two CD56bright NK cell subpopulations in patients with acute myeloid leukemia and supports their activation. Oncotarget 2018; 7:46466-46481. [PMID: 27341131 PMCID: PMC5216810 DOI: 10.18632/oncotarget.10191] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 05/28/2016] [Indexed: 11/25/2022] Open
Abstract
Histamine dihydrochloride (HDC) plus IL-2 has been proposed as a novel maintenance-immunotherapy in acute myeloid leukemia (AML). We analyzed the immunophenotype and function of natural killer (NK) cells in blood of AML patients treated after chemotherapy with HDC plus IL-2. The treatment caused a striking expansion of CD56brightCD16neg and CD56brightCD16low NK cell subpopulations. A reduced NK cell fraction recovered and high proportions of cells expressed the activating receptors NKG2D, NKp30, and NKp46. Concomitantly, KIR-expressing NK cells were reduced and NK cells with inhibitory NKG2A/CD94 receptors increased beyond normal levels. In addition, the immunotherapy-induced NK cells exhibited high capacity to produce IFN-γ and to degranulate. Furthermore, we provide evidence from subsequent in vitro studies that this is caused in part by direct effects of IL-2 on the CD56bright cells. IL-2 specifically induced proliferation of both CD56bright subpopulations, but not of CD56dim cells. It further preserved the expression of activating receptors and the capacity to produce IFN-γ and to degranulate. These data suggest that therapy with HDC plus IL-2 supports the reconstitution of a deficient NK cell fraction through the specific amplification of CD56bright NK cells giving rise to a functional NK cell compartment with high potential to combat leukemic disease.
Collapse
Affiliation(s)
- Angélica Cuapio
- Department of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | - Mirte Post
- Department of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | - Sabine Cerny-Reiterer
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Vienna, Austria.,Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Vienna, Austria
| | - Karoline V Gleixner
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Vienna, Austria.,Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Vienna, Austria
| | - Gabriele Stefanzl
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Jose Basilio
- Department of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | - Susanne Herndlhofer
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Vienna, Austria.,Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Vienna, Austria
| | - Wolfgang R Sperr
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Vienna, Austria.,Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Vienna, Austria
| | - Nicolaas H C Brons
- National Core Facility Cytometry, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Emilio Casanova
- Ludwig Boltzmann Institute of Cancer Research, Vienna, Austria.,Institute of Pharmacology, Center of Physiology and Pharmacology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Jacques Zimmer
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Peter Valent
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Vienna, Austria.,Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Vienna, Austria
| | - Erhard Hofer
- Department of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
86
|
Yang M, Xing S, Ou HL, Zhang L, Shen X, Xiong GL, Wang FM, Xiao H, Tu YH, Cong YW, Wang XR, Yu ZY. Vibsanol A induces differentiation of acute myeloid leukemia cells via activation of the PKC signaling pathway and induction of ROS. Leuk Lymphoma 2018; 59:2414-2422. [PMID: 29334822 DOI: 10.1080/10428194.2017.1421754] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Meng Yang
- Department of Pathophysiology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, China
- Department of Clinical Laboratory, The General Hospital of PLA Rocket Force, Beijing, China
| | - Shuang Xing
- Department of Pathophysiology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Hong-Ling Ou
- Department of Clinical Laboratory, The General Hospital of PLA Rocket Force, Beijing, China
| | - Lu Zhang
- Department of Clinical Laboratory, The General Hospital of PLA Rocket Force, Beijing, China
| | - Xing Shen
- Department of Pathophysiology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Guo-Lin Xiong
- Department of Pathophysiology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Fang-Min Wang
- Department of Pathophysiology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, China
| | - He Xiao
- Department of Molecular Immunology, Institute of Basic Medical Sciences, Beijing, China
| | - Yan-Hong Tu
- Department of Otorhinolaryngology, First Hospital Affiliated to Anhui University of Chinese Medicine, Hefei, China
| | - Yu-Wen Cong
- Department of Pathophysiology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Xin-Ru Wang
- Department of Clinical Laboratory, The General Hospital of PLA Rocket Force, Beijing, China
| | - Zu-Yin Yu
- Department of Pathophysiology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, China
- Department of Graduates, Anhui Medical University, Hefei, China
| |
Collapse
|
87
|
Thivakaran A, Botezatu L, Hönes JM, Schütte J, Vassen L, Al-Matary YS, Patnana P, Zeller A, Heuser M, Thol F, Gabdoulline R, Olberding N, Frank D, Suslo M, Köster R, Lennartz K, Görgens A, Giebel B, Opalka B, Dührsen U, Khandanpour C. Gfi1b: a key player in the genesis and maintenance of acute myeloid leukemia and myelodysplastic syndrome. Haematologica 2018; 103:614-625. [PMID: 29326122 PMCID: PMC5865438 DOI: 10.3324/haematol.2017.167288] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 01/05/2018] [Indexed: 12/22/2022] Open
Abstract
Differentiation of hematopoietic stem cells is regulated by a concert of different transcription factors. Disturbed transcription factor function can be the basis of (pre)malignancies such as myelodysplastic syndrome (MDS) or acute myeloid leukemia (AML). Growth factor independence 1b (Gfi1b) is a repressing transcription factor regulating quiescence of hematopoietic stem cells and differentiation of erythrocytes and platelets. Here, we show that low expression of Gfi1b in blast cells is associated with an inferior prognosis of MDS and AML patients. Using different models of human MDS or AML, we demonstrate that AML development was accelerated with heterozygous loss of Gfi1b, and latency was further decreased when Gfi1b was conditionally deleted. Loss of Gfi1b significantly increased the number of leukemic stem cells with upregulation of genes involved in leukemia development. On a molecular level, we found that loss of Gfi1b led to epigenetic changes, increased levels of reactive oxygen species, as well as alteration in the p38/Akt/FoXO pathways. These results demonstrate that Gfi1b functions as an oncosuppressor in MDS and AML development.
Collapse
Affiliation(s)
- Aniththa Thivakaran
- Department of Haematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Lacramioara Botezatu
- Department of Haematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Judith M Hönes
- Department of Haematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.,Department of Endocrinology, Diabetes and Metabolism, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Judith Schütte
- Department of Haematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Lothar Vassen
- Department of Haematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Yahya S Al-Matary
- Department of Haematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Pradeep Patnana
- Department of Haematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Amos Zeller
- Department of Haematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Michael Heuser
- Department of Haematology, Haemostaseology, Oncology, and Stem Cell Transplantation, Medical University of Hannover, Germany
| | - Felicitas Thol
- Department of Haematology, Haemostaseology, Oncology, and Stem Cell Transplantation, Medical University of Hannover, Germany
| | - Razif Gabdoulline
- Department of Haematology, Haemostaseology, Oncology, and Stem Cell Transplantation, Medical University of Hannover, Germany
| | - Nadine Olberding
- Department of Haematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Daria Frank
- Department of Haematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Marina Suslo
- Department of Haematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Renata Köster
- Department of Haematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Klaus Lennartz
- Institute for Cell Biology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Andre Görgens
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.,Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Bernd Giebel
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Bertram Opalka
- Department of Haematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ulrich Dührsen
- Department of Haematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Cyrus Khandanpour
- Department of Haematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany .,Department of Medicine A, Hematology, Oncology and Pneumology, University Hospital Münster, Germany
| |
Collapse
|
88
|
Zhang J, Lei W, Chen X, Wang S, Qian W. Oxidative stress response induced by chemotherapy in leukemia treatment. Mol Clin Oncol 2018; 8:391-399. [PMID: 29599981 PMCID: PMC5867396 DOI: 10.3892/mco.2018.1549] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 12/06/2017] [Indexed: 12/14/2022] Open
Abstract
Oxidative stress (OS) has been linked to the etiology and development of leukemia as reactive oxygen species (ROS) and free radicals have been implicated in leukemogenesis. OS has beneficial and deleterious effects in the pathogenesis and progression of leukemia. High-dose chemotherapy, which is frequently used in leukemia treatment, is often accompanied by ROS-induced cytotoxicity. Thus, the utilization of chemotherapy in combination with antioxidants may attenuate leukemia progression, particularly for cases of refractory or relapsed neoplasms. The present review focuses on exploring the roles of OS in leukemogenesis and characterizing the associations between ROS and chemotherapy. Certain examples of treatment regimens wherein antioxidants are combined with chemotherapy are presented, in order to highlight the importance of antioxidant application in leukemia treatment, as well as the conflicting opinions regarding this method of therapy. Understanding the underlying mechanisms of OS generation will facilitate the elucidation of novel approaches to leukemia treatment.
Collapse
Affiliation(s)
- Jin Zhang
- Department of Hematology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, P.R. China
| | - Wen Lei
- Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Xiaohui Chen
- Department of Hematology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang 310015, P.R. China
| | - Shibing Wang
- Clinical Research Institute, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| | - Wenbin Qian
- Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| |
Collapse
|
89
|
Moloney JN, Jayavelu AK, Stanicka J, Roche SL, O'Brien RL, Scholl S, Böhmer FD, Cotter TG. Nuclear membrane-localised NOX4D generates pro-survival ROS in FLT3-ITD-expressing AML. Oncotarget 2017; 8:105440-105457. [PMID: 29285262 PMCID: PMC5739649 DOI: 10.18632/oncotarget.22241] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 10/02/2017] [Indexed: 12/21/2022] Open
Abstract
Internal tandem duplication of the juxtamembrane domain of FMS-like tyrosine kinase 3 (FLT3-ITD) is the most prevalent genetic aberration present in 20-30% of acute myeloid leukaemia (AML) cases and is associated with a poor prognosis. FLT3-ITD expressing cells express elevated levels of NADPH oxidase 4 (NOX4)-generated pro-survival hydrogen peroxide (H2O2) contributing to increased levels of DNA oxidation and double strand breaks. NOX4 is constitutively active and has been found to have various isoforms expressed at multiple locations within a cell. The purpose of this study was to investigate the expression, localisation and regulation of NOX4 28 kDa splice variant, NOX4D. NOX4D has previously been shown to localise to the nucleus and nucleolus in various cell types and is implicated in the generation of reactive oxygen species (ROS) and DNA damage. Here, we demonstrate that FLT3-ITD expressing-AML patient samples as well as -cell lines express the NOX4D isoform resulting in elevated H2O2 levels compared to FLT3-WT expressing cells, as quantified by flow cytometry. Cell fractionation indicated that NOX4D is nuclear membrane-localised in FLT3-ITD expressing cells. Treatment of MV4-11 cells with receptor trafficking inhibitors, tunicamycin and brefeldin A, resulted in deglycosylation of NOX4 and NOX4D. Inhibition of the FLT3 receptor revealed that the FLT3-ITD oncogene is responsible for the production of NOX4D-generated H2O2 in AML. We found that inhibition of the PI3K/AKT and STAT5 pathways resulted in down-regulation of NOX4D-generated pro-survival ROS. Taken together these findings indicate that nuclear membrane-localised NOX4D-generated pro-survival H2O2 may be contributing to genetic instability in FLT3-ITD expressing AML.
Collapse
Affiliation(s)
- Jennifer N Moloney
- Tumour Biology Laboratory, School of Biochemistry and Cell Biology, Bioscience Research Institute, University College Cork, Cork, Ireland
| | - Ashok Kumar Jayavelu
- Institute of Molecular Cell Biology, CMB, Jena University Hospital, Jena, Germany.,Current address: Department of Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany
| | - Joanna Stanicka
- Tumour Biology Laboratory, School of Biochemistry and Cell Biology, Bioscience Research Institute, University College Cork, Cork, Ireland
| | - Sarah L Roche
- Tumour Biology Laboratory, School of Biochemistry and Cell Biology, Bioscience Research Institute, University College Cork, Cork, Ireland
| | - Rebecca L O'Brien
- Tumour Biology Laboratory, School of Biochemistry and Cell Biology, Bioscience Research Institute, University College Cork, Cork, Ireland
| | - Sebastian Scholl
- Department of Haematology/Oncology, Clinic for Internal Medicine II, Jena University Hospital, Jena, Germany
| | - Frank-D Böhmer
- Institute of Molecular Cell Biology, CMB, Jena University Hospital, Jena, Germany
| | - Thomas G Cotter
- Tumour Biology Laboratory, School of Biochemistry and Cell Biology, Bioscience Research Institute, University College Cork, Cork, Ireland
| |
Collapse
|
90
|
Liu Y, Jia Q, Guo Q, Jiang A, Zhou J. In Vivo Oxidative Stress Monitoring Through Intracellular Hydroxyl Radicals Detection by Recyclable Upconversion Nanoprobes. Anal Chem 2017; 89:12299-12305. [DOI: 10.1021/acs.analchem.7b03270] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Yuxin Liu
- Department of Chemistry, Capital Normal University, Beijing 100048, China
| | - Qi Jia
- Department of Chemistry, Capital Normal University, Beijing 100048, China
| | - Quanwei Guo
- Department of Chemistry, Capital Normal University, Beijing 100048, China
| | - Anqi Jiang
- Department of Chemistry, Capital Normal University, Beijing 100048, China
| | - Jing Zhou
- Department of Chemistry, Capital Normal University, Beijing 100048, China
| |
Collapse
|
91
|
Novel Cell-Killing Mechanisms of Hydroxyurea and the Implication toward Combination Therapy for the Treatment of Fungal Infections. Antimicrob Agents Chemother 2017; 61:AAC.00734-17. [PMID: 28893786 DOI: 10.1128/aac.00734-17] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 08/28/2017] [Indexed: 01/17/2023] Open
Abstract
We have previously reported that an erg11 mutation affecting ergosterol synthesis and a hem13 mutation in the heme synthesis pathway significantly sensitize the fission yeast Schizosaccharomyces pombe to hydroxyurea (HU) (1, 2). Here we show that treatment with inhibitors of Erg11 and heme biosynthesis phenocopies the two mutations in sensitizing wild-type cells to HU. Importantly, HU synergistically interacts with the heme biosynthesis inhibitor sampangine and several Erg11 inhibitors, the antifungal azoles, in causing cell lethality. Since the synergistic drug interactions are also observed in the phylogenetically divergent Saccharomyces cerevisiae and the opportunistic fungal pathogen Candida albicans, the synergism is likely conserved in eukaryotes. Interestingly, our genetic data for S. pombe has also led to the discovery of a robust synergism between sampangine and the azoles in C. albicans Thus, combinations of HU, sampangine, and the azoles can be further studied as a new method for the treatment of fungal infections.
Collapse
|
92
|
Cherukula K, Nurunnabi M, Jeong YY, Lee YK, Park IK. A targeted graphene nanoplatform carrying histamine dihydrochloride for effective inhibition of leukemia-induced immunosuppression. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2017; 29:734-749. [DOI: 10.1080/09205063.2017.1390382] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Kondareddy Cherukula
- Department of Biomedical Science and BK21 PLUS Centre for Creative Biomedical Scientists, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Md. Nurunnabi
- Department of Green Bioengineering, Korea National University of Transportation, Chungju, Republic of Korea
| | - Yong Yeon Jeong
- Department of Radiology, Chonnam National University Medical School, Chonnam National University Hwasun Hospital, Gwangju, Republic of Korea
| | - Yong-Kyu Lee
- Department of Green Bioengineering, Korea National University of Transportation, Chungju, Republic of Korea
| | - In-Kyu Park
- Department of Biomedical Science and BK21 PLUS Centre for Creative Biomedical Scientists, Chonnam National University Medical School, Gwangju, Republic of Korea
| |
Collapse
|
93
|
Yin HY, Tang J, Zhang JL. Introducing Metallosalens into Biological Studies: The Renaissance of Traditional Coordination Complexes. Eur J Inorg Chem 2017. [DOI: 10.1002/ejic.201700695] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Hao-Yan Yin
- Beijing National Laboratory for Molecular Sciences; State Key Laboratory of Rare Earth Materials Chemistry and Applications; College of Chemistry and Molecular Engineering; Peking University; 100871 Beijing P. R. China
| | - Juan Tang
- Beijing National Laboratory for Molecular Sciences; State Key Laboratory of Rare Earth Materials Chemistry and Applications; College of Chemistry and Molecular Engineering; Peking University; 100871 Beijing P. R. China
| | - Jun-Long Zhang
- Beijing National Laboratory for Molecular Sciences; State Key Laboratory of Rare Earth Materials Chemistry and Applications; College of Chemistry and Molecular Engineering; Peking University; 100871 Beijing P. R. China
| |
Collapse
|
94
|
Should every Patient with MDS get Iron Chelation - Probably Yes. Mediterr J Hematol Infect Dis 2017; 9:e2017055. [PMID: 28894564 PMCID: PMC5584764 DOI: 10.4084/mjhid.2017.055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 07/21/2017] [Indexed: 12/20/2022] Open
|
95
|
Linher-Melville K, Singh G. The complex roles of STAT3 and STAT5 in maintaining redox balance: Lessons from STAT-mediated xCT expression in cancer cells. Mol Cell Endocrinol 2017; 451:40-52. [PMID: 28202313 DOI: 10.1016/j.mce.2017.02.014] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 02/09/2017] [Indexed: 12/12/2022]
Abstract
STAT3 and STAT5 mediate diverse cellular processes, transcriptionally regulating gene expression and interacting with cytoplasmic proteins. Their canonical activity is stimulated by cytokines/growth factors through JAK-STAT signaling. As targets of oncogenes with intrinsic tyrosine kinase activity, STAT3 and STAT5 become constitutively active in hematologic neoplasms and solid tumors, promoting cell proliferation and survival and modulating redox homeostasis. This review summarizes reactive oxygen species (ROS)-regulated STAT activation and how STATs influence ROS production. ROS-induced effects on post-translational modifications are presented, and STAT3/5-mediated regulation of xCT, a redox-sensitive target up-regulated in numerous cancers, is discussed with regard to transcriptional cross-talk.
Collapse
Affiliation(s)
- Katja Linher-Melville
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4L8, Canada
| | - Gurmit Singh
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4L8, Canada.
| |
Collapse
|
96
|
Pilo F, Angelucci E. A storm in the niche: Iron, oxidative stress and haemopoiesis. Blood Rev 2017; 32:29-35. [PMID: 28847531 DOI: 10.1016/j.blre.2017.08.005] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 08/05/2017] [Accepted: 08/11/2017] [Indexed: 12/18/2022]
Abstract
Iron, although essential, is harmful in high amounts. Oxidative stress as a result of excess reactive oxygen species (ROS) and a prooxidative/antioxidative imbalance between ROS production and elimination, play a key role in cellular damage. There is evidence to support the role of ROS in the pathogenesis of a range of diseases including the myelodysplastic syndromes (MDS) and leukaemia. Oxidative stress seems to affect the self-renewal, proliferation and differentiation of haematopoietic stem cells and impair cell growth. Three aspects of these defective haemopoietic mechanisms may be associated with the activities of ROS: clonal evolution, haematological improvement and recovery of haemopoiesis after haematopoietic stem cell transplantation (HSCT). This review aims to provide haematologists with an overview of results from in vitro and murine models and preliminary clinical evidence on the diagnostic, prognostic and therapeutic implications of the complex interactions between the haemopoietic niche, iron, oxidative stress and inadequate haemopoiesis.
Collapse
Affiliation(s)
- Federica Pilo
- Hematology and Transplant Center, Ospedale Oncologico di Riferimento Regionale "Armando Businco", Azienda Ospedaliera Brotzu, Cagliari, Italy..
| | - Emanuele Angelucci
- Hematology and Transplant Center, Ospedale Policlinico San Martino, Genova, Italy.
| |
Collapse
|
97
|
Prokocimer M, Molchadsky A, Rotter V. Dysfunctional diversity of p53 proteins in adult acute myeloid leukemia: projections on diagnostic workup and therapy. Blood 2017; 130:699-712. [PMID: 28607134 PMCID: PMC5659817 DOI: 10.1182/blood-2017-02-763086] [Citation(s) in RCA: 125] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Accepted: 06/06/2017] [Indexed: 12/13/2022] Open
Abstract
The heterogeneous nature of acute myeloid leukemia (AML) and its poor prognosis necessitate therapeutic improvement. Current advances in AML research yield important insights regarding AML genetic, epigenetic, evolutional, and clinical diversity, all in which dysfunctional p53 plays a key role. As p53 is central to hematopoietic stem cell functions, its aberrations affect AML evolution, biology, and therapy response and usually predict poor prognosis. While in human solid tumors TP53 is mutated in more than half of cases, TP53 mutations occur in less than one tenth of de novo AML cases. Nevertheless, wild-type (wt) p53 dysfunction due to nonmutational p53 abnormalities appears to be rather frequent in various AML entities, bearing, presumably, a greater impact than is currently appreciated. Hereby, we advocate assessment of adult AML with respect to coexisting p53 alterations. Accordingly, we focus not only on the effects of mutant p53 oncogenic gain of function but also on the mechanisms underlying nonmutational wtp53 inactivation, which might be of therapeutic relevance. Patient-specific TP53 genotyping with functional evaluation of p53 protein may contribute significantly to the precise assessment of p53 status in AML, thus leading to the tailoring of a rationalized and precision p53-based therapy. The resolution of the mechanisms underlying p53 dysfunction will better address the p53-targeted therapies that are currently considered for AML. Additionally, a suggested novel algorithm for p53-based diagnostic workup in AML is presented, aiming at facilitating the p53-based therapeutic choices.
Collapse
MESH Headings
- Adult
- Animals
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- DNA Damage/drug effects
- Gene Expression Regulation, Leukemic/drug effects
- Genomic Instability/drug effects
- Hematopoiesis/drug effects
- Humans
- Karyopherins/genetics
- Karyopherins/metabolism
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Molecular Targeted Therapy/methods
- Mutation/drug effects
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Nucleophosmin
- Protein Interaction Maps/drug effects
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/metabolism
- Signal Transduction/drug effects
- Translocation, Genetic
- Tumor Suppressor Protein p53/analysis
- Tumor Suppressor Protein p53/genetics
- Tumor Suppressor Protein p53/metabolism
- fms-Like Tyrosine Kinase 3/genetics
- fms-Like Tyrosine Kinase 3/metabolism
- Exportin 1 Protein
Collapse
Affiliation(s)
- Miron Prokocimer
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; and
| | - Alina Molchadsky
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot, Israel
| | - Varda Rotter
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
98
|
Jetly S, Verma N, Naidu K, Faiq MA, Seth T, Saluja D. Alterations in the Reactive Oxygen Species in Peripheral Blood of Chronic Myeloid Leukaemia Patients from Northern India. J Clin Diagn Res 2017; 11:XC01-XC05. [PMID: 28969255 PMCID: PMC5620896 DOI: 10.7860/jcdr/2017/28565.10425] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Accepted: 07/21/2017] [Indexed: 01/09/2023]
Abstract
INTRODUCTION There is a significant difference in the Reactive Oxygen Species (ROS) levels of Chronic Myeloid Leukaemia (CML) patients before and during treatment with Tyrosine Kinase Inhibitors (TKIs). This is because high ROS levels support oncogenic phenotype of CML by inducing proliferation pathway and accumulation of further genetic mutations. Often the measurement is done on WBC or serum for ascertaining one type of ROS species, but measurement of global ROS in fresh whole blood will give more accurate estimation of ROS. AIM To measure global ROS in peripheral blood of CML patients. MATERIALS AND METHODS A case control study was undertaken to measure ROS in peripheral blood of CML patients from Northern India. CML patients on TKIs (n=40 on imatinib herein called treated) and untreated (n=17, who were not on any TKIs or alternative medicine, called as treatment naive) and 52 healthy controls were also enrolled. Chemiluminescent assay was carried out using luminol as signal enhancer in 400 µl of blood to measure ROS. The chemiluminescence was measured as Relative Light Units (RLU)/sec/104 WBC. Data was presented in terms of mean±SE or geometric mean (95% Confidence Interval) for continuous variables and percentage for categorical variables. Groups were compared using two sample t-test for continuous variables and chi-square test for categorical variables. RESULTS The WBC profile and ROS levels of patients taking TKIs were quite similar and showed no significant difference (p<0.999) compared to healthy controls. In contrast, significant increase was observed in the ROS levels of CML patients not on TKIs (untreated) compared to patients under treatment (p<0.029) and healthy controls (p<0.007). We also observed that the absolute ROS values and WBC counts were higher in untreated patients compared to patients on TKIs and healthy controls, even though mean ROS value was less. CONCLUSION To ascertain the alterations in ROS levels of CML patients before and during treatment with TKIs, it is better to measure global ROS in fresh whole blood by chemiluminescent method using luminol. Luminol assay is a quick, easy and inexpensive method to measure global ROS. Patient under treatment with TKIs show significant decrease in ROS levels almost similar to the levels measured in healthy controls yet the mechanisms by which this decrease occurs needs to be elucidated.
Collapse
Affiliation(s)
- Sunita Jetly
- Associate Professor, Department of Biotechnology, Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi, New Delhi, India
| | - Neha Verma
- Project Fellow, Department of Biotechnology, Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi, New Delhi, India
| | - Kumar Naidu
- Statistician, Clinical Research and Development Department, IPCA Laboratories Ltd, Mumbai, India
| | - Muneeb Ahmad Faiq
- Research Fellow, Dr. Rajendra Prasad Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, New Delhi, India
| | - Tulika Seth
- Professor, Department of Hematology, All India Institute of Medical Sciences, New Delhi, India
| | - Daman Saluja
- Professor, Department of Biotechnology, Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi, New Delhi, India
| |
Collapse
|
99
|
Park S, Park JA, Yoo H, Park HB, Lee Y. Proteasome inhibitor-induced cleavage of HSP90 is mediated by ROS generation and caspase 10-activation in human leukemic cells. Redox Biol 2017; 13:470-476. [PMID: 28715732 PMCID: PMC5512190 DOI: 10.1016/j.redox.2017.07.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2017] [Revised: 07/07/2017] [Accepted: 07/11/2017] [Indexed: 12/26/2022] Open
Abstract
Heat shock protein 90 (HSP90) is a molecular chaperone that supports the stability of client proteins. The proteasome is one of the targets for cancer therapy, and studies are underway to use proteasome inhibitors as anti-cancer drugs. In this study, we found that HSP90 was cleaved to a 55 kDa protein after treatment with proteasome inhibitors including MG132 in leukemia cells but was not cleaved in other tissue-derived cells. HSP90 has two major isoforms (HSP90α and HSP90β), and both were cleaved by MG132 treatment. MG132 treatment also induced a decrease in HSP90 client proteins. MG132 treatment generated ROS, and the cleavage of HSP90 was blocked by a ROS scavenger, N-acetylcysteine (NAC). MG132 activated several caspases, and the activation was reduced by pretreatment with NAC. Based on an inhibitor study, the cleavage of HSP90 induced by MG132 was dependent on caspase 10 activation. Furthermore, active recombinant caspase 10 induced HSP90 cleavage in vitro. MG132 upregulated VDUP-1 expression and reduced the GSH levels implying that the regulation of redox-related proteins is involved. Taken all together, our results suggest that the cleavage of HSP90 by MG132 treatment is mediated by ROS generation and caspase 10 activation. HSP90 cleavage may provide an additional mechanism involved in the anti-cancer effects of proteasome inhibitors. Proteasome inhibitors induce cleavage of HSP90. MG132 induces ROS generation via VDUP-1 upregulation and GSH downregulation. ROS-mediated active caspase 10 cleaves HSP90.
Collapse
Affiliation(s)
- Sangkyu Park
- Department of Biochemistry, College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea
| | - Jeong-A Park
- Department of Biochemistry, College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea
| | - Hwanmin Yoo
- Department of Biochemistry, College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea
| | - Han-Bum Park
- Department of Biochemistry, College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea
| | - Younghee Lee
- Department of Biochemistry, College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea.
| |
Collapse
|
100
|
Jin UH, Cheng Y, Zhou B, Safe S. Bardoxolone Methyl and a Related Triterpenoid Downregulate cMyc Expression in Leukemia Cells. Mol Pharmacol 2017; 91:438-450. [PMID: 28275049 PMCID: PMC5399643 DOI: 10.1124/mol.116.106245] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 02/10/2017] [Indexed: 11/22/2022] Open
Abstract
Structurally related pentacyclic triterpenoids methyl 2-cyano-3,12-dioxoolean-1,9-dien-28-oate [bardoxolone-methyl (Bar-Me)] and methyl 2-trifluoromethyl-3,11-dioxoolean-1,12-dien-30-oate (CF3DODA-Me) contain 2-cyano-1-en-3-one and 2-trifluoromethyl-1-en-3-one moieties, respectively, in their A-rings and differ in the position of their en-one structures in ring C. Only Bar-Me forms a Michael addition adduct with glutathione (GSH) and inhibits IKKβ phosphorylation. These differences may be due to steric hindrance by the 11-keto group in CF3DODA-Me, which prevents Michael addition by the conjugated en-one in the A-ring. In contrast, both Bar-Me and CF3DODA-Me induce reactive oxygen species in HL-60 and Jurkat leukemia cells, inhibit cell growth, induce apoptosis and differentiation, and decrease expression of specificity proteins (Sp) 1, 3, and 4, and cMyc, and these effects are significantly attenuated after cotreatment with the antioxidant GSH. In contrast to solid tumor-derived cells, cMyc and Sp transcriptions are regulated independently and cMyc plays a more predominant role than Sp transcription factors in regulating HL-60 or Jurkat cell proliferation and differentiation compared with that observed in cells derived from solid tumors.
Collapse
Affiliation(s)
- Un-Ho Jin
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas
| | - Yating Cheng
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas
| | - Beiyan Zhou
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas
| |
Collapse
|