1
|
Zhang Q, Falqués‐Costa T, Pilheden M, Sturesson H, Ovlund T, Rissler V, Castor A, Marquart HVH, Lausen B, Fioretos T, Hyrenius‐Wittsten A, Hagström‐Andersson AK. Activating mutations remodel the chromatin accessibility landscape to drive distinct regulatory networks in KMT2A-rearranged acute leukemia. Hemasphere 2024; 8:e70006. [PMID: 39329074 PMCID: PMC11426354 DOI: 10.1002/hem3.70006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 08/02/2024] [Accepted: 08/07/2024] [Indexed: 09/28/2024] Open
Abstract
Activating FLT3 and RAS mutations commonly occur in leukemia with KMT2A-gene rearrangements (KMT2A-r). However, how these mutations cooperate with the KMT2A-r to remodel the epigenetic landscape is unknown. Using a retroviral acute myeloid leukemia (AML) mouse model driven by KMT2A::MLLT3, we show that FLT3 ITD , FLT3 N676K , and NRAS G12D remodeled the chromatin accessibility landscape and associated transcriptional networks. Although the activating mutations shared a common core of chromatin changes, each mutation exhibits unique profiles with most opened peaks associating with enhancers in intronic or intergenic regions. Specifically, FLT3 N676K and NRAS G12D rewired similar chromatin and transcriptional networks, distinct from those mediated by FLT3 ITD . Motif analysis uncovered a role for the AP-1 family of transcription factors in KMT2A::MLLT3 leukemia with FLT3 N676K and NRAS G12D , whereas Runx1 and Stat5a/Stat5b were active in the presence of FLT3 ITD . Furthermore, transcriptional programs linked to immune cell regulation were activated in KMT2A-r AML expressing NRAS G12D or FLT3 N676K , and the expression of NKG2D-ligands on KMT2A-r cells rendered them sensitive to CAR T cell-mediated killing. Human KMT2A-r AML cells could be pharmacologically sensitized to NKG2D-CAR T cells by treatment with the histone deacetylase inhibitor LBH589 (panobinostat) which caused upregulation of NKG2D-ligand levels. Co-treatment with LBH589 and NKG2D-CAR T cells enabled robust AML cell killing, and the strongest effect was observed for cells expressing NRAS G12D . Finally, the results were validated and extended to acute leukemia in infancy. Combined, activating mutations induced mutation-specific changes in the epigenetic landscape, leading to changes in transcriptional programs orchestrated by specific transcription factor networks.
Collapse
Affiliation(s)
- Qirui Zhang
- Department of Laboratory Medicine, Division of Clinical GeneticsLund UniversityLundSweden
| | - Ton Falqués‐Costa
- Department of Laboratory Medicine, Division of Clinical GeneticsLund UniversityLundSweden
| | - Mattias Pilheden
- Department of Laboratory Medicine, Division of Clinical GeneticsLund UniversityLundSweden
| | - Helena Sturesson
- Department of Laboratory Medicine, Division of Clinical GeneticsLund UniversityLundSweden
| | - Tina Ovlund
- Department of Laboratory Medicine, Division of Clinical GeneticsLund UniversityLundSweden
| | - Vendela Rissler
- Department of Laboratory Medicine, Division of Clinical GeneticsLund UniversityLundSweden
| | - Anders Castor
- Childhood Cancer CenterSkåne University HospitalLundSweden
| | - Hanne V. H. Marquart
- Department of Clinical ImmunologyNational University HospitalRigshospitalet, CopenhagenDenmark
- Department of Clinical Medicine, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Birgitte Lausen
- Department of Paediatrics and Adolescent Medicine, RigshospitaletUniversity of CopenhagenCopenhagenDenmark
| | - Thoas Fioretos
- Department of Laboratory Medicine, Division of Clinical GeneticsLund UniversityLundSweden
| | - Axel Hyrenius‐Wittsten
- Department of Laboratory Medicine, Division of Clinical GeneticsLund UniversityLundSweden
| | | |
Collapse
|
2
|
Vít O, Petrák J. Autotaxin and Lysophosphatidic Acid Signalling: the Pleiotropic Regulatory Network in Cancer. Folia Biol (Praha) 2023; 69:149-162. [PMID: 38583176 DOI: 10.14712/fb2023069050149] [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: 04/09/2024]
Abstract
Autotaxin, also known as ecto-nucleotide pyrophosphatase/phosphodiesterase family member 2, is a secreted glycoprotein that plays multiple roles in human physiology and cancer pathology. This protein, by converting lysophosphatidylcholine into lysophosphatidic acid, initiates a complex signalling cascade with significant biological implications. The article outlines the autotaxin gene and protein structure, expression regulation and physiological functions, but focuses mainly on the role of autotaxin in cancer development and progression. Autotaxin and lysophosphatidic acid signalling influence several aspects of cancer, including cell proliferation, migration, metastasis, therapy resistance, and interactions with the immune system. The potential of autotaxin as a diagnostic biomarker and promising drug target is also examined.
Collapse
Affiliation(s)
- Ondřej Vít
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czech Republic.
| | - Jiří Petrák
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czech Republic
| |
Collapse
|
3
|
Mieczkowski A, Frączyk T, Psurski M, Wińska P, Siedlecki P, Dziełak M, Trzybiński D, Wilczek M, Bagiński M, Bieszczad B, Woźniak K. Design and in Vitro Characterization of Tricyclic Benzodiazepine Derivatives as Potent and Selective Antileukemic Agents. Chem Biodivers 2020; 18:e2000733. [PMID: 33236468 DOI: 10.1002/cbdv.202000733] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Accepted: 11/24/2020] [Indexed: 12/22/2022]
Abstract
Currently available chemotherapeutic treatments for blood cancers (leukemia) usually have strong side effects. More selective, efficient, and less toxic anticancer agents are needed. We synthesized seven, new, optically pure (12aS)-1,3,4,12a-tetrahydropyrazino[2,1-c][1,4],12(2H,11H)-dione derivatives and examined their cytotoxicity towards eight cancer cell lines, including urinary bladder (TCC-SUP, UM-UC-3, KU-19-9), colon (LoVo), and breast (MCF-7, MDA-MB-231) cancer representatives, as well as two leukemic cell lines (MV-4-11, CCRF-CEM) and normal murine fibroblasts (Balb/3T3) as reference cell line. Three of the seven newly-obtained compounds ((12aS)-8-bromo-2-(3-phenylbenzoyl)-1,3,4,12a-tetrahydropyrazino[2,1-c][1,4],12(2H,11H)-dione, (12aS)-8,9-dimethoxy-2-(4-phenylbenzoyl)-1,3,4,12a-tetrahydropyrazino[2,1-c][1,4],12(2H,11H)-dione and (12aS)-8-nitro-2-(4-phenylbenzoyl)-1,3,4,12a-tetrahydropyrazino[2,1-c][1,4],12(2H,11H)-dione, showed enhanced activity and selectivity toward the leukemic MV-4-11 cell lines when compared to our previously reported compounds, with IC50 values in the range of 2.9-5.6 μM. Additionally, (12aS)-9-nitro-2-(4-phenylbenzoyl)-1,3,4,12a-tetrahydropyrazino[2,1-c][1,4],12(2H,11H)-dione exhibited a strong cytotoxic effect against the leukemic CCRF-CEM (IC50 =6.1 μM) and MV-4-11 (IC50 =11.0 μM) cell lines, a moderate cytotoxic effect toward other tumor lines (IC50 =31.8-55.0 μM) and very weak cytotoxic effect toward the Balb/3T3 reference cell lines. Selected compounds were further evaluated for their potential to induce apoptotic cell death in MV-4-11 cells by measuring caspase-3 activity. We also established the crystal structure of three products and investigated the effect of 22 derivatives of 1,3,4,12a-tetrahydropyrazino[2,1-c][1,4],12(2H,11H)-dione on the activity of the cancer-associated enzyme autotaxin. All compounds proved to be weak inhibitors of autotaxin, although some (R) and (S) enantiomers had Ki values of 10-19 μM. The obtained results showed that the tested compounds exhibited a selective antileukemic effect, which appeared not to be related directly to autotaxin. Molecular targets responsible for this effect remain to be identified. The newly obtained compounds can be used in the search for new, selective anticancer therapies.
Collapse
Affiliation(s)
- Adam Mieczkowski
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawinskiego 5a, 02-106, Warsaw, Poland
| | - Tomasz Frączyk
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawinskiego 5a, 02-106, Warsaw, Poland.,Medical University of Warsaw, Nowogrodzka 59, 02-006, Warsaw, Poland
| | - Mateusz Psurski
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, R., Weigla 12, 53-114, Wroclaw, Poland
| | - Patrycja Wińska
- Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664, Warsaw, Poland
| | - Paweł Siedlecki
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawinskiego 5a, 02-106, Warsaw, Poland
| | - Monika Dziełak
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawinskiego 5a, 02-106, Warsaw, Poland
| | - Damian Trzybiński
- Biological and Chemical Research Centre, University of Warsaw, Żwirki i Wigury 101, 02-089, Warsaw, Poland
| | - Marcin Wilczek
- Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-093, Warsaw, Poland
| | - Maciej Bagiński
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawinskiego 5a, 02-106, Warsaw, Poland.,Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-093, Warsaw, Poland
| | - Bartosz Bieszczad
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawinskiego 5a, 02-106, Warsaw, Poland
| | - Krzysztof Woźniak
- Biological and Chemical Research Centre, University of Warsaw, Żwirki i Wigury 101, 02-089, Warsaw, Poland
| |
Collapse
|
4
|
Lee D, Kim YH, Kim JH. The Role of Lysophosphatidic Acid in Adult Stem Cells. Int J Stem Cells 2020; 13:182-191. [PMID: 32587135 PMCID: PMC7378901 DOI: 10.15283/ijsc20035] [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: 03/04/2020] [Revised: 04/24/2020] [Accepted: 05/21/2020] [Indexed: 01/06/2023] Open
Abstract
Stem cells are undifferentiated multipotent precursor cells that are capable both of perpetuating themselves as stem cells (self-renewal) and of undergoing differentiation into one or more specialized types of cells. And these stem cells have been reported to reside within distinct anatomic locations termed “niches”. The long-term goals of stem cell biology range from an understanding of cell-lineage determination and tissue organization to cellular therapeutics for degenerative diseases. Stem cells maintain tissue function throughout an organism’s lifespan by replacing differentiated cells. To perform this function, stem cells provide a unique combination of multilineage developmental potential and the capacity to undergo self-renewing divisions. The loss of self-renewal capacity in stem cells underlies certain degenerative diseases and the aging process. This self-renewal regulation must balance the regenerative needs of tissues that persist throughout life. Recent evidence suggests lysophosphatidic acid (LPA) signaling pathway plays an important role in the regulation of a variety of stem cells. In this review, we summarize the evidence linking between LPA and stem cell regulation. The LPA-induced signaling pathway regulates the proliferation and survival of stem cells and progenitors, and thus are likely to play a role in the maintenance of stem cell population in the body. This lipid mediator regulatory system can be a novel potential therapeutics for stem cell maintenance.
Collapse
Affiliation(s)
- Dongjun Lee
- Department of Convergence Medicine, Pusan National University School of Medicine, Yangsan, Korea
| | - Yun Hak Kim
- Department of Anatomy, Pusan National University School of Medicine, Yangsan, Korea.,Department of Biomedical Informatics, Pusan National University School of Medicine, Yangsan, Korea
| | - Jae Ho Kim
- Department of Physiology, Pusan National University School of Medicine, Yangsan, Korea
| |
Collapse
|
5
|
Abdelbaset-Ismail A, Cymer M, Borkowska-Rzeszotek S, Brzeźniakiewicz-Janus K, Rameshwar P, Kakar SS, Ratajczak J, Ratajczak MZ. Bioactive Phospholipids Enhance Migration and Adhesion of Human Leukemic Cells by Inhibiting Heme Oxygenase 1 (HO-1) and Inducible Nitric Oxygenase Synthase (iNOS) in a p38 MAPK-Dependent Manner. Stem Cell Rev Rep 2020; 15:139-154. [PMID: 30302660 PMCID: PMC6366663 DOI: 10.1007/s12015-018-9853-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Bioactive phospholipids, including sphingosine-1-phosphate (S1P), ceramide-1-phosphate (C1P), lysophosphatidylcholine (LPC), and its derivative lysophosphatidic acid (LPA), have emerged as important mediators regulating the trafficking of normal and cancer cells. While the role of S1P in regulating migration of hematopoietic cells is well established, in this work we compared its biological effects to the effects of C1P, LPC, and LPA. We employed 10 human myeloid and lymphoid cell lines as well as blasts from AML patients. We observed that human leukemic cells express functional receptors for phospholipids and respond to stimulation by phosphorylation of p42/44 MAPK and AKT. We also found that bioactive phospholipids enhanced cell migration and adhesion of leukemic cells by downregulating expression of HO-1 and iNOS in a p38 MAPK-dependent manner but did not affect cell proliferation. By contrast, downregulation of p38 MAPK by SB203580 enhanced expression of HO-1 and iNOS and decreased migration of leukemic cells in vitro and their seeding efficiency to vital organs in vivo after injection into immunodeficient mice. Based on these findings, we demonstrate that, besides S1P, human leukemic cells also respond to C1P, LPC, and LPA. Since the prometastatic effects of bioactive phospholipids in vivo were mediated, at least in part, by downregulating HO-1 and iNOS expression in a p38 MAPK-dependent manner, we propose that inhibitors of p38 MAPK or stimulators of HO-1 activity will find application in inhibiting the spread of leukemic cells in response to bioactive phospholipids.
Collapse
Affiliation(s)
- Ahmed Abdelbaset-Ismail
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY, 40202, USA.,Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Monika Cymer
- Center for Preclinical Research and Technology, Department of Regenerative Medicine, Warsaw Medical University, Warsaw, Poland
| | | | | | | | - Sham S Kakar
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY, 40202, USA
| | - Janina Ratajczak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY, 40202, USA
| | - Mariusz Z Ratajczak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY, 40202, USA. .,Center for Preclinical Research and Technology, Department of Regenerative Medicine, Warsaw Medical University, Warsaw, Poland.
| |
Collapse
|
6
|
Lin KH, Chiang JC, Ho YH, Yao CL, Lee H. Lysophosphatidic Acid and Hematopoiesis: From Microenvironmental Effects to Intracellular Signaling. Int J Mol Sci 2020; 21:ijms21062015. [PMID: 32188052 PMCID: PMC7139687 DOI: 10.3390/ijms21062015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 03/13/2020] [Accepted: 03/13/2020] [Indexed: 02/06/2023] Open
Abstract
Vertebrate hematopoiesis is a complex physiological process that is tightly regulated by intracellular signaling and extracellular microenvironment. In recent decades, breakthroughs in lineage-tracing technologies and lipidomics have revealed the existence of numerous lipid molecules in hematopoietic microenvironment. Lysophosphatidic acid (LPA), a bioactive phospholipid molecule, is one of the identified lipids that participates in hematopoiesis. LPA exhibits various physiological functions through activation of G-protein-coupled receptors. The functions of these LPARs have been widely studied in stem cells, while the roles of LPARs in hematopoietic stem cells have rarely been examined. Nonetheless, mounting evidence supports the importance of the LPA-LPAR axis in hematopoiesis. In this article, we have reviewed regulation of hematopoiesis in general and focused on the microenvironmental and intracellular effects of the LPA in hematopoiesis. Discoveries in these areas may be beneficial to our understanding of blood-related disorders, especially in the context of prevention and therapy for anemia.
Collapse
Affiliation(s)
- Kuan-Hung Lin
- Department of Life Science, National Taiwan University, Taipei 10617, Taiwan; (K.-H.L.); (J.-C.C.)
| | - Jui-Chung Chiang
- Department of Life Science, National Taiwan University, Taipei 10617, Taiwan; (K.-H.L.); (J.-C.C.)
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ya-Hsuan Ho
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Haematology, University of Cambridge, Cambridge CB2 0AW, UK;
| | - Chao-Ling Yao
- Department of Chemical Engineering and Materials Science, Yuan Ze University, Taoyuan 32003, Taiwan;
| | - Hsinyu Lee
- Department of Life Science, National Taiwan University, Taipei 10617, Taiwan; (K.-H.L.); (J.-C.C.)
- Department of Electrical Engineering, National Taiwan University, Taipei 10617, Taiwan
- Angiogenesis Research Center, National Taiwan University, Taipei 10617, Taiwan
- Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei 10617, Taiwan
- Center for Biotechnology, National Taiwan University, Taipei 10617, Taiwan
- Correspondence: ; Tel.: +8862-3366-2499; Fax: +8862-2363-6837
| |
Collapse
|
7
|
Zhang Q, Wu X, Cao J, Gao F, Huang K. Association between increased mutation rates in DNMT3A and FLT3-ITD and poor prognosis of patients with acute myeloid leukemia. Exp Ther Med 2019; 18:3117-3124. [PMID: 31572552 PMCID: PMC6755468 DOI: 10.3892/etm.2019.7891] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 04/11/2019] [Indexed: 12/17/2022] Open
Abstract
A total of 133 patients with acute myeloid leukemia (AML) were enrolled in the current study and were subdivided into 4 groups: 34 harboring DNA methyltransferase 3 α (DNMT3A) + fms related tyrosine kinase 3-internal tandem duplication (FLT3-ITD) mutations, 37 harboring only FLT3-ITD mutation, 32 harboring only DNMT3A mutation and 30 harboring no mutations in DNMT3A and FLT3-ITD (control). Patients in all groups were administered daunorubicin and cytarabine chemotherapy regimens. The rates of complete remission (CR), 1-year relapse (RR) and 3-year overall survival (OS) were compared. Patients in the DNMT3A + FLT3-ITD mutation group exhibited higher proportions of peripheral white blood cells (WBCs) and myeloid progenitor cells compared with those in DNMT3A mutation only, FLT3-ITD mutation only and control groups (P<0.05). The rates of CD15+ and HLA-DR+ in the DNMT3A + FLT3-ITD mutation and DNMT3A mutation only groups were significantly higher than those in the FLT3-ITD mutation only and control groups (P<0.05); in addition, the rate of CD38+ in the DNMT3A + FLT3-ITD mutation and FLT3-ITD mutation only groups was significantly higher compared with that in the DNMT3A mutation only and control groups (P<0.05). The overall chemotherapy effectiveness rate, CR, 1-year RR and the 3-year OS rates of patients in the DNMT3A + FLT3-ITD mutation group were significantly worse compared with FLT3-ITD mutation only, DNMT3A mutation only and control groups (P<0.05). The results of this study indicated that increased mutation rates in DNMT3A and FLT3-ITD may be associated with increased WBC and myeloid progenitor cell counts, an inferior chemotherapy efficacy and prognosis, a lower CR rate, and higher 1-year RR and mortality rate.
Collapse
Affiliation(s)
- Qiurong Zhang
- Department of Hematology, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou, Jiangsu 215600, P.R. China
| | - Xiao Wu
- Department of Hematology, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou, Jiangsu 215600, P.R. China
| | - Jing Cao
- Department of Hematology, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou, Jiangsu 215600, P.R. China
| | - Feng Gao
- Department of Hematology, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou, Jiangsu 215600, P.R. China
| | - Kun Huang
- Department of Hematology, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou, Jiangsu 215600, P.R. China
| |
Collapse
|
8
|
Zhong W, Bian K, Hu Y, Ji Z, Xu X, Li J, Wu P, Wang X, Zhang Y, Zhang P, Zhang H, Shen Y. Lysophosphatidic acid guides the homing of transplanted olfactory ensheathing cells to the lesion site after spinal cord injury in rats. Exp Cell Res 2019; 379:65-72. [DOI: 10.1016/j.yexcr.2019.03.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 02/21/2019] [Accepted: 03/14/2019] [Indexed: 01/19/2023]
|
9
|
Lidgerwood GE, Pitson SM, Bonder C, Pébay A. Roles of lysophosphatidic acid and sphingosine-1-phosphate in stem cell biology. Prog Lipid Res 2018; 72:42-54. [PMID: 30196008 DOI: 10.1016/j.plipres.2018.09.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 08/15/2018] [Accepted: 09/05/2018] [Indexed: 02/06/2023]
Abstract
Stem cells are unique in their ability to self-renew and differentiate into various cell types. Because of these features, stem cells are key to the formation of organisms and play fundamental roles in tissue regeneration and repair. Mechanisms controlling their fate are thus fundamental to the development and homeostasis of tissues and organs. Lysophosphatidic acid (LPA) and sphingosine-1-phosphate (S1P) are bioactive phospholipids that play a wide range of roles in multiple cell types, during developmental and pathophysiological events. Considerable evidence now demonstrates the potent roles of LPA and S1P in the biology of pluripotent and adult stem cells, from maintenance to repair. Here we review their roles for each main category of stem cells and explore how those effects impact development and physiopathology.
Collapse
Affiliation(s)
- Grace E Lidgerwood
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Australia; Ophthalmology, Department of Surgery, the University of Melbourne, Melbourne, Australia
| | - Stuart M Pitson
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia
| | - Claudine Bonder
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia
| | - Alice Pébay
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Australia; Ophthalmology, Department of Surgery, the University of Melbourne, Melbourne, Australia.
| |
Collapse
|
10
|
Stockard B, Garrett T, Guingab-Cagmat J, Meshinchi S, Lamba J. Distinct Metabolic features differentiating FLT3-ITD AML from FLT3-WT childhood Acute Myeloid Leukemia. Sci Rep 2018; 8:5534. [PMID: 29615816 PMCID: PMC5882915 DOI: 10.1038/s41598-018-23863-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 03/21/2018] [Indexed: 12/14/2022] Open
Abstract
Acute myeloid leukemia (AML) is a heterogeneous disease with dismal response warranting the need for enhancing our understanding of AML biology. One prognostic feature associated with inferior response is the presence of activating mutations in FMS-like tyrosine kinase 3 (FLT3) especially occurrence of internal tandem duplication (FLT3-ITD). Although poorly understood, differential metabolic and signaling pathways associated with FLT3-ITD might contribute towards the observed poor prognosis. We performed a non-targeted global metabolic profiling of matched cell and plasma samples obtained at diagnosis to establish metabolic differences within FLT3-ITD and FLT3-WT pediatric AML. Metabolomic profiling by Ultra-High Performance-Liquid-Chromatography-Mass Spectrometry identified differential abundance of 21 known metabolites in plasma and 33 known metabolites in leukemic cells by FLT3 status. These metabolic features mapped to pathways of significant biological importance. Of interest were metabolites with roles in cancer, cell progression and involvement in purine metabolism and biosynthesis, cysteine/methionine metabolism, tryptophan metabolism, carnitine mediated fatty acid oxidation, and lysophospholipid metabolism. Although validation in a larger cohort is required, our results for the first time investigated global metabolic profile in FLT3-ITD AML.
Collapse
Affiliation(s)
- Bradley Stockard
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Timothy Garrett
- Southeast Center for Integrated Metabolomics, University of Florida, Gainesville, FL, USA
| | - Joy Guingab-Cagmat
- Southeast Center for Integrated Metabolomics, University of Florida, Gainesville, FL, USA
| | - Soheil Meshinchi
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Medicine/Division of Hematology, University of Washington, Seattle, WA, USA
| | - Jatinder Lamba
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
11
|
Quan M, Cui JJ, Feng X, Huang Q. The critical role and potential target of the autotaxin/lysophosphatidate axis in pancreatic cancer. Tumour Biol 2017; 39:1010428317694544. [PMID: 28347252 DOI: 10.1177/1010428317694544] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Autotaxin, an ecto-lysophospholipase D encoded by the human ENNP2 gene, is expressed in multiple tissues, and participates in numerous critical physiologic and pathologic processes including inflammation, pain, obesity, embryo development, and cancer via the generation of the bioactive lipid lysophosphatidate. Overwhelming evidences indicate that the autotaxin/lysophosphatidate signaling axis serves key roles in the numerous processes central to tumorigenesis and progression, including proliferation, survival, migration, invasion, metastasis, cancer stem cell, tumor microenvironment, and treatment resistance by interacting with a series of at least six G-protein-coupled receptors (LPAR1-6). This review provides an overview of the autotaxin/lysophosphatidate axis and collates current knowledge regarding its specific role in pancreatic cancer. With a deeper understanding of the critical role of the autotaxin/lysophosphatidate axis in pancreatic cancer, targeting autotaxin or lysophosphatidate receptor may be a potential and promising strategy for cancer therapy.
Collapse
Affiliation(s)
- Ming Quan
- Cancer Center, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Jiu-Jie Cui
- Cancer Center, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Xiao Feng
- Cancer Center, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Qian Huang
- Cancer Center, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
| |
Collapse
|
12
|
Wobus M, Bornhäuser M, Jacobi A, Kräter M, Otto O, Ortlepp C, Guck J, Ehninger G, Thiede C, Oelschlägel U. Association of the EGF-TM7 receptor CD97 expression with FLT3-ITD in acute myeloid leukemia. Oncotarget 2016; 6:38804-15. [PMID: 26462154 PMCID: PMC4770738 DOI: 10.18632/oncotarget.5661] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 09/25/2015] [Indexed: 12/31/2022] Open
Abstract
Internal tandem duplications within the juxtamembrane region of the FMS-like tyrosine kinase receptor FLT3 (FLT3-ITD) represents one of the most common mutations in patients with acute myeloid leukemia (AML) which results in constitutive aberrant activation, increased proliferation of leukemic progenitors and is associated with an aggressive clinical phenotype. The expression of CD97, an EGF-TM7 receptor, has been linked to invasive behavior in thyroid and colorectal cancer. Here, we have investigated the association of CD97 with FLT3-ITD and its functional consequences in AML.Higher CD97 expression levels have been detected in 208 out of 385 primary AML samples. This was accompanied by a significantly increased bone marrow blast count as well as by mutations in the FLT3 gene. FLT3-ITD expressing cell lines as MV4-11 and MOLM-13 revealed significantly higher CD97 levels than FLT3 wildtype EOL-1, OCI-AML3 and HL-60 cells which were clearly decreased by the tyrosine kinase inhibitors PKC412 and SU5614. CD97 knock down by short hairpin RNA in MV4-11 cells resulted in inhibited trans-well migration towards fetal calf serum (FCS) and lysophosphatidic acid (LPA) being at least in part Rho-A dependent. Moreover, knock down of CD97 led to an altered mechanical phenotype, reduced adhesion to a stromal layer and lower wildtype FLT3 expression.Our results, thus, constitute the first evidence for the functional relevance of CD97 expression in FLT3-ITD AML cells rendering it a potential new theragnostic target.
Collapse
Affiliation(s)
- Manja Wobus
- Division of Hematology, Oncology and Stem Cell Transplantation, Department of Medicine I, University Hospital Carl Gustav Carus, Technische Universität, Dresden, Germany
| | - Martin Bornhäuser
- Division of Hematology, Oncology and Stem Cell Transplantation, Department of Medicine I, University Hospital Carl Gustav Carus, Technische Universität, Dresden, Germany.,German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
| | - Angela Jacobi
- Biotechnology Center, Technische Universität Dresden, Dresden, Germany
| | - Martin Kräter
- Division of Hematology, Oncology and Stem Cell Transplantation, Department of Medicine I, University Hospital Carl Gustav Carus, Technische Universität, Dresden, Germany
| | - Oliver Otto
- Biotechnology Center, Technische Universität Dresden, Dresden, Germany
| | - Claudia Ortlepp
- Division of Hematology, Oncology and Stem Cell Transplantation, Department of Medicine I, University Hospital Carl Gustav Carus, Technische Universität, Dresden, Germany
| | - Jochen Guck
- Biotechnology Center, Technische Universität Dresden, Dresden, Germany
| | - Gerhard Ehninger
- Division of Hematology, Oncology and Stem Cell Transplantation, Department of Medicine I, University Hospital Carl Gustav Carus, Technische Universität, Dresden, Germany
| | - Christian Thiede
- Division of Hematology, Oncology and Stem Cell Transplantation, Department of Medicine I, University Hospital Carl Gustav Carus, Technische Universität, Dresden, Germany
| | - Uta Oelschlägel
- Division of Hematology, Oncology and Stem Cell Transplantation, Department of Medicine I, University Hospital Carl Gustav Carus, Technische Universität, Dresden, Germany
| |
Collapse
|
13
|
Overexpression of autotaxin is associated with human renal cell carcinoma and bladder carcinoma and their progression. Med Oncol 2016; 33:131. [PMID: 27757783 DOI: 10.1007/s12032-016-0836-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 09/24/2016] [Indexed: 12/20/2022]
Abstract
Autotaxin (ATX) as an important tumor cell motility-stimulating factor is upregulated in many different types of cancer. ATX, a member of the ectonucleotide pyrophosphatase and phosphodiesterase family of enzymes, possesses lysophospholipase D activity which hydrolyzes lysophosphatidylcholine to generate the potent tumor growth factor and mitogen lysophosphatidic acid (LPA). LPA acts on specific G-protein-coupled receptors, thereby regulating cell growth, migration, and survival. This study aimed to investigate the differences in gene expression pattern of ATX between cancerous and adjacent normal tissue of human renal cell carcinoma (RCC) and bladder carcinoma (BC) and find the correlation between ATX expression and clinicopathological features of both of these carcinomas. Both the RCC and BC tissues and with the adjacent normal tissues were collected. Immunohistochemistry and Western blotting analysis were used to detect the extent of ATX expression in all of these samples. Immunohistochemistry and Western blot analysis revealed that expression of ATX protein in carcinoma tissues is significantly higher than that in the adjacent normal tissues. Immunohistochemistry analysis showed that ATX is localized in cytoplasm. Western blotting analysis showed that ATX protein is expressed in both RCC and BC, and the expression levels were 69.5 and 48.0 %, respectively, higher in RCC and BC carcinoma tissue samples than in the adjacent normal tissues, which is consistent with the results of immunohistochemistry study. Thus, this study provided the evidence that ATX is highly expressed in both RCC and BC. Further research can be done to identify the diagnosis and treatment significance of both these carcinomas.
Collapse
|
14
|
Lee HJ, Ryu JM, Jung YH, Lee KH, Kim DI, Han HJ. Glycerol-3-phosphate acyltransferase-1 upregulation by O-GlcNAcylation of Sp1 protects against hypoxia-induced mouse embryonic stem cell apoptosis via mTOR activation. Cell Death Dis 2016; 7:e2158. [PMID: 27010859 PMCID: PMC4823928 DOI: 10.1038/cddis.2015.410] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 12/10/2015] [Accepted: 12/14/2015] [Indexed: 12/20/2022]
Abstract
Oxygen signaling is critical for stem cell regulation, and oxidative stress-induced stem cell apoptosis decreases the efficiency of stem cell therapy. Hypoxia activates O-linked β-N-acetyl glucosaminylation (O-GlcNAcylation) of stem cells, which contributes to regulation of cellular metabolism, as well as cell fate. Our study investigated the role of O-GlcNAcylation via glucosamine in the protection of hypoxia-induced apoptosis of mouse embryonic stem cells (mESCs). Hypoxia increased mESCs apoptosis in a time-dependent manner. Moreover, hypoxia also slightly increased the O-GlcNAc level. Glucosamine treatment further enhanced the O-GlcNAc level and prevented hypoxia-induced mESC apoptosis, which was suppressed by O-GlcNAc transferase inhibitors. In addition, hypoxia regulated several lipid metabolic enzymes, whereas glucosamine increased expression of glycerol-3-phosphate acyltransferase-1 (GPAT1), a lipid metabolic enzyme producing lysophosphatidic acid (LPA). In addition, glucosamine-increased O-GlcNAcylation of Sp1, which subsequently leads to Sp1 nuclear translocation and GPAT1 expression. Silencing of GPAT1 by gpat1 siRNA transfection reduced glucosamine-mediated anti-apoptosis in mESCs and reduced mammalian target of rapamycin (mTOR) phosphorylation. Indeed, LPA prevented mESCs from undergoing hypoxia-induced apoptosis and increased phosphorylation of mTOR and its substrates (S6K1 and 4EBP1). Moreover, mTOR inactivation by rapamycin (mTOR inhibitor) increased pro-apoptotic proteins expressions and mESC apoptosis. Furthermore, transplantation of non-targeting siRNA and glucosamine-treated mESCs increased cell survival and inhibited flap necrosis in mouse skin flap model. Conversely, silencing of GPAT1 expression reversed those glucosamine effects. In conclusion, enhancing O-GlcNAcylation of Sp1 by glucosamine stimulates GPAT1 expression, which leads to inhibition of hypoxia-induced mESC apoptosis via mTOR activation.
Collapse
Affiliation(s)
- H J Lee
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science and BK21 Creative Veterinary Research Center, Seoul National University, Seoul, Korea
| | - J M Ryu
- Department of Veterinary Physiology, College of Veterinary Medicine, Chonnam National University, Gwangju 61186, Korea
| | - Y H Jung
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science and BK21 Creative Veterinary Research Center, Seoul National University, Seoul, Korea
| | - K H Lee
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science and BK21 Creative Veterinary Research Center, Seoul National University, Seoul, Korea
| | - D I Kim
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science and BK21 Creative Veterinary Research Center, Seoul National University, Seoul, Korea
| | - H J Han
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science and BK21 Creative Veterinary Research Center, Seoul National University, Seoul, Korea
| |
Collapse
|
15
|
Federico L, Jeong KJ, Vellano CP, Mills GB. Autotaxin, a lysophospholipase D with pleomorphic effects in oncogenesis and cancer progression. J Lipid Res 2016; 57:25-35. [PMID: 25977291 PMCID: PMC4689343 DOI: 10.1194/jlr.r060020] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 05/07/2015] [Indexed: 12/18/2022] Open
Abstract
The ectonucleotide pyrophosphatase/phosphodiesterase type 2, more commonly known as autotaxin (ATX), is an ecto-lysophospholipase D encoded by the human ENNP2 gene. ATX is expressed in multiple tissues and participates in numerous key physiologic and pathologic processes, including neural development, obesity, inflammation, and oncogenesis, through the generation of the bioactive lipid, lysophosphatidic acid. Overwhelming evidence indicates that altered ATX activity leads to oncogenesis and cancer progression through the modulation of multiple hallmarks of cancer pathobiology. Here, we review the structural and catalytic characteristics of the ectoenzyme, how its expression and maturation processes are regulated, and how the systemic integration of its pleomorphic effects on cells and tissues may contribute to cancer initiation, progression, and therapy. Additionally, the up-to-date spectrum of the most frequent ATX genomic alterations from The Cancer Genome Atlas project is reported for a subset of cancers.
Collapse
Affiliation(s)
- Lorenzo Federico
- Department of Systems Biology, University of Texas M. D. Anderson Cancer Center, Houston, TX
| | - Kang Jin Jeong
- Department of Systems Biology, University of Texas M. D. Anderson Cancer Center, Houston, TX
| | - Christopher P Vellano
- Department of Systems Biology, University of Texas M. D. Anderson Cancer Center, Houston, TX
| | - Gordon B Mills
- Department of Systems Biology, University of Texas M. D. Anderson Cancer Center, Houston, TX
| |
Collapse
|
16
|
Ryu JM, Han HJ. Autotaxin-LPA axis regulates hMSC migration by adherent junction disruption and cytoskeletal rearrangement via LPAR1/3-dependent PKC/GSK3β/β-catenin and PKC/Rho GTPase pathways. Stem Cells 2015; 33:819-32. [PMID: 25376707 DOI: 10.1002/stem.1882] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Revised: 09/30/2014] [Accepted: 10/15/2014] [Indexed: 12/29/2022]
Abstract
Bioactive molecules and stem cell-based regenerative engineering is emerging a promising approach for regenerating tissues. Autotaxin (ATX) is a key enzyme that regulates lysophosphatidic acid (LPA) levels in biological fluids, which exerts a wide range of cellular functions. However, the biological role of ATX in human umbilical cord blood-derived mesenchymal stem cells (hMSCs) migration remains to be fully elucidated. In this study, we observed that hMSCs, which were stimulated with LPA, accelerated wound healing, and LPA increased the migration of hMSCs into a wound site in a mouse skin wound healing model. In an experiment to investigate the effect of LPA on hMSC migration, ATX and LPA increased hMSC migration in a dose-dependent manner, and LPA receptor 1/3 siRNA transfections inhibited the ATX-induced cell migration. Furthermore, LPA increased Ca(2+) influx and PKC phosphorylation, which were blocked by Gαi and Gαq knockdown as well as by Ptx pretreatment. LPA increased GSK3β phosphorylation and β-catenin activation. LPA induced the cytosol to nuclear translocation of β-catenin, which was inhibited by PKC inhibitors. LPA stimulated the binding of β-catenin on the E-box located in the promoter of the CDH-1 gene and decreased CDH-1 promoter activity. In addition, the ATX and LPA-induced increase in hMSC migration was blocked by β-catenin siRNA transfection. LPA-induced PKC phosphorylation is also involved in Rac1 and CDC42 activation, and Rac1 and CDC42 knockdown abolished LPA-induced F-actin reorganization. In conclusion, ATX/LPA stimulates the migration of hMSCs through LPAR1/3-dependent E-cadherin reduction and cytoskeletal rearrangement via PKC/GSK3β/β-catenin and PKC/Rho GTPase pathways.
Collapse
Affiliation(s)
- Jung Min Ryu
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, Seoul National University, Seoul, South Korea; BK21 PLUS Creative Veterinary Research Center, Seoul National University, Seoul, South Korea
| | | |
Collapse
|
17
|
Balogh A, Shimizu Y, Lee SC, Norman DD, Gangwar R, Bavaria M, Moon C, Shukla P, Rao R, Ray R, Naren AP, Banerjee S, Banerje S, Miller DD, Balazs L, Pelus L, Tigyi G. The autotaxin-LPA2 GPCR axis is modulated by γ-irradiation and facilitates DNA damage repair. Cell Signal 2015; 27:1751-62. [PMID: 26027517 DOI: 10.1016/j.cellsig.2015.05.015] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 05/15/2015] [Indexed: 12/24/2022]
Abstract
In this study we characterized the effects of radiation injury on the expression and function of the autotaxin (ATX)-LPA2 GPCR axis. In IEC-6 crypt cells and jejunum enteroids quantitative RT-PCR showed a time- and dose-dependent upregulation of lpa2 in response to γ-irradiation that was abolished by mutation of the NF-κB site in the lpa2 promoter or by inhibition of ATM/ATR kinases with CGK-733, suggesting that lpa2 is a DNA damage response gene upregulated by ATM via NF-κB. The resolution kinetics of the DNA damage marker γ-H2AX in LPA-treated IEC-6 cells exposed to γ-irradiation was accelerated compared to vehicle, whereas pharmacological inhibition of LPA2 delayed the resolution of γ-H2AX. In LPA2-reconstituted MEF cells lacking LPA1&3 the levels of γ-H2AX decreased rapidly, whereas in Vector MEF were high and remained sustained. Inhibition of ERK1&2 or PI3K/AKT signaling axis by pertussis toxin or the C311A/C314A/L351A mutation in the C-terminus of LPA2 abrogated the effect of LPA on DNA repair. LPA2 transcripts in Lin(-)Sca-1(+)c-Kit(+) enriched for bone marrow stem cells were 27- and 5-fold higher than in common myeloid or lymphoid progenitors, respectively. Furthermore, after irradiation higher residual γ-H2AX levels were detected in the bone marrow or jejunum of irradiated LPA2-KO mice compared to WT mice. We found that γ-irradiation increases plasma ATX activity and LPA level that is in part due to the previously established radiation-induced upregulation of TNFα. These findings identify ATX and LPA2 as radiation-regulated genes that appear to play a physiological role in DNA repair.
Collapse
Affiliation(s)
- Andrea Balogh
- Department of Physiology, University of Tennessee Health Sciences Center Memphis, 894 Union Avenue, Memphis, TN 38163, USA
| | - Yoshibumi Shimizu
- Department of Physiology, University of Tennessee Health Sciences Center Memphis, 894 Union Avenue, Memphis, TN 38163, USA
| | - Sue Chin Lee
- Department of Physiology, University of Tennessee Health Sciences Center Memphis, 894 Union Avenue, Memphis, TN 38163, USA
| | - Derek D Norman
- Department of Physiology, University of Tennessee Health Sciences Center Memphis, 894 Union Avenue, Memphis, TN 38163, USA
| | - Ruchika Gangwar
- Department of Physiology, University of Tennessee Health Sciences Center Memphis, 894 Union Avenue, Memphis, TN 38163, USA
| | - Mitul Bavaria
- Department of Physiology, University of Tennessee Health Sciences Center Memphis, 894 Union Avenue, Memphis, TN 38163, USA
| | - ChangSuk Moon
- Department of Physiology, University of Tennessee Health Sciences Center Memphis, 894 Union Avenue, Memphis, TN 38163, USA
| | - Pradeep Shukla
- Department of Physiology, University of Tennessee Health Sciences Center Memphis, 894 Union Avenue, Memphis, TN 38163, USA
| | - Radakrishna Rao
- Department of Physiology, University of Tennessee Health Sciences Center Memphis, 894 Union Avenue, Memphis, TN 38163, USA
| | - Ramesh Ray
- Department of Physiology, University of Tennessee Health Sciences Center Memphis, 894 Union Avenue, Memphis, TN 38163, USA
| | - Anjaparavanda P Naren
- Department of Physiology, University of Tennessee Health Sciences Center Memphis, 894 Union Avenue, Memphis, TN 38163, USA
| | | | - Souvik Banerje
- Department of Pharmaceutical Sciences, University of Tennessee Health Sciences Center Memphis, 894 Union Avenue, Memphis, TN 38163, USA
| | - Duane D Miller
- Department of Pharmaceutical Sciences, University of Tennessee Health Sciences Center Memphis, 894 Union Avenue, Memphis, TN 38163, USA
| | - Louisa Balazs
- Department of Pathology and Laboratory Medicine, University of Tennessee Health Sciences Center Memphis, 894 Union Avenue, Memphis, TN 38163, USA
| | - Louis Pelus
- Department of Microbiology & Immunology, Indiana University School of Medicine, 950 West Walnut Street, R2-302, Indianapolis, IN 46202, USA
| | - Gabor Tigyi
- Department of Physiology, University of Tennessee Health Sciences Center Memphis, 894 Union Avenue, Memphis, TN 38163, USA.
| |
Collapse
|
18
|
Augustin E, Skwarska A, Weryszko A, Pelikant I, Sankowska E, Borowa-Mazgaj B. The antitumor compound triazoloacridinone C-1305 inhibits FLT3 kinase activity and potentiates apoptosis in mutant FLT3-ITD leukemia cells. Acta Pharmacol Sin 2015; 36:385-99. [PMID: 25640477 DOI: 10.1038/aps.2014.142] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Accepted: 12/07/2014] [Indexed: 12/20/2022]
Abstract
AIM FMS-like receptor tyrosine kinase (FLT3) is expressed in some normal hematopoietic cell types and plays an important role in the pathogenesis of acute myeloid leukemia (AML). In this study, we examined the effects of triazoloacridinone C-1305, an antitumor compound, on AML cells with different FLT3 status in vitro. METHODS A panel of human leukemic cell lines with different FLT3 status was used, including FLT3 internal tandem duplication mutations (FLT3-ITD, MV-4-11), wild-type FLT3 (RS-4-11) and null-FLT3 (U937) cells. Cell proliferation was estimated using MTT assays, and apoptosis was studied with flow cytometry and fluorescence microscopy. FLT3 kinase activity (phosphorylation of FLT3 at Tyr591) was determined with ELISA and Western blotting. FLT3 downstream signaling proteins involving AKT, MAPK and STAT5 were examined by Western blotting. RNA silencing was used to decrease the endogenous FLT3. RESULTS The mutant FLT3-ITD cells were more sensitive to C-1305 than the wild-type FLT3 and null-FLT3 cells (the IC50 values measured at 24 h were 1.2±0.17, 2.0±09, 7.6±1.6 μmol/L, respectively). C-1305 (1-10 μmol/L) dose-dependently inhibited the kinase activity of FLT3, which was more pronounced in the mutant FLT3-ITD cells than in the wild-type FLT3 cells. Furthermore, C-1305 dose-dependently decreased the phosphorylation of STAT5 and MAPK and the inhibitory phosphorylation of Bad, and induced time- and dose-dependent apoptosis in the 3 cell lines with the null-FLT3 cells being the least susceptible to C-1305-induced apoptosis. Knockdown of FLT3 with siRNA significantly decreased C-1305-induced cytotoxicity in the mutant FLT3-ITD cells. CONCLUSION C-1305 induces apoptosis in FLT3-ITD-expressing human leukemia cells in vitro, suggesting that mutated FLT3 kinase can be a new target for C-1305, and C-1305 may be a drug candidate for the therapeutic intervention in FLT3-associated AML.
Collapse
|
19
|
Patil R, Szabó E, Fells JI, Balogh A, Lim KG, Fujiwara Y, Norman DD, Lee SC, Balazs L, Thomas F, Patil S, Emmons-Thompson K, Boler A, Strobos J, McCool SW, Yates CR, Stabenow J, Byrne GI, Miller DD, Tigyi GJ. Combined mitigation of the gastrointestinal and hematopoietic acute radiation syndromes by an LPA2 receptor-specific nonlipid agonist. ACTA ACUST UNITED AC 2015; 22:206-16. [PMID: 25619933 DOI: 10.1016/j.chembiol.2014.12.009] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Revised: 12/03/2014] [Accepted: 12/15/2014] [Indexed: 02/06/2023]
Abstract
Pharmacological mitigation of injuries caused by high-dose ionizing radiation is an unsolved medical problem. A specific nonlipid agonist of the type 2 G protein coupled receptor for lysophosphatidic acid (LPA2) 2-[4-(1,3-dioxo-1H,3H-benzoisoquinolin-2-yl)butylsulfamoyl]benzoic acid (DBIBB) when administered with a postirradiation delay of up to 72 hr reduced mortality of C57BL/6 mice but not LPA2 knockout mice. DBIBB mitigated the gastrointestinal radiation syndrome, increased intestinal crypt survival and enterocyte proliferation, and reduced apoptosis. DBIBB enhanced DNA repair by augmenting the resolution of γ-H2AX foci, increased clonogenic survival of irradiated IEC-6 cells, attenuated the radiation-induced death of human CD34(+) hematopoietic progenitors and enhanced the survival of the granulocyte/macrophage lineage. DBIBB also increased the survival of mice suffering from the hematopoietic acute radiation syndrome after total-body irradiation. DBIBB represents a drug candidate capable of mitigating acute radiation syndrome caused by high-dose γ-radiation to the hematopoietic and gastrointestinal system.
Collapse
Affiliation(s)
- Renukadevi Patil
- Department of Pharmaceutical Sciences, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Erzsébet Szabó
- Department of Physiology, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - James I Fells
- Department of Physiology, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Andrea Balogh
- Department of Physiology, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Keng G Lim
- Department of Physiology, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Yuko Fujiwara
- Department of Physiology, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Derek D Norman
- Department of Physiology, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Sue-Chin Lee
- Department of Physiology, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Louisa Balazs
- Department of Pathology, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Fridtjof Thomas
- Department of Preventive Medicine, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Shivaputra Patil
- Department of Pharmaceutical Sciences, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | | | | | | | | | | | - Jennifer Stabenow
- The Regional Biocontainment Laboratory, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Gerrald I Byrne
- The Regional Biocontainment Laboratory, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Duane D Miller
- Department of Pharmaceutical Sciences, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Gábor J Tigyi
- Department of Physiology, The University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| |
Collapse
|
20
|
Ho YH, Yao CL, Lin KH, Hou FH, Chen WM, Chiang CL, Lin YN, Li MW, Lin SH, Yang YJ, Lin CC, Lu J, Tigyi G, Lee H. Opposing regulation of megakaryopoiesis by LPA receptors 2 and 3 in K562 human erythroleukemia cells. Biochim Biophys Acta Mol Cell Biol Lipids 2014; 1851:172-83. [PMID: 25463482 DOI: 10.1016/j.bbalip.2014.11.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 11/06/2014] [Accepted: 11/13/2014] [Indexed: 01/10/2023]
Abstract
Erythrocytes and megakaryocytes (MK) are derived from a common progenitor that undergoes lineage specification. Lysophosphatidic acid (LPA), a lipid growth factor was previously shown to be a regulator for erythropoietic process through activating LPA receptor 3 (LPA3). However, whether LPA affects megakaryopoiesis remains unclear. In this study, we used K562 leukemia cell line as a model to investigate the roles of LPA in MK differentiation. We demonstrated that K562 cells express both LPA2 and LPA3, and the expression levels of LPA2 are higher than LPA3. Treatment with phorbol 12-myristate 13-acetate, a commonly used inducer of megakaryopoiesis, reciprocally regulates the expressions of LPA2 and LPA3. By pharmacological blockers and knockdown experiments, we showed that activation of LPA2 suppresses whereas, LPA3 promotes megakaryocytic differentiation in K562. The LPA2-mediated inhibition is dependent on β-catenin translocation, whereas reactive oxygen species (ROS) generation is a downstream signal for activation of LPA3. Furthermore, the hematopoietic transcriptional factors GATA-1 and FLI-1, appear to be involved in these regulatory mechanisms. Taken together, our results suggested that LPA2 and LPA3 may function as a molecular switch and play opposing roles during megakaryopoiesis of K562 cells.
Collapse
Affiliation(s)
- Ya-Hsuan Ho
- Department of Life Science, National Taiwan University, Taipei, Taiwan, ROC
| | - Chao-Ling Yao
- Department of Chemical Engineering and Materials Science, Yuan-Ze University, Chung-Li, Taiwan, ROC
| | - Kuan-Hung Lin
- Department of Life Science, National Taiwan University, Taipei, Taiwan, ROC
| | - Fen-Han Hou
- Department of Life Science, National Taiwan University, Taipei, Taiwan, ROC
| | - Wei-Min Chen
- Department of Life Science, National Taiwan University, Taipei, Taiwan, ROC
| | - Chi-Ling Chiang
- School of Biomedical Science, The Ohio State University, Columbus, USA
| | - Yu-Nung Lin
- Department of Life Science, National Taiwan University, Taipei, Taiwan, ROC
| | - Meng-Wei Li
- Department of Life Science, National Taiwan University, Taipei, Taiwan, ROC
| | - Shi-Hung Lin
- Department of Life Science, National Taiwan University, Taipei, Taiwan, ROC
| | - Ya-Jan Yang
- Department of Life Science, National Taiwan University, Taipei, Taiwan, ROC
| | - Chu-Cheng Lin
- Department of Life Science, National Taiwan University, Taipei, Taiwan, ROC
| | - Jenher Lu
- Department of Pediatrics and Pediatric Cardiology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC.
| | - Gabor Tigyi
- Department of Physiology, University of Tennessee Health Science Center Memphis, Memphis, USA.
| | - Hsinyu Lee
- Department of Life Science, National Taiwan University, Taipei, Taiwan, ROC; Department of Electrical Engineering, National Taiwan University, Taipei, Taiwan, ROC; Angiogenesis Research Center, National Taiwan University, Taipei, Taiwan, ROC; Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan, ROC; Center for Biotechnology, National Taiwan University, Taipei, Taiwan, ROC.
| |
Collapse
|
21
|
Schneider G, Sellers ZP, Abdel-Latif A, Morris AJ, Ratajczak MZ. Bioactive lipids, LPC and LPA, are novel prometastatic factors and their tissue levels increase in response to radio/chemotherapy. Mol Cancer Res 2014; 12:1560-73. [PMID: 25033840 DOI: 10.1158/1541-7786.mcr-14-0188] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
UNLABELLED Bioactive lipids are fundamental mediators of a number of critical biologic processes such as inflammation, proliferation, and apoptosis. Rhabdomyosarcoma (RMS) is common in adolescence with histologic subtypes that favor metastasis. However, the factors that influence metastasis are not well appreciated. Here, it is shown that lysophosphatidylcholine (LPC) and its derivative, lysophosphatidic acid (LPA), strongly enhance motility and adhesion of human RMS cells. Importantly, these metastatic-associated phenotypes were observed at physiologic concentrations of these lipids, which naturally occur in biologic fluids. Moreover, the effects of these bioactive lipids were much stronger as compared with known peptide-based prometastatic factors in RMS, such as stromal-derived factor-1 or hepatocyte growth factor/scatter factor. Finally, both LPC and LPA levels were increased in several organs after γ-irradiation or chemotherapy, supporting the hypothesis that radio/chemotherapy induces an unwanted prometastatic environment in these organs. IMPLICATIONS LPC and LPA play a previously underappreciated role in dissemination of RMS and suggest that antimetastatic treatment with specific molecules blocking LPC/LPA activity should be part of standard radio/chemotherapy arsenal.
Collapse
Affiliation(s)
- Gabriela Schneider
- Stem Cell Institute, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky
| | - Zachariah Payne Sellers
- Stem Cell Institute, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky
| | - Ahmed Abdel-Latif
- Division of Cardiovascular Medicine, Gill Heart Institute, University of Kentucky, Lexington, Kentucky
| | - Andrew J Morris
- Division of Cardiovascular Medicine, Gill Heart Institute, University of Kentucky, Lexington, Kentucky
| | - Mariusz Z Ratajczak
- Stem Cell Institute, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky. Department of Physiology Pomeranian Medical University, Szczecin, Poland.
| |
Collapse
|
22
|
Janke H, Pastore F, Schumacher D, Herold T, Hopfner KP, Schneider S, Berdel WE, Büchner T, Woermann BJ, Subklewe M, Bohlander SK, Hiddemann W, Spiekermann K, Polzer H. Activating FLT3 mutants show distinct gain-of-function phenotypes in vitro and a characteristic signaling pathway profile associated with prognosis in acute myeloid leukemia. PLoS One 2014; 9:e89560. [PMID: 24608088 PMCID: PMC3946485 DOI: 10.1371/journal.pone.0089560] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2013] [Accepted: 01/21/2014] [Indexed: 11/28/2022] Open
Abstract
About 30% of patients with acute myeloid leukemia (AML) harbour mutations of the receptor tyrosine kinase FLT3, mostly internal tandem duplications (ITD) and point mutations of the second tyrosine kinase domain (TKD). It was the aim of this study to comprehensively analyze clinical and functional properties of various FLT3 mutants. In 672 normal karyotype AML patients FLT3-ITD, but not FLT3-TKD mutations were associated with a worse relapse free and overall survival in multivariate analysis. In paired diagnosis-relapse samples FLT3-ITD showed higher stability (70%) compared to FLT3-TKD (30%). In vitro, FLT3-ITD induced a strong activating phenotype in Ba/F3 cells. In contrast, FLT3-TKD mutations and other point mutations – including two novel mutations – showed a weaker but clear gain-of-function phenotype with gradual increase in proliferation and protection from apoptosis. The pro-proliferative capacity of the investigated FLT3 mutants was associated with cell surface expression and tyrosine 591 phosphorylation of the FLT3 receptor. Western blot experiments revealed STAT5 activation only in FLT3-ITD positive cell lines, in contrast to FLT3-non-ITD mutants, which displayed an enhanced signal of AKT and MAPK activation. Gene expression analysis revealed distinct difference between FLT3-ITD and FLT3-TKD for STAT5 target gene expression as well as deregulation of SOCS2, ENPP2, PRUNE2 and ART3. FLT3-ITD and FLT3 point mutations show a gain-of-function phenotype with distinct signalling properties in vitro. Although poor prognosis in AML is only associated with FLT3-ITD, all activating FLT3 mutations can contribute to leukemogenesis and are thus potential targets for therapeutic interventions.
Collapse
Affiliation(s)
- Hanna Janke
- Department of Internal Medicine III, University Hospital Munich, Ludwig-Maximilians-University Munich, Germany
- Clinical Cooperative Group Leukemia, Helmholtz Center Munich, Germany
- * E-mail:
| | - Friederike Pastore
- Department of Internal Medicine III, University Hospital Munich, Ludwig-Maximilians-University Munich, Germany
- Clinical Cooperative Group Leukemia, Helmholtz Center Munich, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Daniela Schumacher
- Department of Internal Medicine III, University Hospital Munich, Ludwig-Maximilians-University Munich, Germany
| | - Tobias Herold
- Department of Internal Medicine III, University Hospital Munich, Ludwig-Maximilians-University Munich, Germany
- Clinical Cooperative Group Leukemia, Helmholtz Center Munich, Germany
| | - Karl-Peter Hopfner
- Department of Biochemistry, Gene Center, Ludwig-Maximilians-University Munich, Germany
| | - Stephanie Schneider
- Department of Internal Medicine III, University Hospital Munich, Ludwig-Maximilians-University Munich, Germany
| | - Wolfgang E. Berdel
- Department of Medicine A, Hematology, Oncology and Pneumology, University Muenster, Germany
| | - Thomas Büchner
- Department of Medicine A, Hematology, Oncology and Pneumology, University Muenster, Germany
| | | | - Marion Subklewe
- Department of Internal Medicine III, University Hospital Munich, Ludwig-Maximilians-University Munich, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Clinical Cooperative Group Immunotherapy, Helmholtz Center Munich, Germany
| | - Stefan K. Bohlander
- Department of Molecular Medicine and Pathology, University of Auckland, New Zealand
| | - Wolfgang Hiddemann
- Department of Internal Medicine III, University Hospital Munich, Ludwig-Maximilians-University Munich, Germany
- Clinical Cooperative Group Leukemia, Helmholtz Center Munich, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Karsten Spiekermann
- Department of Internal Medicine III, University Hospital Munich, Ludwig-Maximilians-University Munich, Germany
- Clinical Cooperative Group Leukemia, Helmholtz Center Munich, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Harald Polzer
- Department of Internal Medicine III, University Hospital Munich, Ludwig-Maximilians-University Munich, Germany
- Clinical Cooperative Group Leukemia, Helmholtz Center Munich, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
23
|
|