1
|
Subas Satish HP, Iyer S, Shi MX, Wong AW, Fischer KC, Wardak AZ, Lio D, Brouwer JM, Uren RT, Czabotar PE, Miller MS, Kluck RM. A novel inhibitory BAK antibody enables assessment of non-activated BAK in cancer cells. Cell Death Differ 2024; 31:711-721. [PMID: 38582955 PMCID: PMC11164899 DOI: 10.1038/s41418-024-01289-3] [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] [Received: 11/13/2023] [Revised: 03/21/2024] [Accepted: 03/22/2024] [Indexed: 04/08/2024] Open
Abstract
BAX and BAK are pro-apoptotic members of the BCL2 family that are required to permeabilize the mitochondrial outer membrane. The proteins can adopt a non-activated monomeric conformation, or an activated conformation in which the exposed BH3 domain facilitates binding either to a prosurvival protein or to another activated BAK or BAX protein to promote pore formation. Certain cancer cells are proposed to have high levels of activated BAK sequestered by MCL1 or BCLXL, thus priming these cells to undergo apoptosis in response to BH3 mimetic compounds that target MCL1 or BCLXL. Here we report the first antibody, 14G6, that is specific for the non-activated BAK conformer. A crystal structure of 14G6 Fab bound to BAK revealed a binding site encompassing both the α1 helix and α5-α6 hinge regions of BAK, two sites involved in the unfolding of BAK during its activation. In mitochondrial experiments, 14G6 inhibited BAK unfolding triggered by three diverse BAK activators, supporting crucial roles for both α1 dissociation and separation of the core (α2-α5) and latch (α6-α9) regions in BAK activation. 14G6 bound the majority of BAK in several leukaemia cell lines, and binding decreased following treatment with BH3 mimetics, indicating only minor levels of constitutively activated BAK in those cells. In summary, 14G6 provides a new means of assessing BAK status in response to anti-cancer treatments.
Collapse
Affiliation(s)
- Hema Preethi Subas Satish
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Sweta Iyer
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Melissa X Shi
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Agnes W Wong
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Karla C Fischer
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Ahmad Z Wardak
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Daisy Lio
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Jason M Brouwer
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Rachel T Uren
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Peter E Czabotar
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Michelle S Miller
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia.
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia.
| | - Ruth M Kluck
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia.
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia.
| |
Collapse
|
2
|
Rahn K, Abdallah AT, Gan L, Herbrich S, Sonntag R, Benitez O, Malaney P, Zhang X, Rodriguez AG, Brottem J, Marx G, Brümmendorf TH, Ostareck DH, Ostareck-Lederer A, Crysandt M, Post SM, Naarmann-de Vries IS. Insight into the mechanism of AML del(9q) progression: hnRNP K targets the myeloid master regulators CEBPA (C/EBPα) and SPI1 (PU.1). BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2024; 1867:195004. [PMID: 38008244 DOI: 10.1016/j.bbagrm.2023.195004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 11/08/2023] [Accepted: 11/20/2023] [Indexed: 11/28/2023]
Abstract
Deletions on the long arm of chromosome 9 (del(9q)) are recurrent abnormalities in about 2 % of acute myeloid leukemia cases, which usually involve HNRNPK and are frequently associated with other known aberrations. Based on an Hnrnpk haploinsufficient mouse model, a recent study demonstrated a function of hnRNP K in pathogenesis of myeloid malignancies via the regulation of cellular proliferation and myeloid differentiation programs. Here, we provide evidence that reduced hnRNP K expression results in the dysregulated expression of C/EBPα and additional transcription factors. CyTOF analysis revealed monocytic skewing with increased levels of mature myeloid cells. To explore the role of hnRNP K during normal and pathological myeloid differentiation in humans, we characterized hnRNP K-interacting RNAs in human AML cell lines. Notably, RNA-sequencing revealed several mRNAs encoding key transcription factors involved in the regulation of myeloid differentiation as targets of hnRNP K. We showed that specific sequence motifs confer the interaction of SPI1 and CEBPA 5' and 3'UTRs with hnRNP K. The siRNA mediated reduction of hnRNP K in human AML cells resulted in an increase of PU.1 and C/EBPα that is most pronounced for the p30 isoform. The combinatorial treatment with the inducer of myeloid differentiation valproic acid resulted in increased C/EBPα expression and myeloid differentiation. Together, our results indicate that hnRNP K post-transcriptionally regulates the expression of myeloid master transcription factors. These novel findings can inaugurate novel options for targeted treatment of AML del(9q) by modulation of hnRNP K function.
Collapse
Affiliation(s)
- Kerstin Rahn
- Department of Intensive Care Medicine, University Hospital RWTH Aachen University, Aachen, Germany; Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ali T Abdallah
- Interdisciplinary Center for Clinical Research (IZKF) Aachen, RWTH Aachen University, Germany; Cluster of Excellence on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Lin Gan
- Interdisciplinary Center for Clinical Research (IZKF) Aachen, RWTH Aachen University, Germany
| | - Shelley Herbrich
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Roland Sonntag
- Department of Internal Medicine III, University Hospital RWTH Aachen University, Aachen, Germany
| | - Oscar Benitez
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Prerna Malaney
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xiaorui Zhang
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ashely G Rodriguez
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jared Brottem
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gernot Marx
- Department of Intensive Care Medicine, University Hospital RWTH Aachen University, Aachen, Germany
| | - Tim H Brümmendorf
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, University Hospital RWTH Aachen University, Aachen, Germany; Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Aachen, Germany
| | - Dirk H Ostareck
- Department of Intensive Care Medicine, University Hospital RWTH Aachen University, Aachen, Germany
| | - Antje Ostareck-Lederer
- Department of Intensive Care Medicine, University Hospital RWTH Aachen University, Aachen, Germany
| | - Martina Crysandt
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, University Hospital RWTH Aachen University, Aachen, Germany; Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Aachen, Germany
| | - Sean M Post
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Isabel S Naarmann-de Vries
- Department of Intensive Care Medicine, University Hospital RWTH Aachen University, Aachen, Germany; Section of Bioinformatics and Systems Cardiology, University Hospital Heidelberg, Heidelberg, Germany.
| |
Collapse
|
3
|
Klein DC, Lardo SM, Hainer SJ. The ncBAF Complex Regulates Transcription in AML Through H3K27ac Sensing by BRD9. CANCER RESEARCH COMMUNICATIONS 2024; 4:237-252. [PMID: 38126767 PMCID: PMC10831031 DOI: 10.1158/2767-9764.crc-23-0382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 11/02/2023] [Accepted: 12/13/2023] [Indexed: 12/23/2023]
Abstract
The non-canonical BAF complex (ncBAF) subunit BRD9 is essential for acute myeloid leukemia (AML) cell viability but has an unclear role in leukemogenesis. Because BRD9 is required for ncBAF complex assembly through its DUF3512 domain, precise bromodomain inhibition is necessary to parse the role of BRD9 as a transcriptional regulator from that of a scaffolding protein. To understand the role of BRD9 bromodomain function in regulating AML, we selected a panel of five AML cell lines with distinct driver mutations, disease classifications, and genomic aberrations and subjected these cells to short-term BRD9 bromodomain inhibition. We examined the bromodomain-dependent growth of these cell lines, identifying a dependency in AML cell lines but not HEK293T cells. To define a mechanism through which BRD9 maintains AML cell survival, we examined nascent transcription, chromatin accessibility, and ncBAF complex binding genome-wide after bromodomain inhibition. We identified extensive regulation of transcription by BRD9 bromodomain activity, including repression of myeloid maturation factors and tumor suppressor genes, while standard AML chemotherapy targets were repressed by inhibition of the BRD9 bromodomain. BRD9 bromodomain activity maintained accessible chromatin at both gene promoters and gene-distal putative enhancer regions, in a manner that qualitatively correlated with enrichment of BRD9 binding. Furthermore, we identified reduced chromatin accessibility at GATA, ETS, and AP-1 motifs and increased chromatin accessibility at SNAIL-, HIC-, and TP53-recognized motifs after BRD9 inhibition. These data suggest a role for BRD9 in regulating AML cell differentiation through modulation of accessibility at hematopoietic transcription factor binding sites. SIGNIFICANCE The bromodomain-containing protein BRD9 is essential for AML cell viability, but it is unclear whether this requirement is due to the protein's role as an epigenetic reader. We inhibited this activity and identified altered gene-distal chromatin regulation and transcription consistent with a more mature myeloid cell state.
Collapse
Affiliation(s)
- David C. Klein
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Santana M. Lardo
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Sarah J. Hainer
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
4
|
Skopek R, Palusińska M, Kaczor-Keller K, Pingwara R, Papierniak-Wyglądała A, Schenk T, Lewicki S, Zelent A, Szymański Ł. Choosing the Right Cell Line for Acute Myeloid Leukemia (AML) Research. Int J Mol Sci 2023; 24:5377. [PMID: 36982453 PMCID: PMC10049680 DOI: 10.3390/ijms24065377] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 03/01/2023] [Accepted: 03/09/2023] [Indexed: 03/14/2023] Open
Abstract
Immortalized cell lines are widely used in vitro tools in oncology and hematology research. While these cell lines represent artificial systems and may accumulate genetic aberrations with each passage, they are still considered valuable models for pilot, preliminary, and screening studies. Despite their limitations, cell lines are cost-effective and provide repeatable and comparable results. Choosing the appropriate cell line for acute myeloid leukemia (AML) research is crucial for obtaining reliable and relevant results. Several factors should be considered when selecting a cell line for AML research, such as specific markers and genetic abnormalities associated with different subtypes of AML. It is also essential to evaluate the karyotype and mutational profile of the cell line, as these can influence the behavior and response to the treatment of the cells. In this review, we evaluate immortalized AML cell lines and discuss the issues surrounding them concerning the revised World Health Organization and the French-American-British classifications.
Collapse
Affiliation(s)
- Rafał Skopek
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Postępu 36A, 05-552 Magdalenka, Poland
| | - Małgorzata Palusińska
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Postępu 36A, 05-552 Magdalenka, Poland
| | - Katarzyna Kaczor-Keller
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Postępu 36A, 05-552 Magdalenka, Poland
| | - Rafał Pingwara
- Department of Physiological Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences-SGGW, 02-787 Warsaw, Poland
| | | | - Tino Schenk
- Department of Hematology and Medical Oncology, Clinic of Internal Medicine II, Jena University Hospital, 07747 Jena, Germany
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine Jena (CMB), Jena University Hospital, 07747 Jena, Germany
| | - Sławomir Lewicki
- Faculty of Medical Sciences and Health Sciences, Kazimierz Pulaski University of Technology and Humanities, 26-600 Radom, Poland
- Institute of Outcomes Research, Maria Sklodowska-Curie Medical Academy, 00-001 Warsaw, Poland
| | - Artur Zelent
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Postępu 36A, 05-552 Magdalenka, Poland
| | - Łukasz Szymański
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Postępu 36A, 05-552 Magdalenka, Poland
| |
Collapse
|
5
|
A pyridinesulfonamide derivative FD268 suppresses cell proliferation and induces apoptosis via inhibiting PI3K pathway in acute myeloid leukemia. PLoS One 2022; 17:e0277893. [PMID: 36413544 PMCID: PMC9681083 DOI: 10.1371/journal.pone.0277893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 11/04/2022] [Indexed: 11/23/2022] Open
Abstract
Aberration of PI3K signaling pathway has been confirmed to be associated with several hematological malignancies including acute myeloid leukemia (AML). FD268, a pyridinesulfonamide derivative characterized by the conjugation of 7-azaindole group, is a newly identified PI3K inhibitor showing high potent enzyme activity at nanomole concentration. In this study, we demonstrated that FD268 dose-dependently inhibits survival of AML cells with the efficacy superior to that of PI-103 (pan-PI3K inhibitor) and CAL-101 (selective PI3Kδ inhibitor) in the tested HL-60, MOLM-16, Mv-4-11, EOL-1 and KG-1 cell lines. Further mechanistic studies focused on HL-60 revealed that FD268 significantly inhibits the PI3K/Akt/mTOR signaling pathway, promotes the activation of pro-apoptotic protein Bad and downregulates the expression of anti-apoptotic protein Mcl-1, thus suppressing the cell proliferation and inducing caspase-3-dependent apoptosis. The bioinformatics analysis of the transcriptome sequencing data also indicated a potential involvement of the PI3K/Akt/mTOR pathway. These studies indicated that FD268 possesses high potent activity toward AML cells via inhibition of PI3K/Akt/mTOR signaling pathway, which sheds some light on the pyridinesulfonamide scaffold for further optimization and investigation.
Collapse
|
6
|
Ramos DFV, Mancuso RI, Contieri B, Duarte A, Paiva L, de Melo Carrilho J, Saad STO, Lazarini M. Rac GTPases in acute myeloid leukemia cells: Expression profile and biological effects of pharmacological inhibition. Toxicol Appl Pharmacol 2022; 442:115990. [PMID: 35331739 DOI: 10.1016/j.taap.2022.115990] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 03/10/2022] [Accepted: 03/17/2022] [Indexed: 12/17/2022]
Abstract
Acute myeloid leukemia (AML) is a highly heterogeneous hematological neoplasm with low survival rates. Thus, the investigation of new therapeutic targets is essential. The Rac subfamily of GTPase proteins has been shown to participate in the physiopathology of hematological malignancies. However, their expression and function in AML remain unclear. In this study, we evaluated Rac1, Rac2 and Rac3 gene expressions in AML and their impact on clinical outcomes. We further investigated the effects of the in vitro treatment with a Rac inhibitor (EHT-1864) on AML cell lines. Rac3 expression was increased in AML derived from myelodysplastic syndromes compared to healthy donors. Rac2 expression did not differ between AML patients and healthy donors, but de novo AML patients with higher Rac2 presented lower overall survival. Oncogenic pathway gene-sets related to AKT/mTOR were identified as associated with Rac1, Rac2 and Rac3 expressions. EHT-1864 treatment reduced the viability of OCI-AML3, KG1 and Kasumi-1 cells in a time and dose-dependent manner. In OCI-AML3 cells, treatment with EHT-1864 induced apoptosis, autophagy, and led to the accumulation of cells in the G1 phase of the cell cycle. These changes were concomitant with alterations in p53 and cyclins. Dowregulation of the PI3K/AKT/mTOR pathway was also observed. Interestingly, the combined treatment of EHT-1864 and low doses of daunorubicin enhanced OCI-AML3 cell apoptosis. In conclusion, Rac2 expression is a prognostic factor in AML and our preclinical results suggest that Rac inhibition may be an attractive mechanism to compose the antineoplastic strategy for this disease.
Collapse
Affiliation(s)
| | - Rubia Isler Mancuso
- Hematology and Bloood Transfusion Center, University of Campinas, Campinas, São Paulo, Brazil
| | - Bruna Contieri
- Department of Pharmaceutical Sciences, Federal University of São Paulo, Diadema, São Paulo, Brazil
| | - Adriana Duarte
- Hematology and Bloood Transfusion Center, University of Campinas, Campinas, São Paulo, Brazil
| | - Luciana Paiva
- Hematology and Bloood Transfusion Center, University of Campinas, Campinas, São Paulo, Brazil
| | | | | | - Mariana Lazarini
- Department of Pharmaceutical Sciences, Federal University of São Paulo, Diadema, São Paulo, Brazil; Hematology and Bloood Transfusion Center, University of Campinas, Campinas, São Paulo, Brazil.
| |
Collapse
|
7
|
Vorobyev PO, Babaeva FE, Panova AV, Shakiba J, Kravchenko SK, Soboleva AV, Lipatova AV. Oncolytic Viruses in the Therapy of Lymphoproliferative Diseases. Mol Biol 2022; 56:684-695. [PMID: 36217339 PMCID: PMC9534467 DOI: 10.1134/s0026893322050144] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 04/28/2022] [Accepted: 05/04/2022] [Indexed: 11/23/2022]
Abstract
Cancer is a leading causes of death. Despite significant success in the treatment of lymphatic system tumors, the problems of relapse, drug resistance and effectiveness of therapy remain relevant. Oncolytic viruses are able to replicate in tumor cells and destroy them without affecting normal, healthy tissues. By activating antitumor immunity, viruses are effective against malignant neoplasms of various nature. In lymphoproliferative diseases with a drug-resistant phenotype, many cases of remissions have been described after viral therapy. The current level of understanding of viral biology and the discovery of host cell interaction mechanisms made it possible to create unique strains with high oncoselectivity widely used in clinical practice in recent years.
Collapse
Affiliation(s)
- P. O. Vorobyev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - F. E. Babaeva
- National Medical Research Center for Hematology, Ministry of Health of Russia, 125167 Moscow, Russia
| | - A. V. Panova
- Vavilov Institute of General Genetics, Russian Academy of Sciences, 117971 Moscow, Russia
| | - J. Shakiba
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | - S. K. Kravchenko
- National Medical Research Center for Hematology, Ministry of Health of Russia, 125167 Moscow, Russia
| | - A. V. Soboleva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - A. V. Lipatova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| |
Collapse
|
8
|
Baba T, Yoshida T, Tanabe Y, Nishimura T, Morishita S, Gotoh N, Hirao A, Hanayama R, Mukaida N. Cytoplasmic DNA accumulation preferentially triggers cell death of myeloid leukemia cells by interacting with intracellular DNA sensing pathway. Cell Death Dis 2021; 12:322. [PMID: 33771977 PMCID: PMC7997981 DOI: 10.1038/s41419-021-03587-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 03/02/2021] [Accepted: 03/04/2021] [Indexed: 12/12/2022]
Abstract
Accumulating evidence indicates the presence of cytoplasmic DNAs in various types of malignant cells, and its involvement in anti-cancer drug- or radiotherapy-mediated DNA damage response and replication stress. However, the pathophysiological roles of cytoplasmic DNAs in leukemias remain largely unknown. We observed that during hematopoietic stem cell transplantation (HSCT) in mouse myeloid leukemia models, double-stranded (ds)DNAs were constitutively secreted in the form of extracellular vesicles (EVs) from myeloid leukemia cells and were transferred to the donor cells to dampen their hematopoietic capabilities. Subsequent analysis of cytoplasmic DNA dynamics in leukemia cells revealed that autophagy regulated cytoplasmic dsDNA accumulation and subsequent redistribution into EVs. Moreover, accumulated cytoplasmic dsDNAs activated STING pathway, thereby reducing leukemia cell viability through reactive oxygen species (ROS) generation. Pharmaceutical inhibition of autophagosome formation induced cytoplasmic DNA accumulation, eventually triggering cytoplasmic DNA sensing pathways to exert cytotoxicity, preferentially in leukemia cells. Thus, manipulation of cytoplasmic dsDNA dynamics can be a novel and potent therapeutic strategy for myeloid leukemias.
Collapse
Affiliation(s)
- Tomohisa Baba
- Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kanazawa, Japan.
| | - Takeshi Yoshida
- Department of Immunology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan.,Nano Life Science Institute, Kanazawa University, Kanazawa, Japan
| | - Yamato Tanabe
- Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Tatsunori Nishimura
- Division of Cancer Cell Biology, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Soji Morishita
- Department of Transfusion Medicine and Stem Cell Regulation, Juntendo University School of Medicine, Tokyo, Japan
| | - Noriko Gotoh
- Division of Cancer Cell Biology, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Atsushi Hirao
- Nano Life Science Institute, Kanazawa University, Kanazawa, Japan.,Division of Molecular Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Rikinari Hanayama
- Department of Immunology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan.,Nano Life Science Institute, Kanazawa University, Kanazawa, Japan
| | - Naofumi Mukaida
- Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
9
|
Mauch-Mücke K, Schön K, Paulus C, Nevels MM. Evidence for Tethering of Human Cytomegalovirus Genomes to Host Chromosomes. Front Cell Infect Microbiol 2020; 10:577428. [PMID: 33117732 PMCID: PMC7561393 DOI: 10.3389/fcimb.2020.577428] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 08/17/2020] [Indexed: 11/27/2022] Open
Abstract
Tethering of viral genomes to host chromosomes has been recognized in a variety of DNA and RNA viruses. It can occur during both the productive cycle and latent infection and may impact viral genomes in manifold ways including their protection, localization, transcription, replication, integration, and segregation. Tethering is typically accomplished by dedicated viral proteins that simultaneously associate with both the viral genome and cellular chromatin via nucleic acid, histone and/or non-histone protein interactions. Some of the most prominent tethering proteins have been identified in DNA viruses establishing sustained latent infections, including members of the papillomaviruses and herpesviruses. Herpesvirus particles have linear genomes that circularize in infected cell nuclei and usually persist as extrachromosomal episomes. In several γ-herpesviruses, tethering facilitates the nuclear retention and faithful segregation of viral episomes during cell division, thus contributing to persistence of these viruses in the absence of infectious particle production. However, it has not been studied whether the genomes of human Cytomegalovirus (hCMV), the prototypical β-herpesvirus, are tethered to host chromosomes. Here we provide evidence by fluorescence in situ hybridization that hCMV genomes associate with the surface of human mitotic chromosomes following infection of both non-permissive myeloid and permissive fibroblast cells. This chromosome association occurs at lower frequency in the absence of the immediate-early 1 (IE1) proteins, which bind to histones and have been implicated in the maintenance of hCMV episomes. Our findings point to a mechanism of hCMV genome maintenance through mitosis and suggest a supporting but non-essential role of IE1 in this process.
Collapse
Affiliation(s)
- Katrin Mauch-Mücke
- Institute for Medical Microbiology and Hygiene, University of Regensburg, Regensburg, Germany
| | - Kathrin Schön
- Institute for Medical Microbiology and Hygiene, University of Regensburg, Regensburg, Germany
| | - Christina Paulus
- Biomedical Sciences Research Complex, University of St Andrews, St Andrews, United Kingdom
| | - Michael M Nevels
- Biomedical Sciences Research Complex, University of St Andrews, St Andrews, United Kingdom
| |
Collapse
|
10
|
Koubi M, Poplineau M, Vernerey J, N'Guyen L, Tiberi G, Garciaz S, El-Kaoutari A, Maqbool MA, Andrau JC, Guillouf C, Saurin AJ, Duprez E. Regulation of the positive transcriptional effect of PLZF through a non-canonical EZH2 activity. Nucleic Acids Res 2019; 46:3339-3350. [PMID: 29425303 PMCID: PMC5909434 DOI: 10.1093/nar/gky080] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 01/31/2018] [Indexed: 11/13/2022] Open
Abstract
The transcription factor PLZF (promyelocytic leukemia zinc finger protein) acts as an epigenetic regulator balancing self-renewal and differentiation of hematopoietic cells through binding to various chromatin-modifying factors. First described as a transcriptional repressor, PLZF is also associated with active transcription, although the molecular bases underlying the differences are unknown. Here, we reveal that in a hematopoietic cell line, PLZF is predominantly associated with transcribed genes. Additionally, we identify a new association between PLZF and the histone methyltransferase, EZH2 at the genomic level. We find that co-occupancy of PLZF and EZH2 on chromatin at PLZF target genes is not associated with SUZ12 or trimethylated lysine 27 of histone H3 (H3K27me3) but with the active histone mark H3K4me3 and active transcription. Removal of EZH2 leads to an increase of PLZF binding and increased gene expression. Our results suggest a new role of EZH2 in restricting PLZF positive transcriptional activity independently of its canonical PRC2 activity.
Collapse
Affiliation(s)
- Myriam Koubi
- Epigenetic Factors in Normal and Malignant Hematopoiesis, Aix Marseille Université, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, 13273 Marseille Cedex 9, France
| | - Mathilde Poplineau
- Epigenetic Factors in Normal and Malignant Hematopoiesis, Aix Marseille Université, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, 13273 Marseille Cedex 9, France
| | - Julien Vernerey
- Epigenetic Factors in Normal and Malignant Hematopoiesis, Aix Marseille Université, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, 13273 Marseille Cedex 9, France
| | - Lia N'Guyen
- Epigenetic Factors in Normal and Malignant Hematopoiesis, Aix Marseille Université, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, 13273 Marseille Cedex 9, France
| | - Guillaume Tiberi
- Epigenetic Factors in Normal and Malignant Hematopoiesis, Aix Marseille Université, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, 13273 Marseille Cedex 9, France
| | - Sylvain Garciaz
- Epigenetic Factors in Normal and Malignant Hematopoiesis, Aix Marseille Université, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, 13273 Marseille Cedex 9, France
| | - Abdessamad El-Kaoutari
- Epigenetic Factors in Normal and Malignant Hematopoiesis, Aix Marseille Université, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, 13273 Marseille Cedex 9, France
| | - Muhammad A Maqbool
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, 34293 Montpellier, Cedex 5, France
| | - Jean-Christophe Andrau
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, 34293 Montpellier, Cedex 5, France
| | - Christel Guillouf
- Gustave Roussy, Université Paris-Saclay, Inserm U1170, CNRS Villejuif, France
| | - Andrew J Saurin
- Aix Marseille Université, CNRS, IBDM, UMR 7288, 13288 Marseille, Cedex 9, France
| | - Estelle Duprez
- Epigenetic Factors in Normal and Malignant Hematopoiesis, Aix Marseille Université, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, 13273 Marseille Cedex 9, France
| |
Collapse
|
11
|
Cluster binding studies with two anti-Thomsen-Friedenreich (anti-core-1, CD176, TF) antibodies: Evidence for a multiple TF epitope. Int Immunopharmacol 2019; 72:186-194. [PMID: 30999209 DOI: 10.1016/j.intimp.2019.03.058] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Revised: 03/04/2019] [Accepted: 03/28/2019] [Indexed: 11/21/2022]
Abstract
Antibodies to carbohydrate epitopes are often of the IgM isotype and require multiple binding for sufficient avidity. Therefore clusters of epitopes are preferred antigenic sites in these cases. We have examined the type of clusters recognized by two anti-Thomsen-Friedenreich (TF, core-1, CD176) IgM antibodies, NM-TF1 and NM-TF2, using several different sets of TF-carrying synthetic glycoconjugates in ELISA experiments. To our surprise, the single most important factor determining binding strength was a close vicinity of several TF glycans at distances of ≤1 nm. Considering the known dimensions of IgM antibodies, our data strongly suggest that a cluster of up to four TF moieties, presenting as a "multiple epitope", is required to attach to a single combining site in order to result in adequate binding strength. This effect can also be achieved by "surrogate-multiple epitopes" consisting of separate TF-carrying molecules in close vicinity. In addition, it was found that serine-linked TFs are stronger bound than threonine-linked TFs by both antibodies. This peculiar type of cluster recognition may contribute to improved avidity and explicit tumor specificity.
Collapse
|
12
|
Hassani S, Ghaffari P, Chahardouli B, Alimoghaddam K, Ghavamzadeh A, Alizadeh S, Ghaffari SH. Disulfiram/copper causes ROS levels alteration, cell cycle inhibition, and apoptosis in acute myeloid leukaemia cell lines with modulation in the expression of related genes. Biomed Pharmacother 2018; 99:561-569. [PMID: 29902866 DOI: 10.1016/j.biopha.2018.01.109] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 01/18/2018] [Accepted: 01/24/2018] [Indexed: 01/13/2023] Open
Abstract
The majority of acute myeloid leukaemia (AML) patients will die from their disease or therapy-related complications. There is an inevitable need to improve the survival of AML patients. Previous studies show that disulfiram (DSF), an anti-alcoholism drug with a low toxicity profile, demonstrates anticancer behaviors. Here, we evaluated the cytotoxicity and mechanistic action of DSF on the AML cell lines KG-1, NB4, and U937. The microculture tetrazolium test revealed that DSF alone or in combination with copper (Cu) is highly toxic to the AML cells at concentrations lower than those achievable in the clinical setting, with Cu increasing the DSF-induced inhibition of metabolic activity. Flow cytometric analysis and QRT-PCR indicated that in the two cell lines, NB4 and U-937, DSF/Cu increased reactive oxygen species (ROS) levels in association with the induction of superoxide dismutase 2 (SOD2) expression and suppression of catalase (CAT). In the KG-1 cell line, DSF/Cu reduced the ROS levels in agreement with the induction of CAT expression. The cell cycle and apoptosis assessment by flow cytometry demonstrated that DSF/Cu induced G0/G1 cell cycle arrest and apoptosis. These were associated with the increased expression of FOXO tumor suppressors, decreased expression of the MYC oncogene and the modulation of their known target genes related to the cell cycle and apoptosis. Therefore, DSF/Cu caused the disturbance of the ROS balance, cell cycle arrest and apoptosis in AML cells in coordination with the modulation in expression of their related genes. These results propose the possible use of DSF in AML therapies.
Collapse
MESH Headings
- Apoptosis/drug effects
- Apoptosis/genetics
- Cell Cycle Checkpoints/drug effects
- Cell Cycle Checkpoints/genetics
- Cell Line, Tumor
- Copper/pharmacology
- Disulfiram/pharmacology
- G1 Phase/drug effects
- G1 Phase/genetics
- Gene Expression Regulation, Leukemic/drug effects
- Humans
- Inhibitory Concentration 50
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Models, Biological
- Reactive Oxygen Species/metabolism
- Resting Phase, Cell Cycle/drug effects
- Resting Phase, Cell Cycle/genetics
Collapse
Affiliation(s)
- Saeed Hassani
- Hematology Department, School of Allied Medicine, Tehran University of Medical Sciences, Tehran, Iran; Hematologic Malignancies Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Parisa Ghaffari
- Hematologic Malignancies Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahram Chahardouli
- Hematologic Malignancies Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Kamran Alimoghaddam
- Hematologic Malignancies Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ardeshir Ghavamzadeh
- Hematologic Malignancies Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Shaban Alizadeh
- Hematology Department, School of Allied Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Seyed H Ghaffari
- Hematologic Malignancies Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
13
|
Cornet-Masana JM, Moreno-Martínez D, Lara-Castillo MC, Nomdedeu M, Etxabe A, Tesi N, Pratcorona M, Esteve J, Risueño RM. Emetine induces chemosensitivity and reduces clonogenicity of acute myeloid leukemia cells. Oncotarget 2018; 7:23239-50. [PMID: 26992240 PMCID: PMC5029623 DOI: 10.18632/oncotarget.8096] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 02/28/2016] [Indexed: 01/22/2023] Open
Abstract
Acute myeloid leukemia (AML) is an hematologic neoplasia characterized by the accumulation of transformed immature myeloid cells in bone marrow. Although the response rate to induction therapy is high, survival rate 5-year after diagnosis is still low, highlighting the necessity of new novel agents. To identify agents with the capability to abolish the self-renewal capacity of AML blasts, an in silico screening was performed to search for small molecules that induce terminal differentiation. Emetine, a hit compound, was validated for its anti-leukemic effect in vitro, ex vivo and in vivo. Emetine, a second-line anti-protozoa drug, differentially reduced cell viability and clonogenic capacity of AML primary patient samples, sparing healthy blood cells. Emetine treatment markedly reduced AML burden in bone marrow of xenotransplanted mice and decreased self-renewal capacity of the remaining engrafted AML cells. Emetine also synergized with commonly used chemotherapeutic agents such as ara-C. At a molecular level, emetine treatment was followed by a reduction in HIF-1α protein levels. This study validated the anti-leukemiceffect of emetine in AML cell lines, a group of diverse AML primary samples, and in a human AML-transplanted murine model, sparing healthy blood cells. The selective anti-leukemic effect of emetine together with the safety of the dose range required to exert this effect support the development of this agent in clinical practice.
Collapse
Affiliation(s)
| | | | | | - Meritxell Nomdedeu
- Josep Carreras Leukaemia Research Institute, Barcelona, Spain.,Department of Hematology, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Amaia Etxabe
- Josep Carreras Leukaemia Research Institute, Barcelona, Spain
| | - Niccolò Tesi
- Josep Carreras Leukaemia Research Institute, Barcelona, Spain
| | - Marta Pratcorona
- Josep Carreras Leukaemia Research Institute, Barcelona, Spain.,Department of Hematology, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Jordi Esteve
- Josep Carreras Leukaemia Research Institute, Barcelona, Spain.,Department of Hematology, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Ruth M Risueño
- Josep Carreras Leukaemia Research Institute, Barcelona, Spain
| |
Collapse
|
14
|
Sarker JM, Pearce SM, Nelson RP, Kinzer-Ursem TL, Umulis DM, Rundell AE. An Integrative multi-lineage model of variation in leukopoiesis and acute myelogenous leukemia. BMC SYSTEMS BIOLOGY 2017; 11:78. [PMID: 28841879 PMCID: PMC5574150 DOI: 10.1186/s12918-017-0469-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 08/11/2017] [Indexed: 12/11/2022]
Abstract
Background Acute myelogenous leukemia (AML) progresses uniquely in each patient. However, patients are typically treated with the same types of chemotherapy, despite biological differences that lead to differential responses to treatment. Results Here we present a multi-lineage multi-compartment model of the hematopoietic system that captures patient-to-patient variation in both the concentration and rates of change of hematopoietic cell populations. By constraining the model against clinical hematopoietic cell recovery data derived from patients who have received induction chemotherapy, we identified trends for parameters that must be met by the model; for example, the mitosis rates and the probability of self-renewal of progenitor cells are inversely related. Within the data-consistent models, we found 22,796 parameter sets that meet chemotherapy response criteria. Simulations of these parameter sets display diverse dynamics in the cell populations. To identify large trends in these model outputs, we clustered the simulated cell population dynamics using k-means clustering and identified thirteen ‘representative patient’ dynamics. In each of these patient clusters, we simulated AML and found that clusters with the greatest mitotic capacity experience clinical cancer outcomes more likely to lead to shorter survival times. Conversely, other parameters, including lower death rates or mobilization rates, did not correlate with survival times. Conclusions Using the multi-lineage model of hematopoiesis, we have identified several key features that determine leukocyte homeostasis, including self-renewal probabilities and mitosis rates, but not mobilization rates. Other influential parameters that regulate AML model behavior are responses to cytokines/growth factors produced in peripheral blood that target the probability of self-renewal of neutrophil progenitors. Finally, our model predicts that the mitosis rate of cancer is the most predictive parameter for survival time, followed closely by parameters that affect the self-renewal of cancer stem cells; most current therapies target mitosis rate, but based on our results, we propose that additional therapeutic targeting of self-renewal of cancer stem cells will lead to even higher survival rates. Electronic supplementary material The online version of this article (doi:10.1186/s12918-017-0469-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Joyatee M Sarker
- Weldon School of Biomedical Engineering, Purdue University, 206 S. Martin Jischke Drive, West Lafayette, 47906, IN, USA
| | - Serena M Pearce
- Weldon School of Biomedical Engineering, Purdue University, 206 S. Martin Jischke Drive, West Lafayette, 47906, IN, USA
| | - Robert P Nelson
- Department of Medicine and Pediatrics, Divisions of Hematology/Oncology, Indiana University School of Medicine, 535 Barnhill Dr., Ste. 473, Indianapolis, 46202, IN, USA
| | - Tamara L Kinzer-Ursem
- Weldon School of Biomedical Engineering, Purdue University, 206 S. Martin Jischke Drive, West Lafayette, 47906, IN, USA
| | - David M Umulis
- Weldon School of Biomedical Engineering, Purdue University, 206 S. Martin Jischke Drive, West Lafayette, 47906, IN, USA. .,Ag. and Biological Engineering, Purdue University, 206 S. Martin Jischke Drive, West Lafayette, 47906, IN, USA.
| | - Ann E Rundell
- Weldon School of Biomedical Engineering, Purdue University, 206 S. Martin Jischke Drive, West Lafayette, 47906, IN, USA
| |
Collapse
|
15
|
Alharbi RA, Pandha HS, Simpson GR, Pettengell R, Poterlowicz K, Thompson A, Harrington K, El-Tanani M, Morgan R. Inhibition of HOX/PBX dimer formation leads to necroptosis in acute myeloid leukemia cells. Oncotarget 2017; 8:89566-89579. [PMID: 29163771 PMCID: PMC5685692 DOI: 10.18632/oncotarget.20023] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 06/26/2017] [Indexed: 12/31/2022] Open
Abstract
The HOX genes encode a family of transcription factors that have key roles in both development and malignancy. Disrupting the interaction between HOX proteins and their binding partner, PBX, has been shown to cause apoptotic cell death in a range of solid tumors. However, despite HOX proteins playing a particularly significant role in acute myeloid leukemia (AML), the relationship between HOX gene expression and patient survival has not been evaluated (with the exception of HOXA9), and the mechanism by which HOX/PBX inhibition induces cell death in this malignancy is not well understood. In this study, we show that the expression of HOXA5, HOXB2, HOXB4, HOXB9, and HOXC9, but not HOXA9, in primary AML samples is significantly related to survival. Furthermore, the previously described inhibitor of HOX/PBX dimerization, HXR9, is cytotoxic to both AML-derived cell lines and primary AML cells from patients. The mechanism of cell death is not dependent on apoptosis but instead involves a regulated form of necrosis referred to as necroptosis. HXR9-induced necroptosis is enhanced by inhibitors of protein kinase C (PKC) signaling, and HXR9 combined with the PKC inhibitor Ro31 causes a significantly greater reduction in tumor growth compared to either reagent alone.
Collapse
Affiliation(s)
- Raed A Alharbi
- Department of Laboratory Medicine, Faculty of Applied Medical Sciences, Albaha University, Albaha, Saudi Arabia.,Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Hardev S Pandha
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Guy R Simpson
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | | | | | - Alexander Thompson
- Division of Cancer and Stem Cells, Centre for Biomolecular Sciences, University of Nottingham, Nottingham, UK
| | - Kevin Harrington
- Targeted Therapy Team, Chester Beatty Laboratories, Institute of Cancer Research, London, UK
| | - Mohamed El-Tanani
- Institute of Cancer Therapeutics, University of Bradford, Bradford, UK
| | - Richard Morgan
- Institute of Cancer Therapeutics, University of Bradford, Bradford, UK
| |
Collapse
|
16
|
Riether C, Schürch CM, Bührer ED, Hinterbrandner M, Huguenin AL, Hoepner S, Zlobec I, Pabst T, Radpour R, Ochsenbein AF. CD70/CD27 signaling promotes blast stemness and is a viable therapeutic target in acute myeloid leukemia. J Exp Med 2016; 214:359-380. [PMID: 28031480 PMCID: PMC5294846 DOI: 10.1084/jem.20152008] [Citation(s) in RCA: 122] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Revised: 09/18/2016] [Accepted: 12/08/2016] [Indexed: 12/23/2022] Open
Abstract
Riether et al. show that CD70/CD27 signaling activates stem cell gene expression programs in acute myeloid leukemia (AML). Blocking the CD70/CD27 interaction inhibits self-renewal and induces differentiation of AML blasts and stem/progenitor cells. Aberrant proliferation, symmetric self-renewal, increased survival, and defective differentiation of malignant blasts are key oncogenic drivers in acute myeloid leukemia (AML). Stem cell gene signatures predict poor prognosis in AML patients; however, with few exceptions, these deregulated molecular pathways cannot be targeted therapeutically. In this study, we demonstrate that the TNF superfamily ligand–receptor pair CD70/CD27 is expressed on AML blasts and AML stem/progenitor cells. CD70/CD27 signaling in AML cells activates stem cell gene expression programs, including the Wnt pathway, and promotes symmetric cell divisions and proliferation. Soluble CD27, reflecting the extent of CD70/CD27 interactions in vivo, was significantly elevated in the sera of newly diagnosed AML patients and is a strong independent negative prognostic biomarker for overall survival. Blocking the CD70/CD27 interaction by mAb induced asymmetric cell divisions and differentiation in AML blasts and AML stem/progenitor cells, inhibited cell growth and colony formation, and significantly prolonged survival in murine AML xenografts. Importantly, hematopoietic stem/progenitor cells from healthy BM donors express neither CD70 nor CD27 and were unaffected by blocking mAb treatment. Therefore, targeting CD70/CD27 signaling represents a promising therapeutic strategy for AML.
Collapse
Affiliation(s)
- Carsten Riether
- Tumor Immunology, Department of Clinical Research, University of Bern, 3008 Bern, Switzerland.,Department of Medical Oncology, Inselspital, University Hospital and University of Bern, 3010 Bern, Switzerland
| | - Christian M Schürch
- Tumor Immunology, Department of Clinical Research, University of Bern, 3008 Bern, Switzerland.,Institute of Pathology, University of Bern, 3008 Bern, Switzerland
| | - Elias D Bührer
- Tumor Immunology, Department of Clinical Research, University of Bern, 3008 Bern, Switzerland
| | | | - Anne-Laure Huguenin
- Tumor Immunology, Department of Clinical Research, University of Bern, 3008 Bern, Switzerland
| | - Sabine Hoepner
- Tumor Immunology, Department of Clinical Research, University of Bern, 3008 Bern, Switzerland
| | - Inti Zlobec
- Institute of Pathology, University of Bern, 3008 Bern, Switzerland
| | - Thomas Pabst
- Department of Medical Oncology, Inselspital, University Hospital and University of Bern, 3010 Bern, Switzerland
| | - Ramin Radpour
- Tumor Immunology, Department of Clinical Research, University of Bern, 3008 Bern, Switzerland
| | - Adrian F Ochsenbein
- Tumor Immunology, Department of Clinical Research, University of Bern, 3008 Bern, Switzerland .,Department of Medical Oncology, Inselspital, University Hospital and University of Bern, 3010 Bern, Switzerland
| |
Collapse
|
17
|
Repositioning of bromocriptine for treatment of acute myeloid leukemia. J Transl Med 2016; 14:261. [PMID: 27604463 PMCID: PMC5015257 DOI: 10.1186/s12967-016-1007-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 08/16/2016] [Indexed: 11/26/2022] Open
Abstract
Background Treatment for acute myeloid leukemia (AML) has not significantly changed in the last decades and new therapeutic approaches are needed to achieve prolonged survival rates. Leukemia stem cells (LSC) are responsible for the initiation and maintenance of AML due to their stem-cell properties. Differentiation therapies aim to abrogate the self-renewal capacity and diminish blast lifespan. Methods An in silico screening was designed to search for FDA-approved small molecules that potentially induce differentiation of AML cells. Bromocriptine was identified and validated in an in vitro screening. Bromocriptine is an approved drug originally indicated for Parkinson’s disease, acromegaly, hyperprolactinemia and galactorrhoea, and recently repositioned for diabetes mellitus. Results Treatment with bromocriptine reduced cell viability of AML cells by activation of the apoptosis program and induction of myeloid differentiation. Moreover, the LSC-enriched primitive AML cell fraction was more sensitive to the presence of bromocriptine. In fact, bromocriptine decreased the clonogenic capacity of AML cells. Interestingly, a negligible effect is observed in healthy blood cells and hematopoietic stem/progenitor cells. Conclusions Our results support the use of bromocriptine as an anti-AML drug in a repositioning setting and the further clinical validation of this preclinical study. Electronic supplementary material The online version of this article (doi:10.1186/s12967-016-1007-5) contains supplementary material, which is available to authorized users.
Collapse
|
18
|
Prevosto C, Usmani MF, McDonald S, Gumienny AM, Key T, Goodman RS, Gaston JSH, Deery MJ, Busch R. Allele-Independent Turnover of Human Leukocyte Antigen (HLA) Class Ia Molecules. PLoS One 2016; 11:e0161011. [PMID: 27529174 PMCID: PMC4987023 DOI: 10.1371/journal.pone.0161011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 07/28/2016] [Indexed: 11/29/2022] Open
Abstract
Major histocompatibility complex class I (MHCI) glycoproteins present cytosolic peptides to CD8+ T cells and regulate NK cell activity. Their heavy chains (HC) are expressed from up to three MHC gene loci (human leukocyte antigen [HLA]-A, -B, and -C in humans), whose extensive polymorphism maps predominantly to the antigen-binding groove, diversifying the bound peptide repertoire. Codominant expression of MHCI alleles is thus functionally critical, but how it is regulated is not fully understood. Here, we have examined the effect of polymorphism on the turnover rates of MHCI molecules in cell lines with functional MHCI peptide loading pathways and in monocyte-derived dendritic cells (MoDCs). Proteins were labeled biosynthetically with heavy water (2H2O), folded MHCI molecules immunoprecipitated, and tryptic digests analysed by mass spectrometry. MHCI-derived peptides were assigned to specific alleles and isotypes, and turnover rates quantified by 2H incorporation, after correcting for cell growth. MHCI turnover half-lives ranged from undetectable to a few hours, depending on cell type, activation state, donor, and MHCI isotype. However, in all settings, the turnover half-lives of alleles of the same isotype were similar. Thus, MHCI protein turnover rates appear to be allele-independent in normal human cells. We propose that this is an important feature enabling the normal function and codominant expression of MHCI alleles.
Collapse
Affiliation(s)
- Claudia Prevosto
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - M. Farooq Usmani
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Sarah McDonald
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | | | - Tim Key
- Tissue Typing Laboratory, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Reyna S. Goodman
- Tissue Typing Laboratory, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - J. S. Hill Gaston
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Michael J. Deery
- Cambridge Centre for Proteomics, University of Cambridge, Cambridge, United Kingdom
| | - Robert Busch
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
- Department of Life Sciences, University of Roehampton, London, United Kingdom
| |
Collapse
|
19
|
Quantitative comparison of DNA methylation assays for biomarker development and clinical applications. Nat Biotechnol 2016; 34:726-37. [PMID: 27347756 DOI: 10.1038/nbt.3605] [Citation(s) in RCA: 226] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Accepted: 05/10/2016] [Indexed: 02/08/2023]
Abstract
DNA methylation patterns are altered in numerous diseases and often correlate with clinically relevant information such as disease subtypes, prognosis and drug response. With suitable assays and after validation in large cohorts, such associations can be exploited for clinical diagnostics and personalized treatment decisions. Here we describe the results of a community-wide benchmarking study comparing the performance of all widely used methods for DNA methylation analysis that are compatible with routine clinical use. We shipped 32 reference samples to 18 laboratories in seven different countries. Researchers in those laboratories collectively contributed 21 locus-specific assays for an average of 27 predefined genomic regions, as well as six global assays. We evaluated assay sensitivity on low-input samples and assessed the assays' ability to discriminate between cell types. Good agreement was observed across all tested methods, with amplicon bisulfite sequencing and bisulfite pyrosequencing showing the best all-round performance. Our technology comparison can inform the selection, optimization and use of DNA methylation assays in large-scale validation studies, biomarker development and clinical diagnostics.
Collapse
|
20
|
Atashrazm F, Lowenthal RM, Woods GM, Holloway AF, Karpiniec SS, Dickinson JL. Fucoidan Suppresses the Growth of Human Acute Promyelocytic Leukemia Cells In Vitro and In Vivo. J Cell Physiol 2016; 231:688-97. [PMID: 26241708 DOI: 10.1002/jcp.25119] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 07/30/2015] [Indexed: 01/10/2023]
Abstract
Fucoidan, a natural component of seaweeds, is reported to have immunomodulatory and anti-tumor effects. The mechanisms underpinning these activities remain poorly understood. In this study, the cytotoxicity and anti-tumor activities of fucoidan were investigated in acute myeloid leukemia (AML) cells. The human AML cell lines NB4, KG1a, HL60, and K562 were treated with fucoidan and cell cycle, cell proliferation, and expression of apoptotic pathways molecules were analyzed. Fucoidan suppressed the proliferation and induced apoptosis through the intrinsic and extrinsic pathways in the acute promyelocytic leukemia (APL) cell lines NB4 and HL60, but not in KG1a and K562 cells. In NB4 cells, apoptosis was caspase-dependent as it was significantly attenuated by pre-treatment with a pan-caspase inhibitor. P21/WAF1/CIP1 was significantly up-regulated leading to cell cycle arrest. Fucoidan decreased the activation of ERK1/2 and down-regulated the activation of AKT through hypo-phosphorylation of Thr(308) residue but not Ser(473). In vivo, a xenograft model using the NB4 cells was employed. Mice were fed with fucoidan and tumor growth was measured following inoculation with NB4 cells. Subsequently, splenic natural killer (NK) cell cytotoxic activity was also examined. Oral doses of fucoidan significantly delayed tumor growth in the xenograft model and increased cytolytic activity of NK cells. Taken together, these data suggest that the selective inhibitory effect of fucoidan on APL cells and its protective effect against APL development in mice warrant further investigation of fucoidan as a useful agent in treatment of certain types of leukemia.
Collapse
Affiliation(s)
- Farzaneh Atashrazm
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Ray M Lowenthal
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Gregory M Woods
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia.,School of Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Adele F Holloway
- School of Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | | | - Joanne L Dickinson
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| |
Collapse
|
21
|
Aasebø E, Forthun RB, Berven F, Selheim F, Hernandez-Valladares M. Global Cell Proteome Profiling, Phospho-signaling and Quantitative Proteomics for Identification of New Biomarkers in Acute Myeloid Leukemia Patients. Curr Pharm Biotechnol 2016; 17:52-70. [PMID: 26306748 PMCID: PMC5388801 DOI: 10.2174/1389201016666150826115626] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 05/29/2015] [Accepted: 07/23/2015] [Indexed: 12/31/2022]
Abstract
The identification of protein biomarkers for acute myeloid leukemia (AML) that could find applications in AML diagnosis and prognosis, treatment and the selection for bone marrow transplant requires substantial comparative analyses of the proteomes from AML patients. In the past years, several studies have suggested some biomarkers for AML diagnosis or AML classification using methods for sample preparation with low proteome coverage and low resolution mass spectrometers. However, most of the studies did not follow up, confirm or validate their candidates with more patient samples. Current proteomics methods, new high resolution and fast mass spectrometers allow the identification and quantification of several thousands of proteins obtained from few tens of μg of AML cell lysate. Enrichment methods for posttranslational modifications (PTM), such as phosphorylation, can isolate several thousands of site-specific phosphorylated peptides from AML patient samples, which subsequently can be quantified with high confidence in new mass spectrometers. While recent reports aiming to propose proteomic or phosphoproteomic biomarkers on the studied AML patient samples have taken advantage of the technological progress, the access to large cohorts of AML patients to sample from and the availability of appropriate control samples still remain challenging.
Collapse
Affiliation(s)
| | | | | | | | - Maria Hernandez-Valladares
- Department of Biomedicine, Faculty of Medicine, Building for Basic Biology, University of Bergen, Jonas Lies vei 91, 5009 Bergen, Norway.
| |
Collapse
|
22
|
Moreno-Martínez D, Nomdedeu M, Lara-Castillo MC, Etxabe A, Pratcorona M, Tesi N, Díaz-Beyá M, Rozman M, Montserrat E, Urbano-Ispizua A, Esteve J, Risueño RM. XIAP inhibitors induce differentiation and impair clonogenic capacity of acute myeloid leukemia stem cells. Oncotarget 2015; 5:4337-46. [PMID: 24952669 PMCID: PMC4147327 DOI: 10.18632/oncotarget.2016] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Acute myeloid leukemia (AML) is a neoplasia characterized by the rapid expansion of immature myeloid blasts in the bone marrow, and marked by poor prognosis and frequent relapse. As such, new therapeutic approaches are required for remission induction and prevention of relapse. Due to the higher chemotherapy sensitivity and limited life span of more differentiated AML blasts, differentiation-based therapies are a promising therapeutic approach. Based on public available gene expression profiles, a myeloid-specific differentiation-associated gene expression pattern was defined as the therapeutic target. A XIAP inhibitor (Dequalinium chloride, DQA) was identified in an in silico screening searching for small molecules that induce similar gene expression regulation. Treatment with DQA, similarly to Embelin (another XIAP inhibitor), induced cytotoxicity and differentiation in AML. XIAP inhibition differentially impaired cell viability of the most primitive AML blasts and reduced clonogenic capacity of AML cells, sparing healthy mature blood and hematopoietic stem cells. Taken together, these results suggest that XIAP constitutes a potential target for AML treatment and support the evaluation of XIAP inhibitors in clinical trials.
Collapse
|
23
|
Boso G, Somia NV. Characterization of resistance to rhabdovirus and retrovirus infection in a human myeloid cell line. PLoS One 2015; 10:e0121455. [PMID: 25811758 PMCID: PMC4374779 DOI: 10.1371/journal.pone.0121455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 02/13/2015] [Indexed: 11/30/2022] Open
Abstract
Viruses interact with various permissive and restrictive factors in host cells throughout their replication cycle. Cell lines that are non-permissive to viral infection have been particularly useful in discovering host cell proteins involved in viral life cycles. Here we describe the characterization of a human myeloid leukemia cell line, KG-1, that is resistant to infection by retroviruses and a Rhabdovirus. We show that KG-1 cells are resistant to infection by Vesicular Stomatits Virus as well as VSV Glycoprotein (VSVG) pseudotyped retroviruses due to a defect in binding. Moreover our results indicate that entry by xenotropic retroviral envelope glycoprotein RD114 is impaired in KG-1 cells. Finally we characterize a post- entry block in the early phase of the retroviral life cycle in KG-1 cells that renders the cell line refractory to infection. This cell line will have utility in discovering proteins involved in infection by VSV and HIV-1.
Collapse
Affiliation(s)
- Guney Boso
- Molecular, Cellular, Developmental Biology and Genetics Graduate Program, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Nikunj V. Somia
- Dept. of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
24
|
Niavarani A, Currie E, Reyal Y, Anjos-Afonso F, Horswell S, Griessinger E, Luis Sardina J, Bonnet D. APOBEC3A is implicated in a novel class of G-to-A mRNA editing in WT1 transcripts. PLoS One 2015; 10:e0120089. [PMID: 25807502 PMCID: PMC4373805 DOI: 10.1371/journal.pone.0120089] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 01/22/2015] [Indexed: 12/18/2022] Open
Abstract
Classic deamination mRNA changes, including cytidine to uridine (C-to-U) and adenosine to inosine (A-to-I), are important exceptions to the central dogma and lead to significant alterations in gene transcripts and products. Although there are a few reports of non-classic mRNA alterations, as yet there is no molecular explanation for these alternative changes. Wilms Tumor 1 (WT1) mutations and variants are implicated in several diseases, including Wilms tumor and acute myeloid leukemia (AML). We observed two alternative G-to-A changes, namely c.1303G>A and c.1586G>A in cDNA clones and found them to be recurrent in a series of 21 umbilical cord blood mononuclear cell (CBMC) samples studied. Two less conserved U-to-C changes were also observed. These alternative changes were found to be significantly higher in non-progenitor as compared to progenitor CBMCs, while they were found to be absent in a series of AML samples studied, indicating they are targeted, cell type-specific mRNA editing modifications. Since APOBEC/ADAR family members are implicated in RNA/DNA editing, we screened them by RNA-interference (RNAi) for WT1-mRNA changes and observed near complete reversal of WT1 c.1303G>A alteration upon APOBEC3A (A3A) knockdown. The role of A3A in mediating this change was confirmed by A3A overexpression in Fujioka cells, which led to a significant increase in WT1 c.1303G>A mRNA editing. Non-progenitor CBMCs showed correspondingly higher levels of A3A-mRNA and protein as compared to the progenitor ones. To our knowledge, this is the first report of mRNA modifying activity for an APOBEC3 protein and implicates A3A in a novel G-to-A form of editing. These findings open the way to further investigations into the mechanisms of other potential mRNA changes, which will help to redefine the RNA editing paradigm in both health and disease.
Collapse
MESH Headings
- Adenosine/metabolism
- Base Sequence
- Cytidine Deaminase/antagonists & inhibitors
- Cytidine Deaminase/genetics
- Cytidine Deaminase/metabolism
- Guanine/metabolism
- Humans
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/pathology
- Leukocytes, Mononuclear/cytology
- Leukocytes, Mononuclear/metabolism
- Molecular Sequence Data
- Mutation
- Proteins/antagonists & inhibitors
- Proteins/genetics
- Proteins/metabolism
- RNA Editing
- RNA Interference
- RNA, Messenger/chemistry
- RNA, Messenger/metabolism
- RNA, Small Interfering/metabolism
- Sequence Analysis, DNA
- Umbilical Cord/cytology
- WT1 Proteins/genetics
- WT1 Proteins/metabolism
- Wilms Tumor/genetics
- Wilms Tumor/pathology
Collapse
Affiliation(s)
- Ahmadreza Niavarani
- Haematopoietic Stem Cell Laboratory, Cancer Research UK, London Research Institute, London, United Kingdom
- Digestive Disease Research Institute (DDRI), Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Erin Currie
- Haematopoietic Stem Cell Laboratory, Cancer Research UK, London Research Institute, London, United Kingdom
| | - Yasmin Reyal
- Department of Haematology, University College London Hospitals NHS Trust, London, United Kingdom
| | - Fernando Anjos-Afonso
- Haematopoietic Stem Cell Laboratory, Cancer Research UK, London Research Institute, London, United Kingdom
| | - Stuart Horswell
- Department of Bioinformatics, Cancer Research UK, London Research Institute, London, United Kingdom
| | - Emmanuel Griessinger
- INSERM U1065, Mediterranean Centre for Molecular Medicine (C3M), Université Nice Sophia Antipolis, Nice, France
| | - Jose Luis Sardina
- Instituto de Biología Funcional y Genómica, CSIC/Universidad de Salamanca, Salamanca, Spain
| | - Dominique Bonnet
- Haematopoietic Stem Cell Laboratory, Cancer Research UK, London Research Institute, London, United Kingdom
- * E-mail:
| |
Collapse
|
25
|
Hematopoietic cancer cell lines can support replication of Sabin poliovirus type 1. BIOMED RESEARCH INTERNATIONAL 2015; 2015:358462. [PMID: 25815312 PMCID: PMC4359862 DOI: 10.1155/2015/358462] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Revised: 02/17/2015] [Accepted: 02/17/2015] [Indexed: 02/02/2023]
Abstract
Viral vaccines can be produced in adherent or in suspension cells. The objective of this work was to screen human suspension cell lines for the capacity to support viral replication. As the first step, it was investigated whether poliovirus can replicate in such cell lines. Sabin poliovirus type 1 was serially passaged on five human cell lines, HL60, K562, KG1, THP-1, and U937. Sabin type 1 was capable of efficiently replicating in three cell lines (K562, KG1, and U937), yielding high viral titers after replication. Expression of CD155, the poliovirus receptor, did not explain susceptibility to replication, since all cell lines expressed CD155. Furthermore, we showed that passaged virus replicated more efficiently than parental virus in KG1 cells, yielding higher virus titers in the supernatant early after infection. Infection of cell lines at an MOI of 0.01 resulted in high viral titers in the supernatant at day 4. Infection of K562 with passaged Sabin type 1 in a bioreactor system yielded high viral titers in the supernatant. Altogether, these data suggest that K562, KG1, and U937 cell lines are useful for propagation of poliovirus.
Collapse
|
26
|
Gañán-Gómez I, Wei Y, Yang H, Pierce S, Bueso-Ramos C, Calin G, Boyano-Adánez MDC, García-Manero G. Overexpression of miR-125a in myelodysplastic syndrome CD34+ cells modulates NF-κB activation and enhances erythroid differentiation arrest. PLoS One 2014; 9:e93404. [PMID: 24690917 PMCID: PMC3972113 DOI: 10.1371/journal.pone.0093404] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Accepted: 03/04/2014] [Indexed: 01/05/2023] Open
Abstract
Myelodysplastic syndromes (MDS) are characterized by impaired proliferation and differentiation of hematopoietic stem cells. The participation of toll-like receptor (TLR)-mediated signaling in MDS is well documented. Increased TLR signaling leads to the constitutive activation of NF-κB, which mediates inflammation, cell proliferation and apoptosis. In addition, the TLR pathway induces the expression of miRNAs which participate in the fine-tuning of the inflammatory response. miRNAs also regulate other biological processes, including hematopoiesis. miR-125a and miR-125b are known modulators of hematopoiesis and are abnormally expressed in several hematologic malignancies. However, little is known about their role in MDS. NF-κB-activating ability has been described for both miRNAs. We studied the role of miR-125a/miR-125b in MDS and their relationship with TLR signaling and hematopoietic differentiation. Our results indicate that miR-125a is significantly overexpressed in MDS patients and correlates negatively with patient survival. Expression of miR-99b, which is clustered with miR-125a, is also directly correlated with prognosis of MDS. Both miR-125a and miR-99b activated NF-κB in vitro; however, we observed a negative correlation between miR-99b expression and the levels of TLR2, TLR7 and two downstream genes, suggesting that NF-κB activation by the miRNA cluster occurs in the absence of TLR signaling. We also show that TLR7 is negatively correlated with patient survival in MDS. In addition, our data suggest that miR-125a may act as an NF-κB inhibitor upon TLR stimulation. These results indicate that miR-125a is involved in the fine-tuning of NF-κB activity and that its effects may depend on the status of the TLR pathway. Furthermore, we observed that miR-125a inhibits erythroid differentiation in leukemia and MDS cell lines. Therefore, this miRNA could serve as a prognostic marker and a potential therapeutic target in MDS.
Collapse
Affiliation(s)
- Irene Gañán-Gómez
- Department of Systems Biology, University of Alcalá, Alcalá de Henares, Madrid, Spain
- * E-mail: (IG-G); (GG-M)
| | - Yue Wei
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Hui Yang
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Sherry Pierce
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Carlos Bueso-Ramos
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - George Calin
- Department of Experimental Therapeutics, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | | | - Guillermo García-Manero
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- * E-mail: (IG-G); (GG-M)
| |
Collapse
|
27
|
Shah K, McCormack CE, Bradbury NA. Do you know the sex of your cells? Am J Physiol Cell Physiol 2014; 306:C3-18. [PMID: 24196532 PMCID: PMC3919971 DOI: 10.1152/ajpcell.00281.2013] [Citation(s) in RCA: 143] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Accepted: 10/31/2013] [Indexed: 11/22/2022]
Abstract
Do you know the sex of your cells? Not a question that is frequently heard around the lab bench, yet thanks to recent research is probably one that should be asked. It is self-evident that cervical epithelial cells would be derived from female tissue and prostate cells from a male subject (exemplified by HeLa and LnCaP, respectively), yet beyond these obvious examples, it would be true to say that the sex of cell lines derived from non-reproductive tissue, such as lung, intestine, kidney, for example, is given minimal if any thought. After all, what possible impact could the presence of a Y chromosome have on the biochemistry and cell biology of tissues such as the exocrine pancreatic acini? Intriguingly, recent evidence has suggested that far from being irrelevant, genes expressed on the sex chromosomes can have a marked impact on the biology of such diverse tissues as neurons and renal cells. It is also policy of AJP-Cell Physiology that the source of all cells utilized (species, sex, etc.) should be clearly indicated when submitting an article for publication, an instruction that is rarely followed (http://www.the-aps.org/mm/Publications/Info-For-Authors/Composition). In this review we discuss recent data arguing that the sex of cells being used in experiments can impact the cell's biology, and we provide a table outlining the sex of cell lines that have appeared in AJP-Cell Physiology over the past decade.
Collapse
Affiliation(s)
- Kalpit Shah
- Department of Physiology and Biophysics, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois
| | | | | |
Collapse
|
28
|
Bade-Döding C, Göttmann W, Baigger A, Farren M, Lee KP, Blasczyk R, Huyton T. Autocrine GM-CSF transcription in the leukemic progenitor cell line KG1a is mediated by the transcription factor ETS1 and is negatively regulated through SECTM1 mediated ligation of CD7. Biochim Biophys Acta Gen Subj 2013; 1840:1004-13. [PMID: 24211252 DOI: 10.1016/j.bbagen.2013.10.043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Revised: 09/17/2013] [Accepted: 10/27/2013] [Indexed: 11/17/2022]
Abstract
BACKGROUND CD7 expression is found on ~30% of acute myeloblastic leukemias (AML). The leukemic progenitor cell line KG1a (CD7+) constitutively expresses GM-CSF while the parental KG1 (CD7-) cell line does not. This study focuses on the molecular basis of CD7 mediated GM-CSF regulation. METHODS KG1a cells were treated with recombinant SECTM1-Fc protein, the PI3K kinase inhibitors wortmannin, LY292004, or PI4K activator spermine. Stable KG1-CD7+, KG1a-shCD7, KG1a-shETS1 as well as KG1a-GFP, KG1a-PKCβII-GFP cell lines were generated and the levels of CD7, GM-CSF and ETS-1 mRNA and protein were compared by real-time-PCR, western blotting, flow cytometry and ELISA. RESULTS SECTM1 is expressed in Human Bone Marrow Endothelial Cells (HBMEC) and its expression can be upregulated by both IFN-γ. KG1a cells demonstrated high expression levels of CD7 and ETS-1 allowing a constitutative signaling through the PI3K/Atk pathway to promote GM-CSF expression, while KG1 cells with low expression of CD7 and ETS-1 showed low GM-CSF expression. On KG1a cells GM-CSF expression could be negatively regulated by PI3K inhibitors or by recombinant SECTM1-Fc. Overexpression of CD7 in KG1 cells was insufficient to promote GM-CSF expression, while silencing of CD7 or ETS-1 resulted in reduced GM-CSF expression levels. Differentiation capable KG1a cells overexpressing PKCβII illustrated complete loss of CD7, but maintained normal levels of both ETS-1 and GM-CSF expression. CONCLUSION These findings add an additional layer to the previously described autocrine/paracrine signaling between leukemic progenitor cells and the bone marrow microenvironment and highlight a role for SECTM1 in both normal and malignant hematopoiesis. GENERAL SIGNIFICANCE This work shows that SECTM1 secreted from bone marrow stromal cells may interact with CD7 to influence GM-CSF expression in leukemic cells.
Collapse
Affiliation(s)
- Christina Bade-Döding
- Institute for Transfusion Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany
| | - Wiebke Göttmann
- Institute for Transfusion Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany
| | - Anja Baigger
- Institute for Transfusion Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany
| | - Matthew Farren
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY 14226, USA
| | - Kelvin P Lee
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY 14226, USA
| | - Rainer Blasczyk
- Institute for Transfusion Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany
| | - Trevor Huyton
- Institute for Transfusion Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany.
| |
Collapse
|
29
|
Morris VA, Zhang A, Yang T, Stirewalt DL, Ramamurthy R, Meshinchi S, Oehler VG. MicroRNA-150 expression induces myeloid differentiation of human acute leukemia cells and normal hematopoietic progenitors. PLoS One 2013; 8:e75815. [PMID: 24086639 PMCID: PMC3782459 DOI: 10.1371/journal.pone.0075815] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Accepted: 08/21/2013] [Indexed: 12/19/2022] Open
Abstract
In acute myeloid leukemia (AML) and blast crisis (BC) chronic myeloid leukemia (CML) normal differentiation is impaired. Differentiation of immature stem/progenitor cells is critical for normal blood cell function. MicroRNAs (miRNAs or miRs) are small non-coding RNAs that interfere with gene expression by degrading messenger RNAs (mRNAs) or blocking protein translation. Aberrant miRNA expression is a feature of leukemia and miRNAs also play a significant role in normal hematopoiesis and differentiation. We have identified miRNAs differentially expressed in AML and BC CML and identified a new role for miR-150 in myeloid differentiation. Expression of miR-150 is low or absent in BC CML and AML patient samples and cell lines. We have found that expression of miR-150 in AML cell lines, CD34+ progenitor cells from healthy individuals, and primary BC CML and AML patient samples at levels similar to miR-150 expression in normal bone marrow promotes myeloid differentiation of these cells. MYB is a direct target of miR-150, and we have identified that the observed phenotype is partially mediated by MYB. In AML cell lines, differentiation of miR-150 expressing cells occurs independently of retinoic acid receptor α (RARA) signaling. High-throughput gene expression profiling (GEP) studies of the AML cell lines HL60, PL21, and THP-1 suggest that activation of CEPBA, CEBPE, and cytokines associated with myeloid differentiation in miR-150 expressing cells as compared to control cells contributes to myeloid differentiation. These data suggest that miR-150 promotes myeloid differentiation, a previously uncharacterized role for this miRNA, and that absent or low miR-150 expression contributes to blocked myeloid differentiation in acute leukemia cells.
Collapse
Affiliation(s)
- Valerie A. Morris
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Division of Hematology, Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Ailin Zhang
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Taimei Yang
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Derek L. Stirewalt
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Ranjani Ramamurthy
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Soheil Meshinchi
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Vivian G. Oehler
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Division of Hematology, Department of Medicine, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
30
|
Fuster O, Llop M, Dolz S, García P, Such E, Ibáñez M, Luna I, Gómez I, López M, Cervera J, Montesinos P, Moscardó F, Cordón L, Solves P, de Juan I, Palanca S, Bolufer P, Sanz MÁ, Barragán E. Adverse prognostic value of MYBL2 overexpression and association with microRNA-30 family in acute myeloid leukemia patients. Leuk Res 2013; 37:1690-6. [PMID: 24199710 DOI: 10.1016/j.leukres.2013.09.015] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Revised: 09/02/2013] [Accepted: 09/13/2013] [Indexed: 11/12/2022]
Abstract
The MYBL2 gene encodes a transcription factor implicated in cell proliferation and maturation whose amplification or overexpression has been associated with different human malignancies, suggesting that it could be implicated in tumorigenesis. We analyzed MYBL2 expression and its prognostic value in 291 patients with de novo acute myeloid leukemia (AML) and we also evaluated its association with microRNAs 29 and 30 families. MYBL2 expression in AML patients was increased relative to CD34+ cells. Moreover, MYBL2 overexpression was associated with lower expression of miR-30a (P=0.024), miR-30b (P=0.021) and miR-30c (P=0.009). Multivariate analysis showed that MYBL2 expression was an independent factor for disease-free survival (HR 3.0, 95% CI 1.5-6.0, P=0.002) and cumulative incidence of relapse (HR 2.6, 95% CI 1.2-5.6, P=0.015) in patients with an intermediate-risk karyotype. In conclusion, our data showed that MYBL2 expression analysis could be useful to define subgroups of patients with poor prognosis.
Collapse
Affiliation(s)
- Oscar Fuster
- Department of Medical Pathology, Hospital Universitario La Fe, Valencia, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Dimicoli S, Wei Y, Bueso-Ramos C, Yang H, DiNardo C, Jia Y, Zheng H, Fang Z, Nguyen M, Pierce S, Chen R, Wang H, Wu C, Garcia-Manero G. Overexpression of the toll-like receptor (TLR) signaling adaptor MYD88, but lack of genetic mutation, in myelodysplastic syndromes. PLoS One 2013; 8:e71120. [PMID: 23976989 PMCID: PMC3744562 DOI: 10.1371/journal.pone.0071120] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Accepted: 06/24/2013] [Indexed: 01/01/2023] Open
Abstract
MYD88 is a key mediator of Toll-like receptor innate immunity signaling. Oncogenically active MYD88 mutations have recently been reported in lymphoid malignancies, but has not been described in MDS. To characterize MYD88 in MDS, we sequenced the coding region of the MYD88 gene in 40 MDS patients. No MYD88 mutation was detected. We next characterized MYD88 expression in bone marrow CD34+ cells (N = 64). Increased MYD88 RNA was detected in 40% of patients. Patients with higher MYD88 expression in CD34+ cells had a tendency for shorter survival compared to the ones with lower MYD88, which was significant when controlled for IPSS and age. We then evaluated effect of MYD88 blockade in the CD34+ cells of patients with lower-risk MDS. Colony formation assays indicated that MYD88 blockade using a MYD88 inhibitor resulted in increased erythroid colony formation. MYD88 blockade also negatively regulated the secretion of interleukin-8. Treatment of MDS CD34+ cells with an IL-8 antibody also increased formation of erythroid colonies. These results indicate that MYD88 plays a role in the pathobiology of MDS and may have prognostic and therapeutic value in the management of patients with this disease.
Collapse
Affiliation(s)
- Sophie Dimicoli
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Yue Wei
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- * E-mail: (GGM); (YW)
| | - Carlos Bueso-Ramos
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Hui Yang
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Courtney DiNardo
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Yu Jia
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Hong Zheng
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Zhihong Fang
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Martin Nguyen
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Sherry Pierce
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Rui Chen
- Department of Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Hui Wang
- Department of Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Chenghua Wu
- Department of Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Guillermo Garcia-Manero
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- * E-mail: (GGM); (YW)
| |
Collapse
|
32
|
Rommer A, Steinleitner K, Hackl H, Schneckenleithner C, Engelmann M, Scheideler M, Vlatkovic I, Kralovics R, Cerny-Reiterer S, Valent P, Sill H, Wieser R. Overexpression of primary microRNA 221/222 in acute myeloid leukemia. BMC Cancer 2013; 13:364. [PMID: 23895238 PMCID: PMC3733744 DOI: 10.1186/1471-2407-13-364] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Accepted: 07/10/2013] [Indexed: 12/21/2022] Open
Abstract
Background Acute myeloid leukemia (AML) is a hematopoietic malignancy with a dismal outcome in the majority of cases. A detailed understanding of the genetic alterations and gene expression changes that contribute to its pathogenesis is important to improve prognostication, disease monitoring, and therapy. In this context, leukemia-associated misexpression of microRNAs (miRNAs) has been studied, but no coherent picture has emerged yet, thus warranting further investigations. Methods The expression of 636 human miRNAs was compared between samples from 52 patients with AML and 13 healthy individuals by highly specific locked nucleic acid (LNA) based microarray technology. The levels of individual mature miRNAs and of primary miRNAs (pri-miRs) were determined by quantitative reverse transcriptase (qRT) PCR. Transfections and infections of human cell lines were performed using standard procedures. Results 64 miRNAs were significantly differentially expressed between AML and controls. Further studies on the clustered miRNAs 221 and 222, already known to act as oncogenes in other tumor types, revealed a deficiency of human myeloid cell lines to process vector derived precursor transcripts. Moreover, endogenous pri-miR-221/222 was overexpressed to a substantially higher extent than its mature products in most primary AML samples, indicating that its transcription was enhanced, but processing was rate limiting, in these cells. Comparison of samples from the times of diagnosis, remission, and relapse of AML demonstrated that pri-miR-221/222 levels faithfully reflected the stage of disease. Conclusions Expression of some miRNAs is strongly regulated at the posttranscriptional level in AML. Pri-miR-221/222 represents a novel molecular marker and putative oncogene in this disease.
Collapse
Affiliation(s)
- Anna Rommer
- Department of Medicine I, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Myeloblastic cell lines mimic some but not all aspects of human cytomegalovirus experimental latency defined in primary CD34+ cell populations. J Virol 2013; 87:9802-12. [PMID: 23824798 DOI: 10.1128/jvi.01436-13] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Human cytomegalovirus (HCMV) is a significant human pathogen that achieves lifelong persistence by establishing latent infections in undifferentiated cells of the myeloid lineage, such as CD34(+) hematopoietic progenitor cells. When latency is established, viral lytic gene expression is silenced in part by a cellular intrinsic defense consisting of Daxx and histone deacetylases (HDACs) because pp71, the tegument transactivator that travels to the nucleus and inactivates this defense at the start of a lytic infection in differentiated cells, remains in the cytoplasm. Because the current in vitro and ex vivo latency models have physiological and practical limitations, we evaluated two CD34(+) myeloblastic cell lines, KG-1 and Kasumi-3, for their ability to establish, maintain, and reactivate HCMV experimental latent infections. Tegument protein pp71 was cytoplasmic, and immediate-early (IE) genes were silenced as in primary CD34(+) cells. However, in contrast to what occurs in primary CD34(+) cells ex vivo or in NT2 and THP-1 in vitro model systems, viral IE gene expression from the laboratory-adapted AD169 genome was not induced in the presence of HDAC inhibitors in either KG-1 or Kasumi-3 cells. Furthermore, while the clinical strain FIX was able to reactivate from Kasumi-3 cells, AD169 was not, and neither strain reactivated from KG-1 cells. Thus, KG-1 and Kasumi-3 experimental latent infections differ in important parameters from those in primary CD34(+) cell populations. Aspects of latency illuminated through the use of these myeloblastoid cell lines should not be considered independently but integrated with results obtained in primary cell systems when paradigms for HCMV latency are proposed.
Collapse
|
34
|
Oraki Kohshour M, Najafi L, Heidari M, Ghaffari Sharaf M. Antiproliferative effect of H2O2 against human acute myelogenous leukemia KG1 cell line. J Acupunct Meridian Stud 2013; 6:134-41. [PMID: 23787282 DOI: 10.1016/j.jams.2012.08.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2011] [Revised: 04/26/2012] [Accepted: 05/03/2012] [Indexed: 01/24/2023] Open
Abstract
It has clearly been established that oxidative stress leads to perturbation of various cellular processes resulting in either inhibition of cell proliferation or cell death. In addition, there is a growing body of evidence indicating that reactive oxygen species (ROS) are required as signal molecules that regulate different physiological processes including survival or death. Free radicals, particularly ROS, have been proposed as general mediators for apoptosis and recent studies have established that the mode of cell death depends on the severity of the oxidative damage. In this study, we determined the effect of oxidative stress on cell proliferation and characterization of cell death in human KG1 cells treated with H2O2. Our results indicated that oxidative stress leads to a significant decrease in cell proliferation and induction of apoptosis. Moreover, our study suggests that antiproliferative and apoptotic cell death effects of H2O2 took place via activation of caspase-3, affecting the expression of Bcl-2 and Bax (an antiapoptotic and a proapoptotic factor, respectively), and through deactivation of catalase enzyme, leading to accumulation of intracellular ROS and depletion of intracellular ATP level.
Collapse
Affiliation(s)
- Mojtaba Oraki Kohshour
- Department of Immunology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | | | | | | |
Collapse
|
35
|
Ulsemer P, Henderson G, Toutounian K, Löffler A, Schmidt J, Karsten U, Blaut M, Goletz S. Specific humoral immune response to the Thomsen-Friedenreich tumor antigen (CD176) in mice after vaccination with the commensal bacterium Bacteroides ovatus D-6. Cancer Immunol Immunother 2013; 62:875-87. [PMID: 23381581 PMCID: PMC11029541 DOI: 10.1007/s00262-013-1394-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Accepted: 01/12/2013] [Indexed: 01/09/2023]
Abstract
The tumor-specific Thomsen-Friedenreich antigen (TFα, CD176) is an attractive target for a cancer vaccine, especially as TF-directed antibodies play an important role in cancer immunosurveillance. However, synthetic TF vaccines have not overcome the low intrinsic immunogenicity of TF. Since natural TF-directed antibodies present in human sera are generated in response to microbes found in the gastrointestinal tract, microbial TF structures are obviously more immunogenic than synthetic TF. We recently isolated a new strain (D-6) of the human gut bacterium Bacteroides ovatus, which carries the true TFα antigen. Here, we present experimental data on the immunogenicity of this strain. Mice immunized with B. ovatus D-6 in the absence of adjuvants developed specific anti-TFα IgM and IgG antibodies which also bound to human cancer cells carrying TFα. Our data suggest that B. ovatus D-6 presents a unique TFα-specific immunogenicity based on a combination of several inherent properties including: expression of the true TFα antigen, clustering and accessible presentation of TFα as repetitive side chains on a capsular polysaccharide, and intrinsic adjuvant properties. Therefore, B. ovatus strain D-6 is an almost perfect candidate for the development of the first adjuvant-free TFα-specific anti-tumor vaccine.
Collapse
Affiliation(s)
- Philippe Ulsemer
- Glycotope GmbH, Robert-Rössle-Str. 10, 13125, Berlin-Buch, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Rosen DB, Harrington KH, Cordeiro JA, Leung LY, Putta S, Lacayo N, Laszlo GS, Gudgeon CJ, Hogge DE, Hawtin RE, Cesano A, Walter RB. AKT signaling as a novel factor associated with in vitro resistance of human AML to gemtuzumab ozogamicin. PLoS One 2013; 8:e53518. [PMID: 23320091 PMCID: PMC3539972 DOI: 10.1371/journal.pone.0053518] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Accepted: 11/29/2012] [Indexed: 01/08/2023] Open
Abstract
Gemtuzumab ozogamicin (GO), an immunoconjugate between an anti-CD33 antibody and a calicheamicin-γ1 derivative, induces remissions and improves survival in a subset of patients with acute myeloid leukemia (AML). As the mechanisms underlying GO and calicheamicin-γ1 resistance are incompletely understood, we herein used flow cytometry-based single cell network profiling (SCNP) assays to study cellular responses of primary human AML cells to GO. Our data indicate that the extent of DNA damage is quantitatively impacted by CD33 expression and drug efflux activity. However, although DNA damage is required for GO-induced cytotoxicity, it is not sufficient for effective cell kill, suggesting that downstream anti-apoptotic pathways may function as relevant resistance mechanisms. Supporting this notion, we found activated PI3K/AKT signaling to be associated with GO resistance in vitro in primary AML cells. Consistently, the investigational AKT inhibitor MK-2206 significantly sensitized various human AML cells to GO or free calicheamicin-γ1 with particularly pronounced effects in otherwise GO or free calicheamicin-γ1 -resistant cells. Likewise, MK-2206 also sensitized primary AML cells to calicheamicin-γ1. Together, our findings illustrate the capacity of SCNP assays to discover chemotherapy-related biological pathways and signaling networks relevant to GO-induced genotoxic stress. The identification of AKT signaling as being associated with GO resistance in vitro may provide a novel approach to improve the in vivo efficacy of GO/calicheamicin-γ1 and, by extrapolation, other DNA damage-based therapeutics.
Collapse
MESH Headings
- Aminoglycosides/pharmacology
- Aminoglycosides/therapeutic use
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antineoplastic Agents/pharmacology
- Cell Line, Tumor
- DNA Damage
- Drug Resistance, Neoplasm
- Enediynes/pharmacology
- Gemtuzumab
- Heterocyclic Compounds, 3-Ring/pharmacology
- Humans
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/therapy
- Phosphatidylinositol 3-Kinases/metabolism
- Protein Kinase Inhibitors/pharmacology
- Proto-Oncogene Proteins c-akt/antagonists & inhibitors
- Proto-Oncogene Proteins c-akt/physiology
- Sialic Acid Binding Ig-like Lectin 3/metabolism
- Signal Transduction/drug effects
- Signal Transduction/physiology
- Single-Cell Analysis
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- David B. Rosen
- Nodality Inc., South San Francisco, California, United States of America
| | - Kimberly H. Harrington
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - James A. Cordeiro
- Nodality Inc., South San Francisco, California, United States of America
| | - Ling Y. Leung
- Nodality Inc., South San Francisco, California, United States of America
| | - Santosh Putta
- Nodality Inc., South San Francisco, California, United States of America
| | - Norman Lacayo
- Division of Pediatric Hematology/Oncology, Stanford University School of Medicine, Palo Alto, California, United States of America
| | - George S. Laszlo
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Chelsea J. Gudgeon
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Donna E. Hogge
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Rachael E. Hawtin
- Nodality Inc., South San Francisco, California, United States of America
| | - Alessandra Cesano
- Nodality Inc., South San Francisco, California, United States of America
- * E-mail:
| | - Roland B. Walter
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Division of Hematology/Department of Medicine, University of Washington, Seattle, Washington, United States of America
- Department of Epidemiology, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
37
|
Gadeock S, Pupovac A, Sluyter V, Spildrejorde M, Sluyter R. P2X7 receptor activation mediates organic cation uptake into human myeloid leukaemic KG-1 cells. Purinergic Signal 2012; 8:669-76. [PMID: 22661222 DOI: 10.1007/s11302-012-9320-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Accepted: 05/22/2012] [Indexed: 01/04/2023] Open
Abstract
The P2X7 purinergic receptor is an ATP-gated cation channel with an emerging role in neoplasia. In this study we demonstrate that the human KG-1 cell line, a model of acute myelogenous leukaemia, expresses functional P2X7. RT-PCR and immunochemical techniques demonstrated the presence of P2X7 mRNA and protein respectively in KG-l cells, as well as in positive control multiple myeloma RPMI 8226 cells. Flow cytometric measurements demonstrated that ATP induced ethidium(+) uptake into KG-l cells suspended in sucrose medium (EC(50) of ≈ 3 μM), but not into cells in NaCl medium. In contrast, ATP induced ethidium(+) uptake into RPMI 8226 cells suspended in either sucrose or NaCl medium (EC(50) of ≈ 3 or ≈ 99 μM, respectively), as well as into RPMI 8226 cells in KCl medium (EC(50) of ≈ 18 μM). BzATP and to a lesser extent ATPγS and αβ-methylene ATP, but not ADP or UTP, also induced ethidium(+) uptake into KG-1 cells. ATP-induced ethidium(+) uptake was completely impaired by the P2X7 antagonists, AZ10606120 and A-438079. ATP-induced ethidium(+) uptake was also impaired by probenecid but not by carbenoxolone, both pannexin-1 antagonists. ATP induced YO-PRO-1(2+) and propidium(2+) uptake into KG-1 cells. Finally, sequencing of full-length P2X7 cDNA identified several single nucleotide polymorphisms (SNPs) in KG-1 cells including H155Y, A348T, T357S and Q460R. RPMI 8226 cells contained A348T, A433V and H521Q SNPs. In conclusion, the KG-1 cell line expresses functional P2X7. This cell line may help elucidate the signalling pathways involved in P2X7-induced survival and invasiveness of myeloid leukaemic cells.
Collapse
Affiliation(s)
- Safina Gadeock
- School of Biological Sciences, University of Wollongong, Wollongong, NSW, 2522, Australia
| | | | | | | | | |
Collapse
|
38
|
Pelliccia F, Ubertini V, Bosco N. The importance of molecular cytogenetic analysis prior to using cell lines in research: The case of the KG-1a leukemia cell line. Oncol Lett 2012; 4:237-240. [PMID: 22844360 DOI: 10.3892/ol.2012.709] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Accepted: 04/18/2012] [Indexed: 11/05/2022] Open
Abstract
KG-1 and its less differentiated subline KG-1a are leukemia cell lines used in research in a number of laboratories. The karyotypes of the two lines were initially identical. In the following years, further analysis revealed that the cell lines had acquired additional karyotypical abnormalities and differed in the presence of certain typical chromosomal rearrangements. To obtain cytogenetic authentication prior to the use of the two cell lines, we analyzed their karyotype by combining DAPI- and CMA-chromosome bandings and a fluorescence in situ hybridization (FISH)-based approach by using BAC clones useful for the identification of chromosome regions of interest. Sequences of the MYC, PLZF, RARA and BCR genes, that are known to play a critical role in leukemogenesis, and certain BAC clones mapped to five known common fragile sites (CFS) were used for the FISH analysis. A telomeric probe (TTAGGG)n and a set of BAC clones were used to characterize the marker chromosome der(1) that was observed in the cell line KG-1a. The existence of notable differences between the karyotype of the KG-1a cell line previously described, and that described in this study, demonstrate that the use of established cancer cell lines should be preceded by cytogenetic and/or molecular characterization.
Collapse
Affiliation(s)
- Franca Pelliccia
- Department of Biology and Biotechnology, Sapienza University, I-00185 Rome, Italy
| | | | | |
Collapse
|
39
|
Human lysosomal α-D-mannosidase regulation in promyelocytic leukaemia cells. Biosci Rep 2012; 31:477-87. [PMID: 21521175 DOI: 10.1042/bsr20110020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Lysosomal α-D-mannosidase is an exoglycosidase involved in the ordered degradation of N-linked oligosaccharides. It is ubiquitously expressed, although the main transcript is more abundant in peripheral blood leucocytes. Here we report that α-D-mannosidase enzyme activity is very high in the promyelocytic leukaemia cell lines HL60 and NB4, as compared with other leukaemic cell lines or cells from different human sources. The MAN2B1 transcript level correlates with enzyme activity, indicating a transcriptional up-regulation of the α-D-mannosidase gene. The promoter was then characterized in HEK-293 cells (human embryonic kidney 293 cells) and HL60 cells; regulatory sequences crucial for its activity were determined by reporter gene assay in HEK-293 cells and located in the region -101/-71 with respect to the first ATG codon. Supershift assay demonstrated that Sp1 (specificity protein 1) bound to this sequence both in HEK-293 and HL60 cells. However, 5'-RACE (5'-rapid amplification of cDNA ends) indicated the use of multiple upstream TSSs (transcription start sites) in HL60 with respect to HEK-293 cells and gel shift analysis of the sequence -373/-269 demonstrated a specific binding by NF-κB (nuclear factor κB) transcription factor in HL60 but not in HEK-293 cells. We concluded that despite the α-D-mannosidase promoter showing typical features of housekeeping gene promoters, α-D-mannosidase transcription is specifically regulated in HL60 by NF-κB transcription factor.
Collapse
|
40
|
Grinstein E, Mahotka C, Borkhardt A. Rb and nucleolin antagonize in controlling human CD34 gene expression. Cell Signal 2011; 23:1358-65. [PMID: 21440621 DOI: 10.1016/j.cellsig.2011.03.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Accepted: 03/17/2011] [Indexed: 01/12/2023]
Abstract
Retinoblastoma protein (Rb) controls cell proliferation, differentiation, survival and gene expression and it has a central role in the signaling network that provides a cell cycle checkpoint in the G1 phase of the cell cycle. Studies in mice have shown that Rb regulates interactions between hematopoietic stem cells and their bone marrow microenvironment and it acts as a critical regulator of hematopoietic stem and progenitor cells under stress. In human hematopoiesis, the CD34 protein is expressed on a subset of progenitor cells capable of self-renewal, multilineage differentiation, and hematopoietic reconstitution, and CD34 has a role in the differentiation of hematopoietic cells. Here we find that, in CD34-positive hematopoietic cells, Rb controls the human CD34 promoter region by antagonizing the CD34 promoter factor nucleolin to provide a mechanism that links expression of endogenous CD34 to cell cycle progression. Our study suggests a direct involvement of Rb in the transcriptional program of human CD34-positive hematopoietic stem/progenitor cells, thus providing further insights into the molecular network relevant to the features of these cells.
Collapse
Affiliation(s)
- Edgar Grinstein
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Center for Child and Adolescent Health, Heinrich Heine University, Düsseldorf, Germany
| | | | | |
Collapse
|
41
|
Jia S, Kaldunski M, Jailwala P, Geoffrey R, Kramer J, Wang X, Hessner MJ. Use of transcriptional signatures induced in lymphoid and myeloid cell lines as an inflammatory biomarker in Type 1 diabetes. Physiol Genomics 2011; 43:697-709. [PMID: 21406607 DOI: 10.1152/physiolgenomics.00235.2010] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Inflammation is common to many disorders and responsible for tissue and organ damage. In many disorders, the associated peripheral cytokine milieu is dilute and difficult to measure, necessitating development of more sensitive and informative biomarkers for mechanistic studies, earlier diagnosis, and monitoring therapeutic interventions. Previously, we have shown that plasma of recent-onset (RO) Type 1 diabetes patients induces a disease-specific proinflammatory transcriptional profile in fresh peripheral blood mononuclear cells (PBMC) compared with that of healthy controls (HC). To eliminate assay variance introduced through the use of multiple donors or multiple draws of the same person over time, we evaluated human leukemia cell lines as potential surrogates for fresh PBMC. We 1) tested seven different cell lines in their power to differentiate RO from HC plasma and 2) compared the similarity of the signatures generated across the seven cell lines to that obtained with fresh PBMC. While each cell line tested exhibited a distinct transcriptional response when cultured with RO or HC plasma, the expression profile induced in any single cell line shared little identity with that of the other cell lines or fresh PBMC. In terms of regulated biological pathways, the transcriptional response of each cell line shared varying degrees of functional identity with fresh PBMC. These results indicate that use of human leukemia cell lines as surrogates for fresh PBMC has potential in detecting perturbations to the peripheral cytokine milieu. However, the response of each is distinct, possessing varying degrees of functional relatedness to that observed with PBMC.
Collapse
Affiliation(s)
- Shuang Jia
- Max McGee National Research Center for Juvenile Diabetes, Department of Pediatrics at the Medical College of Wisconsin, and The Children's Research Institute of Children's Hospital of Wisconsin, Milwaukee, Wisconsin 53226, USA
| | | | | | | | | | | | | |
Collapse
|
42
|
AKAPs in lipid rafts are required for optimal antigen presentation by dendritic cells. Immunol Cell Biol 2011; 89:650-8. [PMID: 21221125 DOI: 10.1038/icb.2010.148] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Dendritic cell (DC) maturation and antigen presentation are regulated by activation of protein kinase A (PKA) signaling pathways, through unknown mechanisms. We have recently shown that interfering with PKA signaling through the use of anchoring inhibitor peptides hinders antigen presentation and DC maturation. These experiments provide evidence that DC maturation and antigen presentation are regulated by A-kinase anchoring proteins (AKAPs). Herein, we determine that the presence of AKAPs and PKA in lipid rafts regulates antigen presentation. Using a combination of western blotting and immuno-cytochemistry, we illustrate the presence of AKAP149, AKAP79, Ezrin and the regulatory subunits of PKA in DC lipid rafts. Incubation of DCs with the type II anchoring inhibitor, AKAP-in silico (AKAP-IS), removes Ezrin and RII from the lipid raft without disrupting raft formation. Addition of a lipid raft disruptor, methyl-β-cyclodextrin, blocks the efficacy of AKAP-IS, suggesting that the lipid raft must be intact for AKAP-IS to inhibit antigen presentation. Ezrin and AKAP79 are present in the lipid raft of stimulated KG1 cells, but Ezrin is not present in the lipid raft of unstimulated KG1 cells and AKAP79 levels are greatly diminished, suggesting that Ezrin and AKAP79 may be the key AKAPs responsible for regulating antigen presentation.
Collapse
|
43
|
Magin AS, Körfer NR, Partenheimer H, Lange C, Zander A, Noll T. Primary cells as feeder cells for coculture expansion of human hematopoietic stem cells from umbilical cord blood--a comparative study. Stem Cells Dev 2010; 18:173-86. [PMID: 18471070 DOI: 10.1089/scd.2007.0273] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Although umbilical cord blood (UCB) has been widely accepted as an alternative source of hematopoietic stem cells (HSC) for transplantation, its use in adults is restricted because of low absolute HSC numbers. To overcome this obstacle, expansion of HSC in coculture with feeder cells is a promising possibility. In this study, we compared the potential of three human primary cell types, namely, mesenchymal stem cells (MSC), human umbilical cord vein endothelial cells (HUVEC), and Wharton's jelly cells (WJC), for use as feeder cells in a potentially clinically applicable coculture system. In first experiments, we evaluated procedures needed to obtain feeder cells, the possibility to separate them from cells derived from CD34(+) cells after coculture, their ability to activate allogeneic T cells, and their survival in CD34(+)-adapted medium. Finally, we compared their support for UCB-derived CD34(+) expansion. MSC and WJC were superior to HUVEC in terms of ease and reliability of isolation procedures needed. None of the potential feeder cells expressed CD34 or CD45, thus providing markers for cell sorting after coculture. Other markers (CD31, CD90, CD105, CD166) were expressed differently on feeder cell types. While MSC in higher concentrations did not activate allogeneic T cells, those were stimulated by lower concentrations of MSC as shown by CD25, CD69, and CD71 expression. In contrast, HUVEC and WJC were proven to activate T cells at all ratios tested. Feeder cells survived a 7-day culture in CD34(+)-adapted medium. In cocultures of UCB CD34(+)cells with primary feeder cells, mononuclear cell expansion was 30- to 60-fold, colony-forming cell expansion 20- to 40-fold, and cobblestone area-forming cell expansion 10- to 50-fold. We conclude that after a careful further evaluation especially of their immunological properties, all three primary cell types might possibly be suitable for use in a potentially clinically applicable system for expansion from UCB CD34(+)cells, with WJC being best choice and MSC still superior to HUVEC.
Collapse
Affiliation(s)
- A S Magin
- Institute of Biotechnology 2, Research Center Juelich GmbH, Juelich, Germany.
| | | | | | | | | | | |
Collapse
|
44
|
Panischeva LA, Kakpakova ES, Rybalkina EY, Stavrovskaya AA. The influence of proteasome inhibitor bortezamib on ABC transporters’ expression and activity in tumor cells. BIOCHEMISTRY MOSCOW SUPPLEMENT SERIES A-MEMBRANE AND CELL BIOLOGY 2010. [DOI: 10.1134/s1990747810020145] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
45
|
Farren MR, Carlson LM, Lee KP. Tumor-mediated inhibition of dendritic cell differentiation is mediated by down regulation of protein kinase C beta II expression. Immunol Res 2010; 46:165-76. [PMID: 19756409 DOI: 10.1007/s12026-009-8118-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Tumor-mediated immune suppression occurs through multiple mechanisms, including dysregulation of dendritic cell differentiation. This block in differentiation results in fewer dendritic cells and an accumulation of immunosuppressive myeloid- derived suppressor cells and is thought to contribute to tumor outgrowth and to act as an impediment to successful anti-cancer immunotherapy. Tumor-mediated myeloid dysregulation is known to be Stat3 dependent; however, the molecular mechanism of this Stat3 signaling remains poorly defined. We have previously shown that PKC betaII is required for dendritic cell differentiation. Here, we describe our finding that tumors mediate both Stat3 activation and PKC betaII down regulation in DC progenitor cells, a process mimicked by the expression of a constitutive active Stat3 mutant. This demonstrates that tumor-mediated myeloid dysregulation may be mediated by Stat3- induced PKC betaII down regulation.
Collapse
Affiliation(s)
- Matthew R Farren
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | | | | |
Collapse
|
46
|
Davies NJ, Hayden RE, Simpson PJ, Birtwistle J, Mayer K, Ride JP, Bunce CM. AKR1C Isoforms Represent a Novel Cellular Target for Jasmonates alongside Their Mitochondrial-Mediated Effects. Cancer Res 2009; 69:4769-75. [DOI: 10.1158/0008-5472.can-08-4533] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
47
|
|
48
|
Xie Y, De Winter JP, Waisfisz Q, Nieuwint AWM, Scheper RJ, Arwert F, Hoatlin ME, Ossenkoppele GJ, Schuurhuis GJ, Joenje H. Aberrant Fanconi anaemia protein profiles in acute myeloid leukaemia cells. Br J Haematol 2008. [DOI: 10.1111/j.1365-2141.2000.02450.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
49
|
Santegoets SJAM, van den Eertwegh AJM, van de Loosdrecht AA, Scheper RJ, de Gruijl TD. Human dendritic cell line models for DC differentiation and clinical DC vaccination studies. J Leukoc Biol 2008; 84:1364-73. [PMID: 18664532 DOI: 10.1189/jlb.0208092] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Dendritic cells (DC) are increasingly applied in the immunotherapy of cancer. As the development of a standardized DC vaccine product is often hampered by the limited availability of DC precursors and inter- and intra-donor variability, and the preparation of individual vaccines is labor-intensive, it would be preferable to use DC from a readily available and unlimited source, such as cell lines can provide. It has been described that leukemia-derived cell lines are able to differentiate into functional DC, creating possibilities for the development of highly reproducible DC vaccines and providing in vitro model systems for in-depth studies about DC physiology. This review discusses the different human DC cell line differentiation models described so far. Based on the available data, characteristics that determine the ability of leukemia cells to differentiate along the different precursor stages into functional DC will be formulated. In addition, evidence will be provided that the human CD34+ acute myeloid leukemia cell line MUTZ-3 provides DC that exhibit the functional properties that are crucial for the in vivo generation of CTL-mediated immunity and thus, currently, represents the most valuable, sustainable model system for myeloid DC differentiation and clinical DC vaccination studies.
Collapse
Affiliation(s)
- Saskia J A M Santegoets
- Department of Pathology, VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
50
|
Nimer SD. Is it important to decipher the heterogeneity of "normal karyotype AML"? Best Pract Res Clin Haematol 2008; 21:43-52. [PMID: 18342811 DOI: 10.1016/j.beha.2007.11.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Almost half of adult acute myelogenous leukemia (AML) is normal cytogenetically, and this subgroup shows a remarkable heterogeneity of genetic mutations at the molecular level and an intermediate response to therapy. The finding of recurrent cytogenetic abnormalities has influenced, in a primary way, the understanding and treatment of leukemias. Yet "normal karyotype AML" lacks such obvious abnormalities, but has a variety of prognostically important genetic abnormalities. Thus, the presence of a FLT3-ITD (internal tandem duplication), MLL-PTD (partial tandem duplication), or the increased expression of ERG or EVI1 mRNAs confer a poor prognosis, and an increased risk of relapse. In contrast, the presence of cytoplasmic nucleophosmin or C/EBPA mutations is associated with lower relapse rates and improved survival. Although resistance to treatment is associated with specific mutations, the degree to which the leukemia resembles a stem cell in its functional properties may provide greater protection from the effects of treatment. Although usually all of the circulating leukemia cells are cleared following treatment, a small residual population of leukemic cells in the bone marrow persists, making this disease hard to eradicate. Increased understanding of the biological consequences of at least some of these mutations in "normal karyotype AML" is leading to more targeted approaches to develop more effective treatments for this disease.
Collapse
Affiliation(s)
- Stephen D Nimer
- Division of Hematologic Oncology, Memorial Sloan Kettering Cancer Center, NY 1275 York Avenue, New York, NY 10021, USA.
| |
Collapse
|