12
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Bone marrow stromal cells reduce low-dose cytarabine-induced differentiation of acute myeloid leukemia

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Low-dose cytarabine (LDAC) is a standard therapy for elderly acute myeloid leukemia (AML) patients unfit for intensive chemotherapy. While high doses of cytarabine induce cytotoxicity, the precise mechanism of action of LDAC in AML remains elusive. In vitro studies have demonstrated LDAC-induced differentiation; however, such differentiation is seldom observed in vivo. We hypothesize that this discrepancy may be attributed to the influence of bone marrow (BM) stromal cells on AML cells. Thus, this study aimed to investigate the impact of BM stromal cells on LDAC-induced differentiation of AML cell lines and primary samples. Our results demonstrate that the presence of MS-5 stromal cells prevented LDAC-induced cell cycle arrest, DNA damage signaling and differentiation of U937 and MOLM-13 cell lines. Although transcriptomic analysis revealed that the stroma reduces the expression of genes involved in cytokine signaling and oxidative stress, data obtained with pharmacological inhibitors and neutralizing antibodies did not support the role for CXCL12, TGF-β1 or reactive oxygen species. The presence of stromal cells reduces LDAC-induced differentiation in primary samples from AML-M4 and myelodysplastic syndrome/AML patients. In conclusion, our study demonstrates that BM stroma reduces differentiation of AML induced by LDAC. These findings provide insights into the limited occurrence of terminal differentiation observed in AML patients, and suggest a potential explanation for this observation.

          Related collections

          Most cited references48

          • Record: found
          • Abstract: found
          • Article: not found

          Enasidenib in mutant IDH2 relapsed or refractory acute myeloid leukemia

          Recurrent mutations in isocitrate dehydrogenase 2 (IDH2) occur in ∼12% of patients with acute myeloid leukemia (AML). Mutated IDH2 proteins neomorphically synthesize 2-hydroxyglutarate resulting in DNA and histone hypermethylation, which leads to blocked cellular differentiation. Enasidenib (AG-221/CC-90007) is a first-in-class, oral, selective inhibitor of mutant-IDH2 enzymes. This first-in-human phase 1/2 study assessed the maximum tolerated dose (MTD), pharmacokinetic and pharmacodynamic profiles, safety, and clinical activity of enasidenib in patients with mutant-IDH2 advanced myeloid malignancies. We assessed safety outcomes for all patients and clinical efficacy in the largest patient subgroup, those with relapsed or refractory AML, from the phase 1 dose-escalation and expansion phases of the study. In the dose-escalation phase, an MTD was not reached at doses ranging from 50 to 650 mg per day. Enasidenib 100 mg once daily was selected for the expansion phase on the basis of pharmacokinetic and pharmacodynamic profiles and demonstrated efficacy. Grade 3 to 4 enasidenib-related adverse events included indirect hyperbilirubinemia (12%) and IDH-inhibitor-associated differentiation syndrome (7%). Among patients with relapsed or refractory AML, overall response rate was 40.3%, with a median response duration of 5.8 months. Responses were associated with cellular differentiation and maturation, typically without evidence of aplasia. Median overall survival among relapsed/refractory patients was 9.3 months, and for the 34 patients (19.3%) who attained complete remission, overall survival was 19.7 months. Continuous daily enasidenib treatment was generally well tolerated and induced hematologic responses in patients for whom prior AML therapy had failed. Inducing differentiation of myeloblasts, not cytotoxicity, seems to drive the clinical efficacy of enasidenib. This trial was registered at www.clinicaltrials.gov as #NCT01915498.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Regulation of the HIF-1alpha level is essential for hematopoietic stem cells.

            Hematopoietic stem cells (HSCs) are sustained in a specific microenvironment known as the stem cell niche. Mammalian HSCs are kept quiescent in the endosteal niche, a hypoxic zone of the bone marrow (BM). In this study, we show that normal HSCs maintain intracellular hypoxia and stabilize hypoxia-inducible factor-1alpha (HIF-1alpha) protein. In HIF-1alpha-deficient mice, the HSCs lost their cell cycle quiescence and HSC numbers decreased during various stress settings including bone marrow transplantation, myelosuppression, or aging, in a p16(Ink4a)/p19(Arf)-dependent manner. Overstabilization of HIF-1alpha by biallelic loss of an E3 ubiquitin ligase for HIF-1alpha (VHL) induced cell cycle quiescence in HSCs and their progenitors but resulted in an impairment in transplantation capacity. In contrast, monoallelic loss of VHL induced cell cycle quiescence and improved BM engraftment during bone marrow transplantation. These data indicate that HSCs maintain cell cycle quiescence through the precise regulation of HIF-1alpha levels. Copyright 2010 Elsevier Inc. All rights reserved.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Protective mitochondrial transfer from bone marrow stromal cells to acute myeloid leukemic cells during chemotherapy.

              Here we demonstrate that in a niche-like coculture system, cells from both primary and cultured acute myeloid leukemia (AML) sources take up functional mitochondria from murine or human bone marrow stromal cells. Using different molecular and imaging approaches, we show that AML cells can increase their mitochondrial mass up to 14%. After coculture, recipient AML cells showed a 1.5-fold increase in mitochondrial adenosine triphosphate production and were less prone to mitochondrial depolarization after chemotherapy, displaying a higher survival. This unidirectional transfer enhanced by some chemotherapeutic agents required cell-cell contacts and proceeded through an endocytic pathway. Transfer was greater in AML blasts compared with normal cord blood CD34(+) cells. Finally, we demonstrate that mitochondrial transfer was observed in vivo in an NSG immunodeficient mouse xenograft model and also occurred in human leukemia initiating cells and progenitors. As mitochondrial transfer provides a clear survival advantage following chemotherapy and a higher leukemic long-term culture initiating cell potential, targeting mitochondrial transfer could represent a future therapeutic target for AML treatment.
                Bookmark

                Author and article information

                Contributors
                URI : https://loop.frontiersin.org/people/1206565/overviewRole: Role: Role: Role: Role:
                URI : https://loop.frontiersin.org/people/2550411/overviewRole: Role: Role: Role: Role:
                URI : https://loop.frontiersin.org/people/1230360/overviewRole: Role: Role: Role: Role:
                URI : https://loop.frontiersin.org/people/1205980/overviewRole: Role: Role: Role: Role:
                URI : https://loop.frontiersin.org/people/2026135/overviewRole: Role: Role:
                Role: Role: Role:
                URI : https://loop.frontiersin.org/people/821127/overviewRole: Role: Role: Role: Role: Role:
                Journal
                Front Pharmacol
                Front Pharmacol
                Front. Pharmacol.
                Frontiers in Pharmacology
                Frontiers Media S.A.
                1663-9812
                26 October 2023
                2023
                : 14
                : 1258151
                Affiliations
                [1] 1 Laboratory for Cell Biology , Department of Physiology , Croatian Institute for Brain Research , University of Zagreb School of Medicine , Zagreb, Croatia
                [2] 2 Division of Hematology , Department of Internal Medicine , University Hospital Centre Zagreb , Zagreb, Croatia
                [3] 3 Clinical Research Division , Fred Hutchinson Cancer Research Centre , Seattle, WA, United States
                Author notes

                Edited by: Yan-Lai Tang, First Affiliated Hospital of Sun Yat-Sen University, China

                Reviewed by: Jennifer Speth, University of Michigan, United States

                Min Hee Kang, Texas Tech University Health Sciences Center, United States

                *Correspondence: Dora Visnjic, visnjic@ 123456mef.hr
                Article
                1258151
                10.3389/fphar.2023.1258151
                10637411
                d123d448-520f-4ed1-a22d-229d83aa4236
                Copyright © 2023 Smoljo, Tomic, Lalic, Dembitz, Batinic, Bedalov and Visnjic.

                This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

                History
                : 13 July 2023
                : 16 October 2023
                Funding
                Funded by: Hrvatska Zaklada za Znanost , doi 10.13039/501100004488;
                Award ID: IP-2016-06-4581
                Funded by: European Social Fund , doi 10.13039/501100004895;
                Award ID: DOK-2018-01-9599
                Funded by: National Institutes of Health , doi 10.13039/100000002;
                Funded by: Lady Tata Memorial Trust , doi 10.13039/501100004101;
                Funded by: European Regional Development Fund , doi 10.13039/501100008530;
                The authors declare financial support was received for the research, authorship, and/or publication of this article. This research was funded by Croatian Science Foundation under the projects IP-2016-06-4581, DOK-2018-01-9599, and DOK-2020-01-2873 by the European Union through the ESF Operational Programme Efficient Human Resources 2014-2020 (to DV), supported by National Institute of Health Grant R01GM117446 to AB, The Lady Tata Memorial Trust (to VD, International Award for Research in Leukaemia), and co-financed by the Scientific Centre of Excellence for Basic, Clinical and Translational Neuroscience (project “Experimental and clinical research of hypoxic-ischemic damage in perinatal and adult brain”; GA KK01.1.1.01.0007) funded by the European Union through the European Regional Development Fund.
                Categories
                Pharmacology
                Original Research
                Custom metadata
                Pharmacology of Anti-Cancer Drugs

                Pharmacology & Pharmaceutical medicine
                cytarabine,differentiation,acute myeloid leukemia,bone marrow stromal cells,cell cycle

                Comments

                Comment on this article