1
|
Patel SB, Moskop DR, Jordan CT, Pietras EM. Understanding MDS stem cells: Advances and limitations. Semin Hematol 2024; 61:409-419. [PMID: 39472255 DOI: 10.1053/j.seminhematol.2024.09.007] [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: 06/25/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 11/10/2024]
Abstract
In work spanning several decades, extensive studies have focused on the properties of malignant stem cells that drive the pathogenesis of acute myeloid leukemia (AML). However, relatively little attention has been devoted to several serious myeloid malignancies that occur prior to the onset of frank leukemia, including myelodysplastic syndrome (MDS). Like leukemia, MDS is hypothesized to arise from a pool of immature malignant stem and progenitor cells (MDS-SCs) that serve as a reservoir for disease evolution and progression1. While multiple studies have sought to identify and characterize the biology and vulnerabilities of MDS-SCs, yet translation of scientific concepts to therapeutically impactful regimens has been limited. Here, we evaluate the currently known properties of MDS-SCs as well as the post-transcriptional mechanisms that drive MDS pathogenesis at a stem and progenitor level. We highlight limits and gaps in our characterization and understanding of MDS-SCs and address the extent to which the properties of MDS-SC are (and can be) inferred from the characterization of LSCs.
Collapse
Affiliation(s)
- Sweta B Patel
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora CO
| | - Daniel R Moskop
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora CO
| | - Craig T Jordan
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora CO.
| | - Eric M Pietras
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora CO.
| |
Collapse
|
2
|
Cheng S, Chen L, Ying J, Wang Y, Jiang W, Zhang Q, Zhang H, Wang J, Wang C, Wu H, Ye J, Zhang L. 20(S)-ginsenoside Rh2 ameliorates ATRA resistance in APL by modulating lactylation-driven METTL3. J Ginseng Res 2024; 48:298-309. [PMID: 38707638 PMCID: PMC11068957 DOI: 10.1016/j.jgr.2023.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/18/2023] [Accepted: 12/20/2023] [Indexed: 05/07/2024] Open
Abstract
Background 20(S)-ginsenoside Rh2(GRh2), an effective natural histone deacetylase inhibitor, can inhibit acute myeloid leukemia (AML) cell proliferation. Lactate regulated histone lactylation, which has different temporal dynamics from acetylation. However, whether the high level of lactylation modification that we first detected in acute promyelocytic leukemia (APL) is associated with all-trans retinoic acid (ATRA) resistance has not been reported. Furthermore, Whether GRh2 can regulate lactylation modification in ATRA-resistant APL remains unknown. Methods Lactylation and METTL3 expression levels in ATRA-sensitive and ATRA-resistant APL cells were detected by Western blot analysis, qRT-PCR and CO-IP. Flow cytometry (FCM) and APL xenograft mouse models were used to determine the effect of METTL3 and GRh2 on ATRA-resistance. Results Histone lactylation and METTL3 expression levels were considerably upregulated in ATRA-resistant APL cells. METTL3 was regulated by histone lactylation and direct lactylation modification. Overexpression of METTL3 promoted ATRA-resistance. GRh2 ameliorated ATRA-resistance by downregulated lactylation level and directly inhibiting METTL3. Conclusions This study suggests that lactylation-modified METTL3 could provide a promising strategy for ameliorating ATRA-resistance in APL, and GRh2 could act as a potential lactylation-modified METTL3 inhibitor to ameliorate ATRA-resistance in APL.
Collapse
Affiliation(s)
- Siyu Cheng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Langqun Chen
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Jiahui Ying
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Ying Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Wenjuan Jiang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Qi Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Hong Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Jiahe Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Chen Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Huimin Wu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Jing Ye
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, China
| | - Liang Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| |
Collapse
|
3
|
van Spronsen MF, Hanekamp D, Westers TM, van Gils N, Vermue E, Rutten A, Jansen JH, Lissenberg-Witte BI, Smit L, Schuurhuis GJ, van de Loosdrecht AA. Immunophenotypic aberrant hematopoietic stem cells in myelodysplastic syndromes: a biomarker for leukemic progression. Leukemia 2023; 37:680-690. [PMID: 36792658 PMCID: PMC9991914 DOI: 10.1038/s41375-023-01811-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 11/06/2022] [Accepted: 01/06/2023] [Indexed: 02/17/2023]
Abstract
Myelodysplastic syndromes (MDS) comprise hematological disorders that originate from the neoplastic transformation of hematopoietic stem cells (HSCs). However, discrimination between HSCs and their neoplastic counterparts in MDS-derived bone marrows (MDS-BMs) remains challenging. We hypothesized that in MDS patients immature CD34+CD38- cells with aberrant expression of immunophenotypic markers reflect neoplastic stem cells and that their frequency predicts leukemic progression. We analyzed samples from 68 MDS patients and 53 controls and discriminated HSCs from immunophenotypic aberrant HSCs (IA-HSCs) expressing membrane aberrancies (CD7, CD11b, CD22, CD33, CD44, CD45RA, CD56, CD123, CD366 or CD371). One-third of the MDS-BMs (23/68) contained IA-HSCs. The presence of IA-HSCs correlated with perturbed hematopoiesis (disproportionally expanded CD34+ subsets beside cytopenias) and an increased hazard of leukemic progression (HR = 25, 95% CI: 2.9-218) that was independent of conventional risk factors. At 2 years follow-up, the sensitivity and specificity of presence of IA-HSCs for predicting leukemic progression was 83% (95% CI: 36-99%) and 71% (95% CI: 58-81%), respectively. In a selected cohort (n = 10), most MDS-BMs with IA-HSCs showed genomic complexity and high human blast counts following xenotransplantation into immunodeficient mice, contrasting MDS-BMs without IA-HSCs. This study demonstrates that the presence of IA-HSCs within MDS-BMs predicts leukemic progression, indicating the clinical potential of IA-HSCs as a prognostic biomarker.
Collapse
Affiliation(s)
- Margot F van Spronsen
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Diana Hanekamp
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Hematology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Theresia M Westers
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Noortje van Gils
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Eline Vermue
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Arjo Rutten
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Joop H Jansen
- Department of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Birgit I Lissenberg-Witte
- Department of Epidemiology and Data Science, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Linda Smit
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Gerrit J Schuurhuis
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Arjan A van de Loosdrecht
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
4
|
Porwit A, Béné MC, Duetz C, Matarraz S, Oelschlaegel U, Westers TM, Wagner-Ballon O, Kordasti S, Valent P, Preijers F, Alhan C, Bellos F, Bettelheim P, Burbury K, Chapuis N, Cremers E, Della Porta MG, Dunlop A, Eidenschink-Brodersen L, Font P, Fontenay M, Hobo W, Ireland R, Johansson U, Loken MR, Ogata K, Orfao A, Psarra K, Saft L, Subira D, Te Marvelde J, Wells DA, van der Velden VHJ, Kern W, van de Loosdrecht AA. Multiparameter flow cytometry in the evaluation of myelodysplasia: Analytical issues: Recommendations from the European LeukemiaNet/International Myelodysplastic Syndrome Flow Cytometry Working Group. CYTOMETRY. PART B, CLINICAL CYTOMETRY 2023; 104:27-50. [PMID: 36537621 PMCID: PMC10107708 DOI: 10.1002/cyto.b.22108] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/20/2022] [Accepted: 11/29/2022] [Indexed: 01/18/2023]
Abstract
Multiparameter flow cytometry (MFC) is one of the essential ancillary methods in bone marrow (BM) investigation of patients with cytopenia and suspected myelodysplastic syndrome (MDS). MFC can also be applied in the follow-up of MDS patients undergoing treatment. This document summarizes recommendations from the International/European Leukemia Net Working Group for Flow Cytometry in Myelodysplastic Syndromes (ELN iMDS Flow) on the analytical issues in MFC for the diagnostic work-up of MDS. Recommendations for the analysis of several BM cell subsets such as myeloid precursors, maturing granulocytic and monocytic components and erythropoiesis are given. A core set of 17 markers identified as independently related to a cytomorphologic diagnosis of myelodysplasia is suggested as mandatory for MFC evaluation of BM in a patient with cytopenia. A myeloid precursor cell (CD34+ CD19- ) count >3% should be considered immunophenotypically indicative of myelodysplasia. However, MFC results should always be evaluated as part of an integrated hematopathology work-up. Looking forward, several machine-learning-based analytical tools of interest should be applied in parallel to conventional analytical methods to investigate their usefulness in integrated diagnostics, risk stratification, and potentially even in the evaluation of response to therapy, based on MFC data. In addition, compiling large uniform datasets is desirable, as most of the machine-learning-based methods tend to perform better with larger numbers of investigated samples, especially in such a heterogeneous disease as MDS.
Collapse
Affiliation(s)
- Anna Porwit
- Division of Oncology and Pathology, Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| | - Marie C Béné
- Hematology Biology, Nantes University Hospital, CRCINA Inserm 1232, Nantes, France
| | - Carolien Duetz
- Department of Hematology, Amsterdam UMC, VU University Medical Center Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Sergio Matarraz
- Cancer Research Center (IBMCC-USAL/CSIC), Department of Medicine and Cytometry Service, Institute for Biomedical Research of Salamanca (IBSAL) and CIBERONC, University of Salamanca, Salamanca, Spain
| | - Uta Oelschlaegel
- Department of Internal Medicine, University Hospital Carl-Gustav-Carus, TU Dresden, Dresden, Germany
| | - Theresia M Westers
- Department of Hematology, Amsterdam UMC, VU University Medical Center Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Orianne Wagner-Ballon
- Department of Hematology and Immunology, Assistance Publique-Hôpitaux de Paris, University Hospital Henri Mondor, Créteil, France
- Inserm U955, Université Paris-Est Créteil, Créteil, France
| | | | - Peter Valent
- Department of Internal Medicine I, Division of Hematology & Hemostaseology and Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
| | - Frank Preijers
- Laboratory of Hematology, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Canan Alhan
- Department of Hematology, Amsterdam UMC, VU University Medical Center Cancer Center Amsterdam, Amsterdam, The Netherlands
| | | | - Peter Bettelheim
- Department of Hematology, Ordensklinikum Linz, Elisabethinen, Linz, Austria
| | - Kate Burbury
- Department of Haematology, Peter MacCallum Cancer Centre, & University of Melbourne, Melbourne, Australia
| | - Nicolas Chapuis
- Laboratory of Hematology, Assistance Publique-Hôpitaux de Paris, Centre-Université de Paris, Cochin Hospital, Paris, France
- Institut Cochin, INSERM U1016, CNRS UMR, Université de Paris, Paris, France
| | - Eline Cremers
- Division of Hematology, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Matteo G Della Porta
- IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Alan Dunlop
- Department of Haemato-Oncology, Royal Marsden Hospital, London, UK
| | | | - Patricia Font
- Department of Hematology, Hospital General Universitario Gregorio Marañon-IiSGM, Madrid, Spain
| | - Michaela Fontenay
- Laboratory of Hematology, Assistance Publique-Hôpitaux de Paris, Centre-Université de Paris, Cochin Hospital, Paris, France
- Institut Cochin, INSERM U1016, CNRS UMR, Université de Paris, Paris, France
| | - Willemijn Hobo
- Department of Internal Medicine I, Division of Hematology & Hemostaseology and Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
| | - Robin Ireland
- Department of Haematology and SE-HMDS, King's College Hospital NHS Foundation Trust, London, UK
| | - Ulrika Johansson
- Laboratory Medicine, SI-HMDS, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, UK
| | | | - Kiyoyuki Ogata
- Metropolitan Research and Treatment Centre for Blood Disorders (MRTC Japan), Tokyo, Japan
| | - Alberto Orfao
- Cancer Research Center (IBMCC-USAL/CSIC), Department of Medicine and Cytometry Service, Institute for Biomedical Research of Salamanca (IBSAL) and CIBERONC, University of Salamanca, Salamanca, Spain
| | - Katherina Psarra
- Department of Immunology - Histocompatibility, Evangelismos Hospital, Athens, Greece
| | - Leonie Saft
- Clinical Pathology and Cancer Diagnostics, Karolinska University Hospital and Institute Solna, Stockholm, Sweden
| | - Dolores Subira
- Department of Hematology, Flow Cytometry Unit, Hospital Universitario de Guadalajara, Guadalajara, Spain
| | - Jeroen Te Marvelde
- Laboratory Medical Immunology, Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | | | - Vincent H J van der Velden
- Laboratory Medical Immunology, Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | | | - Arjan A van de Loosdrecht
- Department of Hematology, Amsterdam UMC, VU University Medical Center Cancer Center Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
5
|
Zhan D, Park CY. Stem Cells in the Myelodysplastic Syndromes. FRONTIERS IN AGING 2021; 2:719010. [PMID: 35822030 PMCID: PMC9261372 DOI: 10.3389/fragi.2021.719010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 07/02/2021] [Indexed: 01/12/2023]
Abstract
The myelodysplastic syndromes (MDS) represent a group of clonal disorders characterized by ineffective hematopoiesis, resulting in peripheral cytopenias and frequent transformation to acute myeloid leukemia (AML). We and others have demonstrated that MDS arises in, and is propagated by malignant stem cells (MDS-SCs), that arise due to the sequential acquisition of genetic and epigenetic alterations in normal hematopoietic stem cells (HSCs). This review focuses on recent advancements in the cellular and molecular characterization of MDS-SCs, as well as their role in mediating MDS clinical outcomes. In addition to discussing the cell surface proteins aberrantly upregulated on MDS-SCs that have allowed the identification and prospective isolation of MDS-SCs, we will discuss the recurrent cytogenetic abnormalities and genetic mutations present in MDS-SCs and their roles in initiating disease, including recent studies demonstrating patterns of clonal evolution and disease progression from pre-malignant HSCs to MDS-SCs. We also will discuss the pathways that have been described as drivers or promoters of disease, including hyperactivated innate immune signaling, and how the identification of these alterations in MDS-SC have led to investigations of novel therapeutic strategies to treat MDS. It is important to note that despite our increasing understanding of the pathogenesis of MDS, the molecular mechanisms that drive responses to therapy remain poorly understood, especially the mechanisms that underlie and distinguish hematologic improvement from reductions in blast burden. Ultimately, such distinctions will be required in order to determine the shared and/or unique molecular mechanisms that drive ineffective hematopoiesis, MDS-SC maintenance, and leukemic transformation.
Collapse
Affiliation(s)
- Di Zhan
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, United States
- Perlmutter Cancer Center, New York University Grossman School of Medicine, New York, NY, United States
| | - Christopher Y. Park
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, United States
- Perlmutter Cancer Center, New York University Grossman School of Medicine, New York, NY, United States
- *Correspondence: Christopher Y. Park,
| |
Collapse
|
6
|
Al Naem M, Bourebaba L, Kucharczyk K, Röcken M, Marycz K. Therapeutic mesenchymal stromal stem cells: Isolation, characterization and role in equine regenerative medicine and metabolic disorders. Stem Cell Rev Rep 2021; 16:301-322. [PMID: 31797146 DOI: 10.1007/s12015-019-09932-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mesenchymal stromal cells (MSC) have become a popular treatment modality in equine orthopaedics. Regenerative therapies are especially interesting for pathologies like complicated tendinopathies of the distal limb, osteoarthritis, osteochondritis dissecans (OCD) and more recently metabolic disorders. Main sources for MSC harvesting in the horse are bone marrow, adipose tissue and umbilical cord blood. While the acquisition of umbilical cord blood is fairly easy and non-invasive, extraction of bone marrow and adipose tissue requires more invasive techniques. Characterization of the stem cells as a result of any isolation method, is also a crucial step for the confirmation of the cells' stemness properties; thus, three main characteristics must be fulfilled by these cells, namely: adherence, expression of a series of well-defined differentiation clusters as well as pluripotency. EVs, resulting from the paracrine action of MSCs, also play a key role in the therapeutic mechanisms mediated by stem cells; MSC-EVs are thus largely implicated in the regulation of proliferation, maturation, polarization and migration of various target cells. Evidence that EVs alone represent a complex network 0involving different soluble factors and could then reflect biophysical characteristics of parent cells has fuelled the importance of developing highly specific techniques for their isolation and analysis. All these aspects related to the functional and technical understanding of MSCs will be discussed and summarized in this review.
Collapse
Affiliation(s)
- Mohamad Al Naem
- Faculty of Veterinary Medicine, Equine Clinic - Equine Surgery, Justus-Liebig-University, 35392, Gießen, Germany
| | - Lynda Bourebaba
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Norwida 27B, 50-375, Wrocław, Poland.,International Institute of Translational Medicine, Jesionowa, 11, Malin, 55-114, Wisznia Mała, Poland
| | - Katarzyna Kucharczyk
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Norwida 27B, 50-375, Wrocław, Poland
| | - Michael Röcken
- Faculty of Veterinary Medicine, Equine Clinic - Equine Surgery, Justus-Liebig-University, 35392, Gießen, Germany
| | - Krzysztof Marycz
- Faculty of Veterinary Medicine, Equine Clinic - Equine Surgery, Justus-Liebig-University, 35392, Gießen, Germany. .,Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Norwida 27B, 50-375, Wrocław, Poland. .,International Institute of Translational Medicine, Jesionowa, 11, Malin, 55-114, Wisznia Mała, Poland.
| |
Collapse
|
7
|
Patnaik MM, Mughal TI, Brooks C, Lindsay R, Pemmaraju N. Targeting CD123 in hematologic malignancies: identifying suitable patients for targeted therapy. Leuk Lymphoma 2021; 62:2568-2586. [PMID: 33999767 DOI: 10.1080/10428194.2021.1927021] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Following the observation of interleukin 3 receptor α chain (IL-3Rα; CD123) upregulation on leukemia stem cells (LSCs) almost two decades ago, targeted treatment via CD123-diptheria toxin conjugates has now been tested in patients with diverse myeloid malignancies. Targeted eradication of LSCs could result in effective treatments for many challenging diseases initiated by these cells. Consequently, considerable effort has been directed toward targeting CD123 as a potential strategy for treating patients with hematologic malignancies in which CD123 is overexpressed. However, these therapies have had limited success so far, highlighting the need for suitable criteria to identify patients who could benefit from them. Given the diversity in CD123 expression across different hematologic malignancies, understanding CD123 expression patterns and the functional pathogenetic significance is crucial. Here, we review the methodologies available for CD123 assessment and discuss the biological and clinical characteristics of patients for whom CD123-targeting therapies may have a clinical impact.
Collapse
Affiliation(s)
- Mrinal M Patnaik
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Tariq I Mughal
- Division of Hematology-Oncology, Tufts University School of Medicine, Boston, MA, USA.,Research & Clinical Drug Development, Stemline Therapeutics, New York, NY, USA
| | - Christopher Brooks
- Research & Clinical Drug Development, Stemline Therapeutics, New York, NY, USA
| | - Ross Lindsay
- Research & Clinical Drug Development, Stemline Therapeutics, New York, NY, USA
| | - Naveen Pemmaraju
- Division of Cancer Medicine, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
8
|
Wang J, Xiang H, Lu Y, Wu T. Role and clinical significance of TGF‑β1 and TGF‑βR1 in malignant tumors (Review). Int J Mol Med 2021; 47:55. [PMID: 33604683 PMCID: PMC7895515 DOI: 10.3892/ijmm.2021.4888] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 01/27/2021] [Indexed: 12/24/2022] Open
Abstract
The appearance and growth of malignant tumors is a complicated process that is regulated by a number of genes. In recent years, studies have revealed that the transforming growth factor-β (TGF-β) signaling pathway serves an important role in cell cycle regulation, growth and development, differentiation, extracellular matrix synthesis and immune response. Notably, two members of the TGF-β signaling pathway, TGF-β1 and TGF-β receptor 1 (TGF-βR1), are highly expressed in a variety of tumors, such as breast cancer, colon cancer, gastric cancer and hepatocellular carcinoma. Moreover, an increasing number of studies have demonstrated that TGF-β1 and TGF-βR1 promote proliferation, migration and epithelial-mesenchymal transition of tumor cells by activating other signaling pathways, signaling molecules or microRNAs (miRs), such as the NF-κB signaling pathway and miR-133b. In addition, some inhibitors targeting TGF-β1 and TGF-βR1 have exhibited positive effects in in vitro experiments. The present review discusses the association between TGF-β1 or TGF-βR1 and tumors, and the development of some inhibitors, hoping to provide more approaches to help identify novel tumor markers to restrain and cure tumors.
Collapse
Affiliation(s)
- Junmin Wang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Hongjiao Xiang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Yifei Lu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Tao Wu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| |
Collapse
|
9
|
Behbehani GK, Finck R, Samusik N, Sridhar K, Fantl WJ, Greenberg PL, Nolan GP. Profiling myelodysplastic syndromes by mass cytometry demonstrates abnormal progenitor cell phenotype and differentiation. CYTOMETRY PART B-CLINICAL CYTOMETRY 2020; 98:131-145. [PMID: 31917512 PMCID: PMC9292828 DOI: 10.1002/cyto.b.21860] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 12/02/2019] [Indexed: 11/25/2022]
Abstract
Background We sought to enhance the cytometric analysis of myelodysplastic syndromes (MDS) by performing a pilot study of a single cell mass cytometry (MCM) assay to more comprehensively analyze patterns of surface marker expression in patients with MDS. Methods Twenty‐three MDS and five healthy donor bone marrow samples were studied using a 34‐parameter mass cytometry panel utilizing barcoding and internal reference standards. The resulting data were analyzed by both traditional gating and high‐dimensional clustering. Results This high‐dimensional assay provided three major benefits relative to traditional cytometry approaches: First, MCM enabled detection of aberrant surface maker at high resolution, detecting aberrancies in 27/31 surface markers, encompassing almost every previously reported MDS surface marker aberrancy. Additionally, three previously unrecognized aberrancies in MDS were detected in multiple samples at least one developmental stage: increased CD321 and CD99; and decreased CD47. Second, analysis of the stem and progenitor cell compartment (HSPCs), demonstrated aberrant expression in 21 of the 23 MDS samples, which were not detected in three samples from patients with idiopathic cytopenia of undetermined significance. These immunophenotypically abnormal HSPCs were also the single most significant distinguishing feature between clinical risk groups. Third, unsupervised clustering of high‐parameter MCM data allowed identification of abnormal differentiation patterns associated with immunophenotypically aberrant myeloid cells similar to myeloid derived suppressor cells. Conclusions These results demonstrate that high‐parameter cytometry methods that enable simultaneous analysis of all bone marrow cell types could enhance the diagnostic utility of immunophenotypic analysis in MDS.
Collapse
Affiliation(s)
- Gregory K. Behbehani
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology & ImmunologyStanford University School of Medicine Stanford California
- Department of Medicine, Division of HematologyStanford University School of Medicine Stanford California
- Stanford Cancer Institute Stanford California
| | - Rachel Finck
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology & ImmunologyStanford University School of Medicine Stanford California
| | - Nikolay Samusik
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology & ImmunologyStanford University School of Medicine Stanford California
| | - Kunju Sridhar
- Department of Medicine, Division of HematologyStanford University School of Medicine Stanford California
| | - Wendy J. Fantl
- Department of Obstetrics and Gynecology, Division of Gynecologic OncologyStanford University School of Medicine Stanford California
| | - Peter L. Greenberg
- Department of Medicine, Division of HematologyStanford University School of Medicine Stanford California
- Stanford Cancer Institute Stanford California
| | - Garry P. Nolan
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology & ImmunologyStanford University School of Medicine Stanford California
- Stanford Cancer Institute Stanford California
- Department of Obstetrics and Gynecology, Division of Gynecologic OncologyStanford University School of Medicine Stanford California
| |
Collapse
|
10
|
Guo D, Xu P, Chen D, Wang L, Zhu Y, Zuo Y, Chen B. Daunorubicin-Loaded CdTe QDs Conjugated with Anti-CD123 mAbs: A Novel Delivery System for Myelodysplastic Syndromes Treatment. Int J Nanomedicine 2020; 15:521-536. [PMID: 32021192 PMCID: PMC6988587 DOI: 10.2147/ijn.s233395] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 01/09/2020] [Indexed: 12/20/2022] Open
Abstract
INTRODUCTION The myelodysplastic syndromes (MDS) are a very heterogeneous group of myeloid disorders characterized by peripheral blood cytopenias and increase risk of transformation to acute myeloid leukemia (AML). Daunorubicin (DNR) is an indispensable drug for the treatment of MDS and AML. However, its side effects including cardiac toxicity and bone marrow suppression severely limit clinical application. Many researches reported high expression of CD123 antigen on high-risk MDS cells, so we constructed a novel drug delivery system comprising daunorubicin-loaded CdTe QDs conjugated with anti-CD123 mAbs (DNR-CdTe-CD123) to develop targeted combination chemotherapy for MDS. METHODS CdTe conjugated antiCD123 through amide bond, co-loaded with DNR with electrostatic bonding. Then, we determined characterization and release rate of DNR-CdTe-CD123. The therapeutic effect and side effect of drug delivery system were evaluated through in vitro and in vivo experiments. RESULTS CdTe showed appropriate diameter and good dispersibility and DNR was loaded into CdTes with high encapsulation efficiency and drug loading. The maximum drug loading and encapsulation efficiency were 42.08 ± 0.64% and 74.52 ± 1.81%, respectively, at DNR concentration of 0.2mg/mL and anti-CD123 mAbs volume of 5ul (100ug/mL). Flow cytometry (FCM) showed that CD123 antigen was highly expressed on MUTZ-1 cells, and its expression rate was 72.89 ± 10.67%. In vitro experiments showed that the inhibition rate and apoptosis rate of MUTZ-1 cells treated with DNR-CdTe-CD123 were higher than those in the other groups (P<0.05). Compared with the other groups, the level of apoptosis-related protein (P53, cleaved caspase-9, Bax and cleaved caspase-3) were upregulated in DNR-CdTe-CD123 group (P<0.05). In vivo experiments, DNR-CdTe-CD123 can effectively inhibit the tumor growth of MDS-bearing nude mice and reduce the side effects of DNR on myocardial cells. CONCLUSION The system of DNR-CdTe-CD123 enhances the therapeutic effects and reduce the side effects of DNR, thus providing a novel platform for MDS treatment.
Collapse
Affiliation(s)
- Dan Guo
- Department of Hematology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu210093, People’s Republic of China
- Department of Hematology, The Affiliated Hospital of Nantong University, Nantong, Jiangsu226001, People’s Republic of China
| | - Peipei Xu
- Department of Hematology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu210093, People’s Republic of China
| | - Dangui Chen
- Department of Hematology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu210093, People’s Republic of China
| | - Lili Wang
- Department of Hematology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu210093, People’s Republic of China
| | - Yudi Zhu
- Department of Hematology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu210093, People’s Republic of China
| | - Yifan Zuo
- Department of Hematology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu210093, People’s Republic of China
| | - Bing Chen
- Department of Hematology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu210093, People’s Republic of China
| |
Collapse
|
11
|
Gardikas N, Vikentiou M, Konsta E, Kontos CK, Papageorgiou SG, Spathis A, Bazani E, Bouchla A, Kapsimali V, Psarra K, Foukas P, Dimitriadis G, Pappa V. Immunophenotypic Profile of CD34+ Subpopulations and Their Role in the Diagnosis and Prognosis of Patients with De-Novo, Particularly Low-Grade Myelodysplastic Syndromes. CYTOMETRY PART B-CLINICAL CYTOMETRY 2018; 96:73-82. [DOI: 10.1002/cyto.b.21725] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 06/20/2018] [Accepted: 06/29/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Nikolaos Gardikas
- Second Department of Internal Medicine and Research Institute; University General Hospital Attikon; Haidari Greece
| | - Myrofora Vikentiou
- Second Department of Internal Medicine and Research Institute; University General Hospital Attikon; Haidari Greece
| | - Evgenia Konsta
- Second Department of Internal Medicine and Research Institute; University General Hospital Attikon; Haidari Greece
| | - Christos K. Kontos
- Department of Biochemistry and Molecular Biology; National and Kapodistrian University of Athens; Panepistimiopolis, Athens Greece
| | - Sotirios G. Papageorgiou
- Second Department of Internal Medicine and Research Institute; University General Hospital Attikon; Haidari Greece
| | - Aris Spathis
- 2nd Department of Pathology, School of Medicine; University of Athens, University General Hospital Attikon; Haidari Greece
| | - Efthimia Bazani
- Second Department of Internal Medicine and Research Institute; University General Hospital Attikon; Haidari Greece
| | - Anthi Bouchla
- Second Department of Internal Medicine and Research Institute; University General Hospital Attikon; Haidari Greece
| | - Violetta Kapsimali
- Department of Dermatology and Venereology; HIV/AIDS Unit, Andreas Syggros Hospital; Athens Greece
| | - Katherina Psarra
- Department of Immunology and Histocompatibility; Evangelismos Hospital; Athens Greece
| | - Periklis Foukas
- 2nd Department of Pathology, School of Medicine; University of Athens, University General Hospital Attikon; Haidari Greece
| | - George Dimitriadis
- Second Department of Internal Medicine and Research Institute; University General Hospital Attikon; Haidari Greece
| | - Vasiliki Pappa
- Second Department of Internal Medicine and Research Institute; University General Hospital Attikon; Haidari Greece
| |
Collapse
|
12
|
Stevens BM, Khan N, D'Alessandro A, Nemkov T, Winters A, Jones CL, Zhang W, Pollyea DA, Jordan CT. Characterization and targeting of malignant stem cells in patients with advanced myelodysplastic syndromes. Nat Commun 2018; 9:3694. [PMID: 30209285 PMCID: PMC6135858 DOI: 10.1038/s41467-018-05984-x] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 07/26/2018] [Indexed: 12/21/2022] Open
Abstract
Myelodysplastic syndrome (MDS) is a chronic hematologic disorder that frequently evolves to more aggressive stages and in some cases leads to acute myeloid leukemia (AML). MDS arises from mutations in hematopoietic stem cells (HSCs). Thus, to define optimal therapies, it is essential to understand molecular events driving HSC pathogenesis. In this study, we report that during evolution of MDS, malignant HSCs activate distinct cellular programs that render such cells susceptible to therapeutic intervention. Specifically, metabolic analyses of the MDS stem cell compartment show a profound activation of protein synthesis machinery and increased oxidative phosphorylation. Pharmacological targeting of protein synthesis and oxidative phosphorylation demonstrated potent and selective eradication of MDS stem cells in primary human patient specimens. Taken together, our findings indicate that MDS stem cells are reliant on specific metabolic events and that such properties can be targeted prior to the onset of clinically significant AML, during antecedent MDS.
Collapse
Affiliation(s)
- Brett M Stevens
- Division of Hematology, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Nabilah Khan
- Division of Hematology, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO, 80045, USA
| | - Travis Nemkov
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO, 80045, USA
| | - Amanda Winters
- Department of Pediatrics, University of Colorado Denver, Aurora, CO, 80045, USA
| | - Courtney L Jones
- Division of Hematology, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Wei Zhang
- Division of Hematology, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Daniel A Pollyea
- Division of Hematology, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Craig T Jordan
- Division of Hematology, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, 80045, USA.
| |
Collapse
|
13
|
Ostendorf BN, Flenner E, Flörcken A, Westermann J. Phenotypic characterization of aberrant stem and progenitor cell populations in myelodysplastic syndromes. PLoS One 2018; 13:e0197823. [PMID: 29799854 PMCID: PMC5969762 DOI: 10.1371/journal.pone.0197823] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 05/09/2018] [Indexed: 01/04/2023] Open
Abstract
Recent reports have revealed myelodysplastic syndromes (MDS) to arise from cancer stem cells phenotypically similar to physiological hematopoietic stem cells. Myelodysplastic hematopoiesis maintains a hierarchical organization, but the proportion of several hematopoietic compartments is skewed and multiple surface markers are aberrantly expressed. These aberrant antigen expression patterns hold diagnostic and therapeutic promise. However, eradication of MDS requires targeting of early myelodysplasia propagating stem cells. This warrants an exact assessment of the differentiation stage at which aberrant expression occurs in transformed hematopoiesis. Here, we report results on the prospective and extensive dissection of the hematopoietic hierarchy in 20 patients with either low-risk MDS or MDS with excess blasts and compare it to hematopoiesis in patients with non-malignancy-associated cytopenia or B cell lymphoma without bone marrow infiltration. We found patients with MDS with excess blasts to exhibit characteristic expansions of specific immature progenitor compartments. We also identified the aberrant expression of several markers including ALDH, CLL-1, CD44, and CD47 to be specific features of hematopoiesis in MDS with excess blasts. We show that amongst these, aberrant CLL-1 expression manifested at the early uncommitted hematopoietic stem cell level, suggesting a potential role as a therapeutic target.
Collapse
Affiliation(s)
- Benjamin N. Ostendorf
- Department of Hematology, Oncology, and Tumor Immunology, Charité –Universitätsmedizin Berlin, Campus Virchow Klinikum, Berlin, Germany
- Labor Berlin Charité Vivantes GmbH, Berlin, Germany
- * E-mail: (BNO); (JW)
| | - Eva Flenner
- Department of Hematology, Oncology, and Tumor Immunology, Charité –Universitätsmedizin Berlin, Campus Virchow Klinikum, Berlin, Germany
- Labor Berlin Charité Vivantes GmbH, Berlin, Germany
| | - Anne Flörcken
- Department of Hematology, Oncology, and Tumor Immunology, Charité –Universitätsmedizin Berlin, Campus Virchow Klinikum, Berlin, Germany
- Labor Berlin Charité Vivantes GmbH, Berlin, Germany
| | - Jörg Westermann
- Department of Hematology, Oncology, and Tumor Immunology, Charité –Universitätsmedizin Berlin, Campus Virchow Klinikum, Berlin, Germany
- Labor Berlin Charité Vivantes GmbH, Berlin, Germany
- * E-mail: (BNO); (JW)
| |
Collapse
|
14
|
Toft-Petersen M, Nederby L, Kjeldsen E, Kerndrup GB, Brown GD, Hokland P, Stidsholt Roug A. Unravelling the relevance of CLEC12A as a cancer stem cell marker in myelodysplastic syndrome. Br J Haematol 2016; 175:393-401. [PMID: 27612176 PMCID: PMC5091626 DOI: 10.1111/bjh.14270] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 06/09/2016] [Indexed: 01/02/2023]
Abstract
Evidence of distinct disease propagating stem cells in myelodysplastic syndrome (MDS) has emerged in recent years. However, immunophenotypic characterization of these cancer stem cells remains sparse. In acute myeloid leukaemia (AML), we have previously described aberrant expression of the C-type lectin domain family 12, member A (CLEC12A) as a stable and reliable marker of leukaemia blasts and as a tool for assessing minimal residual disease. Furthermore, CLEC12A has been proposed as a promising marker of leukaemic stem cells in AML. The role of CLEC12A in MDS, however, remains to be elucidated. In this study, we found CLEC12A aberrantly expressed on the CD34+ CD38- cell compartment in 71% (22/31) of MDS patients, distributed across all Revised International Prognostic Scoring System risk groups. We showed that the CD34+ CD38- CLEC12A+ cells were indeed malignant and possessed functional stem cell properties in the long-term colony-initiating cell assay. As opposed to reported findings in AML, we showed that cancer stem cells from MDS samples derived from both CLEC12A positive and negative CD34+ CD38- subpopulations. Due to the absence of CLEC12A on normal haematopoietic stem cells, CLEC12A stem cell immunophenotyping may contribute to diagnosing and monitoring MDS patients and could furthermore add knowledge about disease propagating cells in MDS.
Collapse
Affiliation(s)
| | - Line Nederby
- Department of Haematology, Aarhus University Hospital, Aarhus, Denmark
| | - Eigil Kjeldsen
- Department of Haematology, Aarhus University Hospital, Aarhus, Denmark
| | - Gitte B Kerndrup
- Department of Pathology, Aarhus University Hospital, Aarhus, Denmark
| | - Gordon D Brown
- Immunity, Infection and Inflammation Programme, Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Peter Hokland
- Department of Haematology, Aarhus University Hospital, Aarhus, Denmark
| | | |
Collapse
|
15
|
Adil MM, Levine RM, Kokkoli E. Increasing Cancer-Specific Gene Expression by Targeting Overexpressed α5β1 Integrin and Upregulated Transcriptional Activity of NF-κB. Mol Pharm 2014; 11:849-58. [DOI: 10.1021/mp400535v] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Maroof M. Adil
- Department
of Chemical Engineering
and Materials Science, University of Minnesota, 421 Washington Ave SE, Minneapolis, Minnesota 55455
| | - Rachel M. Levine
- Department
of Chemical Engineering
and Materials Science, University of Minnesota, 421 Washington Ave SE, Minneapolis, Minnesota 55455
| | - Efrosini Kokkoli
- Department
of Chemical Engineering
and Materials Science, University of Minnesota, 421 Washington Ave SE, Minneapolis, Minnesota 55455
| |
Collapse
|
16
|
Nishioka C, Ikezoe T, Furihata M, Yang J, Serada S, Naka T, Nobumoto A, Kataoka S, Tsuda M, Udaka K, Yokoyama A. CD34⁺/CD38⁻ acute myelogenous leukemia cells aberrantly express CD82 which regulates adhesion and survival of leukemia stem cells. Int J Cancer 2012; 132:2006-19. [PMID: 23055153 DOI: 10.1002/ijc.27904] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Accepted: 09/25/2012] [Indexed: 12/22/2022]
Abstract
To identify molecular targets in leukemia stem cells (LSCs), this study compared the protein expression profile of freshly isolated CD34(+) /CD38(-) cells with that of CD34(+) /CD38(+) counterparts from individuals with acute myelogenous leukemia (n = 2, AML) using isobaric tags for relative and absolute quantitation (iTRAQ). A total of 98 proteins were overexpressed, while six proteins were underexpressed in CD34(+) /CD38(-) AML cells compared with their CD34(+) /CD38(+) counterparts. Proteins overexpressed in CD34(+) /CD38(-) AML cells included a number of proteins involved in DNA repair, cell cycle arrest, gland differentiation, antiapoptosis, adhesion, and drug resistance. Aberrant expression of CD82, a family of adhesion molecules, in CD34(+) /CD38(-) AML cells was noted in additional clinical samples (n = 12) by flow cytometry. Importantly, down-regulation of CD82 in CD34(+) /CD38(-) AML cells by a short hairpin RNA (shRNA) inhibited adhesion to fibronectin via up-regulation of matrix metalloproteinases 9 (MMP9) and colony forming ability of these cells as assessed by transwell assay, real-time RT-PCR, and colony forming assay, respectively. Moreover, we found that down-regulation of CD82 in CD34(+) /CD38(-) AML cells by an shRNA significantly impaired engraftment of these cells in severely immunocompromised mice. Taken together, aberrant expression of CD82 might play a role in adhesion of LSCs to bone marrow microenvironment and survival of LSCs. CD82 could be an attractive molecular target to eradicate LSCs.
Collapse
Affiliation(s)
- Chie Nishioka
- Department of Immunology, Kochi Medical School, Kochi University, Nankoku, Kochi, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Schneider M, Huber J, Hadaschik B, Siegers GM, Fiebig HH, Schüler J. Characterization of colon cancer cells: a functional approach characterizing CD133 as a potential stem cell marker. BMC Cancer 2012; 12:96. [PMID: 22433494 PMCID: PMC3368744 DOI: 10.1186/1471-2407-12-96] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2011] [Accepted: 03/20/2012] [Indexed: 12/13/2022] Open
Abstract
Background Isolation and characterization of tumourigenic colon cancer initiating cells may help to develop novel diagnostic and therapeutic procedures. Methods We characterized a panel of fourteen human colon carcinoma cell lines and their corresponding xenografts for the surface expression of potential stem cell markers CD133, CD24, CD44, CDCP1 and CXCR4. In five cell lines and nine xenografts, mRNA expression of these markers was determined. Tumour growth behaviour of CD133+, CD133- and unsorted SW620 cells was evaluated in vivo. Results All five putative stem cell markers showed distinct expression patterns in the tumours examined. Two patient-derived cell lines highly expressed CD133 (> 85% of positive cells) and three other cell lines had an expression level of about 50% whereas in long-term culture based models CD133 expression ranged only from 0 to 20%. In 8/14 cell lines, more than 80% of the cells were positive for CD24 and 11/14 were over 70% positive for CD44. 10/14 cell lines expressed CDCP1 on ≥ 83% of cells. CXCR4 expression was determined solely on 94 L and SW480. Analyses of the corresponding xenografts revealed a significant reduction of cell numbers expressing the investigated surface markers and showed single cell fractions expressing up to three markers simultaneously. Statistical analysis revealed that the CXCR4 mRNA level correlates negatively with the protein expression of CD133, CD44, CD24 and CDCP1 in cell lines and xenografts. A lower differentiation grade of donor material correlated with a higher CDCP1 mRNA expression level in the respective tumour model. In vivo growth behaviour studies of SW620 revealed significantly higher take rates and shorter doubling times in the tumour growth of CD133 positive subclones in comparison to the unsorted cell line or CD133 negative subclones. Conclusions Our data revealed correlations in the expression of surface markers CD44 and CD24 as well as CD44 and CDCP1 and strongly suggest that CD133 is a stem cell marker within our colon carcinoma panel. Further studies will elucidate its role as a potential therapeutic target.
Collapse
Affiliation(s)
- Meike Schneider
- Department of Urology, University of Heidelberg, Im Neuenheimer Feld 110, D-69120 Heidelberg, Germany.
| | | | | | | | | | | |
Collapse
|
18
|
De Schauwer C, Piepers S, Van de Walle GR, Demeyere K, Hoogewijs MK, Govaere JLJ, Braeckmans K, Van Soom A, Meyer E. In search for cross-reactivity to immunophenotype equine mesenchymal stromal cells by multicolor flow cytometry. Cytometry A 2012; 81:312-23. [DOI: 10.1002/cyto.a.22026] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Revised: 11/23/2011] [Accepted: 01/17/2012] [Indexed: 02/02/2023]
|
19
|
Porwit A. Role of flow cytometry in diagnostics of myelodysplastic syndromes--beyond the WHO 2008 classification. Semin Diagn Pathol 2012; 28:273-82. [PMID: 22195405 DOI: 10.1053/j.semdp.2011.06.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Multiparameter flow cytometry (FCM) is an excellent method to follow the expression patterns of differentiation antigens using monoclonal antibodies to surface and cytoplasmic proteins. Although several authors described various aberrant immunophenotypic features in the bone marrow of patients with myelodysplastic syndromes (MDS), the World Health Organization 2008 classification recommended that, only if 3 or more phenotypic abnormalities are found involving 1 or more of the myeloid lineages can the aberrant FCM findings be considered suggestive of MDS. In the absence of conclusive morphologic and/or cytogenetic features, FCM abnormalities alone were considered not sufficient to establish MDS diagnosis and further follow-up of the patients was recommended. Review of the literature gives accumulating evidence that FCM has become an important part of the integrated diagnostic work-up of patients with suspected MDS. Several studies have also reported FCM findings significant for prognosis and therapy choice in MDS patients. Technical progress in multicolor FCM and new analysis programs, together with ongoing efforts to standardize the methodology, will make it possible to apply FCM in individual risk assessment and choice of best therapy for MDS patients.
Collapse
Affiliation(s)
- Anna Porwit
- Department of Laboratory Hematology, University Health Network, Toronto, Ontario, Canada.
| |
Collapse
|
20
|
Beksac M, Preffer F. Is it time to revisit our current hematopoietic progenitor cell quantification methods in the clinic? Bone Marrow Transplant 2011; 47:1391-6. [PMID: 22139068 DOI: 10.1038/bmt.2011.240] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
In the clinical practice of hematopoietic SCT, the minimum numbers of cells required for a successful engraftment are defined on the basis of their CD45 and CD34 expression profiles. However, the quantity of earlier progenitors or CD34-positive cells at different differentiation stages within stem cell grafts is not generally taken into consideration. During the last decade, various teams have quantified the number of cells expressing various combinations of CD34, CD38, CD133, CD90 co-expression and/or aldehyde dehydrogenase functional capacity using flow cytometry. Some of these studies resulted in the greater appreciation that combinations of these Ags were associated with varied myeloid, erythroid and platelet engraftment rates whereas others showed that the relative absence or presence of these markers could define cells responsible for either short- or long-term engraftment. These findings were also extended to differences between progenitor cell populations found within BM vs peripheral or cord-blood grafts. Cells harvested from donors are also generally frozen and stored; thawed cells have variable levels of viability and functional capacity based on the time tested post thaw, which also can be assessed by flow cytometry. Finally, flow cytometry has the potential for analysis of cells carrying a mesenchymal stem cell phenotype, which may be quiescent within some of the stem cell products. This review will address the need for stem cell subpopulation quantification and summarize existing published data to identify some Ags and functional characteristics that can be applicable to daily clinical practice.
Collapse
Affiliation(s)
- M Beksac
- Ankara University School of Medicine, Department of Hematology, Ankara, Turkey.
| | | |
Collapse
|
21
|
|