1
|
Cieri N, Hookeri N, Stromhaug K, Li L, Keating J, Díaz-Fernández P, Gómez-García de Soria V, Stevens J, Kfuri-Rubens R, Shao Y, Kooshesh KA, Powell K, Ji H, Hernandez GM, Abelin J, Klaeger S, Forman C, Clauser KR, Sarkizova S, Braun DA, Penter L, Kim HT, Lane WJ, Oliveira G, Kean LS, Li S, Livak KJ, Carr SA, Keskin DB, Muñoz-Calleja C, Ho VT, Ritz J, Soiffer RJ, Neuberg D, Stewart C, Getz G, Wu CJ. Systematic identification of minor histocompatibility antigens predicts outcomes of allogeneic hematopoietic cell transplantation. Nat Biotechnol 2024:10.1038/s41587-024-02348-3. [PMID: 39169264 DOI: 10.1038/s41587-024-02348-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 07/02/2024] [Indexed: 08/23/2024]
Abstract
T cell alloreactivity against minor histocompatibility antigens (mHAgs)-polymorphic peptides resulting from donor-recipient (D-R) disparity at sites of genetic polymorphisms-is at the core of the therapeutic effect of allogeneic hematopoietic cell transplantation (allo-HCT). Despite the crucial role of mHAgs in graft-versus-leukemia (GvL) and graft-versus-host disease (GvHD) reactions, it remains challenging to consistently link patient-specific mHAg repertoires to clinical outcomes. Here we devise an analytic framework to systematically identify mHAgs, including their detection on HLA class I ligandomes and functional verification of their immunogenicity. The method relies on the integration of polymorphism detection by whole-exome sequencing of germline DNA from D-R pairs with organ-specific transcriptional- and proteome-level expression. Application of this pipeline to 220 HLA-matched allo-HCT D-R pairs demonstrated that total and organ-specific mHAg load could independently predict the occurrence of acute GvHD and chronic pulmonary GvHD, respectively, and defined promising GvL targets, confirmed in a validation cohort of 58 D-R pairs, for the prevention or treatment of post-transplant disease recurrence.
Collapse
Affiliation(s)
- Nicoletta Cieri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Nidhi Hookeri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Kari Stromhaug
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Liang Li
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Julia Keating
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Paula Díaz-Fernández
- Department of Immunology, Instituto de Investigación Sanitaria Princesa (IIS-IP), Hospital Universitario de La Princesa, Madrid, Spain
| | - Valle Gómez-García de Soria
- Department of Hematology, Instituto de Investigación Sanitaria Princesa (IIS-IP), Hospital Universitario de La Princesa, Madrid, Spain
| | - Jonathan Stevens
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Raphael Kfuri-Rubens
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Yiren Shao
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Kaila Powell
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Helen Ji
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Gabrielle M Hernandez
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Jennifer Abelin
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Susan Klaeger
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
- Department of Proteomic and Genomic Technologies, Genentech Inc, South San Francisco, CA, USA
| | - Cleo Forman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Karl R Clauser
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Siranush Sarkizova
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - David A Braun
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
| | - Livius Penter
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Hematology, Oncology, and Tumorimmunology, Campus Virchow Klinikum, Berlin, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Haesook T Kim
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - William J Lane
- Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Giacomo Oliveira
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Leslie S Kean
- Harvard Medical School, Boston, MA, USA
- Division Hematology/Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
| | - Shuqiang Li
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Translational Immunogenomics Lab, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Kenneth J Livak
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Translational Immunogenomics Lab, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Steven A Carr
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Derin B Keskin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Translational Immunogenomics Lab, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Computer Science, Metropolitan College, Boston University, Boston, MA, USA
- Section for Bioinformatics, Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Cecilia Muñoz-Calleja
- Department of Immunology, Instituto de Investigación Sanitaria Princesa (IIS-IP), Hospital Universitario de La Princesa, Madrid, Spain
- Department of Medicine, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Vincent T Ho
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Jerome Ritz
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Robert J Soiffer
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Donna Neuberg
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Chip Stewart
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Gad Getz
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Catherine J Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
2
|
Nagel S, Meyer C. Identification of Gene Regulatory Networks in B-Cell Progenitor Differentiation and Leukemia. Genes (Basel) 2024; 15:978. [PMID: 39202339 PMCID: PMC11353346 DOI: 10.3390/genes15080978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 07/09/2024] [Accepted: 07/22/2024] [Indexed: 09/03/2024] Open
Abstract
Pro-B- and pre-B-cells are consecutive entities in early B-cell development, representing cells of origin for B-cell precursor acute lymphoid leukemia (BCP-ALL). Normal B-cell differentiation is critically regulated by specific transcription factors (TFs). Accordingly, TF-encoding genes are frequently deregulated or mutated in BCP-ALL. Recently, we described TF-codes which delineate physiological activities of selected groups of TF-encoding genes in hematopoiesis including B-cell development. Here, we exploited these codes to uncover regulatory connections between particular TFs in pro-B- and pre-B-cells via an analysis of developmental TFs encoded by NKL and TALE homeobox genes and by ETS and T-box genes. Comprehensive expression analyses in BCP-ALL cell lines helped identify validated models to study their mutual regulation in vitro. Knockdown and overexpression experiments and subsequent RNA quantification of TF-encoding genes in selected model cell lines revealed activating, inhibitory or absent connections between nine TFs operating in early B-cell development, including HLX, MSX1, IRX1, MEIS1, ETS2, ERG, SPIB, EOMES, and TBX21. In addition, genomic profiling revealed BCP-ALL subtype-specific copy number alterations of ERG at 21q22, while a deletion of the TGFbeta-receptor gene TGFBR2 at 3p24 resulted in an upregulation of EOMES. Finally, we combined the data to uncover gene regulatory networks which control normal differentiation of early B-cells, collectively endorsing more detailed evaluation of BCP-ALL subtypes.
Collapse
Affiliation(s)
- Stefan Nagel
- Department of Human and Animal Cell Lines, Leibniz-Institute DSMZ, 38124 Braunschweig, Germany
| | | |
Collapse
|
3
|
Kasai F, Mizukoshi K, Nakamura Y. Variable characteristics overlooked in human K-562 leukemia cell lines with a common signature. Sci Rep 2024; 14:9619. [PMID: 38671192 PMCID: PMC11053119 DOI: 10.1038/s41598-024-60271-8] [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: 01/30/2024] [Accepted: 04/21/2024] [Indexed: 04/28/2024] Open
Abstract
K-562 is a well-known in vitro cellular model that represents human leukemia cell lines. Although the K-562 cells have been extensively characterized, there are inconsistencies in the data across publications, showing the presence of multiple K-562 cell lines. This suggests that analyzing a single K-562 cell line is insufficient to provide reliable reference data. In this study, we compared three K-562 cell lines with different IDs (RCB0027, RCB1635, and RCB1897) to investigate the fundamental characteristics of K-562 cells. Amplifications of the BCR-ABL1 fusion gene and at 13q31 were detected in all three cell lines, whereas each genome exhibited distinctive features of sequence variants and loss of heterozygosity. This implies that each K-562 cell line can be characterized by common and unique features through a comparison of multiple K-562 cell lines. Variations in transcriptome profiles and hemoglobin synthesis were also observed among the three cell lines, indicating that they should be considered sublines that have diverged from the common ancestral K-562 despite no changes from the original cell name. This leads to unintentional differences in genotypes and/or phenotypes among cell lines that share the same name. These data show that characterizing a single K-562 cell line does not necessarily provide data that are applicable to other K-562 cells. In this context, it is essential to modify cell names in accordance with changes in characteristics during cell culture. Furthermore, our data could serve as a reference for evaluating other K-562 sublines, facilitating the discovery of new K-562 sublines with distinct characteristics. This approach results in the accumulation of K-562 sublines with diverged characteristics and expands the options available, which may help in selecting the most suitable K-562 subline for each experiment.
Collapse
Affiliation(s)
- Fumio Kasai
- RIKEN Cell Bank, Cell Engineering Division, RIKEN BioResource Research Center, Tsukuba, Japan.
| | - Kumiko Mizukoshi
- RIKEN Cell Bank, Cell Engineering Division, RIKEN BioResource Research Center, Tsukuba, Japan
| | - Yukio Nakamura
- RIKEN Cell Bank, Cell Engineering Division, RIKEN BioResource Research Center, Tsukuba, Japan
| |
Collapse
|
4
|
Rouleau M, Villeneuve L, Allain EP, McCabe-Leroux J, Tremblay S, Nguyen Van Long F, Uchil A, Joly-Beauparlant C, Droit A, Guillemette C. Non-canonical transcriptional regulation of the poor prognostic factor UGT2B17 in chronic lymphocytic leukemic and normal B cells. BMC Cancer 2024; 24:410. [PMID: 38566115 PMCID: PMC10985967 DOI: 10.1186/s12885-024-12143-7] [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: 08/22/2023] [Accepted: 03/19/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND High expression of the glycosyltransferase UGT2B17 represents an independent adverse prognostic marker in chronic lymphocytic leukemia (CLL). It also constitutes a predictive marker for therapeutic response and a drug resistance mechanism. The key determinants driving expression of the UGT2B17 gene in normal and leukemic B-cells remain undefined. The UGT2B17 transcriptome is complex and is comprised of at least 10 alternative transcripts, identified by previous RNA-sequencing of liver and intestine. We hypothesized that the transcriptional program regulating UGT2B17 in B-lymphocytes is distinct from the canonical expression previously characterized in the liver. RESULTS RNA-sequencing and genomics data revealed a specific genomic landscape at the UGT2B17 locus in normal and leukemic B-cells. RNA-sequencing and quantitative PCR data indicated that the UGT2B17 enzyme is solely encoded by alternative transcripts expressed in CLL patient cells and not by the canonical transcript widely expressed in the liver and intestine. Chromatin accessible regions (ATAC-Seq) in CLL cells mapped with alternative promoters and non-coding exons, which may be derived from endogenous retrotransposon elements. By luciferase reporter assays, we identified key cis-regulatory STAT3, RELA and interferon regulatory factor (IRF) binding sequences driving the expression of UGT2B17 in lymphoblastoid and leukemic B-cells. Electrophoretic mobility shift assays and pharmacological inhibition demonstrated key roles for the CLL prosurvival transcription factors STAT3 and NF-κB in the leukemic expression of UGT2B17. CONCLUSIONS UGT2B17 expression in B-CLL is driven by key regulators of CLL progression. Our data suggest that a NF-κB/STAT3/IRF/UGT2B17 axis may represent a novel B-cell pathway promoting disease progression and drug resistance.
Collapse
Affiliation(s)
- Michèle Rouleau
- Faculty of Pharmacy, Centre Hospitalier Universitaire de Québec Research Center - Université Laval (CRCHUQc- UL), Université Laval, Québec, QC, Canada
- Cancer research center of Université Laval, Québec, Canada
| | - Lyne Villeneuve
- Faculty of Pharmacy, Centre Hospitalier Universitaire de Québec Research Center - Université Laval (CRCHUQc- UL), Université Laval, Québec, QC, Canada
- Cancer research center of Université Laval, Québec, Canada
| | - Eric P Allain
- Molecular Genetics Laboratory, Vitalité Health Network, Dr. Georges-L.-Dumont University Hospital Center, Moncton, NB, Canada
| | - Jules McCabe-Leroux
- Faculty of Pharmacy, Centre Hospitalier Universitaire de Québec Research Center - Université Laval (CRCHUQc- UL), Université Laval, Québec, QC, Canada
- Cancer research center of Université Laval, Québec, Canada
| | - Sophie Tremblay
- Faculty of Pharmacy, Centre Hospitalier Universitaire de Québec Research Center - Université Laval (CRCHUQc- UL), Université Laval, Québec, QC, Canada
- Cancer research center of Université Laval, Québec, Canada
| | - Flora Nguyen Van Long
- Faculty of Pharmacy, Centre Hospitalier Universitaire de Québec Research Center - Université Laval (CRCHUQc- UL), Université Laval, Québec, QC, Canada
- Cancer research center of Université Laval, Québec, Canada
| | - Ashwini Uchil
- Faculty of Pharmacy, Centre Hospitalier Universitaire de Québec Research Center - Université Laval (CRCHUQc- UL), Université Laval, Québec, QC, Canada
- Cancer research center of Université Laval, Québec, Canada
| | - Charles Joly-Beauparlant
- Cancer research center of Université Laval, Québec, Canada
- CRCHUQc-UL and Faculty of Medicine, Université Laval, Québec, Québec, Canada
| | - Arnaud Droit
- Cancer research center of Université Laval, Québec, Canada
- CRCHUQc-UL and Faculty of Medicine, Université Laval, Québec, Québec, Canada
| | - Chantal Guillemette
- Faculty of Pharmacy, Centre Hospitalier Universitaire de Québec Research Center - Université Laval (CRCHUQc- UL), Université Laval, Québec, QC, Canada.
- Cancer research center of Université Laval, Québec, Canada.
- Canada Research Chair in Pharmacogenomics, Faculty of Pharmacy, Université Laval, Québec, QC, Canada.
| |
Collapse
|
5
|
Nagel S, Rand U, Pommerenke C, Meyer C. Transcriptional Landscape of CUT-Class Homeobox Genes in Blastic Plasmacytoid Dendritic Cell Neoplasm. Int J Mol Sci 2024; 25:2764. [PMID: 38474011 DOI: 10.3390/ijms25052764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 02/20/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
Homeobox genes encode developmental transcription factors regulating tissue-specific differentiation processes and drive cancerogenesis when deregulated. Dendritic cells (DCs) are myeloid immune cells occurring as two types, either conventional or plasmacytoid DCs. Recently, we showed that the expression of NKL-subclass homeobox gene VENTX is restricted to conventional DCs, regulating developmental genes. Here, we identified and investigated homeobox genes specifically expressed in plasmacytoid DCs (pDCs) and derived blastic plasmacytoid dendritic cell neoplasm (BPDCN). We analyzed gene expression data, performed RQ-PCR, protein analyses by Western blot and immuno-cytology, siRNA-mediated knockdown assays and subsequent RNA-sequencing and live-cell imaging. Screening of public gene expression data revealed restricted activity of the CUT-class homeobox gene CUX2 in pDCs. An extended analysis of this homeobox gene class in myelopoiesis showed that additional CUX2 activity was restricted to myeloid progenitors, while BPDCN patients aberrantly expressed ONECUT2, which remained silent in the complete myeloid compartment. ONECUT2 expressing BPDCN cell line CAL-1 served as a model to investigate its regulation and oncogenic activity. The ONECUT2 locus at 18q21 was duplicated and activated by IRF4, AUTS2 and TNF-signaling and repressed by BMP4-, TGFb- and IL13-signalling. Functional analyses of ONECUT2 revealed the inhibition of pDC differentiation and of CDKN1C and CASP1 expression, while SMAD3 and EPAS1 were activated. EPAS1 in turn enhanced survival under hypoxic conditions which thus may support dendritic tumor cells residing in hypoxic skin lesions. Collectively, we revealed physiological and aberrant activities of CUT-class homeobox genes in myelopoiesis including pDCs and in BPDCN, respectively. Our data may aid in the diagnosis of BPDCN patients and reveal novel therapeutic targets for this fatal malignancy.
Collapse
Affiliation(s)
- Stefan Nagel
- Department of Human and Animal Cell Lines, Leibniz-Institute DSMZ, 38124 Braunschweig, Germany
| | - Ulfert Rand
- Department of Human and Animal Cell Lines, Leibniz-Institute DSMZ, 38124 Braunschweig, Germany
| | - Claudia Pommerenke
- Department of Human and Animal Cell Lines, Leibniz-Institute DSMZ, 38124 Braunschweig, Germany
| | - Corinna Meyer
- Department of Human and Animal Cell Lines, Leibniz-Institute DSMZ, 38124 Braunschweig, Germany
| |
Collapse
|
6
|
Pommerenke C, Nagel S, Haake J, Koelz AL, Christgen M, Steenpass L, Eberth S. Molecular Characterization and Subtyping of Breast Cancer Cell Lines Provide Novel Insights into Cancer Relevant Genes. Cells 2024; 13:301. [PMID: 38391914 PMCID: PMC10886524 DOI: 10.3390/cells13040301] [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: 12/21/2023] [Revised: 01/26/2024] [Accepted: 02/02/2024] [Indexed: 02/24/2024] Open
Abstract
Continuous cell lines are important and commonly used in vitro models in breast cancer (BC) research. Selection of the appropriate model cell line is crucial and requires consideration of their molecular characteristics. To characterize BC cell line models in depth, we profiled a panel of 29 authenticated and publicly available BC cell lines by mRNA-sequencing, mutation analysis, and immunoblotting. Gene expression profiles separated BC cell lines in two major clusters that represent basal-like (mainly triple-negative BC) and luminal BC subtypes, respectively. HER2-positive cell lines were located within the luminal cluster. Mutation calling highlighted the frequent aberration of TP53 and BRCA2 in BC cell lines, which, therefore, share relevant characteristics with primary BC. Furthermore, we showed that the data can be used to find novel, potential oncogenic fusion transcripts, e.g., FGFR2::CRYBG1 and RTN4IP1::CRYBG1 in cell line MFM-223, and to elucidate the regulatory circuit of IRX genes and KLF15 as novel candidate tumor suppressor genes in BC. Our data indicated that KLF15 was activated by IRX1 and inhibited by IRX3. Moreover, KLF15 inhibited IRX1 in cell line HCC-1599. Each BC cell line carries unique molecular features. Therefore, the molecular characteristics of BC cell lines described here might serve as a valuable resource to improve the selection of appropriate models for BC research.
Collapse
Affiliation(s)
- Claudia Pommerenke
- Department of Bioinformatics, IT and Databases, Leibniz Institute DSMZ-German Collection of Microorganisms and Cell Cultures, 38124 Braunschweig, Germany;
| | - Stefan Nagel
- Department of Human and Animal Cell Lines, Leibniz Institute DSMZ-German Collection of Microorganisms and Cell Cultures, 38124 Braunschweig, Germany; (S.N.)
| | - Josephine Haake
- Department of Human and Animal Cell Lines, Leibniz Institute DSMZ-German Collection of Microorganisms and Cell Cultures, 38124 Braunschweig, Germany; (S.N.)
| | - Anne Leena Koelz
- Department of Human and Animal Cell Lines, Leibniz Institute DSMZ-German Collection of Microorganisms and Cell Cultures, 38124 Braunschweig, Germany; (S.N.)
| | - Matthias Christgen
- Institute of Pathology, Hannover Medical School, 30625 Hannover, Germany
| | - Laura Steenpass
- Department of Human and Animal Cell Lines, Leibniz Institute DSMZ-German Collection of Microorganisms and Cell Cultures, 38124 Braunschweig, Germany; (S.N.)
- Zoological Institute, Technische Universität Braunschweig, 38106 Braunschweig, Germany
| | - Sonja Eberth
- Department of Human and Animal Cell Lines, Leibniz Institute DSMZ-German Collection of Microorganisms and Cell Cultures, 38124 Braunschweig, Germany; (S.N.)
| |
Collapse
|
7
|
Nagel S, Haake J, Pommerenke C, Meyer C, MacLeod RAF. Establishment of the Myeloid TBX-Code Reveals Aberrant Expression of T-Box Gene TBX1 in Chronic Myeloid Leukemia. Int J Mol Sci 2023; 25:32. [PMID: 38203204 PMCID: PMC10778679 DOI: 10.3390/ijms25010032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 12/08/2023] [Accepted: 12/15/2023] [Indexed: 01/12/2024] Open
Abstract
T-box genes encode transcription factors, which control developmental processes and promote cancer if deregulated. Recently, we described the lymphoid TBX-code, which collates T-box gene activities in normal lymphopoiesis, enabling identification of members deregulated in lymphoid malignancies. Here, we have extended this analysis to cover myelopoiesis, compiling the myeloid TBX-code and, thus, highlighting which of these genes might be deregulated in myeloid tumor types. We analyzed public T-box gene expression datasets bioinformatically for normal and malignant cells. Candidate T-box-gene-expressing model cell lines were identified and examined by RQ-PCR, Western Blotting, genomic profiling, and siRNA-mediated knockdown combined with RNA-seq analysis and live-cell imaging. The established myeloid TBX-code comprised 10 T-box genes, including progenitor-cell-restricted TBX1. Accordingly, we detected aberrant expression of TBX1 in 10% of stem/progenitor-cell-derived chronic myeloid leukemia (CML) patients. The classic CML cell line K-562 expressed TBX1 at high levels and served as a model to identify TBX1 activators, including transcription factor GATA1 and genomic amplification of the TBX1 locus at 22q11; inhibitors, including BCR::ABL1 fusion and downregulated GNAI2, as well as BMP, FGF2, and WNT signaling; and the target genes CDKN1A, MIR17HG, NAV1, and TMEM38A. The establishment of the myeloid TBX-code permitted identification of aberrant TBX1 expression in subsets of CML patients and cell lines. TBX1 forms an integral part of an oncogenic regulatory network impacting proliferation, survival, and differentiation. Thus, the data spotlight novel diagnostic markers and potential therapeutic targets for this malignancy.
Collapse
Affiliation(s)
- Stefan Nagel
- Leibniz-Institute DSMZ, 38124 Braunschweig, Germany
| | | | | | | | | |
Collapse
|
8
|
Bellini C, Vergara E, Bencs F, Fodor K, Bősze S, Krivić D, Bacsa B, Surguta SE, Tóvári J, Reljic R, Horváti K. Design and Characterization of a Multistage Peptide-Based Vaccine Platform to Target Mycobacterium tuberculosis Infection. Bioconjug Chem 2023; 34:1738-1753. [PMID: 37606258 PMCID: PMC10587871 DOI: 10.1021/acs.bioconjchem.3c00273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/09/2023] [Indexed: 08/23/2023]
Abstract
The complex immunopathology ofMycobacterium tuberculosis(Mtb) is one of the main challenges in developing a novel vaccine against this pathogen, particularly regarding eliciting protection against both active and latent stages. Multistage vaccines, which contain antigens expressed in both phases, represent a promising strategy for addressing this issue, as testified by the tuberculosis vaccine clinical pipeline. Given this approach, we designed and characterized a multistage peptide-based vaccine platform containing CD4+ and CD8+ T cell epitopes previously validated for inducing a relevant T cell response against Mtb. After preliminary screening, CFP10 (32-39), GlfT2 (4-12), HBHA (185-194), and PPE15 (1-15) were selected as promising candidates, and we proved that the PM1 pool of these peptides triggered a T cell response in Mtb-sensitized human peripheral blood mononuclear cells (PBMCs). Taking advantage of the use of thiol-maleimide chemoselective ligation, we synthesized a multiepitope conjugate (Ac-CGHP). Our results showed a structure-activity relationship between the conjugation and a higher tendency to fold and assume an ordered secondary structure. Moreover, the palmitoylated conjugate (Pal-CGHP) comprising the same peptide antigens was associated with an enhanced cellular uptake in human and murine antigen-presenting cells and a better immunogenicity profile. Immunization study, conducted in BALB/c mice, showed that Pal-CGHP induced a significantly higher T cell proliferation and production of IFNγ and TNFα over PM1 formulated in the Sigma Adjuvant System.
Collapse
Affiliation(s)
- Chiara Bellini
- MTA-TTK
Lendület “Momentum” Peptide-Based Vaccines Research
Group, Institute of Materials and Environmental Chemistry, Research Centre for Natural Sciences, Budapest 1117, Hungary
- Hevesy
György PhD School of Chemistry, Eötvös
Loránd University, Budapest 1117, Hungary
| | - Emil Vergara
- Institute
for Infection and Immunity, St. George’s,
University of London, London SW17 0RE, U.K.
| | - Fruzsina Bencs
- Hevesy
György PhD School of Chemistry, Eötvös
Loránd University, Budapest 1117, Hungary
- Laboratory
of Structural Chemistry and Biology, Institute of Chemistry, Eötvös Loránd University, Budapest 1117, Hungary
| | - Kinga Fodor
- Department
of Laboratory Animal Science and Animal Protection, University of Veterinary Medicine, Budapest 1078, Hungary
| | - Szilvia Bősze
- ELKH-ELTE
Research Group of Peptide Chemistry, Eötvös Loránd
Research Network (ELKH), Eötvös
Loránd University, Budapest 1117, Hungary
| | - Denis Krivić
- Division
of Medical Physics and Biophysics, Gottfried Schatz Research Center, Medical University of Graz, 8010 Graz, Austria
| | - Bernadett Bacsa
- Division
of Medical Physics and Biophysics, Gottfried Schatz Research Center, Medical University of Graz, 8010 Graz, Austria
| | - Sára Eszter Surguta
- Department
of Experimental Pharmacology and National Tumor Biology Laboratory, National Institute of Oncology, Budapest 1122, Hungary
| | - József Tóvári
- Department
of Experimental Pharmacology and National Tumor Biology Laboratory, National Institute of Oncology, Budapest 1122, Hungary
| | - Rajko Reljic
- Institute
for Infection and Immunity, St. George’s,
University of London, London SW17 0RE, U.K.
| | - Kata Horváti
- MTA-TTK
Lendület “Momentum” Peptide-Based Vaccines Research
Group, Institute of Materials and Environmental Chemistry, Research Centre for Natural Sciences, Budapest 1117, Hungary
| |
Collapse
|
9
|
Nagel S, Fischer A, Bens S, Hauer V, Pommerenke C, Uphoff CC, Zaborski M, Siebert R, Quentmeier H. PI3K/AKT inhibitor BEZ-235 targets CCND2 and induces G1 arrest in breast implant-associated anaplastic large cell lymphoma. Leuk Res 2023; 133:107377. [PMID: 37647808 DOI: 10.1016/j.leukres.2023.107377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/11/2023] [Accepted: 08/25/2023] [Indexed: 09/01/2023]
Abstract
Breast implant-associated anaplastic large cell lymphoma (BIA-ALCL) is a mature, CD30-positive T-cell lymphoma lacking expression of the anaplastic lymphoma kinase (ALK). In contrast to ALK-positive ALCL, BIA-ALCL cells express cyclin D2 (CCND2) which controls cyclin dependent kinases 4 and 6 (CDK4/6). DNA methylation and expression analyses performed with cell lines and primary cells suggest that the expression of CCND2 in BIA-ALCL cell lines conforms to the physiological status of differentiated T-cells, and that it is not the consequence of genomic alterations as observed in other hematopoietic tumors. Using cell line model systems we show that treatment with the CDK4/6 inhibitor palbociclib effects dephosphorylation of the retinoblastoma protein (RB) and causes cell cycle arrest in G1 in BIA-ALCL. Moreover, we show that the PI3K/AKT inhibitor BEZ-235 induces dephosphorylation of the mTORC1 target S6 and of GSK3β, indicators for translational inhibition and proteasomal degradation. Consequently, CCND2 protein levels declined after stimulation with BEZ-235, RB was dephosphorylated and the cell cycle was arrested in G1. Taken together, our data imply potential application of CDK4/6 inhibitors and PI3K/AKT inhibitors for the therapy of BIA-ALCL.
Collapse
Affiliation(s)
- Stefan Nagel
- Leibniz-Institute DSMZ-German Collection of Microorganisms and Cell Cultures, Department of Human and Animal Cell Lines, Braunschweig, Germany.
| | - Anja Fischer
- Ulm University and Ulm University Medical Center, Institute of Human Genetics, Ulm, Germany
| | - Susanne Bens
- Ulm University and Ulm University Medical Center, Institute of Human Genetics, Ulm, Germany
| | - Vivien Hauer
- Leibniz-Institute DSMZ-German Collection of Microorganisms and Cell Cultures, Department of Human and Animal Cell Lines, Braunschweig, Germany
| | - Claudia Pommerenke
- Leibniz-Institute DSMZ-German Collection of Microorganisms and Cell Cultures, Department of Bioinformatics and Databases, Braunschweig, Germany
| | - Cord C Uphoff
- Leibniz-Institute DSMZ-German Collection of Microorganisms and Cell Cultures, Department of Human and Animal Cell Lines, Braunschweig, Germany
| | - Margarete Zaborski
- Leibniz-Institute DSMZ-German Collection of Microorganisms and Cell Cultures, Department of Human and Animal Cell Lines, Braunschweig, Germany
| | - Reiner Siebert
- Ulm University and Ulm University Medical Center, Institute of Human Genetics, Ulm, Germany
| | - Hilmar Quentmeier
- Leibniz-Institute DSMZ-German Collection of Microorganisms and Cell Cultures, Department of Human and Animal Cell Lines, Braunschweig, Germany
| |
Collapse
|
10
|
Oien DB, Sharma S, Hattersley MM, DuPont M, Criscione SW, Prickett L, Goeppert AU, Drew L, Yao Y, Zhang J, Chan HM. BET inhibition targets ABC-DLBCL constitutive B-cell receptor signaling through PAX5. Blood Adv 2023; 7:5108-5121. [PMID: 37184294 PMCID: PMC10477446 DOI: 10.1182/bloodadvances.2022009257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 04/06/2023] [Accepted: 05/04/2023] [Indexed: 05/16/2023] Open
Abstract
B-cell receptor (BCR) signaling is essential for the diffuse large B-cell lymphoma (DLBCL) subtype that originates from activated B-cells (ABCs). ABC-DLBCL cells are sensitive to Bruton tyrosine kinase intervention. However, patients with relapsed or refractory ABC-DLBCL had overall response rates from 33% to 37% for Bruton tyrosine kinase inhibitors, suggesting the evaluation of combination-based treatment for improved efficacy. We investigated the efficacy and mechanism of the bromodomain and extraterminal motif (BET) inhibitor AZD5153 combined with the Bruton tyrosine kinase inhibitor acalabrutinib in ABC-DLBCL preclinical models. AZD5153 is a bivalent BET inhibitor that simultaneously engages the 2 bromodomains of BRD4. Adding AZD5153 to acalabrutinib demonstrated combination benefits in ABC-DLBCL cell line and patient-derived xenograft models. Differential expression analyses revealed PAX5 transcriptional activity as a novel downstream effector of this drug combination. PAX5 is a transcription factor for BCR signaling genes and may be critical for perpetually active BCR signaling in ABC-DLBCL. Our analyses further indicated significant alterations in BCR, RELB/alternative NF-κB, and toll-like receptor/interferon signaling. Validation of these results mapped a positive-feedback signaling loop regulated by PAX5. We demonstrated that AZD5153 decreased PAX5 expression, whereas acalabrutinib disruption of BCR signaling inhibited PAX5 activation. Furthermore, several interferon levels were decreased by AZD5153 and acalabrutinib in tumors. Adding interferon-beta1 (IFNβ1) to cells treated with acalabrutinib partially rescued PAX5 activation. Our results demonstrate that AZD5153 enhances the efficacy of acalabrutinib through PAX5 and BCR mechanisms that are critical for ABC-DLBCL.
Collapse
Affiliation(s)
- Derek B. Oien
- Research and Early Development, Oncology R&D, AstraZeneca, Boston, MA
| | - Samanta Sharma
- Research and Early Development, Oncology R&D, AstraZeneca, Boston, MA
| | | | - Michelle DuPont
- Research and Early Development, Oncology R&D, AstraZeneca, Boston, MA
| | | | - Laura Prickett
- Research and Early Development, Oncology R&D, AstraZeneca, Boston, MA
| | - Anne U. Goeppert
- Research and Early Development, Oncology R&D, AstraZeneca, Boston, MA
| | - Lisa Drew
- Research and Early Development, Oncology R&D, AstraZeneca, Boston, MA
| | - Yi Yao
- Research and Early Development, Oncology R&D, AstraZeneca, Boston, MA
| | - Jingwen Zhang
- Research and Early Development, Oncology R&D, AstraZeneca, Boston, MA
| | - Ho Man Chan
- Research and Early Development, Oncology R&D, AstraZeneca, Boston, MA
| |
Collapse
|
11
|
Nagel S, Meyer C, Pommerenke C. Establishment of the lymphoid ETS-code reveals deregulated ETS genes in Hodgkin lymphoma. PLoS One 2023; 18:e0288031. [PMID: 37428779 DOI: 10.1371/journal.pone.0288031] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 06/16/2023] [Indexed: 07/12/2023] Open
Abstract
The human family of ETS transcription factors numbers 28 genes which control multiple aspects of development, notably the differentiation of blood and immune cells. Otherwise, aberrant expression of ETS genes is reportedly involved in forming leukemia and lymphoma. Here, we comprehensively mapped ETS gene activities in early hematopoiesis, lymphopoiesis and all mature types of lymphocytes using public datasets. We have termed the generated gene expression pattern lymphoid ETS-code. This code enabled identification of deregulated ETS genes in patients with lymphoid malignancies, revealing 12 aberrantly expressed members in Hodgkin lymphoma (HL). For one of these, ETS gene ETV3, expression in stem and progenitor cells in addition to that in developing and mature T-cells was mapped together with downregulation in B-cell differentiation. In contrast, subsets of HL patients aberrantly overexpressed ETV3, indicating oncogenic activity in this B-cell malignancy. Analysis of ETV3-overexpressing HL cell line SUP-HD1 demonstrated genomic duplication of the ETV3 locus at 1q23, GATA3 as mutual activator, and suppressed BMP-signalling as mutual downstream effect. Additional examination of the neighboring ETS genes ETS1 and FLI1 revealed physiological activities in B-cell development and aberrant downregulation in HL patient subsets. SUP-HD1 showed genomic loss on chromosome 11, del(11)(q22q25), targeting both ETS1 and FLI1, underlying their downregulation. Furthermore, in the same cell line we identified PBX1-mediated overexpression of RIOK2 which inhibited ETS1 and activated JAK2 expression. Collectively, we codified normal ETS gene activities in lymphopoiesis and identified oncogenic ETS members in HL.
Collapse
Affiliation(s)
- Stefan Nagel
- Department of Human and Animal Cell Lines, Leibniz-Institute DSMZ, German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| | - Corinna Meyer
- Department of Human and Animal Cell Lines, Leibniz-Institute DSMZ, German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| | - Claudia Pommerenke
- Department of Human and Animal Cell Lines, Leibniz-Institute DSMZ, German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| |
Collapse
|
12
|
Pottosin I, Olivas-Aguirre M, Dobrovinskaya O. In vitro simulation of the acute lymphoblastic leukemia niche: a critical view on the optimal approximation for drug testing. J Leukoc Biol 2023; 114:21-41. [PMID: 37039524 DOI: 10.1093/jleuko/qiad039] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 03/29/2023] [Accepted: 03/31/2023] [Indexed: 04/12/2023] Open
Abstract
Acute lymphoblastic leukemia with the worst prognosis is related to minimal residual disease. Minimal residual disease not only depends on the individual peculiarities of leukemic clones but also reflects the protective role of the acute lymphoblastic leukemia microenvironment. In this review, we discuss in detail cell-to-cell interactions in the 2 leukemic niches, more explored bone marrow and less studied extramedullary adipose tissue. A special emphasis is given to multiple ways of interactions of acute lymphoblastic leukemia cells with the bone marrow or extramedullary adipose tissue microenvironment, indicating observed differences in B- and T-cell-derived acute lymphoblastic leukemia behavior. This analysis argued for the usage of coculture systems for drug testing. Starting with a review of available sources and characteristics of acute lymphoblastic leukemia cells, mesenchymal stromal cells, endothelial cells, and adipocytes, we have then made an update of the available 2-dimensional and 3-dimensional systems, which bring together cellular elements, components of the extracellular matrix, or its imitation. We discussed the most complex available 3-dimensional systems like "leukemia-on-a-chip," which include either a prefabricated microfluidics platform or, alternatively, the microarchitecture, designed by using the 3-dimensional bioprinting technologies. From our analysis, it follows that for preclinical antileukemic drug testing, in most cases, intermediately complex in vitro cell systems are optimal, such as a "2.5-dimensional" coculture of acute lymphoblastic leukemia cells with niche cells (mesenchymal stromal cells, endothelial cells) plus matrix components or scaffold-free mesenchymal stromal cell organoids, populated by acute lymphoblastic leukemia cells. Due to emerging evidence for the correlation of obesity and poor prognosis, a coculture of adipocytes with acute lymphoblastic leukemia cells as a drug testing system is gaining shape.
Collapse
Affiliation(s)
- Igor Pottosin
- Laboratory of Immunobiology and Ionic Transport Regulation, University Center for Biomedical Research, University of Colima, Av. Enrique Arreola Silva 883, Guzmán City, Jalisco, 49000, Mexico
| | - Miguel Olivas-Aguirre
- Laboratory of Immunobiology and Ionic Transport Regulation, University Center for Biomedical Research, University of Colima, Av. Enrique Arreola Silva 883, Guzmán City, Jalisco, 49000, Mexico
- Division of Exact, Natural and Technological Sciences, South University Center (CUSUR), University of Guadalajara, Jalisco, Mexico
| | - Oxana Dobrovinskaya
- Laboratory of Immunobiology and Ionic Transport Regulation, University Center for Biomedical Research, University of Colima, Av. Enrique Arreola Silva 883, Guzmán City, Jalisco, 49000, Mexico
| |
Collapse
|
13
|
Vicente-Garcés C, Maynou J, Fernández G, Esperanza-Cebollada E, Torrebadell M, Català A, Rives S, Camós M, Vega-García N. Fusion InPipe, an integrative pipeline for gene fusion detection from RNA-seq data in acute pediatric leukemia. Front Mol Biosci 2023; 10:1141310. [PMID: 37363396 PMCID: PMC10288994 DOI: 10.3389/fmolb.2023.1141310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 05/30/2023] [Indexed: 06/28/2023] Open
Abstract
RNA sequencing (RNA-seq) is a reliable tool for detecting gene fusions in acute leukemia. Multiple bioinformatics pipelines have been developed to analyze RNA-seq data, but an agreed gold standard has not been established. This study aimed to compare the applicability of 5 fusion calling pipelines (Arriba, deFuse, CICERO, FusionCatcher, and STAR-Fusion), as well as to define and develop an integrative bioinformatics pipeline (Fusion InPipe) to detect clinically relevant gene fusions in acute pediatric leukemia. We analyzed RNA-seq data by each pipeline individually and by Fusion InPipe. Each algorithm individually called most of the fusions with similar sensitivity and precision. However, not all rearrangements were called, suggesting that choosing a single pipeline might cause missing important fusions. To improve this, we integrated the results of the five algorithms in just one pipeline, Fusion InPipe, comparing the output from the agreement of 5/5, 4/5, and 3/5 algorithms. The maximum sensitivity was achieved with the agreement of 3/5 algorithms, with a global sensitivity of 95%, achieving a 100% in patients' data. Furthermore, we showed the necessity of filtering steps to reduce the false positive detection rate. Here, we demonstrate that Fusion InPipe is an excellent tool for fusion detection in pediatric acute leukemia with the best performance when selecting those fusions called by at least 3/5 pipelines.
Collapse
Affiliation(s)
- Clara Vicente-Garcés
- Pediatric Cancer Center Barcelona (PCCB), Institut de Recerca Sant Joan de Déu, Leukemia and Pediatric Hematology Disorders, Developmental Tumors Biology Group, Esplugues de Llobregat, Spain
| | - Joan Maynou
- Hospital Sant Joan de Déu Barcelona, Genetics Medicine Section, Esplugues de Llobregat, Spain
- Institut de Recerca Hospital Sant Joan de Déu, Neurogenetics and Molecular Medicine, Esplugues de Llobregat, Spain
| | - Guerau Fernández
- Hospital Sant Joan de Déu Barcelona, Genetics Medicine Section, Esplugues de Llobregat, Spain
- Institut de Recerca Hospital Sant Joan de Déu, Neurogenetics and Molecular Medicine, Esplugues de Llobregat, Spain
| | - Elena Esperanza-Cebollada
- Pediatric Cancer Center Barcelona (PCCB), Institut de Recerca Sant Joan de Déu, Leukemia and Pediatric Hematology Disorders, Developmental Tumors Biology Group, Esplugues de Llobregat, Spain
| | - Montserrat Torrebadell
- Pediatric Cancer Center Barcelona (PCCB), Institut de Recerca Sant Joan de Déu, Leukemia and Pediatric Hematology Disorders, Developmental Tumors Biology Group, Esplugues de Llobregat, Spain
- Hospital Sant Joan de Déu Barcelona, Hematology Laboratory, Esplugues de Llobregat, Spain
- Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red De Enfermedades Raras (CIBERER), Madrid, Spain
| | - Albert Català
- Pediatric Cancer Center Barcelona (PCCB), Institut de Recerca Sant Joan de Déu, Leukemia and Pediatric Hematology Disorders, Developmental Tumors Biology Group, Esplugues de Llobregat, Spain
- Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red De Enfermedades Raras (CIBERER), Madrid, Spain
- Pediatric Cancer Center Barcelona (PCCB), Hospital Sant Joan De Déu Barcelona, Leukemia and Lymphoma Unit, Barcelona, Spain
| | - Susana Rives
- Pediatric Cancer Center Barcelona (PCCB), Institut de Recerca Sant Joan de Déu, Leukemia and Pediatric Hematology Disorders, Developmental Tumors Biology Group, Esplugues de Llobregat, Spain
- Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red De Enfermedades Raras (CIBERER), Madrid, Spain
- Pediatric Cancer Center Barcelona (PCCB), Hospital Sant Joan De Déu Barcelona, Leukemia and Lymphoma Unit, Barcelona, Spain
| | - Mireia Camós
- Pediatric Cancer Center Barcelona (PCCB), Institut de Recerca Sant Joan de Déu, Leukemia and Pediatric Hematology Disorders, Developmental Tumors Biology Group, Esplugues de Llobregat, Spain
- Hospital Sant Joan de Déu Barcelona, Hematology Laboratory, Esplugues de Llobregat, Spain
- Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red De Enfermedades Raras (CIBERER), Madrid, Spain
| | - Nerea Vega-García
- Pediatric Cancer Center Barcelona (PCCB), Institut de Recerca Sant Joan de Déu, Leukemia and Pediatric Hematology Disorders, Developmental Tumors Biology Group, Esplugues de Llobregat, Spain
- Hospital Sant Joan de Déu Barcelona, Hematology Laboratory, Esplugues de Llobregat, Spain
| |
Collapse
|
14
|
Jayawant E, Pack A, Clark H, Kennedy E, Ghodke A, Jones J, Pepper C, Pepper A, Mitchell S. NF-κB fingerprinting reveals heterogeneous NF-κB composition in diffuse large B-cell lymphoma. Front Oncol 2023; 13:1181660. [PMID: 37333821 PMCID: PMC10272839 DOI: 10.3389/fonc.2023.1181660] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 05/23/2023] [Indexed: 06/20/2023] Open
Abstract
Introduction Improving treatments for Diffuse Large B-Cell Lymphoma (DLBCL) is challenged by the vast heterogeneity of the disease. Nuclear factor-κB (NF-κB) is frequently aberrantly activated in DLBCL. Transcriptionally active NF-κB is a dimer containing either RelA, RelB or cRel, but the variability in the composition of NF-κB between and within DLBCL cell populations is not known. Results Here we describe a new flow cytometry-based analysis technique termed "NF-κB fingerprinting" and demonstrate its applicability to DLBCL cell lines, DLBCL core-needle biopsy samples, and healthy donor blood samples. We find each of these cell populations has a unique NF-κB fingerprint and that widely used cell-of-origin classifications are inadequate to capture NF-κB heterogeneity in DLBCL. Computational modeling predicts that RelA is a key determinant of response to microenvironmental stimuli, and we experimentally identify substantial variability in RelA between and within ABC-DLBCL cell lines. We find that when we incorporate NF-κB fingerprints and mutational information into computational models we can predict how heterogeneous DLBCL cell populations respond to microenvironmental stimuli, and we validate these predictions experimentally. Discussion Our results show that the composition of NF-κB is highly heterogeneous in DLBCL and predictive of how DLBCL cells will respond to microenvironmental stimuli. We find that commonly occurring mutations in the NF-κB signaling pathway reduce DLBCL's response to microenvironmental stimuli. NF-κB fingerprinting is a widely applicable analysis technique to quantify NF-κB heterogeneity in B cell malignancies that reveals functionally significant differences in NF-κB composition within and between cell populations.
Collapse
|
15
|
Vincken R, Ruiz-Saenz A. A co-culture model system to quantify antibody-dependent cellular cytotoxicity in human breast cancer cells using an engineered natural killer cell line. STAR Protoc 2023; 4:102224. [PMID: 37071532 PMCID: PMC10323021 DOI: 10.1016/j.xpro.2023.102224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/20/2023] [Accepted: 03/15/2023] [Indexed: 04/19/2023] Open
Abstract
Current protocols measure antibody-dependent cellular cytotoxicity (ADCC) in vitro using peripheral blood mononuclear cells (PBMCs), but isolation and variability among donors limit the viability and reproducibility of this approach. Here, we present a standardized co-culture model system to quantify ADCC on human breast cancer cells. We describe steps to engineer a natural killer cell line that stably expresses FCγRIIIa (CD16), required to mediate ADCC. We then detail the steps for the cancer-immune co-culture setup, followed by cytotoxicity measurement and analysis.
Collapse
Affiliation(s)
- Roos Vincken
- Department of Cell Biology, Erasmus University Medical Center Rotterdam, 3015 CN Rotterdam, the Netherlands.
| | - Ana Ruiz-Saenz
- Department of Cell Biology, Erasmus University Medical Center Rotterdam, 3015 CN Rotterdam, the Netherlands; Department of Medical Oncology, Erasmus University Medical Center Rotterdam, 3015 GD Rotterdam, the Netherlands.
| |
Collapse
|
16
|
Leserer S, Graf T, Franke M, Bogdanov R, Arrieta-Bolaños E, Buttkereit U, Leimkühler N, Fleischhauer K, Reinhardt HC, Beelen DW, Turki AT. Time series clustering of T cell subsets dissects heterogeneity in immune reconstitution and clinical outcomes among MUD-HCT patients receiving ATG or PTCy. Front Immunol 2023; 14:1082727. [PMID: 37020562 PMCID: PMC10067907 DOI: 10.3389/fimmu.2023.1082727] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 03/02/2023] [Indexed: 03/22/2023] Open
Abstract
IntroductionAnti-T-lymphocyte globulin (ATG) or post-transplant cyclophosphamide (PTCy) prevent graft-versus-host disease (GVHD) after hematopoietic cell transplantation (HCT), yet individual patients benefit differentially.MethodsGiven the sparse comparative data on the impact of cellular immune reconstitution in this setting, we studied flow cytometry and clinical outcomes in 339 recipients of 10/10 matched-unrelated donor (MUD) HCT using either ATG (n=304) or PTCy (n=35) for in vivo T cell manipulation along with a haploidentical PTCy control cohort (n=45). Longitudinal cellular immune reconstitution data were analyzed conventionally and with a data science approach using clustering with dynamic time warping to determine the similarity between time-series of T cell subsets.ResultsConsistent with published studies, no significant differences in clinical outcomes were observed at the cohort level between MUD-ATG and MUD-PTCy. However, cellular reconstitution revealed preferences for distinct T cell subpopulations associating with GVHD protection in each setting. Starting early after HCT, MUD-PTCy patients had higher regulatory T cell levels after HCT (p <0.0001), while MUD-ATG patients presented with higher levels of γδ T- or NKT cells (both p <0.0001). Time-series clustering further dissected the patient population’s heterogeneity revealing distinct immune reconstitution clusters. Importantly, it identified phenotypes that reproducibly associated with impaired clinical outcomes within the same in vivo T cell manipulation platform. Exemplarily, patients with lower activated- and αβ T cell counts had significantly higher NRM (p=0.032) and relapse rates (p =0.01).DiscussionThe improved understanding of the heterogeneity of cellular reconstitution in MUD patients with T cell manipulation both at the cohort and individual level may support clinicians in managing HCT complications.
Collapse
Affiliation(s)
- Saskia Leserer
- Department of Hematology and Stem Cell Transplantation, West-German Cancer Center, University Hospital Essen, Essen, Germany
- Computational Hematology Lab, Department of Hematology and Stem Cell Transplantation, West-German Cancer Center, University Hospital Essen, Essen, Germany
- Institute for Experimental Cellular Therapy, West-German Cancer Center, University Hospital Essen, Essen, Germany
| | - Theresa Graf
- Department of Hematology and Stem Cell Transplantation, West-German Cancer Center, University Hospital Essen, Essen, Germany
- Computational Hematology Lab, Department of Hematology and Stem Cell Transplantation, West-German Cancer Center, University Hospital Essen, Essen, Germany
| | - Martina Franke
- Department of Hematology and Stem Cell Transplantation, West-German Cancer Center, University Hospital Essen, Essen, Germany
| | - Rashit Bogdanov
- Department of Hematology and Stem Cell Transplantation, West-German Cancer Center, University Hospital Essen, Essen, Germany
- Computational Hematology Lab, Department of Hematology and Stem Cell Transplantation, West-German Cancer Center, University Hospital Essen, Essen, Germany
| | - Esteban Arrieta-Bolaños
- Institute for Experimental Cellular Therapy, West-German Cancer Center, University Hospital Essen, Essen, Germany
- German Cancer Consortium Deutsches Konsortium für Translationale Krebsforschung (DKTK), Partner site Essen/Düsseldorf, Essen, Germany
| | - Ulrike Buttkereit
- Department of Hematology and Stem Cell Transplantation, West-German Cancer Center, University Hospital Essen, Essen, Germany
| | - Nils Leimkühler
- Department of Hematology and Stem Cell Transplantation, West-German Cancer Center, University Hospital Essen, Essen, Germany
| | - Katharina Fleischhauer
- Institute for Experimental Cellular Therapy, West-German Cancer Center, University Hospital Essen, Essen, Germany
- German Cancer Consortium Deutsches Konsortium für Translationale Krebsforschung (DKTK), Partner site Essen/Düsseldorf, Essen, Germany
| | - Hans Christian Reinhardt
- Department of Hematology and Stem Cell Transplantation, West-German Cancer Center, University Hospital Essen, Essen, Germany
- Cancer Research Center Cologne Essen (CCCE), Essen, Germany
| | - Dietrich W. Beelen
- Department of Hematology and Stem Cell Transplantation, West-German Cancer Center, University Hospital Essen, Essen, Germany
| | - Amin T. Turki
- Department of Hematology and Stem Cell Transplantation, West-German Cancer Center, University Hospital Essen, Essen, Germany
- Computational Hematology Lab, Department of Hematology and Stem Cell Transplantation, West-German Cancer Center, University Hospital Essen, Essen, Germany
- Institute for Experimental Cellular Therapy, West-German Cancer Center, University Hospital Essen, Essen, Germany
- *Correspondence: Amin T. Turki,
| |
Collapse
|
17
|
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
|
18
|
Sher S, Whipp E, Walker J, Zhang P, Beaver L, Williams K, Orwick S, Ravikrishnan J, Walker B, Perry E, Gregory C, Purcell M, Pan A, Yan P, Alinari L, Johnson AJ, Frigault MM, Greer JM, Hamdy A, Izumi R, Mo X, Sampath D, Woyach J, Blachly J, Byrd JC, Lapalombella R. VIP152 is a selective CDK9 inhibitor with pre-clinical in vitro and in vivo efficacy in chronic lymphocytic leukemia. Leukemia 2023; 37:326-338. [PMID: 36376377 PMCID: PMC9898036 DOI: 10.1038/s41375-022-01758-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 10/25/2022] [Accepted: 11/02/2022] [Indexed: 11/16/2022]
Abstract
Chronic lymphocytic leukemia (CLL) is effectively treated with targeted therapies including Bruton tyrosine kinase inhibitors and BCL2 antagonists. When these become ineffective, treatment options are limited. Positive transcription elongation factor complex (P-TEFb), a heterodimeric protein complex composed of cyclin dependent kinase 9 (CDK9) and cyclin T1, functions to regulate short half-life transcripts by phosphorylation of RNA Polymerase II (POLII). These transcripts are frequently dysregulated in hematologic malignancies; however, therapies targeting inhibition of P-TEFb have not yet achieved approval for cancer treatment. VIP152 kinome profiling revealed CDK9 as the main enzyme inhibited at 100 nM, with over a 10-fold increase in potency compared with other inhibitors currently in development for this target. VIP152 induced cell death in CLL cell lines and primary patient samples. Transcriptome analysis revealed inhibition of RNA degradation through the AU-Rich Element (ARE) dysregulation. Mechanistically, VIP152 inhibits the assembly of P-TEFb onto the transcription machinery and disturbs binding partners. Finally, immune competent mice engrafted with CLL-like cells of Eµ-MTCP1 over-expressing mice and treated with VIP152 demonstrated reduced disease burden and improvement in overall survival compared to vehicle-treated mice. These data suggest that VIP152 is a highly selective inhibitor of CDK9 that represents an attractive new therapy for CLL.
Collapse
Affiliation(s)
- Steven Sher
- Division of Hematology, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Ethan Whipp
- Division of Hematology, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Janek Walker
- Division of Hematology, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Pu Zhang
- Division of Hematology, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Larry Beaver
- Division of Hematology, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Katie Williams
- Division of Hematology, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Shelley Orwick
- Division of Hematology, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Janani Ravikrishnan
- Division of Hematology, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Brandi Walker
- Division of Hematology, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Elizabeth Perry
- Division of Hematology, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Charles Gregory
- Division of Hematology, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Matthew Purcell
- Division of Hematology, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Alexander Pan
- Division of Hematology, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Pearlly Yan
- Division of Hematology, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Lapo Alinari
- Division of Hematology, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH, USA
| | | | | | | | | | | | - Xiaokui Mo
- Division of Hematology, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Deepa Sampath
- Department of Hematopoietic Biology & Malignancy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jennifer Woyach
- Division of Hematology, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH, USA
| | - James Blachly
- Division of Hematology, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH, USA
| | - John C Byrd
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| | - Rosa Lapalombella
- Division of Hematology, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH, USA.
| |
Collapse
|
19
|
Brophy S, Amet R, Foy-Stones H, Gardiner N, McElligott AM. Isolation and Cryopreservation of Mononuclear Cells from Peripheral Blood and Bone Marrow of Blood Cancer Patients. Methods Mol Biol 2023; 2645:179-187. [PMID: 37202619 DOI: 10.1007/978-1-0716-3056-3_10] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Peripheral blood and bone marrow aspirates are routinely obtained from blood cancer patients for diagnostic investigations and provide an accessible source of patient-specific cancer cells, as well as non-malignant cells, for research proposes. The simple and reproducible method presented here allows isolation of viable mononuclear cells, including malignant cells, from fresh peripheral blood or bone marrow aspirates using density gradient centrifugation. The cells obtained using the protocol described can be further purified for a variety of cellular, immunological, molecular, and functional assays. In addition, these cells can be cryopreserved and bio-banked for future research studies.
Collapse
Affiliation(s)
- Sarah Brophy
- The John Durkan Leukaemia Laboratory, Trinity Translational Medicine Institute, Trinity College and St James's Hospital, Dublin, Ireland
| | - Rebecca Amet
- The John Durkan Leukaemia Laboratory, Trinity Translational Medicine Institute, Trinity College and St James's Hospital, Dublin, Ireland
| | - Hayley Foy-Stones
- Cryobiology Laboratory Stem Cell Facility, St James's Hospital, Dublin, Ireland
| | - Nicola Gardiner
- Cryobiology Laboratory Stem Cell Facility, St James's Hospital, Dublin, Ireland
| | - Anthony M McElligott
- The John Durkan Leukaemia Laboratory, Trinity Translational Medicine Institute, Trinity College and St James's Hospital, Dublin, Ireland.
- Trinity St. James's Cancer Institute, Trinity College and St James's Hospital, Dublin, Ireland.
| |
Collapse
|
20
|
Welters C, Lammoglia Cobo MF, Stein CA, Hsu MT, Ben Hamza A, Penter L, Chen X, Buccitelli C, Popp O, Mertins P, Dietze K, Bullinger L, Moosmann A, Blanc E, Beule D, Gerbitz A, Strobel J, Hackstein H, Rahn HP, Dornmair K, Blankenstein T, Hansmann L. Immune Phenotypes and Target Antigens of Clonally Expanded Bone Marrow T Cells in Treatment-Naïve Multiple Myeloma. Cancer Immunol Res 2022; 10:1407-1419. [PMID: 36122410 PMCID: PMC9627264 DOI: 10.1158/2326-6066.cir-22-0434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/23/2022] [Accepted: 09/09/2022] [Indexed: 01/07/2023]
Abstract
Multiple myeloma is a hematologic malignancy of monoclonal plasma cells that accumulate in the bone marrow. Despite their clinical and pathophysiologic relevance, the roles of bone marrow-infiltrating T cells in treatment-naïve patients are incompletely understood. We investigated whether clonally expanded T cells (i) were detectable in multiple myeloma bone marrow, (ii) showed characteristic immune phenotypes, and (iii) whether dominant clones recognized antigens selectively presented on multiple myeloma cells. Single-cell index sorting and T-cell receptor (TCR) αβ sequencing of bone marrow T cells from 13 treatment-naïve patients showed dominant clonal expansion within CD8+ cytolytic effector compartments, and only a minority of expanded T-cell clones expressed the classic immune-checkpoint molecules PD-1, CTLA-4, or TIM-3. To identify their molecular targets, TCRs of 68 dominant bone marrow clones from five selected patients were reexpressed and incubated with multiple myeloma and non-multiple myeloma cells from corresponding patients. Only 1 of 68 TCRs recognized antigen presented on multiple myeloma cells. This TCR was HLA-C-restricted, self-peptide-specific and could be activated by multiple myeloma cells of multiple patients. The remaining dominant T-cell clones did not recognize multiple myeloma cells and were, in part, specific for antigens associated with chronic viral infections. In conclusion, we showed that dominant bone marrow T-cell clones in treatment-naïve patients rarely recognize antigens presented on multiple myeloma cells and exhibit low expression of classic immune-checkpoint molecules. Our data provide experimental context for experiences from clinical immune-checkpoint inhibition trials and will inform future T cell-dependent therapeutic strategies.
Collapse
Affiliation(s)
- Carlotta Welters
- Department of Hematology, Oncology, and Tumor Immunology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - María Fernanda Lammoglia Cobo
- Department of Hematology, Oncology, and Tumor Immunology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Christian Alexander Stein
- Department of Hematology, Oncology, and Tumor Immunology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Meng-Tung Hsu
- Molecular Immunology and Gene Therapy, Max-Delbrück-Center for Molecular Medicine (MDC) Berlin, Germany
| | - Amin Ben Hamza
- Department of Hematology, Oncology, and Tumor Immunology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Livius Penter
- Department of Hematology, Oncology, and Tumor Immunology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Xiaojing Chen
- Molecular Immunology and Gene Therapy, Max-Delbrück-Center for Molecular Medicine (MDC) Berlin, Germany
| | - Christopher Buccitelli
- Proteomics Platform, Max-Delbrück-Center for Molecular Medicine and Berlin Institute of Health, Berlin, Germany
| | - Oliver Popp
- Proteomics Platform, Max-Delbrück-Center for Molecular Medicine and Berlin Institute of Health, Berlin, Germany
| | - Philipp Mertins
- Proteomics Platform, Max-Delbrück-Center for Molecular Medicine and Berlin Institute of Health, Berlin, Germany
| | - Kerstin Dietze
- Department of Hematology, Oncology, and Tumor Immunology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Lars Bullinger
- Department of Hematology, Oncology, and Tumor Immunology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Andreas Moosmann
- Department of Medicine III, Klinikum der Universität München, Munich, Germany.,German Center for Infection Research (DZIF), Munich, Germany
| | - Eric Blanc
- Core Unit Bioinformatics, Berlin Institute of Health, Berlin, Germany
| | - Dieter Beule
- Core Unit Bioinformatics, Berlin Institute of Health, Berlin, Germany
| | - Armin Gerbitz
- Hans Messner Allogeneic Stem Cell Transplant Program, Princess Margaret Cancer Centre, Toronto, Canada
| | - Julian Strobel
- Department of Transfusion Medicine and Hemostaseology, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Holger Hackstein
- Department of Transfusion Medicine and Hemostaseology, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Hans-Peter Rahn
- Preparative Flow Cytometry, Max-Delbrück-Centrum für Molekulare Medizin, Berlin, Germany
| | - Klaus Dornmair
- Institute of Clinical Neuroimmunology, University Hospital and Biomedical Center, LMU Munich, Germany
| | - Thomas Blankenstein
- Molecular Immunology and Gene Therapy, Max-Delbrück-Center for Molecular Medicine (MDC) Berlin, Germany
| | - Leo Hansmann
- Department of Hematology, Oncology, and Tumor Immunology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Corresponding Author: Leo Hansmann, Charité–Universitätsmedizin Berlin (CVK), Department of Hematology, Oncology, and Tumor Immunology, Augustenburger Platz 1, 13353 Berlin, Germany. Phone: 49-(0)30-450-665238; Fax: 49-(0)30-450-553914; E-mail:
| |
Collapse
|
21
|
Jorge J, Magalhães N, Alves R, Lapa B, Gonçalves AC, Sarmento-Ribeiro AB. Antitumor Effect of Brusatol in Acute Lymphoblastic Leukemia Models Is Triggered by Reactive Oxygen Species Accumulation. Biomedicines 2022; 10:biomedicines10092207. [PMID: 36140308 PMCID: PMC9496058 DOI: 10.3390/biomedicines10092207] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 08/31/2022] [Accepted: 09/02/2022] [Indexed: 11/16/2022] Open
Abstract
Acute lymphoblastic leukemia (ALL) is one of the most common hematological malignancies at pediatric ages and is characterized by different chromosomal rearrangements and genetic abnormalities involved in the differentiation and proliferation of lymphoid precursor cells. Brusatol is a quassinoid plant extract extensively studied due to its antineoplastic effect through global protein synthesis and nuclear factor erythroid 2-related factor-2 (NRF2) signaling inhibition. NRF2 is the main regulator of cellular antioxidant response and reactive oxygen species (ROS), which plays an important role in oxidative stress regulation. This study aimed to evaluate the effect of brusatol in in vitro models of ALL. KOPN-8 (B-ALL), CEM (T-ALL), and MOLT-4 (T-ALL) cell lines were incubated with increasing concentrations of brusatol, and the metabolic activity was evaluated using the resazurin assay. Flow cytometry was used to evaluate cell death, cell cycle, mitochondrial membrane potential (Δψmit), and to measure ROS and reduced glutathione (GSH) levels. Our results show that brusatol promoted a decrease in metabolic activity in ALL cell lines in a time-, dose-, and cell-line-dependent manner. Brusatol induced a cytostatic effect by cell cycle arrest in G0/G1 in all cell lines; however, cell death mediated by apoptosis was only observed in T-ALL cells. Brusatol leads to an oxidative stress imbalance by the increase in ROS levels, namely, superoxide anion. Redox imbalance and cellular apoptosis induced by brusatol are highly modulated by mitochondria disruption as a decrease in mitochondrial membrane potential is detected. These data suggest that brusatol might represent a new therapeutic approach for acute lymphoblastic leukemia, particularly for ALL T-cell lineage.
Collapse
Affiliation(s)
- Joana Jorge
- Laboratory of Oncobiology and Hematology (LOH) and University Clinic of Hematology, Faculty of Medicine University of Coimbra (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR)—Group of Environmental Genetics of Oncobiology (CIMAGO), Faculty of Medicine University of Coimbra (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), 3004-504 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3000-061 Coimbra, Portugal
| | - Nisa Magalhães
- Laboratory of Oncobiology and Hematology (LOH) and University Clinic of Hematology, Faculty of Medicine University of Coimbra (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal
| | - Raquel Alves
- Laboratory of Oncobiology and Hematology (LOH) and University Clinic of Hematology, Faculty of Medicine University of Coimbra (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR)—Group of Environmental Genetics of Oncobiology (CIMAGO), Faculty of Medicine University of Coimbra (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), 3004-504 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3000-061 Coimbra, Portugal
| | - Beatriz Lapa
- Laboratory of Oncobiology and Hematology (LOH) and University Clinic of Hematology, Faculty of Medicine University of Coimbra (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR)—Group of Environmental Genetics of Oncobiology (CIMAGO), Faculty of Medicine University of Coimbra (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), 3004-504 Coimbra, Portugal
| | - Ana Cristina Gonçalves
- Laboratory of Oncobiology and Hematology (LOH) and University Clinic of Hematology, Faculty of Medicine University of Coimbra (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR)—Group of Environmental Genetics of Oncobiology (CIMAGO), Faculty of Medicine University of Coimbra (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), 3004-504 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3000-061 Coimbra, Portugal
- Correspondence: (A.C.G.); (A.B.S.-R.); Tel.: +351-239-480-024 (A.C.G.)
| | - Ana Bela Sarmento-Ribeiro
- Laboratory of Oncobiology and Hematology (LOH) and University Clinic of Hematology, Faculty of Medicine University of Coimbra (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR)—Group of Environmental Genetics of Oncobiology (CIMAGO), Faculty of Medicine University of Coimbra (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), 3004-504 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3000-061 Coimbra, Portugal
- Hematology Service, Centro Hospitalar e Universitário de Coimbra (CHUC), 3000-061 Coimbra, Portugal
- Correspondence: (A.C.G.); (A.B.S.-R.); Tel.: +351-239-480-024 (A.C.G.)
| |
Collapse
|
22
|
Paulmann C, Spallek R, Karpiuk O, Heider M, Schäffer I, Zecha J, Klaeger S, Walzik M, Öllinger R, Engleitner T, Wirth M, Keller U, Krönke J, Rudelius M, Kossatz S, Rad R, Kuster B, Bassermann F. The OTUD6B-LIN28B-MYC axis determines the proliferative state in multiple myeloma. EMBO J 2022; 41:e110871. [PMID: 36059274 PMCID: PMC9574752 DOI: 10.15252/embj.2022110871] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 07/27/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
Deubiquitylases (DUBs) are therapeutically amenable components of the ubiquitin machinery that stabilize substrate proteins. Their inhibition can destabilize oncoproteins that may otherwise be undruggable. Here, we screened for DUB vulnerabilities in multiple myeloma, an incurable malignancy with dependency on the ubiquitin proteasome system and identified OTUD6B as an oncogene that drives the G1/S‐transition. LIN28B, a suppressor of microRNA biogenesis, is specified as a bona fide cell cycle‐specific substrate of OTUD6B. Stabilization of LIN28B drives MYC expression at G1/S, which in turn allows for rapid S‐phase entry. Silencing OTUD6B or LIN28B inhibits multiple myeloma outgrowth in vivo and high OTUD6B expression evolves in patients that progress to symptomatic multiple myeloma and results in an adverse outcome of the disease. Thus, we link proteolytic ubiquitylation with post‐transcriptional regulation and nominate OTUD6B as a potential mediator of the MGUS‐multiple myeloma transition, a central regulator of MYC, and an actionable vulnerability in multiple myeloma and other tumors with an activated OTUD6B‐LIN28B axis.
Collapse
Affiliation(s)
- Carmen Paulmann
- Department of Medicine III, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany.,TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, Munich, Germany
| | - Ria Spallek
- Department of Medicine III, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany.,TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, Munich, Germany
| | - Oleksandra Karpiuk
- Department of Medicine III, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany.,TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, Munich, Germany
| | - Michael Heider
- Department of Medicine III, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany.,TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, Munich, Germany
| | - Isabell Schäffer
- Department of Medicine III, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany.,TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, Munich, Germany
| | - Jana Zecha
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany
| | - Susan Klaeger
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany
| | - Michaela Walzik
- Department of Medicine III, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany.,TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, Munich, Germany
| | - Rupert Öllinger
- TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, Munich, Germany.,Department of Medicine II, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany.,Institute of Molecular Oncology and Functional Genomics, Technical University of Munich, Munich, Germany
| | - Thomas Engleitner
- TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, Munich, Germany.,Department of Medicine II, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany.,Institute of Molecular Oncology and Functional Genomics, Technical University of Munich, Munich, Germany
| | - Matthias Wirth
- Department of Hematology, Oncology and Cancer Immunology, Campus Benjamin Franklin Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ulrich Keller
- Department of Hematology, Oncology and Cancer Immunology, Campus Benjamin Franklin Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,Deutsches Konsortium für Translationale Krebsforschung (DKTK), Heidelberg, Germany.,Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Jan Krönke
- Department of Hematology, Oncology and Cancer Immunology, Campus Benjamin Franklin Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,Deutsches Konsortium für Translationale Krebsforschung (DKTK), Heidelberg, Germany
| | - Martina Rudelius
- Institute of Pathology, Ludwigs Maximilians University, Munich, Germany
| | - Susanne Kossatz
- TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, Munich, Germany.,Department of Nuclear Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Roland Rad
- TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, Munich, Germany.,Department of Medicine II, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany.,Institute of Molecular Oncology and Functional Genomics, Technical University of Munich, Munich, Germany.,Deutsches Konsortium für Translationale Krebsforschung (DKTK), Heidelberg, Germany
| | - Bernhard Kuster
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany.,Deutsches Konsortium für Translationale Krebsforschung (DKTK), Heidelberg, Germany
| | - Florian Bassermann
- Department of Medicine III, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany.,TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, Munich, Germany.,Deutsches Konsortium für Translationale Krebsforschung (DKTK), Heidelberg, Germany
| |
Collapse
|
23
|
Nagel S, Pommerenke C, Quentmeier H, Meyer C, Kaufmann M, MacLeod RAF. Genomic Aberrations Generate Fusion Gene FOXK2::TP63 and Activate NFKB1 in Cutaneous T-Cell Lymphoma. Biomedicines 2022; 10:biomedicines10082038. [PMID: 36009586 PMCID: PMC9406051 DOI: 10.3390/biomedicines10082038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/16/2022] [Accepted: 08/19/2022] [Indexed: 11/21/2022] Open
Abstract
Cutaneous T-cell lymphoma (CTCL) is a severe lymphoid malignancy with a worse prognosis lacking curative treatment regimens. Several gene mutations and deregulated pathways, including NFkB signaling, have been implicated in its pathogenesis. Accordingly, CTCL cell line HUT-78 reportedly contains mutated NFKB2, which is constitutively activated via partial gene deletion, also demonstrating that genomic rearrangements cause driving mutations in this malignancy. Here, along with HUT-78, we analyzed CTCL cell line HH to identify additional aberrations underlying gene deregulation. Karyotyping and genomic profiling of HH showed several rearrangements worthy of detailed investigation. Corresponding to the established karyotype, RNA-seq data and PCR analysis confirmed the presence of t(3;17)(q28;q25), generating a novel fusion gene, FOXK2::TP63. Furthermore, chromosomal rearrangement t(1;4)(p32;q25) was connected to amplification at 4q24–26, affecting aberrant NFKB1 overexpression thereat. Transcription factor binding-site analysis and knockdown experiments demonstrated that IRF4 contributed to NFKB1 expression. Within the same amplicon, we identified amplification and overexpression of NFkB signaling activator CAMK2D (4q26) and p53-inhibitor UBE2D3 (4q24). Genomic profiling data for HUT-78 detailed a deletion at 10q25 underlying reported NFKB2 activation. Moreover, amplifications of ID1 (20q11) and IKZF2 (2q34) in this cell line drove overexpression of these NK cell differentiation factors and possibly thus formed corresponding lineage characteristics. Target gene analysis for NFKB1 via siRNA-mediated knockdown in HH revealed activation of TP63, MIR155, and NOTCH pathway component RBPJ. Finally, treatment of HH with NFkB inhibitor demonstrated a role for NFkB in supporting proliferation, while usage of inhibitor DAPT showed significant survival effects via the NOTCH pathway. Collectively, our data suggest that NFkB and/or NOTCH inhibitors may represent reasonable treatment options for subsets of CTCL patients.
Collapse
|
24
|
Koblitz J, Dirks WG, Eberth S, Nagel S, Steenpass L, Pommerenke C. DSMZCellDive: Diving into high-throughput cell line data. F1000Res 2022; 11:420. [PMID: 35949917 PMCID: PMC9334839 DOI: 10.12688/f1000research.111175.2] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/18/2022] [Indexed: 01/08/2023] Open
Abstract
Human and animal cell lines serve as model systems in a wide range of life sciences such as cancer and infection research or drug screening. Reproducible data are highly dependent on authenticated, contaminant-free cell lines, no better delivered than by the official and certified biorepositories. Offering a web portal to high-throughput information on these model systems will facilitate working with and comparing to these references by data otherwise dispersed at different sources. We here provide DSMZCellDive to access a comprehensive data source on human and animal cell lines, freely available at celldive.dsmz.de. A wide variety of data sources are generated such as RNA-seq transcriptome data and STR (short tandem repeats) profiles. Several starting points ease entering the database via browsing, searching or visualising. This web tool is designed for further expansion on meta and high-throughput data to be generated in future. Explicated examples for the power of this novel tool include analysis of B-cell differentiation markers, homeo-oncogene expression, and measurement of genomic loss of heterozygosities by an enlarged STR panel of 17 loci. Sharing the data on cell lines by the biorepository itself will be of benefit to the scientific community since it (1) supports the selection of appropriate model cell lines, (2) ensures reliability, (3) avoids misleading data, (4) saves on additional experimentals, and (5) serves as reference for genomic and gene expression data.
Collapse
Affiliation(s)
- Julia Koblitz
- Leibniz Institute DSMZ - German Collection of Microorganisms and Cell Cultures, Braunschweig, 38124, Germany
| | - Wilhelm G. Dirks
- Leibniz Institute DSMZ - German Collection of Microorganisms and Cell Cultures, Braunschweig, 38124, Germany
| | - Sonja Eberth
- Leibniz Institute DSMZ - German Collection of Microorganisms and Cell Cultures, Braunschweig, 38124, Germany
| | - Stefan Nagel
- Leibniz Institute DSMZ - German Collection of Microorganisms and Cell Cultures, Braunschweig, 38124, Germany
| | - Laura Steenpass
- Leibniz Institute DSMZ - German Collection of Microorganisms and Cell Cultures, Braunschweig, 38124, Germany
| | - Claudia Pommerenke
- Leibniz Institute DSMZ - German Collection of Microorganisms and Cell Cultures, Braunschweig, 38124, Germany
| |
Collapse
|
25
|
Smith AL, Eiken AP, Skupa SA, Moore DY, Umeta LT, Smith LM, Lyden ER, D’Angelo CR, Kallam A, Vose JM, Kutateladze TG, El-Gamal D. A Novel Triple-Action Inhibitor Targeting B-Cell Receptor Signaling and BRD4 Demonstrates Preclinical Activity in Chronic Lymphocytic Leukemia. Int J Mol Sci 2022; 23:6712. [PMID: 35743155 PMCID: PMC9224275 DOI: 10.3390/ijms23126712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/10/2022] [Accepted: 06/13/2022] [Indexed: 11/30/2022] Open
Abstract
B-cell chronic lymphocytic leukemia (CLL) results from intrinsic genetic defects and complex microenvironment stimuli that fuel CLL cell growth through an array of survival signaling pathways. Novel small-molecule agents targeting the B-cell receptor pathway and anti-apoptotic proteins alone or in combination have revolutionized the management of CLL, yet combination therapy carries significant toxicity and CLL remains incurable due to residual disease and relapse. Single-molecule inhibitors that can target multiple disease-driving factors are thus an attractive approach to combat both drug resistance and combination-therapy-related toxicities. We demonstrate that SRX3305, a novel small-molecule BTK/PI3K/BRD4 inhibitor that targets three distinctive facets of CLL biology, attenuates CLL cell proliferation and promotes apoptosis in a dose-dependent fashion. SRX3305 also inhibits the activation-induced proliferation of primary CLL cells in vitro and effectively blocks microenvironment-mediated survival signals, including stromal cell contact. Furthermore, SRX3305 blocks CLL cell migration toward CXCL-12 and CXCL-13, which are major chemokines involved in CLL cell homing and retention in microenvironment niches. Importantly, SRX3305 maintains its anti-tumor effects in ibrutinib-resistant CLL cells. Collectively, this study establishes the preclinical efficacy of SRX3305 in CLL, providing significant rationale for its development as a therapeutic agent for CLL and related disorders.
Collapse
Affiliation(s)
- Audrey L. Smith
- Eppley Institute for Research in Cancer and Allied Diseases, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA; (A.L.S.); (A.P.E.); (S.A.S.); (D.Y.M.); (L.T.U.)
| | - Alexandria P. Eiken
- Eppley Institute for Research in Cancer and Allied Diseases, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA; (A.L.S.); (A.P.E.); (S.A.S.); (D.Y.M.); (L.T.U.)
| | - Sydney A. Skupa
- Eppley Institute for Research in Cancer and Allied Diseases, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA; (A.L.S.); (A.P.E.); (S.A.S.); (D.Y.M.); (L.T.U.)
| | - Dalia Y. Moore
- Eppley Institute for Research in Cancer and Allied Diseases, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA; (A.L.S.); (A.P.E.); (S.A.S.); (D.Y.M.); (L.T.U.)
| | - Lelisse T. Umeta
- Eppley Institute for Research in Cancer and Allied Diseases, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA; (A.L.S.); (A.P.E.); (S.A.S.); (D.Y.M.); (L.T.U.)
| | - Lynette M. Smith
- Department of Biostatistics, College of Public Health, University of Nebraska Medical Center, Omaha, NE 68198, USA; (L.M.S.); (E.R.L.)
| | - Elizabeth R. Lyden
- Department of Biostatistics, College of Public Health, University of Nebraska Medical Center, Omaha, NE 68198, USA; (L.M.S.); (E.R.L.)
| | - Christopher R. D’Angelo
- Division of Hematology and Oncology, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA; (C.R.D.); (A.K.); (J.M.V.)
| | - Avyakta Kallam
- Division of Hematology and Oncology, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA; (C.R.D.); (A.K.); (J.M.V.)
| | - Julie M. Vose
- Division of Hematology and Oncology, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA; (C.R.D.); (A.K.); (J.M.V.)
| | - Tatiana G. Kutateladze
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA;
| | - Dalia El-Gamal
- Eppley Institute for Research in Cancer and Allied Diseases, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA; (A.L.S.); (A.P.E.); (S.A.S.); (D.Y.M.); (L.T.U.)
| |
Collapse
|
26
|
Bein J, Flinner N, Häupl B, Mathur A, Schneider O, Abu‐Ayyad M, Hansmann M, Piel M, Oellerich T, Hartmann S. T-cell-derived Hodgkin lymphoma has motility characteristics intermediate between Hodgkin and anaplastic large cell lymphoma. J Cell Mol Med 2022; 26:3495-3505. [PMID: 35586951 PMCID: PMC9189347 DOI: 10.1111/jcmm.17389] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/21/2022] [Accepted: 04/29/2022] [Indexed: 01/19/2023] Open
Abstract
Classic Hodgkin lymphoma (cHL) is usually characterized by a low tumour cell content, derived from crippled germinal centre B cells. Rare cases have been described in which the tumour cells show clonal T-cell receptor rearrangements. From a clinicopathological perspective, it is unclear if these cases should be classified as cHL or anaplastic large T-cell lymphoma (ALCL). Since we recently observed differences in the motility of ALCL and cHL tumour cells, here, we aimed to obtain a better understanding of T-cell-derived cHL by investigating their global proteomic profiles and their motility. In a proteomics analysis, when only motility-associated proteins were regarded, T-cell-derived cHL cell lines showed the highest similarity to ALK- ALCL cell lines. In contrast, T-cell-derived cHL cell lines presented a very low overall motility, similar to that observed in conventional cHL. Whereas all ALCL cell lines, as well as T-cell-derived cHL, predominantly presented an amoeboid migration pattern with uropod at the rear, conventional cHL never presented with uropods. The migration of ALCL cell lines was strongly impaired upon application of different inhibitors. This effect was less pronounced in cHL cell lines and almost invisible in T-cell-derived cHL. In summary, our cell line-derived data suggest that based on proteomics and migration behaviour, T-cell-derived cHL is a neoplasm that shares features with both cHL and ALCL and is not an ALCL with low tumour cell content. Complementary clinical studies on this lymphoma are warranted.
Collapse
Affiliation(s)
- Julia Bein
- Dr. Senckenberg Institute of PathologyGoethe University Frankfurt am MainFrankfurt am MainGermany
| | - Nadine Flinner
- Dr. Senckenberg Institute of PathologyGoethe University Frankfurt am MainFrankfurt am MainGermany
- Frankfurt Cancer InstituteGoethe UniversityFrankfurt am MainGermany
- University Cancer Center (UCT) FrankfurtUniversity HospitalGoethe UniversityFrankfurt am MainGermany
- Frankfurt Institute for Advanced StudiesFrankfurt am MainGermany
| | - Björn Häupl
- Frankfurt Cancer InstituteGoethe UniversityFrankfurt am MainGermany
- Department of Internal Medicine 2Goethe University HospitalFrankfurtGermany
- German Cancer Consortium/German Cancer Research CenterHeidelbergGermany
| | - Aastha Mathur
- Institut Curie and Institut Pierre Gilles de GennesPSL Research UniversityCNRSUMR 144ParisFrance
| | - Olga Schneider
- Dr. Senckenberg Institute of PathologyGoethe University Frankfurt am MainFrankfurt am MainGermany
| | - Marwa Abu‐Ayyad
- Dr. Senckenberg Institute of PathologyGoethe University Frankfurt am MainFrankfurt am MainGermany
| | - Martin‐Leo Hansmann
- Frankfurt Institute for Advanced StudiesFrankfurt am MainGermany
- Institute of General Pharmacology and ToxicologyGoethe University Frankfurt am MainFrankfurt am MainGermany
| | - Matthieu Piel
- Institut Curie and Institut Pierre Gilles de GennesPSL Research UniversityCNRSUMR 144ParisFrance
| | - Thomas Oellerich
- Frankfurt Cancer InstituteGoethe UniversityFrankfurt am MainGermany
- Department of Internal Medicine 2Goethe University HospitalFrankfurtGermany
- German Cancer Consortium/German Cancer Research CenterHeidelbergGermany
| | - Sylvia Hartmann
- Dr. Senckenberg Institute of PathologyGoethe University Frankfurt am MainFrankfurt am MainGermany
| |
Collapse
|
27
|
Teo WW, Cao X, Wu CS, Tan HK, Zhou Q, Gao C, Vanuytsel K, Kumar SS, Murphy GJ, Yang H, Chai L, Tenen DG. Non-coding RNA LEVER sequestration of PRC2 can mediate long range gene regulation. Commun Biol 2022; 5:343. [PMID: 35411071 PMCID: PMC9001699 DOI: 10.1038/s42003-022-03250-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 03/09/2022] [Indexed: 11/20/2022] Open
Abstract
Polycomb Repressive Complex 2 (PRC2) is an epigenetic regulator required for gene silencing during development. Although PRC2 is a well-established RNA-binding complex, the biological function of PRC2-RNA interaction has been controversial. Here, we study the gene-regulatory role of the inhibitory PRC2-RNA interactions. We report a nuclear long non-coding RNA, LEVER, which mapped 236 kb upstream of the β-globin cluster as confirmed by Nanopore sequencing. LEVER RNA interacts with PRC2 in its nascent form, and this prevents the accumulation of the H3K27 repressive histone marks within LEVER locus. Interestingly, the accessible LEVER chromatin, in turn, suppresses the chromatin interactions between the ε-globin locus and β-globin locus control region (LCR), resulting in a repressive effect on ε-globin gene expression. Our findings validate that the nascent RNA-PRC2 interaction inhibits local PRC2 function in situ. More importantly, we demonstrate that such a local process can in turn regulate the expression of neighboring genes. Identification of a long non-coding RNA LEVER, that inhibits the Polycomb Repressive Complex 2 (PRC2) and controls nearby embryonic form of beta-globin gene, provides additional evidence for PRC2-RNA functional interaction.
Collapse
Affiliation(s)
- Wei Wen Teo
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Xinang Cao
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.,Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Chan-Shuo Wu
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Hong Kee Tan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.,National University of Singapore, Graduate School for Integrative Sciences and Engineering, Singapore, Singapore
| | - Qiling Zhou
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Chong Gao
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Kim Vanuytsel
- Section of Hematology and Medical Oncology, School of Medicine, Boston University, Boston, MA, USA.,Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA, USA
| | - Sara S Kumar
- Section of Hematology and Medical Oncology, School of Medicine, Boston University, Boston, MA, USA.,Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA, USA
| | - George J Murphy
- Section of Hematology and Medical Oncology, School of Medicine, Boston University, Boston, MA, USA.,Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA, USA
| | - Henry Yang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Li Chai
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA.
| | - Daniel G Tenen
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore. .,Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA. .,Harvard Initiative for RNA Medicine, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
28
|
Sun W, Guo J, McClellan D, Poeschla A, Bareyan D, Casey MJ, Cairns BR, Tantin D, Engel ME. GFI1 Cooperates with IKZF1/IKAROS to Activate Gene Expression in T-cell Acute Lymphoblastic Leukemia. Mol Cancer Res 2022; 20:501-514. [PMID: 34980595 PMCID: PMC8983472 DOI: 10.1158/1541-7786.mcr-21-0352] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 10/04/2021] [Accepted: 12/21/2021] [Indexed: 11/16/2022]
Abstract
Growth factor independence-1 (GFI1) is a transcriptional repressor and master regulator of normal and malignant hematopoiesis. Repression by GFI1 is attributable to recruitment of LSD1-containing protein complexes via its SNAG domain. However, the full complement of GFI1 partners in transcriptional control is not known. We show that in T-acute lymphoblastic leukemia (ALL) cells, GFI1 and IKAROS are transcriptional partners that co-occupy regulatory regions of hallmark T-cell development genes. Transcriptional profiling reveals a subset of genes directly transactivated through the GFI1-IKAROS partnership. Among these is NOTCH3, a key factor in T-ALL pathogenesis. Surprisingly, NOTCH3 expression by GFI1 and IKAROS requires the GFI1 SNAG domain but occurs independent of SNAG-LSD1 binding. GFI1 variants deficient in LSD1 binding fail to activate NOTCH3, but conversely, small molecules that disrupt the SNAG-LSD1 interaction while leaving the SNAG primary structure intact stimulate NOTCH3 expression. These results identify a noncanonical transcriptional control mechanism in T-ALL which supports GFI1-mediated transactivation in partnership with IKAROS and suggest competition between LSD1-containing repressive complexes and others favoring transactivation. IMPLICATIONS Combinatorial diversity and cooperation between DNA binding proteins and complexes assembled by them can direct context-dependent transcriptional outputs to control cell fate and may offer new insights for therapeutic targeting in cancer.
Collapse
Affiliation(s)
- Wenxiang Sun
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Jingtao Guo
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
- Department of Oncological Sciences, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - David McClellan
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
- Department of Oncological Sciences, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Alexandra Poeschla
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Diana Bareyan
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Mattie J. Casey
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
- Department of Oncological Sciences, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Bradley R. Cairns
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
- Department of Oncological Sciences, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
- Howard Hughes Medical Institute, University of Utah School of Medicine, Salt Lake City, Utah
| | - Dean Tantin
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Michael E. Engel
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
- Department of Oncological Sciences, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
- Primary Children’s Hospital, Salt Lake City, UT 84112, USA
| |
Collapse
|
29
|
The Hematopoietic TALE-Code Shows Normal Activity of IRX1 in Myeloid Progenitors and Reveals Ectopic Expression of IRX3 and IRX5 in Acute Myeloid Leukemia. Int J Mol Sci 2022; 23:ijms23063192. [PMID: 35328612 PMCID: PMC8952210 DOI: 10.3390/ijms23063192] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/11/2022] [Accepted: 03/14/2022] [Indexed: 12/10/2022] Open
Abstract
Homeobox genes encode transcription factors that control basic developmental decisions. Knowledge of their hematopoietic activities casts light on normal and malignant immune cell development. Recently, we constructed the so-called lymphoid TALE-code that codifies expression patterns of all active TALE class homeobox genes in early hematopoiesis and lymphopoiesis. Here, we present the corresponding myeloid TALE-code to extend this gene signature, covering the entire hematopoietic system. The collective data showed expression patterns for eleven TALE homeobox genes and highlighted the exclusive expression of IRX1 in megakaryocyte-erythroid progenitors (MEPs), implicating this TALE class member in a specific myeloid differentiation process. Analysis of public profiling data from acute myeloid leukemia (AML) patients revealed aberrant activity of IRX1 in addition to IRX3 and IRX5, indicating an oncogenic role for these TALE homeobox genes when deregulated. Screening of RNA-seq data from 100 leukemia/lymphoma cell lines showed overexpression of IRX1, IRX3, and IRX5 in megakaryoblastic and myelomonocytic AML cell lines, chosen as suitable models for studying the regulation and function of these homeo-oncogenes. Genomic copy number analysis of IRX-positive cell lines demonstrated chromosomal amplification of the neighboring IRX3 and IRX5 genes at position 16q12 in MEGAL, underlying their overexpression in this cell line model. Comparative gene expression analysis of these cell lines revealed candidate upstream factors and target genes, namely the co-expression of GATA1 and GATA2 together with IRX1, and of BMP2 and HOXA10 with IRX3/IRX5. Subsequent knockdown and stimulation experiments in AML cell lines confirmed their activating impact in the corresponding IRX gene expression. Furthermore, we demonstrated that IRX1 activated KLF1 and TAL1, while IRX3 inhibited GATA1, GATA2, and FST. Accordingly, we propose that these regulatory relationships may represent major physiological and oncogenic activities of IRX factors in normal and malignant myeloid differentiation, respectively. Finally, the established myeloid TALE-code is a useful tool for evaluating TALE homeobox gene activities in AML.
Collapse
|
30
|
Farooq A, Trøen G, Delabie J, Wang J. Integrating whole genome sequencing, methylation, gene expression, topological associated domain information in regulatory mutation prediction: a study of follicular lymphoma. Comput Struct Biotechnol J 2022; 20:1726-1742. [PMID: 35495111 PMCID: PMC9024376 DOI: 10.1016/j.csbj.2022.03.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 03/22/2022] [Accepted: 03/22/2022] [Indexed: 11/13/2022] Open
Abstract
A major challenge in human genetics is of the analysis of the interplay between genetic and epigenetic factors in a multifactorial disease like cancer. Here, a novel methodology is proposed to investigate genome-wide regulatory mechanisms in cancer, as studied with the example of follicular Lymphoma (FL). In a first phase, a new machine-learning method is designed to identify Differentially Methylated Regions (DMRs) by computing six attributes. In a second phase, an integrative data analysis method is developed to study regulatory mutations in FL, by considering differential methylation information together with DNA sequence variation, differential gene expression, 3D organization of genome (e.g., topologically associated domains), and enriched biological pathways. Resulting mutation block-gene pairs are further ranked to find out the significant ones. By this approach, BCL2 and BCL6 were identified as top-ranking FL-related genes with several mutation blocks and DMRs acting on their regulatory regions. Two additional genes, CDCA4 and CTSO, were also found in top rank with significant DNA sequence variation and differential methylation in neighboring areas, pointing towards their potential use as biomarkers for FL. This work combines both genomic and epigenomic information to investigate genome-wide gene regulatory mechanisms in cancer and contribute to devising novel treatment strategies.
Collapse
|
31
|
Prajapati SC, Dunham N, Fan H, Garrett-Bakelman FE. Validation of CRISPR targeting for proliferation and cytarabine resistance control genes in the acute myeloid leukemia cell line MOLM-13. Biotechniques 2022; 72:81-84. [PMID: 35119307 PMCID: PMC9413368 DOI: 10.2144/btn-2021-0089] [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] [Indexed: 11/23/2022] Open
Abstract
Acute myeloid leukemia patients with FMS-like tyrosine kinase 3–internal tandem duplications and mixed lineage leukemia–protein AF9 fusion proteins suffer from poor clinical outcomes. The MOLM-13 acute myeloid leukemia cell line harbors both of these abnormalities and is used in CRISPR experiments to identify disease drivers. However, experimental observations may be biased or inconclusive in the absence of experimentally validated positive control genes. We validated sgRNAs for knockdown of TP53 for cell proliferation and for DCK knockdown and CDA upregulation for cytarabine resistance control genes in MOLM-13 cells. We have provided a detailed CRISPR protocol applicable to both gene knockdown or activation experiments and downstream leukemic phenotype analyses. Inclusion of these controls in CRISPR experiments will enhance the capacity to identify novel myeloid leukemia drivers in MOLM-13 cells. CRISPR-Cas9 knockdown and upregulation approaches were used for changing the expression of control genes TP53, DCK, and CDA in the acute myeloid leukemia cell line MOLM-13. Lentivirus transduced cells were selected (sorted for green fluorescent protein positive cells or selected by antibiotic treatment), and knockdown or upregulation of genes in the stable cells was confirmed by real-time quantitative PCR and western blot analyses. EdU incorporation assay was used to measure cell proliferation. Colorimetric proliferation/survival assay and flow cytometric cell counting were used to measure cell growth and survival. Results from the experimental and control groups were compared using Student's t-test.
Collapse
Affiliation(s)
- Subhash C Prajapati
- Department of Biochemistry & Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Nicholas Dunham
- Department of Biochemistry & Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Hao Fan
- Department of Biochemistry & Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Francine E Garrett-Bakelman
- Department of Biochemistry & Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA 22908, USA.,Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA 22908, USA.,University of Virginia Cancer Center, Charlottesville, VA 22903, USA
| |
Collapse
|
32
|
Inhibition of MCL1 induces apoptosis in anaplastic large cell lymphoma and in primary effusion lymphoma. Sci Rep 2022; 12:1085. [PMID: 35058488 PMCID: PMC8776734 DOI: 10.1038/s41598-022-04916-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 12/17/2021] [Indexed: 12/12/2022] Open
Abstract
AbstractOverexpression of antiapoptotic BCL2 family proteins occurs in various hematologic malignancies and contributes to tumorigenesis by inhibiting the apoptotic machinery of the cells. Antagonizing BH3 mimetics provide an option for medication, with venetoclax as the first drug applied for chronic lymphocytic leukemia and for acute myeloid leukemia. To find additional hematologic entities with ectopic expression of BCL2 family members, we performed expression screening of cell lines applying the LL-100 panel. Anaplastic large cell lymphoma (ALCL) and primary effusion lymphoma (PEL), 2/22 entities covered by this panel, stood out by high expression of MCL1 and low expression of BCL2. The MCL1 inhibitor AZD-5991 induced apoptosis in cell lines from both malignancies, suggesting that this BH3 mimetic might be efficient as drug for these diseases. The ALCL cell lines also expressed BCLXL and BCL2A1, both contributing to survival of the cells. The combination of specific BH3 mimetics yielded synergistic effects, pointing to a novel strategy for the treatment of ALCL. The PI3K/mTOR inhibitor BEZ-235 could also efficiently be applied in combination with AZD-5991, offering an alternative to avoid thrombocytopenia which is associated with the use of BCLXL inhibitors.
Collapse
|
33
|
Abstract
Most non-Hodgkin's lymphomas grow exclusively in the lymph node compartment protected by the tumor microenvironment. To better understand the cellular heterogeneity and the complex interaction between malignant and non-malignant cells, experiments with primary, patient-derived samples are often indispensable. Here, we describe a time-efficient but gentle protocol to process human lymph node samples. This protocol avoids enzymatic or mechanical stress and was optimized for the purpose of generating single-cell suspension suitable for delicate assays, such as single-cell RNA sequencing. For complete details on the use and execution of this protocol, please refer to Roider et al. (2020).
Collapse
Affiliation(s)
- Tobias Roider
- European Molecular Biology Laboratory (EMBL), 69120 Heidelberg, Germany.,Molecular Medicine Partnership Unit (MMPU), 69120 Heidelberg, Germany.,Department of Medicine V, Hematology, Oncology and Rheumatology, University of Heidelberg, 69120 Heidelberg, Germany
| | - Berit J Brinkmann
- European Molecular Biology Laboratory (EMBL), 69120 Heidelberg, Germany.,Molecular Medicine Partnership Unit (MMPU), 69120 Heidelberg, Germany.,Department of Medicine V, Hematology, Oncology and Rheumatology, University of Heidelberg, 69120 Heidelberg, Germany.,Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Sascha Dietrich
- European Molecular Biology Laboratory (EMBL), 69120 Heidelberg, Germany.,Molecular Medicine Partnership Unit (MMPU), 69120 Heidelberg, Germany.,Department of Medicine V, Hematology, Oncology and Rheumatology, University of Heidelberg, 69120 Heidelberg, Germany
| |
Collapse
|
34
|
Characterization of a novel glucocorticoid-resistant human B-cell acute lymphoblastic leukemia cell line, with AMPK, mTOR and fatty acid synthesis pathway inhibition. Cancer Cell Int 2021; 21:623. [PMID: 34823530 PMCID: PMC8614043 DOI: 10.1186/s12935-021-02335-7] [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: 07/22/2021] [Accepted: 11/11/2021] [Indexed: 02/08/2023] Open
Abstract
Background Acquired glucocorticoid (GC) resistance remains the main obstacle in acute lymphoblastic leukemia (ALL) therapy. The aim of the present study was to establish a novel GC-resistant B-ALL cell line and investigate its biological characteristics. Methods A cell culture technique was used to establish the GC-resistant cell line from the parental cell, NALM-6. Molecular and cellular biological techniques including flow cytometry, MTT assay, western blotting, DNA fingerprinting analysis and whole transcriptome sequencing (WTS) were used to characterize the GC-resistant cell lines. Nude mice were used for xenograft studies. Results The GC-resistant cell line, NALM-6/HDR, was established by culturing NALM-6 cells under hypoxia for 5 weeks with a single dexamethasone (Dex) treatment. We subcloned the NALM-6/HDR cell lines, and got 6 monoclone Dex-resistant cell lines, NALM-6/HDR-C1, C3, C4, C5, C6 and C9 with resistance index (RI) ranging from 20,000–50,000. NALM-6/HDR and its monoclone cell line, NALM-6/HDR-C5, exhibited moderate (RI 5–15) to high resistance (RI > 20) to Ara-c; low or no cross-resistance to L-Asp, VCR, DNR, and MTX (RI < 5). STR analysis confirmed that NALM-6/HDR and NALM-6/H were all derived from NALM-6. All these cells derived from NALM-6 showed similar morphology, growth curves, immunophenotype, chromosomal karyotype and tumorigenicity. WTS analysis revealed that the main metabolic differences between NALM-6 or NALM-6/H (GC-sensitive) and NALM-6/HDR (GC-resistant) were lipid and carbohydrates metabolism. Western blotting analysis showed that NALM-6/HDR cells had a low expression of GR and p-GR. Moreover, AMPK, mTORC1, glycolysis and de novo fatty acid synthesis (FAS) pathway were inhibited in NALM-6/HDR when compared with NALM-6. Conclusions NALM-6/HDR cell line may represent a subtype of B-ALL cells in patients who acquired GC and Ara-c resistance during the treatment. These patients may get little benefit from the available therapy target of AMPK, mTORC1, glycolysis and FAS pathway. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-021-02335-7.
Collapse
|
35
|
Nagel S, Meyer C. Establishment of the TBX-code reveals aberrantly activated T-box gene TBX3 in Hodgkin lymphoma. PLoS One 2021; 16:e0259674. [PMID: 34807923 PMCID: PMC8608327 DOI: 10.1371/journal.pone.0259674] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 10/22/2021] [Indexed: 11/23/2022] Open
Abstract
T-box genes encode transcription factors which control basic processes in development of several tissues including cell differentiation in the hematopoietic system. Here, we analyzed the physiological activities of all 17 human T-box genes in early hematopoiesis and in lymphopoiesis including developing and mature B-cells, T-cells, natural killer (NK)-cells and innate lymphoid cells. The resultant expression pattern comprised six genes, namely EOMES, MGA, TBX1, TBX10, TBX19 and TBX21. We termed this gene signature TBX-code which enables discrimination of normal and aberrant activities of T-box genes in lymphoid malignancies. Accordingly, expression analysis of T-box genes in Hodgkin lymphoma (HL) patients using a public profiling dataset revealed overexpression of EOMES, TBX1, TBX2, TBX3, TBX10, TBX19, TBX21 and TBXT while MGA showed aberrant downregulation. Analysis of T-cell acute lymphoid leukemia patients indicated aberrant overexpression of six T-box genes while no deregulated T-box genes were detected in anaplastic large cell lymphoma patients. As a paradigm we focused on TBX3 which was ectopically activated in about 6% of HL patients analyzed. Normally, TBX3 is expressed in tissues like lung, adrenal gland and retina but not in hematopoiesis. HL cell line KM-H2 expressed enhanced TBX3 levels and was used as an in vitro model to identify upstream regulators and downstream targets in this malignancy. Genomic studies of this cell line showed focal amplification of the TBX3 locus at 12q24 which may underlie its aberrant expression. In addition, promoter analysis and comparative expression profiling of HL cell lines followed by knockdown experiments revealed overexpressed transcription factors E2F4 and FOXC1 and chromatin modulator KDM2B as functional activators. Furthermore, we identified repressed target genes of TBX3 in HL including CDKN2A, NFKBIB and CD19, indicating its respective oncogenic function in proliferation, NFkB-signaling and B-cell differentiation. Taken together, we have revealed a lymphoid TBX-code and used it to identify an aberrant network around deregulated T-box gene TBX3 in HL which promotes hallmark aberrations of this disease. These findings provide a framework for future studies to evaluate deregulated T-box genes in lymphoid malignancies.
Collapse
Affiliation(s)
- Stefan Nagel
- Department of Human and Animal Cell Lines, Leibniz-Institute DSMZ–German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
- * E-mail:
| | - Corinna Meyer
- Department of Human and Animal Cell Lines, Leibniz-Institute DSMZ–German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| |
Collapse
|
36
|
Nagel S. The Role of NKL Homeobox Genes in T-Cell Malignancies. Biomedicines 2021; 9:biomedicines9111676. [PMID: 34829904 PMCID: PMC8615965 DOI: 10.3390/biomedicines9111676] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/08/2021] [Accepted: 11/10/2021] [Indexed: 11/16/2022] Open
Abstract
Homeobox genes encode transcription factors controlling basic developmental processes. The homeodomain is encoded by the homeobox and mediates sequence-specific DNA binding and interaction with cofactors, thus operating as a basic regulatory platform. Similarities in their homeobox sequences serve to arrange these genes in classes and subclasses, including NKL homeobox genes. In accordance with their normal functions, deregulated homeobox genes contribute to carcinogenesis along with hematopoietic malignancies. We have recently described the physiological expression of eleven NKL homeobox genes in the course of hematopoiesis and termed this gene expression pattern NKL-code. Due to the developmental impact of NKL homeobox genes these data suggest a key role for their activity in the normal regulation of hematopoietic cell differentiation including T-cells. On the other hand, aberrant overexpression of NKL-code members or ectopical activation of non-code members has been frequently reported in lymphoid and myeloid leukemia/lymphoma, demonstrating their oncogenic impact in the hematopoietic compartment. Here, we provide an overview of the NKL-code in normal hematopoiesis and discuss the oncogenic role of deregulated NKL homeobox genes in T-cell malignancies.
Collapse
Affiliation(s)
- Stefan Nagel
- Department of Human and Animal Cell Lines, Leibniz-Institute DSMZ, 38124 Braunschweig, Germany
| |
Collapse
|
37
|
NKL Homeobox Genes NKX2-3 and NKX2-4 Deregulate Megakaryocytic-Erythroid Cell Differentiation in AML. Int J Mol Sci 2021; 22:ijms222111434. [PMID: 34768865 PMCID: PMC8583893 DOI: 10.3390/ijms222111434] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/13/2021] [Accepted: 10/18/2021] [Indexed: 12/18/2022] Open
Abstract
NKL homeobox genes encode transcription factors that impact normal development and hematopoietic malignancies if deregulated. Recently, we established an NKL-code that describes the physiological expression pattern of eleven NKL homeobox genes in the course of hematopoiesis, allowing evaluation of aberrantly activated NKL genes in leukemia/lymphoma. Here, we identify ectopic expression of NKL homeobox gene NKX2-4 in an erythroblastic acute myeloid leukemia (AML) cell line OCI-M2 and describe investigation of its activating factors and target genes. Comparative expression profiling data of AML cell lines revealed in OCI-M2 an aberrantly activated program for endothelial development including master factor ETV2 and the additional endothelial signature genes HEY1, IRF6, and SOX7. Corresponding siRNA-mediated knockdown experiments showed their role in activating NKX2-4 expression. Furthermore, the ETV2 locus at 19p13 was genomically amplified, possibly underlying its aberrant expression. Target gene analyses of NKX2-4 revealed activated ETV2, HEY1, and SIX5 and suppressed FLI1. Comparative expression profiling analysis of public datasets for AML patients and primary megakaryocyte–erythroid progenitor cells showed conspicuous similarities to NKX2-4 activating factors and the target genes we identified, supporting the clinical relevance of our findings and developmental disturbance by NKX2-4. Finally, identification and target gene analysis of aberrantly expressed NKX2-3 in AML patients and a megakaryoblastic AML cell line ELF-153 showed activation of FLI1, contrasting with OCI-M2. FLI1 encodes a master factor for myelopoiesis, driving megakaryocytic differentiation and suppressing erythroid differentiation, thus representing a basic developmental target of these homeo-oncogenes. Taken together, we have identified aberrantly activated NKL homeobox genes NKX2-3 and NKX2-4 in AML, deregulating genes involved in megakaryocytic and erythroid differentiation processes, and thereby contributing to the formation of specific AML subtypes.
Collapse
|
38
|
Sidhu I, Barwe SP, Pillai RK, Gopalakrishnapillai A. Harnessing the Power of Induced Pluripotent Stem Cells and Gene Editing Technology: Therapeutic Implications in Hematological Malignancies. Cells 2021; 10:2698. [PMID: 34685678 PMCID: PMC8534597 DOI: 10.3390/cells10102698] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 10/06/2021] [Accepted: 10/07/2021] [Indexed: 12/16/2022] Open
Abstract
In vitro modeling of hematological malignancies not only provides insights into the influence of genetic aberrations on cellular and molecular mechanisms involved in disease progression but also aids development and evaluation of therapeutic agents. Owing to their self-renewal and differentiation capacity, induced pluripotent stem cells (iPSCs) have emerged as a potential source of short in supply disease-specific human cells of the hematopoietic lineage. Patient-derived iPSCs can recapitulate the disease severity and spectrum of prognosis dictated by the genetic variation among patients and can be used for drug screening and studying clonal evolution. However, this approach lacks the ability to model the early phases of the disease leading to cancer. The advent of genetic editing technology has promoted the generation of precise isogenic iPSC disease models to address questions regarding the underlying genetic mechanism of disease initiation and progression. In this review, we discuss the use of iPSC disease modeling in hematological diseases, where there is lack of patient sample availability and/or difficulty of engraftment to generate animal models. Furthermore, we describe the power of combining iPSC and precise gene editing to elucidate the underlying mechanism of initiation and progression of various hematological malignancies. Finally, we discuss the power of iPSC disease modeling in developing and testing novel therapies in a high throughput setting.
Collapse
Affiliation(s)
- Ishnoor Sidhu
- Nemours Centers for Childhood Cancer Research and Cancer & Blood Disorders, Nemours Children’s Health, Wilmington, DE 19803, USA; (I.S.); (S.P.B.)
- Department of Biological Sciences, University of Delaware, Newark, DE 19711, USA
| | - Sonali P. Barwe
- Nemours Centers for Childhood Cancer Research and Cancer & Blood Disorders, Nemours Children’s Health, Wilmington, DE 19803, USA; (I.S.); (S.P.B.)
- Department of Biological Sciences, University of Delaware, Newark, DE 19711, USA
| | - Raju K. Pillai
- National Medical Center, Department of Pathology, City of Hope, Duarte, CA 91105, USA;
| | - Anilkumar Gopalakrishnapillai
- Nemours Centers for Childhood Cancer Research and Cancer & Blood Disorders, Nemours Children’s Health, Wilmington, DE 19803, USA; (I.S.); (S.P.B.)
- Department of Biological Sciences, University of Delaware, Newark, DE 19711, USA
| |
Collapse
|
39
|
Siraki AG. The many roles of myeloperoxidase: From inflammation and immunity to biomarkers, drug metabolism and drug discovery. Redox Biol 2021; 46:102109. [PMID: 34455146 PMCID: PMC8403760 DOI: 10.1016/j.redox.2021.102109] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 08/12/2021] [Accepted: 08/16/2021] [Indexed: 12/11/2022] Open
Abstract
This review provides a practical guide to myeloperoxidase (MPO) and presents to the reader the diversity of its presence in biology. The review provides a historical background, from peroxidase activity to the discovery of MPO, to its role in disease and drug development. MPO is discussed in terms of its necessity, as specific individuals lack MPO expression. An underlying theme presented throughout brings up the question of the benefit and burden of MPO activity. Enzyme structure is discussed, including accurate masses and glycosylation sites. The catalytic cycle of MPO and its corresponding pathways are presented, with a discussion of the importance of the redox couples of the different states of MPO. Cell lines expressing MPO are discussed and practically summarized for the reader, and locations of MPO (primary and secondary) are provided. Useful methods of MPO detection are discussed, and how these can be used for studying disease processes are implied through the presentation of MPO as a biomarker. The presence of MPO in neutrophil extracellular traps is presented, and the activators of the former are provided. Lastly, the transition from drug metabolism to a target for drug development is where the review concludes.
Collapse
Affiliation(s)
- Arno G Siraki
- Faculty of Pharmacy & Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
40
|
Pommerenke C, Rand U, Uphoff CC, Nagel S, Zaborski M, Hauer V, Kaufmann M, Meyer C, Denkmann SA, Riese P, Eschke K, Kim Y, Safranko ZM, Kurolt IC, Markotic A, Cicin-Sain L, Steenpass L. Identification of cell lines CL-14, CL-40 and CAL-51 as suitable models for SARS-CoV-2 infection studies. PLoS One 2021; 16:e0255622. [PMID: 34339474 PMCID: PMC8328321 DOI: 10.1371/journal.pone.0255622] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 07/20/2021] [Indexed: 02/07/2023] Open
Abstract
The SARS-CoV-2 pandemic is a major global threat that sparked global research efforts. Pre-clinical and biochemical SARS-CoV-2 studies firstly rely on cell culture experiments where the importance of choosing an appropriate cell culture model is often underestimated. We here present a bottom-up approach to identify suitable permissive cancer cell lines for drug screening and virus research. Human cancer cell lines were screened for the SARS-CoV-2 cellular entry factors ACE2 and TMPRSS2 based on RNA-seq data of the Cancer Cell Line Encyclopedia (CCLE). However, experimentally testing permissiveness towards SARS-CoV-2 infection, we found limited correlation between receptor expression and permissiveness. This underlines that permissiveness of cells towards viral infection is determined not only by the presence of entry receptors but is defined by the availability of cellular resources, intrinsic immunity, and apoptosis. Aside from established cell culture infection models CACO-2 and CALU-3, three highly permissive human cell lines, colon cancer cell lines CL-14 and CL-40 and the breast cancer cell line CAL-51 and several low permissive cell lines were identified. Cell lines were characterised in more detail offering a broader choice of non-overexpression in vitro infection models to the scientific community. For some cell lines a truncated ACE2 mRNA and missense variants in TMPRSS2 might hint at disturbed host susceptibility towards viral entry.
Collapse
Affiliation(s)
- Claudia Pommerenke
- Department of Bioinformatics and Databases, Leibniz Institute DSMZ - German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
- * E-mail:
| | - Ulfert Rand
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Cord C. Uphoff
- Department of Human and Animal Cell Lines, Leibniz Institute DSMZ - German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| | - Stefan Nagel
- Department of Human and Animal Cell Lines, Leibniz Institute DSMZ - German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| | - Margarete Zaborski
- Department of Human and Animal Cell Lines, Leibniz Institute DSMZ - German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| | - Vivien Hauer
- Department of Human and Animal Cell Lines, Leibniz Institute DSMZ - German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| | - Maren Kaufmann
- Department of Human and Animal Cell Lines, Leibniz Institute DSMZ - German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| | - Corinna Meyer
- Department of Human and Animal Cell Lines, Leibniz Institute DSMZ - German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| | - Sabine A. Denkmann
- Department of Human and Animal Cell Lines, Leibniz Institute DSMZ - German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| | - Peggy Riese
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Kathrin Eschke
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Yeonsu Kim
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | | | | | - Alemka Markotic
- Dr. Fran Mihaljević University Hospital for Infectious Diseases, Zagreb, Croatia
| | - Luka Cicin-Sain
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Centre for Individualized Infection Medicine, a Joint Venture of Helmholtz Centre for Infection Research and Medical School Hannover, Hannover, Germany
| | - Laura Steenpass
- Department of Human and Animal Cell Lines, Leibniz Institute DSMZ - German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| |
Collapse
|
41
|
Sallustio F, Curci C, Solimando AG, Leone P, Pontrelli P, Gesualdo L, Vacca A, Racanelli V, Gallone A. Identification and monitoring of Copy Number Variants (CNV) in monoclonal gammopathy. Cancer Biol Ther 2021; 22:404-412. [PMID: 34288806 DOI: 10.1080/15384047.2021.1946458] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Monoclonal gammopathy of undetermined significance (MGUS) represents the pre-clinical stage of Multiple Myeloma (MM) with the 5% of MGUS progresses to MM. Although the progression from MGUS to MM has not been completely characterized, it is possible to monitor the DNA modifications of patients diagnosed with MGUS to detect early specific genomic abnormalities, including copy number variations (CNV). The CNVs of chromosome 1q and chromosome 13q are associated with a worse prognosis in MM.In the present study, we showed that it is possible to monitor the 1q21 gain and 13q deletion frequencies in gDNA using digital PCR. The CNV analysis of three cell lines with a well-characterized cytogenetic profile were compared with measures performed by a real-time PCR approach and with a digital PCR approach. Then, we analyzed CNVs in CD138+ plasma cells isolated from bone marrow of MGUS and MM patients.Our results show that digital PCR and targeted DNA monitoring represent a specific and accurate technique for the early detection of specific genomic abnormalities both in MM and in MGUS patients.Our results could represent a remarkable advancement in MM and MGUS diagnosis and in CNV analysis for the evaluation of the risk of progression from MGUS to MM.
Collapse
Affiliation(s)
- Fabio Sallustio
- Interdisciplinary Department of Medicine, University of Bari Aldo Moro, Bari, Italy
| | - Claudia Curci
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari Aldo Moro, Bari, Italy
| | - Antonio Giovanni Solimando
- Department of Biomedical Sciences and Human Oncology, Internal Medicine Unit "G. Baccelli", University of Bari Aldo Moro, Bari, Italy.,IRCCS Istituto Tumori Giovanni Paolo II of Bari, Italy
| | - Patrizia Leone
- Department of Biomedical Sciences and Human Oncology, Internal Medicine Unit "G. Baccelli", University of Bari Aldo Moro, Bari, Italy
| | - Paola Pontrelli
- Department of Emergency and Organ Transplantation, Nephrology, Dialysis and Transplantation Unit, University of Bari Aldo Moro, Bari, Italy
| | - Loreto Gesualdo
- Department of Emergency and Organ Transplantation, Nephrology, Dialysis and Transplantation Unit, University of Bari Aldo Moro, Bari, Italy
| | - Angelo Vacca
- Department of Biomedical Sciences and Human Oncology, Internal Medicine Unit "G. Baccelli", University of Bari Aldo Moro, Bari, Italy
| | - Vito Racanelli
- Department of Biomedical Sciences and Human Oncology, Internal Medicine Unit "G. Baccelli", University of Bari Aldo Moro, Bari, Italy
| | - Anna Gallone
- Interdisciplinary Department of Medicine, University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
42
|
An Overview on Diffuse Large B-Cell Lymphoma Models: Towards a Functional Genomics Approach. Cancers (Basel) 2021; 13:cancers13122893. [PMID: 34207773 PMCID: PMC8226720 DOI: 10.3390/cancers13122893] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/04/2021] [Accepted: 06/08/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Lymphoma research is a paradigm of integrating basic and applied research within the fields of molecular marker-based diagnosis and therapy. In recent years, major advances in next-generation sequencing have substantially improved the understanding of the genomics underlying diffuse large B-cell lymphoma (DLBCL), the most frequent type of B-cell lymphoma. This review addresses the various approaches that have helped unveil the biology and intricate alterations in this pathology, from cell lines to more sophisticated last-generation experimental models, such as organoids. We also provide an overview of the most recent findings in the field, their potential relevance for designing targeted therapies and the corresponding applicability to personalized medicine. Abstract Lymphoma research is a paradigm of the integration of basic and clinical research within the fields of diagnosis and therapy. Clinical, phenotypic, and genetic data are currently used to predict which patients could benefit from standard treatment. However, alternative therapies for patients at higher risk from refractoriness or relapse are usually empirically proposed, based on trial and error, without considering the genetic complexity of aggressive B-cell lymphomas. This is primarily due to the intricate mosaic of genetic and epigenetic alterations in lymphomas, which are an obstacle to the prediction of which drug will work for any given patient. Matching a patient’s genes to drug sensitivity by directly testing live tissues comprises the “precision medicine” concept. However, in the case of lymphomas, this concept should be expanded beyond genomics, eventually providing better treatment options for patients in need of alternative therapeutic approaches. We provide an overview of the most recent findings in diffuse large B-cell lymphomas genomics, from the classic functional models used to study tumor biology and the response to experimental treatments using cell lines and mouse models, to the most recent approaches with spheroid/organoid models. We also discuss their potential relevance and applicability to daily clinical practice.
Collapse
|
43
|
Nagel S, Pommerenke C, Meyer C, Drexler HG. NKL Homeobox Gene VENTX Is Part of a Regulatory Network in Human Conventional Dendritic Cells. Int J Mol Sci 2021; 22:ijms22115902. [PMID: 34072771 PMCID: PMC8198381 DOI: 10.3390/ijms22115902] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/10/2021] [Accepted: 05/27/2021] [Indexed: 01/09/2023] Open
Abstract
Recently, we documented a hematopoietic NKL-code mapping physiological expression patterns of NKL homeobox genes in human myelopoiesis including monocytes and their derived dendritic cells (DCs). Here, we enlarge this map to include normal NKL homeobox gene expressions in progenitor-derived DCs. Analysis of public gene expression profiling and RNA-seq datasets containing plasmacytoid and conventional dendritic cells (pDC and cDC) demonstrated HHEX activity in both entities while cDCs additionally expressed VENTX. The consequent aim of our study was to examine regulation and function of VENTX in DCs. We compared profiling data of VENTX-positive cDC and monocytes with VENTX-negative pDC and common myeloid progenitor entities and revealed several differentially expressed genes encoding transcription factors and pathway components, representing potential VENTX regulators. Screening of RNA-seq data for 100 leukemia/lymphoma cell lines identified prominent VENTX expression in an acute myelomonocytic leukemia cell line, MUTZ-3 containing inv(3)(q21q26) and t(12;22)(p13;q11) and representing a model for DC differentiation studies. Furthermore, extended gene analyses indicated that MUTZ-3 is associated with the subtype cDC2. In addition to analysis of public chromatin immune-precipitation data, subsequent knockdown experiments and modulations of signaling pathways in MUTZ-3 and control cell lines confirmed identified candidate transcription factors CEBPB, ETV6, EVI1, GATA2, IRF2, MN1, SPIB, and SPI1 and the CSF-, NOTCH-, and TNFa-pathways as VENTX regulators. Live-cell imaging analyses of MUTZ-3 cells treated for VENTX knockdown excluded impacts on apoptosis or induced alteration of differentiation-associated cell morphology. In contrast, target gene analysis performed by expression profiling of knockdown-treated MUTZ-3 cells revealed VENTX-mediated activation of several cDC-specific genes including CSFR1, EGR2, and MIR10A and inhibition of pDC-specific genes like RUNX2. Taken together, we added NKL homeobox gene activities for progenitor-derived DCs to the NKL-code, showing that VENTX is expressed in cDCs but not in pDCs and forms part of a cDC-specific gene regulatory network operating in DC differentiation and function.
Collapse
|
44
|
Drexler HG, Nagel S, Quentmeier H. Leukemia Cell Lines: In Vitro Models for the Study of Chronic Neutrophilic Leukemia. ACTA ACUST UNITED AC 2021; 28:1790-1794. [PMID: 34068566 PMCID: PMC8161829 DOI: 10.3390/curroncol28030166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 04/27/2021] [Accepted: 05/06/2021] [Indexed: 11/16/2022]
Abstract
Chronic neutrophilic leukemia (CNL) is a rare myeloproliferative neoplasm that is genetically characterized by the absence of both the Philadelphia chromosome and BCR-ABL1 fusion gene and the high prevalence of mutations in the colony-stimulating factor 3 receptor (CSF3R). Additional disease-modifying mutations have been recognized in CNL samples, portraying a distinct mutational landscape. Despite the growing knowledge base on genomic aberrations, further progress could be gained from the availability of representative models of CNL. To address this gap, we screened a large panel of available leukemia cell lines, followed by a detailed mutational investigation with focus on the CNL-associated candidate driver genes. The sister cell lines CNLBC-1 and MOLM-20 were derived from a patient with CNL and carry CNL-typical molecular hallmarks, namely mutations in several genes, such as CSF3R, ASXL1, EZH2, NRAS, and SETBP1. The use of these validated and comprehensively characterized models will benefit the understanding of the pathobiology of CNL and help inform therapeutic strategies.
Collapse
Affiliation(s)
- Hans G. Drexler
- Faculty of Life Sciences, Technical University of Braunschweig, 38106 Braunschweig, Germany
- Correspondence:
| | - Stefan Nagel
- Department of Human and Animal Cell Lines, Leibniz-Institute DSMZ-German Collection of Microorganisms and Cell Cultures, 38124 Braunschweig, Germany; (S.N.); (H.Q.)
| | - Hilmar Quentmeier
- Department of Human and Animal Cell Lines, Leibniz-Institute DSMZ-German Collection of Microorganisms and Cell Cultures, 38124 Braunschweig, Germany; (S.N.); (H.Q.)
| |
Collapse
|
45
|
Engel K, Wieland L, Krüger A, Volkmer I, Cynis H, Emmer A, Staege MS. Identification of Differentially Expressed Human Endogenous Retrovirus Families in Human Leukemia and Lymphoma Cell Lines and Stem Cells. Front Oncol 2021; 11:637981. [PMID: 33996550 PMCID: PMC8117144 DOI: 10.3389/fonc.2021.637981] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 04/13/2021] [Indexed: 12/29/2022] Open
Abstract
Endogenous retroviruses (ERVs) are becoming more and more relevant in cancer research and might be potential targets. The oncogenic potential of human ERVs (HERVs) has been recognized and includes immunosuppression, cell fusion, antigenicity of viral proteins, and regulation of neighboring genes. To decipher the role of HERVs in human cancers, we used a bioinformatics approach and analyzed RNA sequencing data from the LL-100 panel, covering 22 entities of hematopoietic neoplasias including T cell, B cell and myeloid malignancies. We compared HERV expression in this panel with hematopoietic stem cells (HSCs), embryonic stem cells (ESCs) and normal blood cells. RNA sequencing data were mapped against a comprehensive synthetic viral metagenome with 116 HERV sequences from 14 different HERV families. Of these, 13 HERV families and elements were differently expressed in malignant hematopoietic cells and stem cells. We found transcriptional upregulation of HERVE family in acute megakaryocytic and erythroid leukemia and of HERVFc family in multiple myeloma/plasma cell leukemia (PCL). The HERVFc member HERVFc-1 was found transcriptionally active in the multiple myeloma cell line OPM-2 and also in the Hodgkin lymphoma cell line L-428. The expression of HERVFc-1 in L-428 cells was validated by qRT-PCR. We also confirm transcriptional downregulation of ERV3 in acute megakaryocytic and erythroid leukemia, and HERVK in acute monocytic and myelocytic leukemia and a depression of HERVF in all malignant entities. Most of the higher expressed HERV families could be detected in stem cells including HERVK (HML-2), HERV-like, HERVV, HERVT, ERV9, HERVW, HERVF, HERVMER, ERV3, HERVH and HERVPABLB.
Collapse
Affiliation(s)
- Kristina Engel
- Department of Surgical and Conservative Pediatrics and Adolescent Medicine, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Lisa Wieland
- Department of Surgical and Conservative Pediatrics and Adolescent Medicine, Martin Luther University Halle-Wittenberg, Halle, Germany.,Department of Neurology, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Anna Krüger
- Department of Surgical and Conservative Pediatrics and Adolescent Medicine, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Ines Volkmer
- Department of Surgical and Conservative Pediatrics and Adolescent Medicine, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Holger Cynis
- Department of Drug Design and Target Validation, Fraunhofer Institute for Cell Therapy and Immunology, Halle, Germany
| | - Alexander Emmer
- Department of Neurology, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Martin S Staege
- Department of Surgical and Conservative Pediatrics and Adolescent Medicine, Martin Luther University Halle-Wittenberg, Halle, Germany
| |
Collapse
|
46
|
NKL-Code in Normal and Aberrant Hematopoiesis. Cancers (Basel) 2021; 13:cancers13081961. [PMID: 33921702 PMCID: PMC8073162 DOI: 10.3390/cancers13081961] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/13/2021] [Accepted: 04/15/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Gene codes represent expression patterns of closely related genes in particular tissues, organs or body parts. The NKL-code describes the activity of NKL homeobox genes in the hematopoietic system. NKL homeobox genes encode transcription factors controlling basic developmental processes. Therefore, aberrations of this code may contribute to deregulated hematopoiesis including leukemia and lymphoma. Normal and abnormal activities of NKL homeobox genes are described and mechanisms of (de)regulation, function, and diseases exemplified. Abstract We have recently described physiological expression patterns of NKL homeobox genes in early hematopoiesis and in subsequent lymphopoiesis and myelopoiesis, including terminally differentiated blood cells. We thereby systematized differential expression patterns of eleven such genes which form the so-called NKL-code. Due to the developmental impact of NKL homeobox genes, these data suggest a key role for their activity in normal hematopoietic differentiation processes. On the other hand, the aberrant overexpression of NKL-code-members or the ectopical activation of non-code members have been frequently reported in lymphoid and myeloid leukemia/lymphoma, revealing the oncogenic potential of these genes in the hematopoietic compartment. Here, I provide an overview of the NKL-code in normal hematopoiesis and instance mechanisms of deregulation and oncogenic functions of selected NKL genes in hematologic cancers. As well as published clinical studies, our conclusions are based on experimental work using hematopoietic cell lines which represent useful models to characterize the role of NKL homeobox genes in specific tumor types.
Collapse
|
47
|
Highly efficient CRISPR-Cas9-mediated gene knockout in primary human B cells for functional genetic studies of Epstein-Barr virus infection. PLoS Pathog 2021; 17:e1009117. [PMID: 33857265 PMCID: PMC8078793 DOI: 10.1371/journal.ppat.1009117] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 04/27/2021] [Accepted: 03/04/2021] [Indexed: 02/07/2023] Open
Abstract
Gene editing is now routine in all prokaryotic and metazoan cells but has not received much attention in immune cells when the CRISPR-Cas9 technology was introduced in the field of mammalian cell biology less than ten years ago. This versatile technology has been successfully adapted for gene modifications in human myeloid cells and T cells, among others, but applications to human primary B cells have been scarce and limited to activated B cells. This limitation has precluded conclusive studies into cell activation, differentiation or cell cycle control in this cell type. We report on highly efficient, simple and rapid genome engineering in primary resting human B cells using nucleofection of Cas9 ribonucleoprotein complexes, followed by EBV infection or culture on CD40 ligand feeder cells to drive in vitro B cell survival. We provide proof-of-principle of gene editing in quiescent human B cells using two model genes: CD46 and CDKN2A. The latter encodes the cell cycle regulator p16INK4a which is an important target of Epstein-Barr virus (EBV). Infection of B cells carrying a knockout of CDKN2A with wildtype and EBNA3 oncoprotein mutant strains of EBV allowed us to conclude that EBNA3C controls CDKN2A, the only barrier to B cell proliferation in EBV infected cells. Together, this approach enables efficient targeting of specific gene loci in quiescent human B cells supporting basic research as well as immunotherapeutic strategies. Human hematopoietic stem cells and their derivatives of the myeloid and lymphoid lineages are important targets for gene correction or modifications using the CRISPR-Cas9 technology. Among others, this approach can support site-specific insertion of chimeric antigen receptors (CARs) or T cell receptors (TCRs) into primary T cells. Their subsequent adoptive transfer to patient donors is a promising immunotherapeutic concept that may control chronic infection or certain types of cancer. Human B cells have a similar potential but, in contrast to T cells, they are very sensitive, difficult to handle, and short-lived ex vivo precluding their genetic modification. Here, we provide efficient means to manipulate primary human B cells genetically using in vitro assembled Cas9 ribonucleoprotein complexes. Subsequently, we used Epstein-Barr virus (EBV) infection to ensure the cells’ in vitro survival for long-term investigations. Our study demonstrates near-to-complete loss of a model target gene and provides examples to evaluate a cellular gene with a critical role during viral infection.
Collapse
|
48
|
Lara S, Anania JC, Virtanen A, Stenhammar V, Kleinau S. Importance of antibody isotypes in antitumor immunity by monocytes and complement using human-immune tumor models. Eur J Immunol 2021; 51:1218-1233. [PMID: 33533020 DOI: 10.1002/eji.202048885] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 12/31/2020] [Indexed: 02/01/2023]
Abstract
Monoclonal antibodies (mAbs) have revolutionized clinical medicine, especially in the field of cancer immunotherapy. The challenge now is to improve the response rates, as immunotherapy still fails for many patients. Strategies to enhance tumor cell death is a fundamental aim, but relevant model systems for human tumor immunology are lacking. Herein, we have developed a preclinical human immune - three-dimensional (3D) tumor model (spheroids) to map the efficiency of tumor-specific isotypes for improved tumor cell killing. Different anti-CD20 Rituximab (RTX) isotypes alone or in combination, were evaluated for mediating complement-dependent cytotoxicity and antibody-dependent phagocytosis by human monocytic cells in 3D spheroids, in parallel with monolayer cultures, of human CD20+ B-cell lymphomas. We demonstrate that the IgG3 variant of RTX has the greatest tumoricidal effect over other isotypes, and when combined with apoptosis-inducing RTX-IgG2 isotype the therapeutic effect can be substantially enhanced. The results show further that the treatment outcome by RTX isotypes is influenced by tumor morphology and expression of the complement inhibitor CD59. Hence, the human immune-3D tumor model is a clinical relevant and attractive ex vivo system to predict mAbs for best efficacy in cancer immunotherapy.
Collapse
Affiliation(s)
- Sandra Lara
- Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| | - Jessica C Anania
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden.,Center for Cancer Immunology, University of Southampton, Southampton, UK
| | - Alexander Virtanen
- Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| | - Viktoria Stenhammar
- Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| | - Sandra Kleinau
- Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
49
|
Nagel S, Pommerenke C, Meyer C, MacLeod RAF, Drexler HG. Establishment of the TALE-code reveals aberrantly activated homeobox gene PBX1 in Hodgkin lymphoma. PLoS One 2021; 16:e0246603. [PMID: 33539429 PMCID: PMC7861379 DOI: 10.1371/journal.pone.0246603] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 01/21/2021] [Indexed: 12/26/2022] Open
Abstract
Homeobox genes encode transcription factors which regulate basic processes in development and cell differentiation and are grouped into classes and subclasses according to sequence similarities. Here, we analyzed the activities of the 20 members strong TALE homeobox gene class in early hematopoiesis and in lymphopoiesis including developing and mature B-cells, T-cells, natural killer (NK)-cells and innate lymphoid cells (ILC). The resultant expression pattern comprised eleven genes and which we termed TALE-code enables discrimination of normal and aberrant activities of TALE homeobox genes in lymphoid malignancies. Subsequent expression analysis of TALE homeobox genes in public datasets of Hodgkin lymphoma (HL) patients revealed overexpression of IRX3, IRX4, MEIS1, MEIS3, PBX1, PBX4 and TGIF1. As paradigm we focused on PBX1 which was deregulated in about 17% HL patients. Normal PBX1 expression was restricted to hematopoietic stem cells and progenitors of T-cells and ILCs but absent in B-cells, reflecting its roles in stemness and early differentiation. HL cell line SUP-HD1 expressed enhanced PBX1 levels and served as an in vitro model to identify upstream regulators and downstream targets in this malignancy. Genomic studies of this cell line therein showed a gain of the PBX1 locus at 1q23 which may underlie its aberrant expression. Comparative expression profiling analyses of HL patients and cell lines followed by knockdown experiments revealed NFIB and TLX2 as target genes activated by PBX1. HOX proteins operate as cofactors of PBX1. Accordingly, our data showed that HOXB9 overexpressed in HL coactivated TLX2 but not NFIB while activating TNFRSF9 without PBX1. Further downstream analyses showed that TLX2 activated TBX15 which operated anti-apoptotically. Taken together, we discovered a lymphoid TALE-code and identified an aberrant network around deregulated TALE homeobox gene PBX1 which may disturb B-cell differentiation in HL by reactivation of progenitor-specific genes. These findings may provide the framework for future studies to exploit possible vulnerabilities of malignant cells in therapeutic scenarios.
Collapse
Affiliation(s)
- Stefan Nagel
- Department of Human and Animal Cell Lines, Leibniz-Institute DSMZ – German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| | - Claudia Pommerenke
- Department of Human and Animal Cell Lines, Leibniz-Institute DSMZ – German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| | - Corinna Meyer
- Department of Human and Animal Cell Lines, Leibniz-Institute DSMZ – German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| | - Roderick A. F. MacLeod
- Department of Human and Animal Cell Lines, Leibniz-Institute DSMZ – German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| | - Hans G. Drexler
- Department of Human and Animal Cell Lines, Leibniz-Institute DSMZ – German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| |
Collapse
|
50
|
Human leukocyte antigen class II quantification by targeted mass spectrometry in dendritic-like cell lines and monocyte-derived dendritic cells. Sci Rep 2021; 11:1028. [PMID: 33441579 PMCID: PMC7807004 DOI: 10.1038/s41598-020-77024-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 10/26/2020] [Indexed: 11/08/2022] Open
Abstract
The major histocompatibility complex II (HLA-II) facilitates the presentation of antigen-derived peptides to CD4+ T-cells. Antigen presentation is not only affected by peptide processing and intracellular trafficking, but also by mechanisms that govern HLA-II abundance such as gene expression, biosynthesis and degradation. Herein we describe a mass spectrometry (MS) based HLA-II-protein quantification method, applied to dendritic-like cells (KG-1 and MUTZ-3) and human monocyte-derived dendritic cells (DCs). This method monitors the proteotypic peptides VEHWGLDKPLLK, VEHWGLDQPLLK and VEHWGLDEPLLK, mapping to the α-chains HLA-DQA1, -DPA1 and -DRA1/DQA2, respectively. Total HLA-II was detected at 176 and 248 fmol per million unstimulated KG-1 and MUTZ-3 cells, respectively. In contrast, TNF- and LPS-induced MUTZ-3 cells showed a 50- and 200-fold increase, respectively, of total α-chain as measured by MS. HLA-II protein levels in unstimulated DCs varied significantly between donors ranging from ~ 4 to ~ 50 pmol per million DCs. Cell surface HLA-DR levels detected by flow cytometry increased 2- to 3-fold after DC activation with lipopolysaccharide (LPS), in contrast to a decrease or no change in total HLA α-chain as determined by MS. HLA-DRA1 was detected as the predominant variant, representing > 90% of total α-chain, followed by DPA1 and DQA1 at 3-7% and ≤ 1%, respectively.
Collapse
|