1
|
Manivannan MS, Yang X, Patel N, Peters A, Johnston JB, Gibson SB. Lysosome-Disrupting Agents in Combination with Venetoclax Increase Apoptotic Response in Primary Chronic Lymphocytic Leukemia (CLL) Cells Mediated by Lysosomal Cathepsin D Release and Inhibition of Autophagy. Cells 2024; 13:1041. [PMID: 38920669 PMCID: PMC11202145 DOI: 10.3390/cells13121041] [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: 03/18/2024] [Revised: 06/10/2024] [Accepted: 06/13/2024] [Indexed: 06/27/2024] Open
Abstract
Venetoclax and obinutuzumab are becoming frontline therapies for chronic lymphocytic leukemia (CLL) patients. Unfortunately, drug resistance still occurs, and the combination could be immunosuppressive. Lysosomes have previously been identified as a target for obinutuzumab cytotoxicity in CLL cells, but the mechanism remains unclear. In addition, studies have shown that lysosomotropic agents can cause synergistic cell death in vitro when combined with the BTK inhibitor, ibrutinib, in primary CLL cells. This indicates that targeting lysosomes could be a treatment strategy for CLL. In this study, we have shown that obinutuzumab induces lysosome membrane permeabilization (LMP) and cathepsin D release in CLL cells. Inhibition of cathepsins reduced obinutuzumab-induced cell death in CLL cells. We further determined that the lysosomotropic agent siramesine in combination with venetoclax increased cell death in primary CLL cells through an increase in reactive oxygen species (ROS) and cathepsin release. Siramesine treatment also induced synergistic cytotoxicity when combined with venetoclax. Microenvironmental factors IL4 and CD40L or incubation with HS-5 stromal cells failed to significantly protect CLL cells from siramesine- and venetoclax-induced apoptosis. We also found that siramesine treatment inhibited autophagy through reduced autolysosomes. Finally, the autophagy inhibitor chloroquine failed to further increase siramesine-induced cell death. Taken together, lysosome-targeting drugs could be an effective strategy in combination with venetoclax to overcome drug resistance in CLL.
Collapse
MESH Headings
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Sulfonamides/pharmacology
- Lysosomes/metabolism
- Lysosomes/drug effects
- Apoptosis/drug effects
- Autophagy/drug effects
- Cathepsin D/metabolism
- Reactive Oxygen Species/metabolism
- Drug Synergism
- Cell Line, Tumor
Collapse
Affiliation(s)
- Madhumita S. Manivannan
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2R3, Canada; (M.S.M.); (X.Y.); (N.P.); (A.P.)
| | - Xiaoyan Yang
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2R3, Canada; (M.S.M.); (X.Y.); (N.P.); (A.P.)
| | - Nirav Patel
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2R3, Canada; (M.S.M.); (X.Y.); (N.P.); (A.P.)
| | - Anthea Peters
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2R3, Canada; (M.S.M.); (X.Y.); (N.P.); (A.P.)
- Cross Cancer Institute, Alberta Health Services, Edmonton, AB T5J 3E4, Canada
| | - James B. Johnston
- CancerCare Manitoba Research Institute, Hematologist/Oncologist, CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada;
| | - Spencer B. Gibson
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2R3, Canada; (M.S.M.); (X.Y.); (N.P.); (A.P.)
- Department of Internal Medicine, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
- Department of Biochemistry and Medical Genetics, Faculty of Medicine, University of Manitoba, Winnipeg, MB R3T 2N2, USA
| |
Collapse
|
2
|
Mateos-Jaimez J, Mangolini M, Vidal A, Kulis M, Colomer D, Campo E, Ringshausen I, Martin-Subero JI, Maiques-Diaz A. Robust CRISPR-Cas9 Genetic Editing of Primary Chronic Lymphocytic Leukemia and Mantle Cell Lymphoma Cells. Hemasphere 2023; 7:e909. [PMID: 37304935 PMCID: PMC10249715 DOI: 10.1097/hs9.0000000000000909] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 05/03/2023] [Indexed: 06/13/2023] Open
Affiliation(s)
- Judith Mateos-Jaimez
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Maurizio Mangolini
- Department of Hematology and Cambridge Stem Cell Institute, University of Cambridge, United Kingdom
| | - Anna Vidal
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Marta Kulis
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Dolors Colomer
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Hematopathology Unit, Pathology Department, Hospital Clinic, Barcelona, Spain
- University of Barcelona, Spain
| | - Elias Campo
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Hematopathology Unit, Pathology Department, Hospital Clinic, Barcelona, Spain
- University of Barcelona, Spain
| | - Ingo Ringshausen
- Department of Hematology and Cambridge Stem Cell Institute, University of Cambridge, United Kingdom
| | - Jose I. Martin-Subero
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Alba Maiques-Diaz
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| |
Collapse
|
3
|
Mangolini M, Maiques-Diaz A, Charalampopoulou S, Gerhard-Hartmann E, Bloehdorn J, Moore A, Giachetti G, Lu J, Roamio Franklin VN, Chilamakuri CSR, Moutsopoulos I, Rosenwald A, Stilgenbauer S, Zenz T, Mohorianu I, D'Santos C, Deaglio S, Hodson DJ, Martin-Subero JI, Ringshausen I. Viral transduction of primary human lymphoma B cells reveals mechanisms of NOTCH-mediated immune escape. Nat Commun 2022; 13:6220. [PMID: 36266281 PMCID: PMC9585083 DOI: 10.1038/s41467-022-33739-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 09/30/2022] [Indexed: 12/24/2022] Open
Abstract
Hotspot mutations in the PEST-domain of NOTCH1 and NOTCH2 are recurrently identified in B cell malignancies. To address how NOTCH-mutations contribute to a dismal prognosis, we have generated isogenic primary human tumor cells from patients with Chronic Lymphocytic Leukemia (CLL) and Mantle Cell Lymphoma (MCL), differing only in their expression of the intracellular domain (ICD) of NOTCH1 or NOTCH2. Our data demonstrate that both NOTCH-paralogs facilitate immune-escape of malignant B cells by up-regulating PD-L1, partly dependent on autocrine interferon-γ signaling. In addition, NOTCH-activation causes silencing of the entire HLA-class II locus via epigenetic regulation of the transcriptional co-activator CIITA. Notably, while NOTCH1 and NOTCH2 govern similar transcriptional programs, disease-specific differences in their expression levels can favor paralog-specific selection. Importantly, NOTCH-ICD also strongly down-regulates the expression of CD19, possibly limiting the effectiveness of immune-therapies. These NOTCH-mediated immune escape mechanisms are associated with the expansion of exhausted CD8+ T cells in vivo.
Collapse
Affiliation(s)
- Maurizio Mangolini
- Wellcome/MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, CB2 0AW, UK
- Department of Haematology, University of Cambridge, Cambridge, CB2 0AH, UK
| | - Alba Maiques-Diaz
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | | | | | - Johannes Bloehdorn
- Department of Internal Medicine III, Division of CLL, Ulm University, Ulm, Germany
| | - Andrew Moore
- Wellcome/MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, CB2 0AW, UK
- Department of Haematology, University of Cambridge, Cambridge, CB2 0AH, UK
| | - Giorgia Giachetti
- Wellcome/MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, CB2 0AW, UK
- Department of Haematology, University of Cambridge, Cambridge, CB2 0AH, UK
| | - Junyan Lu
- European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
- Molecular Medicine Partnership Unit (MMPU), Heidelberg, Germany
| | | | | | - Ilias Moutsopoulos
- Wellcome/MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Andreas Rosenwald
- Pathologisches Institut Universität Würzburg, 97080, Würzburg, Germany
| | - Stephan Stilgenbauer
- Department of Internal Medicine III, Division of CLL, Ulm University, Ulm, Germany
| | - Thorsten Zenz
- Department of Medical Oncology and Hematology, University Hospital Zürich and University of Zürich, Zürich, Switzerland
- Molecular Therapy in Hematology and Oncology, National Center for Tumor Diseases and German Cancer, Research Centre, Heidelberg, Germany
| | - Irina Mohorianu
- Wellcome/MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Clive D'Santos
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Silvia Deaglio
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Daniel J Hodson
- Wellcome/MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, CB2 0AW, UK
- Department of Haematology, University of Cambridge, Cambridge, CB2 0AH, UK
| | - Jose I Martin-Subero
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Ingo Ringshausen
- Wellcome/MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, CB2 0AW, UK.
- Department of Haematology, University of Cambridge, Cambridge, CB2 0AH, UK.
| |
Collapse
|
4
|
Guo W, Liang D, Wang P, Yin L, Zhang H, Xing C, Huang Z, Wu Y, Li H, Cheng Z, Xiao X, Liu J, Wang Z, Peng H. HIF-PH Encoded by EGLN1 Is a Potential Therapeutic Target for Chronic Lymphocytic Leukemia. Pharmaceuticals (Basel) 2022; 15:ph15060734. [PMID: 35745653 PMCID: PMC9229586 DOI: 10.3390/ph15060734] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 05/28/2022] [Accepted: 06/08/2022] [Indexed: 01/27/2023] Open
Abstract
Owing to the recent emergence of drug resistance to Bruton's tyrosine kinase inhibitors (BTK) in chronic lymphocytic leukemia (CLL) treatment, it is crucial to identify alternative therapeutic targets. Therefore, we aimed to identify therapeutic options for CLL besides BTK. We identified that HIF1A expression was higher in CLL patients than in controls, which may suggest good prognosis. We used a lentiviral knockdown of EGLN1 (encoding hypoxia-inducible factor prolyl hydroxylase [HIF-PH]) and found that the growth of MEC-1 cells slowed in the knockdown group. Treatment of CLL cell lines MEC-1 and HG3 with the HIF-PH inhibitor molidustat showed that molidustat could induce apoptosis in a concentration-dependent manner in CLL cells and had low cytotoxicity at this concentration. CXCR4, HIF1A, SLC2AI, and VEGF, the downstream molecules of the HIF pathway, were upregulated after molidustat treatment. Western blotting results indicated that molidustat increased HIF1A expression in CLL cell lines and cells from CLL patients, and sequencing/quantitative PCR analysis demonstrated that the ribosome biogenesis pathway was inhibited in MEC-1 cells after molidustat treatment. We further identified synergistic cytotoxicity of molidustat in combination with ibrutinib on the MEC-1 and HG3 cell lines at certain concentrations. Therefore, molidustat is a potential therapeutic option for CLL.
Collapse
Affiliation(s)
- Wancheng Guo
- Department of Hematology, The Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, China; (W.G.); (P.W.); (L.Y.); (H.Z.); (C.X.); (Z.H.); (H.L.); (Z.C.); (X.X.); (J.L.)
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha 410011, China
- Xiangya School of Medicine, Central South University, Changsha 410013, China;
| | - Daomiao Liang
- The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People’s Hospital), Changsha 410005, China;
| | - Peilong Wang
- Department of Hematology, The Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, China; (W.G.); (P.W.); (L.Y.); (H.Z.); (C.X.); (Z.H.); (H.L.); (Z.C.); (X.X.); (J.L.)
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha 410011, China
| | - Le Yin
- Department of Hematology, The Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, China; (W.G.); (P.W.); (L.Y.); (H.Z.); (C.X.); (Z.H.); (H.L.); (Z.C.); (X.X.); (J.L.)
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha 410011, China
| | - Huifang Zhang
- Department of Hematology, The Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, China; (W.G.); (P.W.); (L.Y.); (H.Z.); (C.X.); (Z.H.); (H.L.); (Z.C.); (X.X.); (J.L.)
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha 410011, China
| | - Cheng Xing
- Department of Hematology, The Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, China; (W.G.); (P.W.); (L.Y.); (H.Z.); (C.X.); (Z.H.); (H.L.); (Z.C.); (X.X.); (J.L.)
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha 410011, China
| | - Zineng Huang
- Department of Hematology, The Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, China; (W.G.); (P.W.); (L.Y.); (H.Z.); (C.X.); (Z.H.); (H.L.); (Z.C.); (X.X.); (J.L.)
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha 410011, China
| | - Yinghua Wu
- Xiangya School of Medicine, Central South University, Changsha 410013, China;
| | - Heng Li
- Department of Hematology, The Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, China; (W.G.); (P.W.); (L.Y.); (H.Z.); (C.X.); (Z.H.); (H.L.); (Z.C.); (X.X.); (J.L.)
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha 410011, China
| | - Zhao Cheng
- Department of Hematology, The Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, China; (W.G.); (P.W.); (L.Y.); (H.Z.); (C.X.); (Z.H.); (H.L.); (Z.C.); (X.X.); (J.L.)
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha 410011, China
| | - Xiaojuan Xiao
- Department of Hematology, The Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, China; (W.G.); (P.W.); (L.Y.); (H.Z.); (C.X.); (Z.H.); (H.L.); (Z.C.); (X.X.); (J.L.)
| | - Jing Liu
- Department of Hematology, The Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, China; (W.G.); (P.W.); (L.Y.); (H.Z.); (C.X.); (Z.H.); (H.L.); (Z.C.); (X.X.); (J.L.)
| | - Zhihua Wang
- Department of Hematology, The Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, China; (W.G.); (P.W.); (L.Y.); (H.Z.); (C.X.); (Z.H.); (H.L.); (Z.C.); (X.X.); (J.L.)
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha 410011, China
- Correspondence: (Z.W.); (H.P.); Tel.: +86-155-7588-6164 (Z.W.); +86-731-8529-5296 (H.P.)
| | - Hongling Peng
- Department of Hematology, The Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, China; (W.G.); (P.W.); (L.Y.); (H.Z.); (C.X.); (Z.H.); (H.L.); (Z.C.); (X.X.); (J.L.)
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha 410011, China
- Correspondence: (Z.W.); (H.P.); Tel.: +86-155-7588-6164 (Z.W.); +86-731-8529-5296 (H.P.)
| |
Collapse
|
5
|
Benatti S, Atene CG, Fiorcari S, Mesini N, Martinelli S, Zucchini P, Bacchelli F, Maccaferri M, Debbia G, Potenza L, Rossi D, Vallisa D, Trentin L, Gaidano G, Luppi M, Marasca R, Maffei R. IRF4 L116R mutation promotes proliferation of chronic lymphocytic leukemia B cells inducing MYC. Hematol Oncol 2021; 39:707-711. [PMID: 34431535 DOI: 10.1002/hon.2915] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Stefania Benatti
- Division of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Claudio Giacinto Atene
- Division of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Stefania Fiorcari
- Division of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Nicolò Mesini
- Division of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Silvia Martinelli
- Division of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Patrizia Zucchini
- Division of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Francesca Bacchelli
- Division of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Monica Maccaferri
- Department of Oncology and Hematology, Hematology Division, Azienda Ospedaliero-Universitaria of Modena-Policlinico, Modena, Italy
| | - Giulia Debbia
- Division of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Leonardo Potenza
- Division of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Davide Rossi
- Hematology, Oncology Institute of Southern Switzerland and Institute of Oncology Research, Bellinzona, Switzerland
| | - Daniele Vallisa
- Division of Hematology, Guglielmo da Saliceto Hospital, Piacenza, Italy
| | - Livio Trentin
- Hematology and Clinical Immunology, Department of Medicine, University of Padua, Padua, Italy
| | - Gianluca Gaidano
- Division of Hematology, Department of Translational Medicine, University of Eastern Piedmont, Novara, Italy
| | - Mario Luppi
- Division of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Roberto Marasca
- Division of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Rossana Maffei
- Department of Oncology and Hematology, Hematology Division, Azienda Ospedaliero-Universitaria of Modena-Policlinico, Modena, Italy
| |
Collapse
|
6
|
Keim D, Gollner K, Gollner U, Jérôme V, Freitag R. Generation of Recombinant Primary Human B Lymphocytes Using Non-Viral Vectors. Int J Mol Sci 2021; 22:8239. [PMID: 34361005 PMCID: PMC8347318 DOI: 10.3390/ijms22158239] [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: 07/14/2021] [Revised: 07/29/2021] [Accepted: 07/29/2021] [Indexed: 12/26/2022] Open
Abstract
Although the development of gene delivery systems based on non-viral vectors is advancing, it remains a challenge to deliver plasmid DNA into human blood cells. The current "gold standard", namely linear polyethyleneimine (l-PEI 25 kDa), in particular, is unable to produce transgene expression levels >5% in primary human B lymphocytes. Here, it is demonstrated that a well-defined 24-armed poly(2-dimethylamino) ethyl methacrylate (PDMAEMA, 755 kDa) nano-star is able to reproducibly elicit high transgene expression (40%) at sufficient residual viability (69%) in primary human B cells derived from tonsillar tissue. Moreover, our results indicate that the length of the mitogenic stimulation prior to transfection is an important parameter that must be established during the development of the transfection protocol. In our hands, four days of stimulation with rhCD40L post-thawing led to the best transfection results in terms of TE and cell survival. Most importantly, our data argue for an impact of the B cell subsets on the transfection outcomes, underlining that the complexity and heterogeneity of a given B cell population pre- and post-transfection is a critical parameter to consider in the multiparametric approach required for the implementation of the transfection protocol.
Collapse
Affiliation(s)
- Daniel Keim
- Process Biotechnology, University of Bayreuth, 95447 Bayreuth, Germany; (D.K.); (V.J.)
| | - Katrin Gollner
- Praxis am Schießgraben, Schießgraben 21, 95326 Kulmbach, Germany; (K.G.); (U.G.)
| | - Ulrich Gollner
- Praxis am Schießgraben, Schießgraben 21, 95326 Kulmbach, Germany; (K.G.); (U.G.)
| | - Valérie Jérôme
- Process Biotechnology, University of Bayreuth, 95447 Bayreuth, Germany; (D.K.); (V.J.)
| | - Ruth Freitag
- Process Biotechnology, University of Bayreuth, 95447 Bayreuth, Germany; (D.K.); (V.J.)
| |
Collapse
|
7
|
IRF4 modulates the response to BCR activation in chronic lymphocytic leukemia regulating IKAROS and SYK. Leukemia 2021; 35:1330-1343. [PMID: 33623139 DOI: 10.1038/s41375-021-01178-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 01/18/2021] [Accepted: 02/01/2021] [Indexed: 01/31/2023]
Abstract
Interferon regulatory factor 4 (IRF4) is a transcriptional regulator of immune system development and function. Here, we investigated the role of IRF4 in controlling responsiveness to B-cell receptor (BCR) stimulation in chronic lymphocytic leukemia (CLL). We modulated IRF4 levels by transfecting CLL cells with an IRF4 vector or by silencing using small-interfering RNAs. Higher IRF4 levels attenuated BCR signaling by reducing AKT and ERK phosphorylation and calcium release. Conversely, IRF4 reduction improved the strength of the intracellular cascade activated by BCR engagement. Our results also indicated that IRF4 negatively regulates the expression of the spleen tyrosine kinase SYK, a crucial protein for propagation of BCR signaling, and the zinc finger DNA-binding protein IKAROS. We modulated IKAROS protein levels both by genetic manipulation and pharmacologically by treating CLL cells with lenalidomide and avadomide (IMIDs). IKAROS promoted BCR signaling by reducing the expression of inositol 5-phosphatase SHIP1. Lastly, IMIDs induced IRF4 expression, while down-regulating IKAROS and interfered with survival advantage mediated by BCR triggering, also in combination with ibrutinib. Overall, our findings elucidate the mechanism by which IRF4 tunes BCR signaling in CLL cells. Low IRF4 levels allow an efficient transmission of BCR signal throughout the accumulation of SYK and IKAROS.
Collapse
|
8
|
Palacios F, Yan XJ, Ferrer G, Chen SS, Vergani S, Yang X, Gardner J, Barrientos JC, Rock P, Burack R, Kolitz JE, Allen SL, Kharas MG, Abdel-Wahab O, Rai KR, Chiorazzi N. Musashi 2 influences chronic lymphocytic leukemia cell survival and growth making it a potential therapeutic target. Leukemia 2021; 35:1037-1052. [PMID: 33504942 PMCID: PMC8024198 DOI: 10.1038/s41375-020-01115-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 11/04/2020] [Accepted: 12/14/2020] [Indexed: 01/30/2023]
Abstract
Progression of chronic lymphocytic leukemia (CLL) results from the expansion of a small fraction of proliferating leukemic B cells. When comparing the global gene expression of recently divided CLL cells with that of previously divided cells, we found higher levels of genes involved in regulating gene expression. One of these was the oncogene Musashi 2 (MSI2), an RNA-binding protein that induces or represses translation. While there is an established role for MSI2 in normal and malignant stem cells, much less is known about its expression and role in CLL. Here we report for the first time ex vivo and in vitro experiments that MSI2 protein levels are higher in dividing and recently divided leukemic cells and that downregulating MSI2 expression or blocking its function eliminates primary human and murine CLL and mature myeloid cells. Notably, mature T cells and hematopoietic stem and progenitor cells are not affected. We also confirm that higher MSI2 levels correlate with poor outcome markers, shorter time-to-first-treatment, and overall survival. Thus, our data highlight an important role for MSI2 in CLL-cell survival and proliferation and associate MSI2 with poor prognosis in CLL patients. Collectively, these findings pinpoint MSI2 as a potentially valuable therapeutic target in CLL.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents
- Apoptosis/drug effects
- Biomarkers, Tumor
- Caspase 3/metabolism
- Cell Cycle Checkpoints/drug effects
- Cell Line, Tumor
- Cell Survival/genetics
- Cyclin-Dependent Kinase Inhibitor p27/metabolism
- Disease Models, Animal
- Gene Expression
- Gene Expression Profiling
- Gene Expression Regulation, Leukemic
- Gene Knockdown Techniques
- Humans
- Immunophenotyping
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/mortality
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Mice
- Molecular Targeted Therapy
- Prognosis
- RNA, Small Interfering
- RNA-Binding Proteins/genetics
- RNA-Binding Proteins/metabolism
- Tumor Suppressor Protein p53/metabolism
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Florencia Palacios
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Xiao-Jie Yan
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Gerardo Ferrer
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Shih-Shih Chen
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Stefano Vergani
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Xuejing Yang
- Molecular Pharmacology Program, Center for Cell Engineering, Center for Stem Cell Biology, Center for Experimental Therapeutics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jeffrey Gardner
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jaqueline C Barrientos
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
- Department of Medicine, Northwell Health, Manhasset and New Hyde Park, New York, NY, USA
- Department of Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Philip Rock
- Department of Pathology, University of Rochester, Rochester, NY, USA
| | - Richard Burack
- Department of Pathology, University of Rochester, Rochester, NY, USA
| | - Jonathan E Kolitz
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
- Department of Medicine, Northwell Health, Manhasset and New Hyde Park, New York, NY, USA
- Department of Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Steven L Allen
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
- Department of Medicine, Northwell Health, Manhasset and New Hyde Park, New York, NY, USA
- Department of Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Michael G Kharas
- Molecular Pharmacology Program, Center for Cell Engineering, Center for Stem Cell Biology, Center for Experimental Therapeutics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Omar Abdel-Wahab
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kanti R Rai
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
- Department of Medicine, Northwell Health, Manhasset and New Hyde Park, New York, NY, USA
- Department of Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Nicholas Chiorazzi
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA.
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Medicine, Northwell Health, Manhasset and New Hyde Park, New York, NY, USA.
| |
Collapse
|
9
|
Resveratrol Enhances mRNA and siRNA Lipid Nanoparticles Primary CLL Cell Transfection. Pharmaceutics 2020; 12:pharmaceutics12060520. [PMID: 32517377 PMCID: PMC7355647 DOI: 10.3390/pharmaceutics12060520] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 05/23/2020] [Accepted: 06/05/2020] [Indexed: 11/20/2022] Open
Abstract
Chronic lymphocytic leukemia (CLL) is the most common adult leukemia in Western populations. Therapies such as mRNA and siRNA encapsulated in lipid nanoparticles (LNPs) represent a clinically advanced platform and are utilized for a wide variety of applications. Unfortunately, transfection of RNA into CLL cells remains a formidable challenge and a bottleneck for developing targeted therapies for this disease. Therefore, we aimed to elucidate the barriers to efficient transfection of RNA-encapsulated LNPs into primary CLL cells to advance therapies in the future. To this end, we transfected primary CLL patient samples with mRNA and siRNA payloads encapsulated in an FDA-approved LNP formulation and characterized the transfection. Additionally, we tested the potential of repurposing caffeic acid, curcumin and resveratrol to enhance the transfection of nucleic acids into CLL cells. The results demonstrate that the rapid uptake of LNPs is required for successful transfection. Furthermore, we demonstrate that resveratrol enhances the delivery of both mRNA and siRNA encapsulated in LNPs into primary CLL patient samples, overcoming inter-patient heterogeneity. This study points out the important challenges to consider for efficient RNA therapeutics for CLL patients and advocates the use of resveratrol in combination with RNA lipid nanoparticles to enhance delivery into CLL cells.
Collapse
|
10
|
Huang RS, Shih HA, Lai MC, Chang YJ, Lin S. Enhanced NK-92 Cytotoxicity by CRISPR Genome Engineering Using Cas9 Ribonucleoproteins. Front Immunol 2020; 11:1008. [PMID: 32528479 PMCID: PMC7256201 DOI: 10.3389/fimmu.2020.01008] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 04/28/2020] [Indexed: 12/26/2022] Open
Abstract
Natural killer (NK) cells are an attractive cell-type for adoptive immunotherapy, but challenges in preparation of therapeutic primary NK cells restrict patient accessibility to NK cell immunotherapy. NK-92 is a well-characterized human NK cell line that has demonstrated promising anti-cancer activities in clinical trials. Unlimited proliferation of NK-92 cells provides a consistent supply of cells for the administration and development of NK cell immunotherapy. However, the clinical efficacy of NK-92 cells has not reached its full potential due to reduced immune functions as compared to primary NK cells. Improvements of NK-92 functions currently rely on conventional transgene delivery by mRNA, plasmid and viral vector with limited efficiencies. To enable precise genetic modifications, we have established a robust CRISPR genome engineering platform for NK-92 based on the nucleofection of Cas9 ribonucleoprotein. To demonstrate the versatility of the platform, we have performed cell-based screening of Cas9 guide RNA, multiplex gene knockout of activating and inhibitory receptors, knock-in of a fluorescent gene, and promoter insertion to reactivate endogenous CD16 and DNAM-1. The CRISPR-engineered NK-92 demonstrated markedly enhanced cytotoxicity and could mediate antibody-dependent cellular cytotoxicity against hard to kill cancer cell lines. Our genome editing platform is straightforward and robust for both functional studies and therapeutic engineering of NK-92 cells.
Collapse
MESH Headings
- Antibody-Dependent Cell Cytotoxicity
- Antigens, Differentiation, T-Lymphocyte/genetics
- Antigens, Differentiation, T-Lymphocyte/metabolism
- CRISPR-Associated Protein 9/genetics
- CRISPR-Associated Protein 9/metabolism
- CRISPR-Cas Systems
- Cell Survival
- Clustered Regularly Interspaced Short Palindromic Repeats
- GPI-Linked Proteins/genetics
- GPI-Linked Proteins/metabolism
- Gene Expression Regulation
- Gene Targeting
- HEK293 Cells
- HeLa Cells
- Humans
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Neoplasms/immunology
- Neoplasms/metabolism
- Neoplasms/pathology
- Neoplasms/therapy
- RNA, Guide, CRISPR-Cas Systems/genetics
- RNA, Guide, CRISPR-Cas Systems/metabolism
- Receptors, IgG/genetics
- Receptors, IgG/metabolism
Collapse
Affiliation(s)
- Rih-Sheng Huang
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Hsin-An Shih
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
- Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| | - Min-Chi Lai
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
- Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| | - Yao-Jen Chang
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Steven Lin
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
- Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
11
|
Man T, Zhu X, Chow YT, Dawson ER, Wen X, Patananan AN, Liu TL, Zhao C, Wu C, Hong JS, Chung PS, Clemens DL, Lee BY, Weiss PS, Teitell MA, Chiou PY. Intracellular Photothermal Delivery for Suspension Cells Using Sharp Nanoscale Tips in Microwells. ACS NANO 2019; 13:10835-10844. [PMID: 31487464 DOI: 10.1021/acsnano.9b06025] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Efficient intracellular delivery of biomolecules into cells that grow in suspension is of great interest for biomedical research, such as for applications in cancer immunotherapy. Although tremendous effort has been expended, it remains challenging for existing transfer platforms to deliver materials efficiently into suspension cells. Here, we demonstrate a high-efficiency photothermal delivery approach for suspension cells using sharp nanoscale metal-coated tips positioned at the edge of microwells, which provide controllable membrane disruption for each cell in an array. Self-aligned microfabrication generates a uniform microwell array with three-dimensional nanoscale metallic sharp tip structures. Suspension cells self-position by gravity within each microwell in direct contact with eight sharp tips, where laser-induced cavitation bubbles generate transient pores in the cell membrane to facilitate intracellular delivery of extracellular cargo. A range of cargo sizes were tested on this platform using Ramos suspension B cells with an efficiency of >84% for Calcein green (0.6 kDa) and >45% for FITC-dextran (2000 kDa), with retained viability of >96% and a throughput of >100 000 cells delivered per minute. The bacterial enzyme β-lactamase (29 kDa) was delivered into Ramos B cells and retained its biological activity, whereas a green fluorescence protein expression plasmid was delivered into Ramos B cells with a transfection efficiency of >58%, and a viability of >89% achieved.
Collapse
Affiliation(s)
- Tianxing Man
- Department of Mechanical and Aerospace Engineering , University of California, Los Angeles , Los Angeles , California 90095 , United States
| | - Xiongfeng Zhu
- Department of Mechanical and Aerospace Engineering , University of California, Los Angeles , Los Angeles , California 90095 , United States
| | - Yu Ting Chow
- Department of Mechanical and Aerospace Engineering , University of California, Los Angeles , Los Angeles , California 90095 , United States
| | - Emma R Dawson
- Department of Pathology and Laboratory Medicine , University of California, Los Angeles , Los Angeles , California 90095 , United States
| | - Ximiao Wen
- Department of Mechanical and Aerospace Engineering , University of California, Los Angeles , Los Angeles , California 90095 , United States
| | - Alexander N Patananan
- Department of Pathology and Laboratory Medicine , University of California, Los Angeles , Los Angeles , California 90095 , United States
| | - Tingyi Leo Liu
- Department of Mechanical and Aerospace Engineering , University of California, Los Angeles , Los Angeles , California 90095 , United States
| | - Chuanzhen Zhao
- California NanoSystems Institute , University of California, Los Angeles , Los Angeles , California 90095 , United States
- Department of Chemistry and Biochemistry , University of California, Los Angeles , Los Angeles , California 90095 , United States
| | - Cong Wu
- Department of Mechanical and Aerospace Engineering , University of California, Los Angeles , Los Angeles , California 90095 , United States
| | - Jason S Hong
- Department of Pathology and Laboratory Medicine , University of California, Los Angeles , Los Angeles , California 90095 , United States
| | - Pei-Shan Chung
- Department of Bioengineering , University of California, Los Angeles , Los Angeles , California 90095 , United States
| | - Daniel L Clemens
- Division of Infectious Diseases, Department of Medicine , University of California, Los Angeles , Los Angeles , California 90095 , United States
| | - Bai-Yu Lee
- Division of Infectious Diseases, Department of Medicine , University of California, Los Angeles , Los Angeles , California 90095 , United States
| | - Paul S Weiss
- California NanoSystems Institute , University of California, Los Angeles , Los Angeles , California 90095 , United States
- Department of Chemistry and Biochemistry , University of California, Los Angeles , Los Angeles , California 90095 , United States
- Department of Materials Science and Engineering , University of California, Los Angeles , Los Angeles , California 90095 , United States
| | - Michael A Teitell
- Department of Pathology and Laboratory Medicine , University of California, Los Angeles , Los Angeles , California 90095 , United States
- Department of Bioengineering , University of California, Los Angeles , Los Angeles , California 90095 , United States
- California NanoSystems Institute , University of California, Los Angeles , Los Angeles , California 90095 , United States
- Molecular Biology Institute, Department of Pathology and Laboratory Medicine, Department of Pediatrics, Jonsson Comprehensive Cancer Center, Broad Center of Regenerative Medicine and Stem Cell Research , University of California, Los Angeles , Los Angeles , California 90095 , United States
| | - Pei-Yu Chiou
- Department of Mechanical and Aerospace Engineering , University of California, Los Angeles , Los Angeles , California 90095 , United States
- Department of Bioengineering , University of California, Los Angeles , Los Angeles , California 90095 , United States
- California NanoSystems Institute , University of California, Los Angeles , Los Angeles , California 90095 , United States
| |
Collapse
|
12
|
Shih T, De S, Barnes BJ. RNAi Transfection Optimized in Primary Naïve B Cells for the Targeted Analysis of Human Plasma Cell Differentiation. Front Immunol 2019; 10:1652. [PMID: 31396212 PMCID: PMC6664017 DOI: 10.3389/fimmu.2019.01652] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 07/03/2019] [Indexed: 12/25/2022] Open
Abstract
Upon antigen recognition, naïve B cells undergo rapid proliferation followed by differentiation to specialized antibody secreting cells (ASCs), called plasma cells. Increased circulating plasma cells are reported in patients with B cell-associated malignancies, chronic graft-vs.-host disease, and autoimmune disorders. Our aim was to optimize an RNAi-based method that efficiently and reproducibly knocks-down genes of interest in human primary peripheral B cells for the targeted analysis of ASC differentiation. The unique contributions of transcriptional diversity in species-specific regulatory networks and the mechanisms of gene function need to be approached directly in human B cells with tools to hone our basic inferences from animal models to human biology. To date, methods for gene knockdown in human primary B cells, which tend to be more refractory to transfection than immortalized B cell lines, have been limited by losses in cell viability and ineffective penetrance. Our single-step siRNA nucleofector-based approach for human primary naïve B cells demonstrates reproducible knockdown efficiency (~40–60%). We focused on genes already known to play key roles in murine ASC differentiation, such as interferon regulatory factor 4 (IRF4) and AID. This study reports a validated non-viral method of siRNA delivery into human primary B cells that can be applied to study gene regulatory networks that control human ASC differentiation.
Collapse
Affiliation(s)
- Tiffany Shih
- Center for Autoimmune Musculoskeletal and Hematopoietic Disease, Northwell Health, The Feinstein Institute for Medical Research, Manhasset, NY, United States
| | - Saurav De
- Center for Autoimmune Musculoskeletal and Hematopoietic Disease, Northwell Health, The Feinstein Institute for Medical Research, Manhasset, NY, United States.,Graduate School of Biomedical Sciences Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Betsy J Barnes
- Center for Autoimmune Musculoskeletal and Hematopoietic Disease, Northwell Health, The Feinstein Institute for Medical Research, Manhasset, NY, United States.,Departments of Molecular Medicine and Pediatrics, Zucker School of Medicine at Hofstra/Northwell Health, Hempstead, NY, United States
| |
Collapse
|
13
|
Therapeutic mRNA delivery to leukocytes. J Control Release 2019; 305:165-175. [DOI: 10.1016/j.jconrel.2019.05.032] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 05/15/2019] [Accepted: 05/19/2019] [Indexed: 12/14/2022]
|
14
|
Schulze-Edinghausen L, Dürr C, Öztürk S, Zucknick M, Benner A, Kalter V, Ohl S, Close V, Wuchter P, Stilgenbauer S, Lichter P, Seiffert M. Dissecting the Prognostic Significance and Functional Role of Progranulin in Chronic Lymphocytic Leukemia. Cancers (Basel) 2019; 11:E822. [PMID: 31200555 PMCID: PMC6627891 DOI: 10.3390/cancers11060822] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 05/30/2019] [Accepted: 06/05/2019] [Indexed: 12/11/2022] Open
Abstract
Chronic lymphocytic leukemia (CLL) is known for its strong dependency on the tumor microenvironment. We found progranulin (GRN), a protein that has been linked to inflammation and cancer, to be upregulated in the serum of CLL patients compared to healthy controls, and increased GRN levels to be associated with an increased hazard for disease progression and death. This raised the question of whether GRN is a functional driver of CLL. We observed that recombinant GRN did not directly affect viability, activation, or proliferation of primary CLL cells in vitro. However, GRN secretion was induced in co-cultures of CLL cells with stromal cells that enhanced CLL cell survival. Gene expression profiling and protein analyses revealed that primary mesenchymal stromal cells (MSCs) in co-culture with CLL cells acquire a cancer-associated fibroblast-like phenotype. Despite its upregulation in the co-cultures, GRN treatment of MSCs did not mimic this effect. To test the relevance of GRN for CLL in vivo, we made use of the Eμ-TCL1 CLL mouse model. As we detected strong GRN expression in myeloid cells, we performed adoptive transfer of Eμ-TCL1 leukemia cells to bone marrow chimeric Grn-/- mice that lack GRN in hematopoietic cells. Thereby, we observed that CLL-like disease developed comparable in Grn-/- chimeras and respective control mice. In conclusion, serum GRN is found to be strongly upregulated in CLL, which indicates potential use as a prognostic marker, but there is no evidence that elevated GRN functionally drives the disease.
Collapse
Affiliation(s)
- Lena Schulze-Edinghausen
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| | - Claudia Dürr
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| | - Selcen Öztürk
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| | - Manuela Zucknick
- Oslo Centre for Biostatistics and Epidemiology, Department of Biostatistics, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway.
| | - Axel Benner
- Division of Biostatistics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| | - Verena Kalter
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| | - Sibylle Ohl
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| | - Viola Close
- Internal Medicine III, University of Ulm, 89081 Ulm, Germany, and Cooperation Unit Mechanisms of Leukemogenesis, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| | - Patrick Wuchter
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, German Red Cross Blood Service Baden-Württemberg-Hessen, 68167 Mannheim, Germany.
| | - Stephan Stilgenbauer
- Internal Medicine III, University of Ulm, 89081 Ulm, Germany, and Department of Internal Medicine I, Saarland University, 66421 Homburg, Germany.
| | - Peter Lichter
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
- German Cancer Research Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| | - Martina Seiffert
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| |
Collapse
|
15
|
De S, Zhang B, Shih T, Singh S, Winkler A, Donnelly R, Barnes BJ. B Cell-Intrinsic Role for IRF5 in TLR9/BCR-Induced Human B Cell Activation, Proliferation, and Plasmablast Differentiation. Front Immunol 2018; 8:1938. [PMID: 29367853 PMCID: PMC5768180 DOI: 10.3389/fimmu.2017.01938] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 12/15/2017] [Indexed: 12/25/2022] Open
Abstract
Upon recognition of antigen, B cells undergo rapid proliferation followed by differentiation to specialized antibody secreting cells (ASCs). During this transition, B cells are reliant upon a multilayer transcription factor network to achieve a dramatic remodeling of the B cell transcriptional landscape. Increased levels of ASCs are often seen in autoimmune diseases and it is believed that altered expression of regulatory transcription factors play a role in this imbalance. The transcription factor interferon regulatory factor 5 (IRF5) is one such candidate as polymorphisms in IRF5 associate with risk of numerous autoimmune diseases and correlate with elevated IRF5 expression. IRF5 genetic risk has been widely replicated in systemic lupus erythematosus (SLE), and loss of Irf5 ameliorates disease in murine lupus models, in part, through the lack of pathogenic autoantibody secretion. It remains unclear, however, whether IRF5 is contributing to autoantibody production through a B cell-intrinsic function. To date, IRF5 function in healthy human B cells has not been characterized. Using human primary naive B cells, we define a critical intrinsic role for IRF5 in B cell activation, proliferation, and plasmablast differentiation. Targeted IRF5 knockdown resulted in significant immunoglobulin (Ig) D retention, reduced proliferation, plasmablast differentiation, and IgG secretion. The observed decreases were due to impaired B cell activation and clonal expansion. Distinct from murine studies, we identify and confirm new IRF5 target genes, IRF4, ERK1, and MYC, and pathways that mediate IRF5 B cell-intrinsic function. Together, these results identify IRF5 as an early regulator of human B cell activation and provide the first dataset in human primary B cells to map IRF5 dysfunction in SLE.
Collapse
Affiliation(s)
- Saurav De
- Rutgers Graduate School of Biomedical Sciences, Newark, NJ, United States.,Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institute for Medical Research, Manhasset, NY, United States
| | - Baohong Zhang
- Clinical Genetics and Bioinformatics, Pfizer Inc., Cambridge, MA, United States
| | - Tiffany Shih
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institute for Medical Research, Manhasset, NY, United States
| | - Sukhwinder Singh
- Department of Pathology and Laboratory Medicine, Rutgers Biomedical and Health Sciences, New Jersey Medical School, Newark, NJ, United States
| | - Aaron Winkler
- Department of Inflammation and Immunology, Pfizer Inc., Cambridge, MA, United States
| | - Robert Donnelly
- Department of Pathology and Laboratory Medicine, Rutgers Biomedical and Health Sciences, New Jersey Medical School, Newark, NJ, United States
| | - Betsy J Barnes
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institute for Medical Research, Manhasset, NY, United States.,Rutgers Biomedical and Health Sciences, New Jersey Medical School-Cancer Center, Newark, NJ, United States
| |
Collapse
|
16
|
Patel S, Ashwanikumar N, Robinson E, DuRoss A, Sun C, Murphy-Benenato KE, Mihai C, Almarsson Ö, Sahay G. Boosting Intracellular Delivery of Lipid Nanoparticle-Encapsulated mRNA. NANO LETTERS 2017; 17:5711-5718. [PMID: 28836442 PMCID: PMC5623340 DOI: 10.1021/acs.nanolett.7b02664] [Citation(s) in RCA: 162] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
Intracellular delivery of mRNA holds great potential for vaccine1-3 and therapeutic4 discovery and development. Despite increasing recognition of the utility of lipid-based nanoparticles (LNPs) for intracellular delivery of mRNA, particle engineering is hindered by insufficient understanding of endosomal escape, which is believed to be a main limiter of cytosolic availability and activity of the nucleic acid inside the cell. Using a series of CRISPR-based genetic perturbations of the lysosomal pathway, we have identified that late endosome/lysosome (LE/Ly) formation is essential for functional delivery of exogenously presented mRNA. Lysosomes provide a spatiotemporal hub to orchestrate mTOR signaling and are known to control cell proliferation, nutrient sensing, ribosomal biogenesis, and mRNA translation. Through modulation of the mTOR pathway we were able to enhance or inhibit LNP-mediated mRNA delivery. To further boost intracellular delivery of mRNA, we screened 212 bioactive lipid-like molecules that are either enriched in vesicular compartments or modulate cell signaling. Surprisingly, we have discovered that leukotriene-antagonists, clinically approved for treatment of asthma and other lung diseases, enhance intracellular mRNA delivery in vitro (over 3-fold, p < 0.005) and in vivo (over 2-fold, p < 0.005). Understanding LNP-mediated intracellular delivery will inspire the next generation of RNA therapeutics that have high potency and limited toxicity.
Collapse
Affiliation(s)
- Siddharth Patel
- Department of Pharmaceutical Sciences, College of Pharmacy, Collaborative Life Science Building, 2730 SW Moody Ave, Oregon State University, Portland, OR, 97201
| | - N Ashwanikumar
- Department of Pharmaceutical Sciences, College of Pharmacy, Collaborative Life Science Building, 2730 SW Moody Ave, Oregon State University, Portland, OR, 97201
| | - Emily Robinson
- Department of Pharmaceutical Sciences, College of Pharmacy, Collaborative Life Science Building, 2730 SW Moody Ave, Oregon State University, Portland, OR, 97201
| | - Allison DuRoss
- Department of Pharmaceutical Sciences, College of Pharmacy, Collaborative Life Science Building, 2730 SW Moody Ave, Oregon State University, Portland, OR, 97201
| | - Conroy Sun
- Department of Pharmaceutical Sciences, College of Pharmacy, Collaborative Life Science Building, 2730 SW Moody Ave, Oregon State University, Portland, OR, 97201
- Department of Radiation Medicine, School of Medicine, 3181 S.W. Sam Jackson Park Road, Oregon Health Science University, Portland, OR, 97239
| | | | - Cosmin Mihai
- Moderna Therapeutics, 200 Technology Square, Cambridge, MA, 02139
| | - Örn Almarsson
- Moderna Therapeutics, 200 Technology Square, Cambridge, MA, 02139
| | - Gaurav Sahay
- Department of Pharmaceutical Sciences, College of Pharmacy, Collaborative Life Science Building, 2730 SW Moody Ave, Oregon State University, Portland, OR, 97201
- Department of Biomedical Engineering, Collaborative Life Science Building, 2730 SW Moody Ave, Oregon Health Science University, Portland, OR, 97201
| |
Collapse
|
17
|
Vaisitti T, Gaudino F, Ouk S, Moscvin M, Vitale N, Serra S, Arruga F, Zakrzewski JL, Liou HC, Allan JN, Furman RR, Deaglio S. Targeting metabolism and survival in chronic lymphocytic leukemia and Richter syndrome cells by a novel NF-κB inhibitor. Haematologica 2017; 102:1878-1889. [PMID: 28860341 PMCID: PMC5664392 DOI: 10.3324/haematol.2017.173419] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 08/28/2017] [Indexed: 02/04/2023] Open
Abstract
IT-901 is a novel and selective NF-κB inhibitor with promising activity in pre-clinical models. Here we show that treatment of chronic lymphocytic leukemia cells (CLL) with IT-901 effectively interrupts NF-κB transcriptional activity. CLL cells exposed to the drug display elevated mitochondrial reactive oxygen species, which damage mitochondria, limit oxidative phosphorylation and ATP production, and activate intrinsic apoptosis. Inhibition of NF-κB signaling in stromal and myeloid cells, both tumor-supportive elements, fails to induce apoptosis, but impairs NF-κB-driven expression of molecules involved in cell-cell contacts and immune responses, essential elements in creating a pro-leukemic niche. The consequence is that accessory cells do not protect CLL cells from IT-901-induced apoptosis. In this context, IT-901 shows synergistic activity with ibrutinib, arguing in favor of combination strategies. IT-901 is also effective in primary cells from patients with Richter syndrome (RS). Its anti-tumor properties are confirmed in xenograft models of CLL and in RS patient-derived xenografts, with documented NF-κB inhibition and significant reduction of tumor burden. Together, these results provide pre-clinical proof of principle for IT-901 as a potential new drug in CLL and RS.
Collapse
Affiliation(s)
- Tiziana Vaisitti
- Department of Medical Sciences, University of Turin, Italy .,Italian Institute for Genomic Medicine, Turin, Italy
| | - Federica Gaudino
- Department of Medical Sciences, University of Turin, Italy.,Italian Institute for Genomic Medicine, Turin, Italy
| | | | - Maria Moscvin
- Italian Institute for Genomic Medicine, Turin, Italy
| | - Nicoletta Vitale
- Department of Molecular Biotechnologies and Health Sciences, University of Turin, Italy
| | - Sara Serra
- Department of Medical Sciences, University of Turin, Italy.,Italian Institute for Genomic Medicine, Turin, Italy
| | | | | | | | - John N Allan
- CLL Research Center, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY, USA
| | - Richard R Furman
- CLL Research Center, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY, USA
| | - Silvia Deaglio
- Department of Medical Sciences, University of Turin, Italy .,Italian Institute for Genomic Medicine, Turin, Italy
| |
Collapse
|
18
|
Cornwall S, Cull G, Joske D, Ghassemifar R. Green tea polyphenol "epigallocatechin-3-gallate", differentially induces apoptosis in CLL B-and T-Cells but not in healthy B-and T-Cells in a dose dependant manner. Leuk Res 2016; 51:56-61. [PMID: 27855324 DOI: 10.1016/j.leukres.2016.10.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 10/24/2016] [Accepted: 10/31/2016] [Indexed: 01/18/2023]
Abstract
B-cell chronic lymphocytic leukaemia (CLL) is characterized by an accumulation of CD5-positive monoclonal B-cells due in large part to a failure of apoptosis. The ability to study CLL B-cells in vitro has always been a challenge and hampered by the low viability of the CLL B-cells in cell culture systems. In this study, we present a multicellular cell culture system to maintain CLL B-cells viable in culture for 60h in the presence of a stromal cell feeder layer in combination with a whole white blood cell preparation. Using this optimized system, we tested and showed that the addition of epigallocatechin-3-gallate (EGCG) at concentrations ranging from 25 to 100μg/ml induced apoptosis in CLL B-cells whilst not affecting healthy control B-cells. Moreover, the results showed that in contrast to healthy controls, T-cells from CLL patients underwent apoptosis in the presence of EGCG. This study demonstrated that the combination of a cell feeder layer with a whole white blood cell preparation maintained B-cell viability in vitro over an extended period of time. In addition, the study showed that EGCG differentially induces apoptosis in CLL B-and T-Cells but not in healthy B-and T-Cells in a dose dependent manner.
Collapse
Affiliation(s)
- Scott Cornwall
- Department of Haematology, PathWest Laboratory Medicine, Queen Elizabeth II Medical Centre, Nedlands, Western Australia, Australia
| | - Gavin Cull
- Department of Haematology, PathWest Laboratory Medicine, Queen Elizabeth II Medical Centre, Nedlands, Western Australia, Australia; Haematology Department, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia; School of Pathology and Laboratory Medicine, University of Western Australia, Nedlands, Western Australia, Australia
| | - David Joske
- Department of Haematology, PathWest Laboratory Medicine, Queen Elizabeth II Medical Centre, Nedlands, Western Australia, Australia; Haematology Department, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia; School of Medicine and Pharmacology, University of Western Australia, Nedlands, Western Australia, Australia
| | - Reza Ghassemifar
- Department of Haematology, PathWest Laboratory Medicine, Queen Elizabeth II Medical Centre, Nedlands, Western Australia, Australia; School of Pathology and Laboratory Medicine, University of Western Australia, Nedlands, Western Australia, Australia.
| |
Collapse
|
19
|
Diener Y, Bosio A, Bissels U. Delivery of RNA-based molecules to human hematopoietic stem and progenitor cells for modulation of gene expression. Exp Hematol 2016; 44:991-1001. [PMID: 27576131 DOI: 10.1016/j.exphem.2016.08.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 08/01/2016] [Accepted: 08/18/2016] [Indexed: 12/26/2022]
Abstract
Gene modulation of human hematopoietic stem and progenitor cells (HSPCs) harbors great potential for therapeutic application of these cells and presents a versatile tool in basic research to enhance our understanding of HSPC biology. However, stable genetic modification might be adverse, particularly in clinical settings. Here, we review a broad range of approaches to transient, nonviral modulation of protein expression with a focus on RNA-based methods. We compare different delivery methods and describe the usefulness of RNA molecules for overexpression as well as downregulation of proteins in HSPCs.
Collapse
Affiliation(s)
| | | | - Ute Bissels
- Miltenyi Biotec GmbH, Bergisch Gladbach, Germany.
| |
Collapse
|
20
|
Sprouty 2: a novel attenuator of B-cell receptor and MAPK-Erk signaling in CLL. Blood 2016; 127:2310-21. [PMID: 26809508 DOI: 10.1182/blood-2015-09-669317] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 01/13/2016] [Indexed: 12/19/2022] Open
Abstract
Clinical heterogeneity is a major barrier to effective treatment of chronic lymphocytic leukemia (CLL). Emerging evidence suggests that constitutive activation of various signaling pathways like mitogen-activated protein kinase-extracellular signal-regulated kinase (MAPK-Erk) signaling plays a role in the heterogeneous clinical outcome of CLL patients. In this study, we have investigated the role of Sprouty (SPRY)2 as a negative regulator of receptor and nonreceptor tyrosine kinase signaling in the pathogenesis of CLL. We show that SPRY2 expression is significantly decreased in CLL cells, particularly from poor-prognosis patients compared with those from good-prognosis patients. Overexpression of SPRY2 in CLL cells from poor-prognosis patients increased their apoptosis. Conversely, downregulation of SPRY2 in CLL cells from good-prognosis patients resulted in increased proliferation. Furthermore, CLL cells with low SPRY2 expression grew more rapidly in a xenograft model of CLL. Strikingly, B-cell-specific transgenic overexpression of spry2 in mice led to a decrease in the frequency of B1 cells, the precursors of CLL cells in rodents. Mechanistically, we show that SPRY2 attenuates the B-cell receptor (BCR) and MAPK-Erk signaling by binding to and antagonizing the activities of RAF1, BRAF, and spleen tyrosine kinase (SYK) in normal B cells and CLL cells. We also show that SPRY2 is targeted by microRNA-21, which in turn leads to increased activity of Syk and Erk in CLL cells. Taken together, these results establish SPRY2 as a critical negative regulator of BCR-mediated MAPK-Erk signaling in CLL, thereby providing one of the molecular mechanisms to explain the clinical heterogeneity of CLL.
Collapse
|
21
|
Göckeritz E, Kerwien S, Baumann M, Wigger M, Vondey V, Neumann L, Landwehr T, Wendtner CM, Klein C, Liu N, Hallek M, Frenzel LP, Krause G. Efficacy of phosphatidylinositol-3 kinase inhibitors with diverse isoform selectivity profiles for inhibiting the survival of chronic lymphocytic leukemia cells. Int J Cancer 2015; 137:2234-42. [DOI: 10.1002/ijc.29579] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Accepted: 03/13/2015] [Indexed: 01/08/2023]
Affiliation(s)
- Elisa Göckeritz
- Department I of Internal Medicine and Center of Integrated Oncology Cologne Bonn; Cologne Germany
- Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases University of Cologne; Cologne Germany
| | - Susan Kerwien
- Department I of Internal Medicine and Center of Integrated Oncology Cologne Bonn; Cologne Germany
| | - Michael Baumann
- Department I of Internal Medicine and Center of Integrated Oncology Cologne Bonn; Cologne Germany
| | - Marion Wigger
- Department I of Internal Medicine and Center of Integrated Oncology Cologne Bonn; Cologne Germany
| | - Verena Vondey
- Department I of Internal Medicine and Center of Integrated Oncology Cologne Bonn; Cologne Germany
- Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases University of Cologne; Cologne Germany
| | - Lars Neumann
- Department I of Internal Medicine and Center of Integrated Oncology Cologne Bonn; Cologne Germany
- Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases University of Cologne; Cologne Germany
| | - Thomas Landwehr
- Department I of Internal Medicine and Center of Integrated Oncology Cologne Bonn; Cologne Germany
| | - Clemens M. Wendtner
- Department I of Internal Medicine and Center of Integrated Oncology Cologne Bonn; Cologne Germany
| | - Christian Klein
- Roche Pharmaceutical Research & Early Development, Roche Innovation Center Zurich; Switzerland
| | - Ningshu Liu
- Bayer HealthCare Pharmaceuticals, Global Drug Discovery, TRG Oncology; Berlin Germany
| | - Michael Hallek
- Department I of Internal Medicine and Center of Integrated Oncology Cologne Bonn; Cologne Germany
- Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases University of Cologne; Cologne Germany
| | - Lukas P. Frenzel
- Department I of Internal Medicine and Center of Integrated Oncology Cologne Bonn; Cologne Germany
- Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases University of Cologne; Cologne Germany
| | - Günter Krause
- Department I of Internal Medicine and Center of Integrated Oncology Cologne Bonn; Cologne Germany
- Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases University of Cologne; Cologne Germany
| |
Collapse
|
22
|
Mamidi S, Höne S, Teufel C, Sellner L, Zenz T, Kirschfink M. Neutralization of membrane complement regulators improves complement-dependent effector functions of therapeutic anticancer antibodies targeting leukemic cells. Oncoimmunology 2015; 4:e979688. [PMID: 25949896 PMCID: PMC4404820 DOI: 10.4161/2162402x.2014.979688] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 10/16/2014] [Accepted: 10/16/2014] [Indexed: 01/12/2023] Open
Abstract
Complement-dependent cytotoxicity (CDC) is one of the effector mechanisms mediated by therapeutic anticancer monoclonal antibodies (mAbs). However, the efficacy of antibodies is limited by the resistance of malignant cells to complement attack, primarily due to the over-expression of one or more membrane complement regulatory proteins (mCRPs) CD46, CD55, and CD59. CD20-positive Burkitt lymphoma Raji cells and primary CLL cells are resistant to rituximab (RTX)-induced CDC whereas ofatumumab (OFA) proved to be more efficient in cell killing. Primary CLL cells but not CD52-positive acute lymphoblastic leukemia (ALL) REH cells were sensitive to alemtuzumab (ALM)-induced CDC. Upon combined inhibition on Raji and CLL cells by mCRPs-specific siRNAs or neutralizing antibodies, CDC induced by RTX and by OFA was augmented. Similarly, CDC of REH cells was enhanced after mCRPs were inhibited upon treatment with ALM. All mAbs induced C3 opsonization, which was significantly augmented upon blocking mCRPs. C3 opsonization led to enhanced cell-mediated cytotoxicity of leukemia cells exposed to PBLs or macrophages. Furthermore, opsonized CLL cells were efficiently phagocytized by macrophages. Our results provide conclusive evidence that inhibition of mCRPs expression sensitizes leukemic cells to complement attack thereby enhancing the therapeutic effect of mAbs targeting leukemic cells.
Collapse
Key Words
- ADCC, antibody-dependent cellular cytotoxicity
- ALM, Alemtuzumab
- CDC, complement-dependent cytotoxicity
- CDCC, complement-dependent cellular cytotoxicity
- MAC, membrane attack complex
- NHS, Normal Human Serum
- OFA, Ofatumumab
- PBLs, peripheral blood leukocytes
- RTX, Rituximab
- TRX, Trastuzumab
- alemtuzumab
- chronic lymphocytic leukemia
- complement regulatory proteins
- complement-dependent cytotoxicity
- mCRP, membrane-bound complement regulatory protein
- ofatumumab
- opsonization
- rituximab
- siRNA, small interfering RNA
Collapse
Affiliation(s)
- Srinivas Mamidi
- Institute for Immunology; University of Heidelberg; Heidelberg, Germany
| | - Simon Höne
- Institute for Immunology; University of Heidelberg; Heidelberg, Germany
| | - Claudia Teufel
- Institute for Immunology; University of Heidelberg; Heidelberg, Germany
| | - Leopold Sellner
- Department of Translational Oncology; National Center for Tumour Diseases (NCT) and German Cancer Research Center (DKFZ); Heidelberg, Germany
- Department of Medicine V; University of Heidelberg; Heidelberg, Germany
| | - Thorsten Zenz
- Department of Translational Oncology; National Center for Tumour Diseases (NCT) and German Cancer Research Center (DKFZ); Heidelberg, Germany
- Department of Medicine V; University of Heidelberg; Heidelberg, Germany
| | | |
Collapse
|
23
|
Bhattacharya N, Reichenzeller M, Caudron-Herger M, Haebe S, Brady N, Diener S, Nothing M, Döhner H, Stilgenbauer S, Rippe K, Mertens D. Loss of cooperativity of secreted CD40L and increased dose-response to IL4 on CLL cell viability correlates with enhanced activation of NF-kB and STAT6. Int J Cancer 2014; 136:65-73. [PMID: 24828787 DOI: 10.1002/ijc.28974] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 04/17/2014] [Indexed: 02/02/2023]
Abstract
Chronic lymphocytic leukemia (CLL) cells fail to enter apoptosis in vivo as opposed to their non-malignant B-lymphocyte counterparts. The ability of CLL cells to escape apoptosis is highly dependent on their microenvironment. Compared to non-malignant B cells, CLL cells are more responsive to complex stimuli that can be reproduced in vitro by the addition of cytokines. To understand the molecular mechanism of the environment-dependent anti-apoptotic signaling circuitry of CLL cells, we quantified the effect of the SDF-1, BAFF, APRIL, anti-IgM, interleukin-4 (IL4) and secreted CD40L (sCD40L) on the survival of in vitro cultured CLL cells and found IL4 and sCD40L to be most efficient in rescuing CLL cells from apoptosis. In quantitative dose-response experiments using cell survival as readout, the binding affinity of IL4 to its receptor was similar between malignant and non-malignant cells. However, the downstream signaling in terms of the amount of STAT6 and its degree of phosphorylation was highly stimulated in CLL cells. In contrast, the response to sCD40L showed a loss of cooperative binding in CLL cells but displayed a largely increased ligand binding affinity. Although a high-throughput microscopy analysis did not reveal a significant difference in the spatial CD40 receptor organization, the downstream signaling showed an enhanced activation of the NF-kB pathway in the malignant cells. Thus, we propose that the anti-apoptotic phenotype of CLL involves a sensitized response for IL4 dependent STAT6 phosphorylation, and an activation of NF-kB signaling due to an increased affinity of sCD40L to its receptor.
Collapse
Affiliation(s)
- Nupur Bhattacharya
- Cooperation Unit, "Mechanisms of Leukemogenesis", University of Ulm, Ulm, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Knudsen PB, Hanna B, Ohl S, Sellner L, Zenz T, Döhner H, Stilgenbauer S, Larsen TO, Lichter P, Seiffert M. Chaetoglobosin A preferentially induces apoptosis in chronic lymphocytic leukemia cells by targeting the cytoskeleton. Leukemia 2013; 28:1289-98. [PMID: 24280868 DOI: 10.1038/leu.2013.360] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Revised: 11/08/2013] [Accepted: 11/20/2013] [Indexed: 12/19/2022]
Abstract
Chronic lymphocytic leukemia (CLL) is an incurable malignancy of mature B cells. One of the major challenges in treatment of CLL is the achievement of a complete remission to prevent relapse of disease originating from cells within lymphoid tissues and subsequent chemoresistance. In search for novel drugs that target CLL cells in protective microenvironments, we performed a fungal extract screen using cocultures of primary CLL cells with bone marrow-derived stromal cells. A secondary metabolite produced by Penicillium aquamarinium was identified as Chaetoglobosin A (ChA), a member of the cytochalasan family that showed preferential induction of apoptosis in CLL cells, even under culture conditions that mimic lymphoid tissues. In vitro testing of 89 CLL cases revealed effective targeting of CLL cells by ChA, independent of bad prognosis characteristics, like 17p deletion or TP53 mutation. To provide insight into its mechanism of action, we showed that ChA targets filamentous actin in CLL cells and thereby induces cell-cycle arrest and inhibits membrane ruffling and cell migration. Our data further revealed that ChA prevents CLL cell activation and sensitizes them for treatment with PI3K and BTK inhibitors, suggesting this compound as a novel potential drug for CLL.
Collapse
Affiliation(s)
- P B Knudsen
- 1] Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany [2] Department of Systems Biology, Technical University of Denmark (DTU), Lyngby, Denmark
| | - B Hanna
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - S Ohl
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - L Sellner
- 1] Department of Translational Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany [2] Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | - T Zenz
- 1] Department of Translational Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany [2] Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | - H Döhner
- Internal Medicine III, University of Ulm, Ulm, Germany
| | | | - T O Larsen
- Department of Systems Biology, Technical University of Denmark (DTU), Lyngby, Denmark
| | - P Lichter
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - M Seiffert
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
25
|
Bladt TT, Dürr C, Knudsen PB, Kildgaard S, Frisvad JC, Gotfredsen CH, Seiffert M, Larsen TO. Bio-activity and dereplication-based discovery of ophiobolins and other fungal secondary metabolites targeting leukemia cells. Molecules 2013; 18:14629-50. [PMID: 24287995 PMCID: PMC6290568 DOI: 10.3390/molecules181214629] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2013] [Revised: 11/15/2013] [Accepted: 11/21/2013] [Indexed: 12/27/2022] Open
Abstract
The purpose of this study was to identify and characterize fungal natural products (NPs) with in vitro bioactivity towards leukemia cells. We based our screening on a combined analytical and bio-guided approach of LC-DAD-HRMS dereplication, explorative solid-phase extraction (E-SPE), and a co-culture platform of CLL and stromal cells. A total of 289 fungal extracts were screened and we tracked the activity to single compounds in seven of the most active extracts. The novel ophiobolin U was isolated together with the known ophiobolins C, H, K as well as 6-epiophiobolins G, K and N from three fungal strains in the Aspergillus section Usti. Ophiobolins A, B, C and K displayed bioactivity towards leukemia cells with induction of apoptosis at nanomolar concentrations. The remaining ophiobolins were mainly inactive or only slightly active at micromolar concentrations. Dereplication of those ophiobolin derivatives possessing different activity in combination with structural analysis allowed a correlation of the chemical structure and conformation with the extent of bioactivity, identifying the hydroxy group at C3 and an aldehyde at C21, as well as the A/B-cis ring structure, as indispensible for the strong activity of the ophiobolins. The known compounds penicillic acid, viridicatumtoxin, calbistrin A, brefeldin A, emestrin A, and neosolaniol monoacetate were identified from the extracts and also found generally cytotoxic.
Collapse
Affiliation(s)
- Tanja Thorskov Bladt
- Department of Systems Biology, Technical University of Denmark, Søltofts Plads, Building 221, Kgs. Lyngby DK-2800, Denmark; E-Mails: (T.T.B.); (P.B.K.); (S.K.); (J.C.F.)
| | - Claudia Dürr
- German Cancer Research Center, Molecular Genetics, Im Neuenheimer Feld 280, Heidelberg D-69120, Germany; E-Mail:
| | - Peter Boldsen Knudsen
- Department of Systems Biology, Technical University of Denmark, Søltofts Plads, Building 221, Kgs. Lyngby DK-2800, Denmark; E-Mails: (T.T.B.); (P.B.K.); (S.K.); (J.C.F.)
| | - Sara Kildgaard
- Department of Systems Biology, Technical University of Denmark, Søltofts Plads, Building 221, Kgs. Lyngby DK-2800, Denmark; E-Mails: (T.T.B.); (P.B.K.); (S.K.); (J.C.F.)
| | - Jens Christian Frisvad
- Department of Systems Biology, Technical University of Denmark, Søltofts Plads, Building 221, Kgs. Lyngby DK-2800, Denmark; E-Mails: (T.T.B.); (P.B.K.); (S.K.); (J.C.F.)
| | - Charlotte Held Gotfredsen
- Department of Chemistry, Technical University of Denmark, Kemitorvet, Building 201, Kgs. Lyngby DK-2800, Denmark; E-Mail:
| | - Martina Seiffert
- German Cancer Research Center, Molecular Genetics, Im Neuenheimer Feld 280, Heidelberg D-69120, Germany; E-Mail:
- Authors to whom correspondence should be addressed; E-Mails: (M.S.); (T.O.L.); Tel.: +49-6221-42-4586 (M.S.); Fax: +49-6221-42-2995 (M.S.); Tel.: +45-4525-2632 (T.O.L.); Fax: +45-4588-4148 (T.O.L.)
| | - Thomas Ostenfeld Larsen
- Department of Systems Biology, Technical University of Denmark, Søltofts Plads, Building 221, Kgs. Lyngby DK-2800, Denmark; E-Mails: (T.T.B.); (P.B.K.); (S.K.); (J.C.F.)
- Authors to whom correspondence should be addressed; E-Mails: (M.S.); (T.O.L.); Tel.: +49-6221-42-4586 (M.S.); Fax: +49-6221-42-2995 (M.S.); Tel.: +45-4525-2632 (T.O.L.); Fax: +45-4588-4148 (T.O.L.)
| |
Collapse
|
26
|
Defective DROSHA processing contributes to downregulation of MiR-15/-16 in chronic lymphocytic leukemia. Leukemia 2013; 28:98-107. [PMID: 23974981 DOI: 10.1038/leu.2013.246] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Revised: 08/07/2013] [Accepted: 08/08/2013] [Indexed: 12/21/2022]
Abstract
The MIR-15A/-16-1 tumor suppressor microRNAs (miRNAs) are deleted in leukemic cells from more than 50% of patients with chronic lymphocytic leukemia (CLL). As these miRNAs are also less abundant in patients without genomic deletion, their downregulation in CLL is likely to be caused by additional mechanisms. We found the primary transcripts (pri-miRNAs) of MIR-15a/-16/-15b to be elevated and processing intermediates (precursor miRNAs) to be reduced in cells from CLL patients (22/38) compared with non-malignant B-cells (n=14), indicating a block of miRNA maturation at the DROSHA processing step. Using a luciferase reporter assay for pri-miR processing we validated the defect in primary CLL cells. The block of miRNA maturation is restricted to specific miRNAs and can be found in the cell line MEC-2, but not in MEC-1, even though both are derived from the same CLL patient. In these cells, the RNA-specific deaminase ADARB1 leads to reduced pri-miRNA processing, but full processing efficiency is recovered upon deletion of the RNA-binding domains or nuclear localization of ADARB1. Thus, we show that, apart from genomic deletion or transcriptional downregulation, aberrant processing of miRNA leads to specific reduction of miRNAs in leukemic cells. This represents a novel oncogenic mechanism in the pathogenesis of CLL.
Collapse
|
27
|
Shalek AK, Gaublomme JT, Wang L, Yosef N, Chevrier N, Andersen MS, Robinson JT, Pochet N, Neuberg D, Gertner R, Amit I, Brown JR, Hacohen N, Regev A, Wu CJ, Park H. Nanowire-mediated delivery enables functional interrogation of primary immune cells: application to the analysis of chronic lymphocytic leukemia. NANO LETTERS 2012; 12:6498-504. [PMID: 23190424 PMCID: PMC3573729 DOI: 10.1021/nl3042917] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Indexed: 05/10/2023]
Abstract
A circuit level understanding of immune cells and hematological cancers has been severely impeded by a lack of techniques that enable intracellular perturbation without significantly altering cell viability and function. Here, we demonstrate that vertical silicon nanowires (NWs) enable gene-specific manipulation of diverse murine and human immune cells with negligible toxicity. To illustrate the power of the technique, we then apply NW-mediated gene silencing to investigate the role of the Wnt signaling pathway in chronic lymphocytic leukemia (CLL). Remarkably, CLL-B cells from different patients exhibit tremendous heterogeneity in their response to the knockdown of a single gene, LEF1. This functional heterogeneity defines three distinct patient groups not discernible by conventional CLL cytogenetic markers and provides a prognostic indicator for patients' time to first therapy. Analyses of gene expression signatures associated with these functional patient subgroups reveal unique insights into the underlying molecular basis for disease heterogeneity. Overall, our findings suggest a functional classification that can potentially guide the selection of patient-specific therapies in CLL and highlight the opportunities for nanotechnology to drive biological inquiry.
Collapse
Affiliation(s)
- Alex K. Shalek
- Department
of Chemistry and Chemical Biology and Department of Physics, Harvard University, 12 Oxford Street,
Cambridge, Massachusetts 02138, United States
| | - Jellert T. Gaublomme
- Department
of Chemistry and Chemical Biology and Department of Physics, Harvard University, 12 Oxford Street,
Cambridge, Massachusetts 02138, United States
| | - Lili Wang
- Department
of Medicine, Harvard Medical School and Cancer Vaccine Center and Department of
Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
02115, United States
| | - Nir Yosef
- Broad Institute of MIT
and Harvard, 7 Cambridge Center, Cambridge, Massachusetts
02142, United States
| | - Nicolas Chevrier
- Broad Institute of MIT
and Harvard, 7 Cambridge Center, Cambridge, Massachusetts
02142, United States
| | - Mette S. Andersen
- Department
of Chemistry and Chemical Biology and Department of Physics, Harvard University, 12 Oxford Street,
Cambridge, Massachusetts 02138, United States
| | - Jacob T. Robinson
- Department
of Chemistry and Chemical Biology and Department of Physics, Harvard University, 12 Oxford Street,
Cambridge, Massachusetts 02138, United States
| | - Nathalie Pochet
- Broad Institute of MIT
and Harvard, 7 Cambridge Center, Cambridge, Massachusetts
02142, United States
| | - Donna Neuberg
- Department
of Medicine, Harvard Medical School and Cancer Vaccine Center and Department of
Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
02115, United States
| | - Rona
S. Gertner
- Department
of Chemistry and Chemical Biology and Department of Physics, Harvard University, 12 Oxford Street,
Cambridge, Massachusetts 02138, United States
| | - Ido Amit
- Broad Institute of MIT
and Harvard, 7 Cambridge Center, Cambridge, Massachusetts
02142, United States
| | - Jennifer R. Brown
- Department
of Medicine, Harvard Medical School and Cancer Vaccine Center and Department of
Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
02115, United States
| | - Nir Hacohen
- Broad Institute of MIT
and Harvard, 7 Cambridge Center, Cambridge, Massachusetts
02142, United States
| | - Aviv Regev
- Broad Institute of MIT
and Harvard, 7 Cambridge Center, Cambridge, Massachusetts
02142, United States
- Howard
Hughes Medical Institute,
Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02140, United States
| | - Catherine J. Wu
- Department
of Medicine, Harvard Medical School and Cancer Vaccine Center and Department of
Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
02115, United States
| | - Hongkun Park
- Department
of Chemistry and Chemical Biology and Department of Physics, Harvard University, 12 Oxford Street,
Cambridge, Massachusetts 02138, United States
| |
Collapse
|
28
|
Fang Y, Mo X, Luo Y, Lu Y. BAX gene over-expression via nucleofection to induce apoptosis in human lens epithelial cells. Exp Biol Med (Maywood) 2012; 237:1000-6. [PMID: 22946090 DOI: 10.1258/ebm.2012.012108] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Despite significant advances in cataract surgery techniques, posterior capsule opacification (PCO) remains a common complication. In PCO, remaining epithelial cells cloud the lens capsule and impair postoperative vision. This in vitro study was designed to investigate the potential of a gene-based approach, specifically over-expression of the proapoptotic BAX gene, to prevent PCO. Human lens epithelial cells (HLECs) were transfected by nucleofection with a plasmid encoding a fusion protein of green fluorescent protein and human BAX. The expression levels of BAX and its antiapoptotic counterpart BCL2 were determined by realtime reverse transcription polymerase chain reaction, Western blotting and immunofluorescence. BAX over-expression-induced cell death was analyzed by fluorescence-activated cell sorting using the Annexin V antibody. Fluorescence microscopy and transmission electron microscopy were used to assess changes in morphology and ultrastructure. Differential expression of the downstream apoptosis-related factor, caspase 3, was detected by Western blotting. Nucleofection efficiency was high (nearly 80%). BAX-transfected HLECs showed remarkably enhanced BAX gene expression and BAX:BCL2 ratio, but relatively little change in endogenous BCL2 expression. BAX over-expression also led to significant cytotoxicity, induction of apoptosis-related characteristics and activation of caspase 3. In conclusion, our results indicate that BAX gene over-expression can trigger cell death in HLECs via an apoptotic pathway. Thus, BAX may be a promising candidate for human gene therapy to treat PCO.
Collapse
Affiliation(s)
- Yanwen Fang
- Department of Ophthalmology, Eye and Ear, Nose and Throat Hospital, Fudan University, 83 Fenyang Road, Shanghai 200031, China
| | | | | | | |
Collapse
|
29
|
Höfig I, Ehrhardt H, Jeremias I. Efficient RNA interference in patients' acute lymphoblastic leukemia cells amplified as xenografts in mice. Cell Commun Signal 2012; 10:8. [PMID: 22448764 PMCID: PMC3349556 DOI: 10.1186/1478-811x-10-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2011] [Accepted: 03/26/2012] [Indexed: 12/23/2022] Open
Abstract
Background Signaling studies in cell lines are hampered by non-physiological alterations obtained in vitro. Physiologic primary tumor cells from patients with leukemia require passaging through immune-compromised mice for amplification. The aim was to enable molecular work in patients' ALL cells by establishing siRNA transfection into cells amplified in mice. Results We established delivering siRNA into these cells without affecting cell viability. Knockdown of single or multiple genes reduced constitutive or induced protein expression accompanied by marked signaling alterations. Conclusion Our novel technique allows using patient-derived tumor cells instead of cell lines for signaling studies in leukemia.
Collapse
Affiliation(s)
- Ines Höfig
- Department of Gene Vectors, Helmholtz Center Munich - German Research Center for Environmental Health, Marchioninistr, 25, 81377 Munich, Germany.
| | | | | |
Collapse
|
30
|
Kovaleva V, Mora R, Park YJ, Plass C, Chiramel AI, Bartenschlager R, Döhner H, Stilgenbauer S, Pscherer A, Lichter P, Seiffert M. miRNA-130a targets ATG2B and DICER1 to inhibit autophagy and trigger killing of chronic lymphocytic leukemia cells. Cancer Res 2012; 72:1763-72. [PMID: 22350415 DOI: 10.1158/0008-5472.can-11-3671] [Citation(s) in RCA: 154] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Toxicity and relapses from the immunochemotherapy used to treat chronic lymphocytic leukemia (CLL) prompt continued interest in gentle but effective targeted treatment options for the mainly elderly population suffering from this disease. Here, we report the definition of critical CLL cell survival pathways that can be targeted by ectopic reexpression of the miRNA genes miR-130a and miR-143 which are widely downregulated in CLL. Notably, miR-130a inhibited autophagy by reducing autophagosome formation, an effect mediated by downregulation of the genes ATG2B and DICER1, the latter of which is a major component of the miRNA silencing machinery. In support of the concept of a fundamental connection between miRNA disregulation and altered autophagic flux in this cancer, we showed that RNA interference-mediated knockdown of DICER1 expression was sufficient to reduce autophagy in primary or established cultures of CLL cells. Together, our findings show that miR-130a modulates cell survival programs by regulating autophagic flux, and they define roles for miR-130a and Dicer1 in a regulatory feedback loop that mediates CLL cell survival.
Collapse
|
31
|
Seiffert M, Dietrich S, Jethwa A, Glimm H, Lichter P, Zenz T. Exploiting biological diversity and genomic aberrations in chronic lymphocytic leukemia. Leuk Lymphoma 2011; 53:1023-31. [PMID: 22023519 DOI: 10.3109/10428194.2011.631638] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
There is remarkable heterogeneity in the clinical course and biological characteristics of patient subgroups with chronic lymphocytic leukemia (CLL). Mutations of key tumor suppressors (ATM, miR-15a/16-1 and TP53) have been identified in CLL, and these aberrations are important "drivers" of the disease and some of its clinical characteristics. While some mutations are associated with poor outcome [particularly del(17p) and TP53 mutation], others are linked to a favorable clinical course [e.g. del(13q) as sole aberration]. In addition to genetic aberrations, antigen drive and microenvironmental interactions contribute to the pathogenesis of CLL. How the genetic aberrations impact on the process of antigen drive or microenvironmental interactions is currently unclear. Our improved understanding of the biology and clinical course of specific genetic subgroups is beginning to be translated into more specific and targeted treatment approaches. As a result, genetic subgroups are treated in distinct protocols. This review summarizes the contribution of the microenvironment and the most important genetic aberrations in CLL and how our improved knowledge of the biology of CLL may translate into improved treatment results.
Collapse
Affiliation(s)
- Martina Seiffert
- Division of Molecular Genetics, German Cancer Research Center, Heidelberg, Germany
| | | | | | | | | | | |
Collapse
|
32
|
Kardava L, Moir S, Wang W, Ho J, Buckner CM, Posada JG, O'Shea MA, Roby G, Chen J, Sohn HW, Chun TW, Pierce SK, Fauci AS. Attenuation of HIV-associated human B cell exhaustion by siRNA downregulation of inhibitory receptors. J Clin Invest 2011; 121:2614-24. [PMID: 21633172 DOI: 10.1172/jci45685] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2010] [Accepted: 04/06/2011] [Indexed: 01/08/2023] Open
Abstract
Chronic immune activation in HIV-infected individuals leads to accumulation of exhausted tissue-like memory B cells. Exhausted lymphocytes display increased expression of multiple inhibitory receptors, which may contribute to the inefficiency of HIV-specific antibody responses. Here, we show that downregulation of B cell inhibitory receptors in primary human B cells led to increased tissue-like memory B cell proliferation and responsiveness against HIV. In human B cells, siRNA knockdown of 9 known and putative B cell inhibitory receptors led to enhanced B cell receptor-mediated (BCR-mediated) proliferation of tissue-like memory but not other B cell subpopulations. The strongest effects were observed with the putative inhibitory receptors Fc receptor-like-4 (FCRL4) and sialic acid-binding Ig-like lectin 6 (Siglec-6). Inhibitory receptor downregulation also led to increased levels of HIV-specific antibody-secreting cells and B cell-associated chemokines and cytokines. The absence of known ligands for FCRL4 and Siglec-6 suggests these receptors may regulate BCR signaling through their own constitutive or tonic signaling. Furthermore, the extent of FCLR4 knockdown effects on BCR-mediated proliferation varied depending on the costimulatory ligand, suggesting that inhibitory receptors may engage specific pathways in inhibiting B cell proliferation. These findings on HIV-associated B cell exhaustion define potential targets for reversing the deleterious effect of inhibitory receptors on immune responses against persistent viral infections.
Collapse
Affiliation(s)
- Lela Kardava
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland 20892-1576, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Martins LR, Lúcio P, Silva MC, Gameiro P, Silva MG, Barata JT. On CK2 regulation of chronic lymphocytic leukemia cell viability. Mol Cell Biochem 2011; 356:51-5. [PMID: 21750986 DOI: 10.1007/s11010-011-0947-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2011] [Accepted: 06/24/2011] [Indexed: 01/12/2023]
Abstract
Specific inhibition of signaling elements essential for the viability of B-cell chronic lymphocytic leukemia (CLL) cells offers great promise for the design of more efficient therapies. The protein serine/threonine kinase CK2 is frequently upregulated in cancer, and it is overexpressed and hyperactivated in primary CLL cells from untreated patients. We have shown that inhibition of CK2 induces apoptosis of CLL cells, whereas it does not significantly impact normal lymphocytes, demonstrating the selectivity of the CK2 inhibitors toward leukemia cells. Notably, although co-culture with OP9 stromal cells and BCR stimulation both promote leukemia cell survival in vitro, they do not prevent apoptosis of CLL cells treated with CK2 inhibitors. PI3K signaling pathway was previously shown to be essential for CLL cell viability, an observation we confirmed in all patient samples analyzed. Further, we observed that CK2 blockade decreases PTEN phosphorylation, leading to PTEN activation, and that apoptosis of CLL cells upon CK2 inhibition is mediated by PKC inactivation. This suggests that activation of PI3K/PKC signaling pathway is involved in the pro-survival effects of CK2 in CLL cells. Sensitivity to CK2 inhibition does not correlate with expression of ZAP-70 or CD38, or with IGVH mutation status. However, it positively correlates with the percentage of CLL cells in the peripheral blood, β2 microglobulin levels, and Binet clinical stage. CK2 appears to play an important role in the biology of CLL and constitutes a promising target for the development of leukemia-specific therapies.
Collapse
Affiliation(s)
- Leila R Martins
- Cancer Biology Unit, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Av. Prof. Egas Moniz, Lisbon 1649-028, Portugal
| | | | | | | | | | | |
Collapse
|
34
|
Anti-CD22 antibody targeting of pH-responsive micelles enhances small interfering RNA delivery and gene silencing in lymphoma cells. Mol Ther 2011; 19:1529-37. [PMID: 21629223 DOI: 10.1038/mt.2011.104] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The application of small interfering RNA (siRNA) for cancer treatment is a promising strategy currently being explored in early phase clinical trials. However, efficient systemic delivery limits clinical implementation. We developed and tested a novel delivery system comprised of (i) an internalizing streptavidin-conjugated monoclonal antibody (mAb-SA) directed against CD22 and (ii) a biotinylated diblock copolymer containing both a positively charged siRNA condensing block and a pH-responsive block to facilitate endosome release. The modular design of the carrier facilitates the exchange of different targeting moieties and siRNAs to permit its usage in a variety of tumor types. The polymer was synthesized using the reversible addition fragmentation chain transfer (RAFT) technique and formed micelles capable of binding siRNA and mAb-SA. A hemolysis assay confirmed the predicted membrane destabilizing activity of the polymer under acidic conditions typical of the endosomal compartment. Enhanced siRNA uptake was demonstrated in DoHH2 lymphoma and transduced HeLa-R cells expressing CD22 but not in CD22 negative HeLa-R cells. Gene knockdown was significantly improved with CD22-targeted vs. nontargeted polymeric micelles. Treatment of DoHH2 cells with CD22-targeted polymeric micelles containing 15 nmol/l siRNA produced 70% reduction of gene expression. This CD22-targeted polymer carrier may be useful for siRNA delivery to lymphoma cells.
Collapse
|
35
|
Kardava L, Yang Q, St Leger A, Foon KA, Lentzsch S, Vallejo AN, Milcarek C, Borghesi L. The B lineage transcription factor E2A regulates apoptosis in chronic lymphocytic leukemia (CLL) cells. Int Immunol 2011; 23:375-84. [PMID: 21551245 DOI: 10.1093/intimm/dxr027] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Chronic lymphocytic leukemia (CLL) is a common malignancy characterized by the accumulation of B lymphocytes with an antigen-experienced activated CD19(+)CD5(+) clonal phenotype. Clinically, ∼50% of cases will behave more aggressively. Here, we investigate the role of the major B-cell transcription factor E2A, a known regulator of B-cell survival and proliferation, to CLL persistence. We show that E2A is elevated at the mRNA and protein levels relative to normal B-cell subsets. E2A silencing in primary CLL cells leads to a significant increase in spontaneous apoptosis in both CD38(+) (aggressive) and CD38(-) (indolent) cases. Moreover, E2A knockdown synergizes with the immunomodulatory drug lenalidomide to reduce CLL viability. E2A is known to restrain the proliferation of primary B and T lymphocytes at multiple stages of maturation and we report that targeted E2A disruption increases the frequency of Ki-67(+) CLL cells in the absence of effects on de novo proliferation. At the molecular level, E2A siRNA-treated CLL cells display reduced expression of key genes associated with survival and cell cycling including p27, p21 and mcl-1, of which the former two are known E2A target genes. Thus, E2A, a key transcription factor associated with the B-cell activation profile, regulates apoptosis in CLL and may contribute to disease pathology.
Collapse
Affiliation(s)
- Lela Kardava
- Department of Immunology, University of Pittsburgh School of Medicine, PA 15261, USA
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Gehrke I, Gandhirajan RK, Poll-Wolbeck SJ, Hallek M, Kreuzer KA. Bone marrow stromal cell-derived vascular endothelial growth factor (VEGF) rather than chronic lymphocytic leukemia (CLL) cell-derived VEGF is essential for the apoptotic resistance of cultured CLL cells. Mol Med 2011; 17:619-27. [PMID: 21519633 DOI: 10.2119/molmed.2010.00210] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2010] [Accepted: 04/13/2011] [Indexed: 12/16/2022] Open
Abstract
Chronic lymphocytic leukemia (CLL) cells feature a pronounced apoptotic resistance. The vascular endothelial growth factor (VEGF) possesses a role in this apoptotic block, although underlying functional mechanisms and the involvement of the microenvironment are unclear. In this study, the VEGF status in CLL was assessed by enzyme-linked immunosorbent assay and immunofluorescence. VEGF receptor 2 (VEGFR2) phosphorylation was determined flow cytometrically and by immunofluorescence. For co-culture, CLL cells were cultivated on a monolayer of the bone marrow-derived stromal cell (BMSC) line HS5. Secreted VEGF was neutralized using the monoclonal antibody mAb293 (R&D Systems, Minneapolis, MN, USA). To block protein secretion, we used Brefeldin A. VEGF was downregulated in BMSCs by small interfering RNA (siRNA), and we assessed survival by annexin V-fluorescein isothiocyanate (FITC)/propidium iodide (PI) staining. CLL cells express and secrete VEGF and possess phosphorylated VEGFR2. This positive VEGF status is not sufficient to prevent spontaneous apoptosis in vitro. Coculture with BMSCs, which secrete vast amounts of VEGF, maintains in vitro CLL cell survival. Blockage of secreted VEGF using the monoclonal antibody mAb293 significantly reduced the survival support for cocultured CLL cells. Both general blockage of protein secretion by Brefeldin A in BMSCs, but not in CLL cells, and siRNA-mediated downregulation of VEGF in BMSCs, significantly reduced the coculture-mediated survival support for CLL cells. It can be concluded that BMSC-derived proteins and VEGF, in particular, but not CLL cell-derived VEGF, is essentially involved in the coculture-mediated survival support for CLL cells. Hence, therapeutic targeting of VEGF signaling might be a promising approach to overcome the apoptotic resistance CLL cells feature within their natural microenvironment.
Collapse
Affiliation(s)
- Iris Gehrke
- Department I of Internal Medicine, Center for Integrated Oncology, University at Cologne, Cologne, Germany
| | | | | | | | | |
Collapse
|
37
|
In vitro and in vivo model of a novel immunotherapy approach for chronic lymphocytic leukemia by anti-CD23 chimeric antigen receptor. Blood 2011; 117:4736-45. [PMID: 21406718 DOI: 10.1182/blood-2010-10-311845] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Chronic lymphocytic leukemia (CLL) is characterized by an accumulation of mature CD19(+)CD5(+)CD20(dim) B lymphocytes that typically express the B-cell activation marker CD23. In the present study, we cloned and expressed in T lymphocytes a novel chimeric antigen receptor (CAR) targeting the CD23 antigen (CD23.CAR). CD23.CAR(+) T cells showed specific cytotoxic activity against CD23(+) tumor cell lines (average lysis 42%) and primary CD23(+) CLL cells (average lysis 58%). This effect was obtained without significant toxicity against normal B lymphocytes, in contrast to CARs targeting CD19 or CD20 antigens, which are also expressed physiologically by normal B lymphocytes. Moreover, CLL-derived CD23.CAR(+) T cells released inflammatory cytokines (1445-fold more TNF-β, 20-fold more TNF-α, and 4-fold more IFN-γ). IL-2 was also produced (average release 2681 pg/mL) and sustained the antigen-dependent proliferation of CD23.CAR(+) T cells. Redirected T cells were also effective in vivo in a CLL Rag2(-/-)γ(c)(-/-) xenograft mouse model. Compared with mice treated with control T cells, the infusion of CD23.CAR(+) T cells resulted in a significant delay in the growth of the MEC-1 CLL cell line. These data suggest that CD23.CAR(+) T cells represent a selective immunotherapy for the elimination of CD23(+) leukemic cells in patients with CLL.
Collapse
|
38
|
Dürig J, Dührsen U, Klein-Hitpass L, Worm J, Hansen JBR, Ørum H, Wissenbach M. The novel antisense Bcl-2 inhibitor SPC2996 causes rapid leukemic cell clearance and immune activation in chronic lymphocytic leukemia. Leukemia 2011; 25:638-47. [PMID: 21358717 DOI: 10.1038/leu.2010.322] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
SPC2996 is a novel locked nucleic acid phosphorothioate antisense molecule targeting the mRNA of the Bcl-2 oncoprotein. We investigated the mechanism of action of SPC2996 and the basis for its clinically observed immunostimulatory effects in chronic lymphocytic leukemia (CLL). Patients with relapsed CLL were treated with a maximum of six doses of SPC2996 (0.2-6 mg/kg) in a multicenter phase I trial. Microarray-based transcriptional profiling of circulating CLL cells was carried out before and after the first infusion of SPC2996 in 18 patients. Statistically significant transcriptomic changes were observed at doses 4 mg/kg and occurred as early as 24 h after the first infusion of the oligonucleotide. SPC2996 induced the upregulation of 466 genes including a large number of immune response and apoptotic regulator molecules, which were enriched for Toll-like receptor response genes. Serum measurements confirmed the release of pro-inflammatory cytokines including chemokine (C-C motif) ligand 3 (macrophage inflammatory protein 1α) and tumor necrosis factor-α, thereby validating the in vivo transcriptomic data at the protein level. SPC2996 caused a 50% reduction of circulating lymphocytes in five of 18 (28%) patients, which was found to be independent of its immunostimulatory and anti-Bcl-2 effects.
Collapse
Affiliation(s)
- J Dürig
- Department of Hematology, University Hospital, University of Duisburg-Essen, Essen, Germany.
| | | | | | | | | | | | | |
Collapse
|
39
|
Ryan J, Ludbrook L, Wilhelm D, Sinclair A, Koopman P, Bernard P, Harley V. Analysis of Gene Function in Cultured Embryonic Mouse Gonads Using Nucleofection. Sex Dev 2011; 5:7-15. [DOI: 10.1159/000322162] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/24/2010] [Indexed: 12/13/2022] Open
|
40
|
Schulz A, Toedt G, Zenz T, Stilgenbauer S, Lichter P, Seiffert M. Inflammatory cytokines and signaling pathways are associated with survival of primary chronic lymphocytic leukemia cells in vitro: a dominant role of CCL2. Haematologica 2010; 96:408-16. [PMID: 21134984 DOI: 10.3324/haematol.2010.031377] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Chronic lymphocytic leukemia cells show prolonged survival in vivo, but rapidly die by spontaneous apoptosis in vitro, unless they are co-cultured with stromal cells or non-malignant leukocytes. The objective of this study was to characterize the survival-inducing cross-talk of chronic lymphocytic leukemia cells with their microenvironment to identify novel therapeutic targets. DESIGN AND METHODS We analyzed and compared microarray-based expression profiles of chronic lymphocytic leukemia cells before and after three different survival-inducing culture conditions: (i) stromal cell co-culture, (ii) stromal cell conditioned medium and (iii) high cell density cultures of unsorted peripheral blood mononuclear cells. Cytokine antibody arrays were applied to study the composition of soluble factors present in these cultures. RESULTS The different survival-supportive culture conditions induced distinct gene expression changes, the majority of which were common to all three conditions. Pathway analyses identified - in addition to known signaling networks in chronic lymphocytic leukemia - novel pathways, of which Toll-like receptor signaling, nuclear respiratory factor-2 (NRF2)-mediated oxidative stress response, and signaling via triggering receptor expressed on myeloid cells-1 (TREM1) were the most relevant. A high proportion of up-regulated genes were inflammatory cytokines, of which chemokine (C-C motif) ligand 2 (CCL2) was shown to be induced in monocytes by the presence of chronic lymphocytic leukemia cells in vitro. In addition, increased serum levels of this chemokine were detected in patients with chronic lymphocytic leukemia. CONCLUSIONS Our data provide several lines of evidence that an inflammatory microenvironment is induced in survival-supportive cultures of chronic lymphocytic leukemia cells which might be directly or indirectly involved in the prolonged survival of the malignant cells.
Collapse
Affiliation(s)
- Angela Schulz
- German Cancer Research Center, Dept. for Molecular Genetics, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | | | | | | | | | | |
Collapse
|
41
|
Soluble CD14 is a novel monocyte-derived survival factor for chronic lymphocytic leukemia cells, which is induced by CLL cells in vitro and present at abnormally high levels in vivo. Blood 2010; 116:4223-30. [DOI: 10.1182/blood-2010-05-284505] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Abstract
Accumulation of leukemic cells in patients with chronic lymphocytic leukemia (CLL) is due to prolonged cell survival rather than increased proliferation. Survival of CLL cells depends on microenvironmental factors. Even though long-lived in vivo, CLL cells rapidly die by spontaneous apoptosis in vitro unless cocultured with stromal cells or their conditioned medium. In the present study, we show that survival of CLL cells is maintained in high cell density cultures, where the main prosurvival activity is delivered by monocytes. Cytokine array and enzyme-linked immunosorbent assay studies revealed increased expression of soluble CD14 by monocytes in the presence of CLL cells. The addition of recombinant soluble CD14 to primary CLL cells resulted in significantly increased cell survival rates, which were associated with higher activity nuclear factor κB. Quantification of serum levels of soluble CD14 revealed abnormally high levels of this protein in CLL patients, indicating a potential role of soluble CD14 in vivo. In summary, the presented data show that monocytes help in the survival of CLL cells by secreting soluble CD14, which induces nuclear factor κB activation in these cells, and that CLL cells actively shape their microenvironment by inducing CD14 secretion in accessory monocytes.
Collapse
|
42
|
Elliott SL, Crawford C, Mulligan E, Summerfield G, Newton P, Wallis J, Mainou-Fowler T, Evans P, Bedwell C, Durkacz BW, Willmore E. Mitoxantrone in combination with an inhibitor of DNA-dependent protein kinase: a potential therapy for high risk B-cell chronic lymphocytic leukaemia. Br J Haematol 2010; 152:61-71. [PMID: 21083655 DOI: 10.1111/j.1365-2141.2010.08425.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Defects in the DNA damage response pathway [e.g. del(17p)] are associated with drug-resistant B-cell chronic lymphocytic leukaemia (CLL). We previously demonstrated that over-expression of DNA-dependent protein kinase (DNA-PK) correlates with chemo-resistance and that inhibition of DNA-PK sensitizes CLL cells to chemotherapeutics. Here, we investigated expression of DNA-PK and other proteins that impact on drug resistance, and evaluated the effects of a DNA-PK inhibitor (NU7441) on mitoxantrone-induced cytotoxicity in CLL cells. NU7441 sensitized cells from 42/49 CLL samples to mitoxantrone, with sensitization ranging from 2- to 200-fold Co-culture of CLL cells in conditioned stromal medium increased chemoresistance but did not reduce sensitization by NU7441. Mitoxantrone treatment induced γH2AX foci and NU7441 increased their longevity (24 h). NU7441 prevented mitoxantrone-induced autophosphorylation of the DNA-PK catalytic subunit (DNA-PKcs) at Ser 2056, confirming that DNA-PK participates in repair of mitoxantrone-induced DNA damage. del(17p) cases were more resistant to mitoxantrone than del(13q) cases, but were resensitized (7-16 fold) by co-incubation with NU7441. Expression of DNA-PKcs, Ku80, P-glycoprotein and topoisomerase IIβ were significantly higher in del(17p) cases. PRKDC mRNA levels correlated with DNA-PKcs protein expression, which predicted shorter survival. These data confirm the potential of DNA-PK as a therapeutic target in poor prognosis CLL.
Collapse
Affiliation(s)
- Sarah L Elliott
- Newcastle Cancer Centre at the NorthernInstitute for Cancer Research, Medical School, Newcastle University, Newcastle upon Tyne, UK
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Buggins AGS, Pepper C, Patten PEM, Hewamana S, Gohil S, Moorhead J, Folarin N, Yallop D, Thomas NSB, Mufti GJ, Fegan C, Devereux S. Interaction with vascular endothelium enhances survival in primary chronic lymphocytic leukemia cells via NF-kappaB activation and de novo gene transcription. Cancer Res 2010; 70:7523-33. [PMID: 20736369 DOI: 10.1158/0008-5472.can-10-1634] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Chronic lymphocytic leukemia (CLL) cells rapidly undergo apoptosis in vitro, suggesting that the in vivo microenvironment provides crucial antiapoptotic signals. Overexpression of the antiapoptotic proteins Bcl-2 and Mcl-1 is a hallmark of CLL, and their expression is further enhanced in the lymphoid tissues. However, the high levels of Mcl-1 found in peripheral blood samples, coupled with its short half-life, led us to hypothesize that it must be actively maintained in the peripheral circulation. Coculture of CLL cells with human vascular endothelial cells significantly enhanced tumor cell survival, an effect that was not observed with normal B cells. This was associated with elevated levels of the antiapoptotic proteins Bcl-2, Mcl-1, and Bcl-X(L) and marked increased expression of CD38 and CD49d, both of which are associated with clinically aggressive disease. Because CD38, CD49d, and some Bcl-2 family genes are transcriptional targets for NF-κB, we assessed NF-κB activation following coculture with endothelial cells. DNA binding of the NF-κB subunit Rel A was significantly increased and strongly correlated with changes in transcription of CD38, CD49d, BCL2, MCL1, and BCLXL, effects that were reversed by a peptide inhibitor of Rel A. These effects were not observed following coculture with nonendothelial cell lines. Therefore, CLL cells receive specific survival signals following interaction with endothelial cells mediated through the activation of NF-κB and the induction of downstream target genes. This type of interaction in the peripheral vasculature may explain the constitutive NF-κB activation and the overexpression of Bcl-2 family proteins commonly seen in this disease.
Collapse
Affiliation(s)
- Andrea G S Buggins
- Department of Haematological Medicine, King's College London, London, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Astrocytic Responses to DNA Delivery Using Nucleofection. Neurochem Res 2010; 35:1771-9. [DOI: 10.1007/s11064-010-0243-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/03/2010] [Indexed: 01/10/2023]
|
45
|
Inhibition of GLI, but not Smoothened, induces apoptosis in chronic lymphocytic leukemia cells. Oncogene 2010; 29:4885-95. [PMID: 20603613 DOI: 10.1038/onc.2010.243] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The Hedgehog (Hh) pathway regulates cell proliferation and survival and contributes to tumorigenesis. We investigated the expression and function of this pathway in B-cell chronic lymphocytic leukemia (CLL) cells and in healthy B lymphocytes. Profiling of cognate Hh pathway members revealed reduced expression of two key Hh signaling effectors, Smoothened (SMOH) and GLI, in CLL cells, whereas transcription levels of other investigated members resembled normal B-lymphocyte levels. Examining the functional role of SMOH and GLI in cell survival, we found that CLL cells were hardly sensitive toward specific SMOH inhibition, but showed an unspecific decline in cell viability in response to high concentrations of the SMOH antagonist cyclopamine. In contrast, treatment with the novel GLI antagonist GANT61 reduced expression of the target gene Patched and preferentially decreased the viability of malignant cells. Specific RNA interference knockdown experiments in a CLL-derived cell line confirmed the autonomous role of GLI in malignant cell survival. GANT61-induced apoptosis in primary leukemic cells was partly attenuated by protective stromal cells, but not soluble sonic hedgehog ligand. In summary, our data show a downregulation of the classical Hh pathway in CLL and suggest an intrinsic SMOH-independent role of GLI in the ex vivo survival of CLL cells.
Collapse
|
46
|
Fabani MM, Abreu-Goodger C, Williams D, Lyons PA, Torres AG, Smith KGC, Enright AJ, Gait MJ, Vigorito E. Efficient inhibition of miR-155 function in vivo by peptide nucleic acids. Nucleic Acids Res 2010; 38:4466-75. [PMID: 20223773 PMCID: PMC2910044 DOI: 10.1093/nar/gkq160] [Citation(s) in RCA: 161] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2010] [Revised: 02/24/2010] [Accepted: 02/24/2010] [Indexed: 12/17/2022] Open
Abstract
MicroRNAs (miRNAs) play an important role in diverse physiological processes and are potential therapeutic agents. Synthetic oligonucleotides (ONs) of different chemistries have proven successful for blocking miRNA expression. However, their specificity and efficiency have not been fully evaluated. Here, we show that peptide nucleic acids (PNAs) efficiently block a key inducible miRNA expressed in the haematopoietic system, miR-155, in cultured B cells as well as in mice. Remarkably, miR-155 inhibition by PNA in primary B cells was achieved in the absence of any transfection agent. In mice, the high efficiency of the treatment was demonstrated by a strong overlap in global gene expression between B cells isolated from anti-miR-155 PNA-treated and miR-155-deficient mice. Interestingly, PNA also induced additional changes in gene expression. Our analysis provides a useful platform to aid the design of efficient and specific anti-miRNA ONs for in vivo use.
Collapse
Affiliation(s)
- Martin M. Fabani
- Medical Research Council, Laboratory of Molecular Biology, Cambridge CB2 0QH, European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Cambridge CB10 1SD, Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 0XY and Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Cei Abreu-Goodger
- Medical Research Council, Laboratory of Molecular Biology, Cambridge CB2 0QH, European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Cambridge CB10 1SD, Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 0XY and Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Donna Williams
- Medical Research Council, Laboratory of Molecular Biology, Cambridge CB2 0QH, European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Cambridge CB10 1SD, Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 0XY and Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Paul A. Lyons
- Medical Research Council, Laboratory of Molecular Biology, Cambridge CB2 0QH, European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Cambridge CB10 1SD, Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 0XY and Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Adrian G. Torres
- Medical Research Council, Laboratory of Molecular Biology, Cambridge CB2 0QH, European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Cambridge CB10 1SD, Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 0XY and Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Kenneth G. C. Smith
- Medical Research Council, Laboratory of Molecular Biology, Cambridge CB2 0QH, European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Cambridge CB10 1SD, Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 0XY and Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Anton J. Enright
- Medical Research Council, Laboratory of Molecular Biology, Cambridge CB2 0QH, European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Cambridge CB10 1SD, Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 0XY and Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Michael J. Gait
- Medical Research Council, Laboratory of Molecular Biology, Cambridge CB2 0QH, European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Cambridge CB10 1SD, Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 0XY and Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Elena Vigorito
- Medical Research Council, Laboratory of Molecular Biology, Cambridge CB2 0QH, European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Cambridge CB10 1SD, Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 0XY and Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| |
Collapse
|
47
|
Buggins AGS, Pepper CJ. The role of Bcl-2 family proteins in chronic lymphocytic leukaemia. Leuk Res 2010; 34:837-42. [PMID: 20359747 DOI: 10.1016/j.leukres.2010.03.011] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2010] [Revised: 01/25/2010] [Accepted: 03/06/2010] [Indexed: 02/05/2023]
Abstract
Bcl-2 family proteins have long been implicated in the pathology of chronic lymphocytic leukaemia (CLL). Indeed, a number of these proteins have been shown to have prognostic importance in this disease. The precise ways in which these proteins impact upon CLL and the ways in which they are regulated remain incompletely resolved. However, significant advances have been recently made in our understanding of how these proteins are controlled by genetic, epigenetic and microenvironmental cues. Furthermore, major progress has been made in trying to target these proteins therapeutically. Here we review the current knowledge about this family of apoptosis-regulating proteins and how they impact upon drug resistance and disease progression. We also summarise evolution in the development of Bcl-2 family inhibitors for the treatment of CLL and other cancers.
Collapse
Affiliation(s)
- Andrea G S Buggins
- Department of Haematological Medicine, King's College London, London, UK
| | | |
Collapse
|
48
|
Königs SK, Pallasch CP, Lindner LH, Schwamb J, Schulz A, Brinker R, Claasen J, Veldurthy A, Eibl H, Hallek M, Wendtner CM. Erufosine, a novel alkylphosphocholine, induces apoptosis in CLL through a caspase-dependent pathway. Leuk Res 2010; 34:1064-9. [PMID: 20092894 DOI: 10.1016/j.leukres.2009.12.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2009] [Revised: 12/11/2009] [Accepted: 12/12/2009] [Indexed: 10/19/2022]
Abstract
The alkylphosphocholine (APC) erufosine is a synthetic phospholipid analogue with antineoplastic activity. APC are known to interact with lipid metabolism and modulate cellular signaling pathways, particularly the phosphorylation of Akt. Here, in primary CLL cells induction of apoptosis was detected with an IC50 of 22muM whereas healthy donor PBMC were less sensitive towards erufosine. Treatment with erufosine caused dose-dependent cleavage of PARP, co-incubation with caspase inhibitor z-VAD almost completely abrogated the cytotoxic effect of erufosine indicating a caspase-dependent mechanism of erufosine. Erufosine was shown to induce apoptosis in primary CLL cells and merits further investigation regarding therapeutic options in CLL.
Collapse
|
49
|
Farfsing A, Engel F, Seiffert M, Hartmann E, Ott G, Rosenwald A, Stilgenbauer S, Döhner H, Boutros M, Lichter P, Pscherer A. Gene knockdown studies revealed CCDC50 as a candidate gene in mantle cell lymphoma and chronic lymphocytic leukemia. Leukemia 2009; 23:2018-26. [PMID: 19641524 DOI: 10.1038/leu.2009.144] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The two B-cell non-Hodgkin's lymphoma entities, chronic lymphocytic leukemia (CLL) and mantle cell lymphoma (MCL), show recurrent chromosomal gains of 3q25-q29, 12q13-q14 and 18q21-q22. The pathomechanisms affected by these aberrations are not understood. The aim of this study was to identify genes, located within these gained regions, which control cell death and cell survival of MCL and CLL cancer cells. Blood samples collected from 18 patients with CLL and 6 patients with MCL, as well as 6 cell lines representing both malignancies were analyzed by gene expression profiling. By a comparison of genomic DNA and gene expression, 72 candidate genes were identified. We performed a limited RNA interference screening with these candidates to identify genes affecting cell survival. CCDC50 (coiled coil domain containing protein 50), SERPINI2 and SMARCC2 mediated a reduction of cell viability in primary CLL cells as well as in cell lines. Gene knockdown and a nuclear factor kappa B (NFkappaB) reporter gene assay revealed that CCDC50 is required for survival in MCL and CLL cells and controls NFkappaB signaling.
Collapse
Affiliation(s)
- A Farfsing
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Pros E, Fernández-Rodríguez J, Canet B, Benito L, Sánchez A, Benavides A, Ramos FJ, López-Ariztegui MA, Capellá G, Blanco I, Serra E, Lázaro C. Antisense therapeutics for neurofibromatosis type 1 caused by deep intronic mutations. Hum Mutat 2009; 30:454-62. [PMID: 19241459 DOI: 10.1002/humu.20933] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Neurofibromatosis type 1 (NF1) is an autosomal dominant disorder affecting 1:3,500 individuals. Disease expression is highly variable and complications are diverse. However, currently there is no specific treatment for the disease. NF1 is caused by mutations in the NF1 gene, approximately 2.1% of constitutional mutations identified in our population are deep intronic mutations producing the insertion of a cryptic exon into the mature mRNA. We used antisense morpholino oligomers (AMOs) to restore normal splicing in primary fibroblast and lymphocyte cell lines derived from six NF1 patients bearing three deep intronic mutations in the NF1 gene (c.288+2025T>G, c.5749+332A>G, and c.7908-321C>G). AMOs were designed to target the newly created 5' splice sites to prevent the incorporation of cryptic exons. Our results demonstrate that AMO treatment effectively restored normal NF1 splicing at the mRNA level for the three mutations studied in the different cell lines analyzed. We also found that AMOs had a rapid effect that lasted for several days, acting in a sequence-specific manner and interfering with the splicing mechanism. Finally, to test whether the correction of aberrant NF1 splicing also restored neurofibromin function to wild-type levels, we measured the amount of Ras-GTP after AMO treatment in primary fibroblasts. The results clearly show an AMO-dependent decrease in Ras-GTP levels, which is consistent with the restoration of neurofibromin function. To our knowledge this is the first time that an antisense technique has been used successfully to correct NF1 mutations opening the possibility of a therapeutic strategy for this type of mutation not only for NF1 but for other genetic disorders.
Collapse
Affiliation(s)
- Eva Pros
- Laboratori de Recerca Translacional, Institut Català d'Oncologia-Institut d'Investigació Biomèdica de Bellvitge, L'Hospitalet de Llobregat, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|