1
|
Zylbersztejn F, Byelinska I, Jeanpierre S, Barral L, Geistlich K, Flores-Violante M, Voeltzel T, Paubelle E, Heiblig M, Alcazer V, Le Meur G, Fossard G, Belhabri A, Cruz-Moura I, Hermine O, Lefort S, Maguer-Satta V. Human myeloid differentiation by BMP4 signaling through the VDR pathway in acute myeloid leukemia. Cell Death Discov 2024; 10:325. [PMID: 39013874 PMCID: PMC11252393 DOI: 10.1038/s41420-024-02090-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 07/03/2024] [Accepted: 07/04/2024] [Indexed: 07/18/2024] Open
Affiliation(s)
- Florence Zylbersztejn
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 69000, Lyon, France
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69000, Lyon, France
- Department of Cancer Initiation and Tumor Cell Identity, Lyon, France
- Université Claude Bernard Lyon 1, CRCL, 69000, Lyon, France
- Necker Hospital, Imagine Institute, Inserm U116 CNRS ERL 8654, 75015, Paris, France
| | - Iryna Byelinska
- Department of Clinical Medicine, Educational and Scientific Center "Institute of Biology and Medicine", Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Sandrine Jeanpierre
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 69000, Lyon, France
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69000, Lyon, France
- Department of Cancer Initiation and Tumor Cell Identity, Lyon, France
- Université Claude Bernard Lyon 1, CRCL, 69000, Lyon, France
- Centre Léon Bérard, 69000, Lyon, France
| | - Léa Barral
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 69000, Lyon, France
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69000, Lyon, France
- Department of Cancer Initiation and Tumor Cell Identity, Lyon, France
- Université Claude Bernard Lyon 1, CRCL, 69000, Lyon, France
| | - Kevin Geistlich
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 69000, Lyon, France
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69000, Lyon, France
- Department of Cancer Initiation and Tumor Cell Identity, Lyon, France
- Université Claude Bernard Lyon 1, CRCL, 69000, Lyon, France
- Necker Hospital, Imagine Institute, Inserm U116 CNRS ERL 8654, 75015, Paris, France
| | - Mario Flores-Violante
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 69000, Lyon, France
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69000, Lyon, France
- Department of Cancer Initiation and Tumor Cell Identity, Lyon, France
- Université Claude Bernard Lyon 1, CRCL, 69000, Lyon, France
| | - Thibault Voeltzel
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 69000, Lyon, France
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69000, Lyon, France
- Department of Cancer Initiation and Tumor Cell Identity, Lyon, France
- Université Claude Bernard Lyon 1, CRCL, 69000, Lyon, France
| | - Etienne Paubelle
- Hospices Civils de Lyon, Hematology Department, Centre Hospitalier Lyon Sud, 69495, Pierre Bénite, France
| | - Mael Heiblig
- Hospices Civils de Lyon, Hematology Department, Centre Hospitalier Lyon Sud, 69495, Pierre Bénite, France
| | - Vincent Alcazer
- Hospices Civils de Lyon, Hematology Department, Centre Hospitalier Lyon Sud, 69495, Pierre Bénite, France
| | - Gregoire Le Meur
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 69000, Lyon, France
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69000, Lyon, France
- Department of Cancer Initiation and Tumor Cell Identity, Lyon, France
- Université Claude Bernard Lyon 1, CRCL, 69000, Lyon, France
- Hospices Civils de Lyon, Hematology Department, Centre Hospitalier Lyon Sud, 69495, Pierre Bénite, France
| | - Gaelle Fossard
- Hospices Civils de Lyon, Hematology Department, Centre Hospitalier Lyon Sud, 69495, Pierre Bénite, France
| | - Amine Belhabri
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 69000, Lyon, France
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69000, Lyon, France
- Department of Cancer Initiation and Tumor Cell Identity, Lyon, France
- Université Claude Bernard Lyon 1, CRCL, 69000, Lyon, France
- Centre Léon Bérard, 69000, Lyon, France
| | - Ivan Cruz-Moura
- Necker Hospital, Imagine Institute, Inserm U116 CNRS ERL 8654, 75015, Paris, France
| | - Olivier Hermine
- Necker Hospital, Imagine Institute, Inserm U116 CNRS ERL 8654, 75015, Paris, France
| | - Sylvain Lefort
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 69000, Lyon, France.
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69000, Lyon, France.
- Department of Cancer Initiation and Tumor Cell Identity, Lyon, France.
- Université Claude Bernard Lyon 1, CRCL, 69000, Lyon, France.
| | - Véronique Maguer-Satta
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 69000, Lyon, France.
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69000, Lyon, France.
- Department of Cancer Initiation and Tumor Cell Identity, Lyon, France.
- Université Claude Bernard Lyon 1, CRCL, 69000, Lyon, France.
- Centre Léon Bérard, 69000, Lyon, France.
| |
Collapse
|
2
|
Zhou W, Yan K, Xi Q. BMP signaling in cancer stemness and differentiation. CELL REGENERATION (LONDON, ENGLAND) 2023; 12:37. [PMID: 38049682 PMCID: PMC10695912 DOI: 10.1186/s13619-023-00181-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 11/06/2023] [Indexed: 12/06/2023]
Abstract
The BMP (Bone morphogenetic protein) signaling pathway plays a central role in metazoan biology, intricately shaping embryonic development, maintaining tissue homeostasis, and influencing disease progression. In the context of cancer, BMP signaling exhibits context-dependent dynamics, spanning from tumor suppression to promotion. Cancer stem cells (CSCs), a modest subset of neoplastic cells with stem-like attributes, exert substantial influence by steering tumor growth, orchestrating therapy resistance, and contributing to relapse. A comprehensive grasp of the intricate interplay between CSCs and their microenvironment is pivotal for effective therapeutic strategies. Among the web of signaling pathways orchestrating cellular dynamics within CSCs, BMP signaling emerges as a vital conductor, overseeing CSC self-renewal, differentiation dynamics, and the intricate symphony within the tumor microenvironment. Moreover, BMP signaling's influence in cancer extends beyond CSCs, intricately regulating cellular migration, invasion, and metastasis. This multifaceted role underscores the imperative of comprehending BMP signaling's contributions to cancer, serving as the foundation for crafting precise therapies to navigate multifaceted challenges posed not only by CSCs but also by various dimensions of cancer progression. This article succinctly encapsulates the diverse roles of the BMP signaling pathway across different cancers, spanning glioblastoma multiforme (GBM), diffuse intrinsic pontine glioma (DIPG), colorectal cancer, acute myeloid leukemia (AML), lung cancer, prostate cancer, and osteosarcoma. It underscores the necessity of unraveling underlying mechanisms and molecular interactions. By delving into the intricate tapestry of BMP signaling's engagement in cancers, researchers pave the way for meticulously tailored therapies, adroitly leveraging its dualistic aspects-whether as a suppressor or promoter-to effectively counter the relentless march of tumor progression.
Collapse
Affiliation(s)
- Wei Zhou
- State Key Laboratory of Molecular Oncology, MOE Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Kun Yan
- Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Qiaoran Xi
- State Key Laboratory of Molecular Oncology, MOE Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China.
- Joint Graduate Program of Peking-Tsinghua-NIBS, Tsinghua University, Beijing, China.
| |
Collapse
|
3
|
Luis TC, Barkas N, Carrelha J, Giustacchini A, Mazzi S, Norfo R, Wu B, Aliouat A, Guerrero JA, Rodriguez-Meira A, Bouriez-Jones T, Macaulay IC, Jasztal M, Zhu G, Ni H, Robson MJ, Blakely RD, Mead AJ, Nerlov C, Ghevaert C, Jacobsen SEW. Perivascular niche cells sense thrombocytopenia and activate hematopoietic stem cells in an IL-1 dependent manner. Nat Commun 2023; 14:6062. [PMID: 37770432 PMCID: PMC10539537 DOI: 10.1038/s41467-023-41691-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 09/11/2023] [Indexed: 09/30/2023] Open
Abstract
Hematopoietic stem cells (HSCs) residing in specialized niches in the bone marrow are responsible for the balanced output of multiple short-lived blood cell lineages in steady-state and in response to different challenges. However, feedback mechanisms by which HSCs, through their niches, sense acute losses of specific blood cell lineages remain to be established. While all HSCs replenish platelets, previous studies have shown that a large fraction of HSCs are molecularly primed for the megakaryocyte-platelet lineage and are rapidly recruited into proliferation upon platelet depletion. Platelets normally turnover in an activation-dependent manner, herein mimicked by antibodies inducing platelet activation and depletion. Antibody-mediated platelet activation upregulates expression of Interleukin-1 (IL-1) in platelets, and in bone marrow extracellular fluid in vivo. Genetic experiments demonstrate that rather than IL-1 directly activating HSCs, activation of bone marrow Lepr+ perivascular niche cells expressing IL-1 receptor is critical for the optimal activation of quiescent HSCs upon platelet activation and depletion. These findings identify a feedback mechanism by which activation-induced depletion of a mature blood cell lineage leads to a niche-dependent activation of HSCs to reinstate its homeostasis.
Collapse
Affiliation(s)
- Tiago C Luis
- Haematopoietic Stem Cell Biology Laboratory, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, OX3 9DS, Oxford, UK.
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, OX3 9DS, Oxford, UK.
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, W12 0NN, London, UK.
- Department of Life Sciences, Imperial College London, SW7 2AZ, London, UK.
| | - Nikolaos Barkas
- Haematopoietic Stem Cell Biology Laboratory, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, OX3 9DS, Oxford, UK
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, OX3 9DS, Oxford, UK
| | - Joana Carrelha
- Haematopoietic Stem Cell Biology Laboratory, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, OX3 9DS, Oxford, UK
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, OX3 9DS, Oxford, UK
| | - Alice Giustacchini
- Haematopoietic Stem Cell Biology Laboratory, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, OX3 9DS, Oxford, UK
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, OX3 9DS, Oxford, UK
- Molecular and Cellular Immunology Section, UCL Great Ormond Street Institute of Child Health, London, UK
- Human Technopole, Viale Rita Levi-Montalcini 1, 20157, Milan, Italy
| | - Stefania Mazzi
- Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, SE-141 86, Stockholm, Sweden
| | - Ruggiero Norfo
- Haematopoietic Stem Cell Biology Laboratory, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, OX3 9DS, Oxford, UK
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, OX3 9DS, Oxford, UK
| | - Bishan Wu
- Haematopoietic Stem Cell Biology Laboratory, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, OX3 9DS, Oxford, UK
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, OX3 9DS, Oxford, UK
| | - Affaf Aliouat
- Haematopoietic Stem Cell Biology Laboratory, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, OX3 9DS, Oxford, UK
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, OX3 9DS, Oxford, UK
| | - Jose A Guerrero
- Department of Haematology, University of Cambridge, Cambridge, UK
- National Health Service (NHS) Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK
| | - Alba Rodriguez-Meira
- Haematopoietic Stem Cell Biology Laboratory, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, OX3 9DS, Oxford, UK
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, OX3 9DS, Oxford, UK
| | - Tiphaine Bouriez-Jones
- Haematopoietic Stem Cell Biology Laboratory, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, OX3 9DS, Oxford, UK
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, OX3 9DS, Oxford, UK
| | - Iain C Macaulay
- Haematopoietic Stem Cell Biology Laboratory, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, OX3 9DS, Oxford, UK
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, OX3 9DS, Oxford, UK
- Earlham Institute, Norwich Research Park, NR4 7UZ, Norwich, UK
| | - Maria Jasztal
- Department of Haematology, University of Cambridge, Cambridge, UK
- National Health Service (NHS) Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK
| | - Guangheng Zhu
- Toronto Platelet Immunobiology Group and Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, M5B 1W8, Canada
- CCOA Therapeutics Inc, Toronto, ON, M5B 1T8, Canada
| | - Heyu Ni
- Toronto Platelet Immunobiology Group and Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, M5B 1W8, Canada
- CCOA Therapeutics Inc, Toronto, ON, M5B 1T8, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A1, Canada
- Canadian Blood Services Centre for Innovation, Toronto, ON, M5B 1W8, Canada
| | - Matthew J Robson
- Department of Biomedical Science, Charles E. Schmidt College of Medicine and Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, 33458, USA
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, 45267, USA
| | - Randy D Blakely
- Department of Biomedical Science, Charles E. Schmidt College of Medicine and Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, 33458, USA
| | - Adam J Mead
- Haematopoietic Stem Cell Biology Laboratory, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, OX3 9DS, Oxford, UK
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, OX3 9DS, Oxford, UK
| | - Claus Nerlov
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, OX3 9DS, Oxford, UK
| | - Cedric Ghevaert
- Department of Haematology, University of Cambridge, Cambridge, UK
- National Health Service (NHS) Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK
| | - Sten Eirik W Jacobsen
- Haematopoietic Stem Cell Biology Laboratory, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, OX3 9DS, Oxford, UK.
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, OX3 9DS, Oxford, UK.
- Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, SE-141 86, Stockholm, Sweden.
- Department of Cell and Molecular Biology, Karolinska Institutet, SE-171 77, Stockholm, Sweden.
- Department of Hematology, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
4
|
The Hematopoietic TALE-Code Shows Normal Activity of IRX1 in Myeloid Progenitors and Reveals Ectopic Expression of IRX3 and IRX5 in Acute Myeloid Leukemia. Int J Mol Sci 2022; 23:ijms23063192. [PMID: 35328612 PMCID: PMC8952210 DOI: 10.3390/ijms23063192] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/11/2022] [Accepted: 03/14/2022] [Indexed: 12/10/2022] Open
Abstract
Homeobox genes encode transcription factors that control basic developmental decisions. Knowledge of their hematopoietic activities casts light on normal and malignant immune cell development. Recently, we constructed the so-called lymphoid TALE-code that codifies expression patterns of all active TALE class homeobox genes in early hematopoiesis and lymphopoiesis. Here, we present the corresponding myeloid TALE-code to extend this gene signature, covering the entire hematopoietic system. The collective data showed expression patterns for eleven TALE homeobox genes and highlighted the exclusive expression of IRX1 in megakaryocyte-erythroid progenitors (MEPs), implicating this TALE class member in a specific myeloid differentiation process. Analysis of public profiling data from acute myeloid leukemia (AML) patients revealed aberrant activity of IRX1 in addition to IRX3 and IRX5, indicating an oncogenic role for these TALE homeobox genes when deregulated. Screening of RNA-seq data from 100 leukemia/lymphoma cell lines showed overexpression of IRX1, IRX3, and IRX5 in megakaryoblastic and myelomonocytic AML cell lines, chosen as suitable models for studying the regulation and function of these homeo-oncogenes. Genomic copy number analysis of IRX-positive cell lines demonstrated chromosomal amplification of the neighboring IRX3 and IRX5 genes at position 16q12 in MEGAL, underlying their overexpression in this cell line model. Comparative gene expression analysis of these cell lines revealed candidate upstream factors and target genes, namely the co-expression of GATA1 and GATA2 together with IRX1, and of BMP2 and HOXA10 with IRX3/IRX5. Subsequent knockdown and stimulation experiments in AML cell lines confirmed their activating impact in the corresponding IRX gene expression. Furthermore, we demonstrated that IRX1 activated KLF1 and TAL1, while IRX3 inhibited GATA1, GATA2, and FST. Accordingly, we propose that these regulatory relationships may represent major physiological and oncogenic activities of IRX factors in normal and malignant myeloid differentiation, respectively. Finally, the established myeloid TALE-code is a useful tool for evaluating TALE homeobox gene activities in AML.
Collapse
|
5
|
Guyot B, Lefort S, Voeltzel T, Pécheur EI, Maguer-Satta V. Altered BMP2/4 Signaling in Stem Cells and Their Niche: Different Cancers but Similar Mechanisms, the Example of Myeloid Leukemia and Breast Cancer. Front Cell Dev Biol 2022; 9:787989. [PMID: 35047500 PMCID: PMC8762220 DOI: 10.3389/fcell.2021.787989] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 12/14/2021] [Indexed: 12/31/2022] Open
Abstract
Understanding mechanisms of cancer development is mandatory for disease prevention and management. In healthy tissue, the microenvironment or niche governs stem cell fate by regulating the availability of soluble molecules, cell-cell contacts, cell-matrix interactions, and physical constraints. Gaining insight into the biology of the stem cell microenvironment is of utmost importance, since it plays a role at all stages of tumorigenesis, from (stem) cell transformation to tumor escape. In this context, BMPs (Bone Morphogenetic Proteins), are key mediators of stem cell regulation in both embryonic and adult organs such as hematopoietic, neural and epithelial tissues. BMPs directly regulate the niche and stem cells residing within. Among them, BMP2 and BMP4 emerged as master regulators of normal and tumorigenic processes. Recently, a number of studies unraveled important mechanisms that sustain cell transformation related to dysregulations of the BMP pathway in stem cells and their niche (including exposure to pollutants such as bisphenols). Furthermore, a direct link between BMP2/BMP4 binding to BMP type 1 receptors and the emergence and expansion of cancer stem cells was unveiled. In addition, a chronic exposure of normal stem cells to abnormal BMP signals contributes to the emergence of cancer stem cells, or to disease progression independently of the initial transforming event. In this review, we will illustrate how the regulation of stem cells and their microenvironment becomes dysfunctional in cancer via the hijacking of BMP signaling with main examples in myeloid leukemia and breast cancers.
Collapse
Affiliation(s)
- Boris Guyot
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Université de Lyon, Lyon, France
- Department of Cancer Initiation and Tumor Cell Identity, Lyon, France
- Université de Lyon 1, Lyon, France
- Centre Leon Bérard, Lyon, France
| | - Sylvain Lefort
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Université de Lyon, Lyon, France
- Department of Cancer Initiation and Tumor Cell Identity, Lyon, France
- Université de Lyon 1, Lyon, France
- Centre Leon Bérard, Lyon, France
| | - Thibault Voeltzel
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Université de Lyon, Lyon, France
- Department of Cancer Initiation and Tumor Cell Identity, Lyon, France
- Université de Lyon 1, Lyon, France
- Centre Leon Bérard, Lyon, France
| | - Eve-Isabelle Pécheur
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Université de Lyon, Lyon, France
- Department of Cancer Initiation and Tumor Cell Identity, Lyon, France
- Université de Lyon 1, Lyon, France
- Centre Leon Bérard, Lyon, France
| | - Véronique Maguer-Satta
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Université de Lyon, Lyon, France
- Department of Cancer Initiation and Tumor Cell Identity, Lyon, France
- Université de Lyon 1, Lyon, France
| |
Collapse
|
6
|
Wang R, Hu X, Wang J, Zhou L, Hong Y, Zhang Y, Xiong F, Zhang X, Ye WC, Wang H. Proanthocyanidin A1 promotes the production of platelets to ameliorate chemotherapy-induced thrombocytopenia through activating JAK2/STAT3 pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 95:153880. [PMID: 34906892 DOI: 10.1016/j.phymed.2021.153880] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 11/22/2021] [Accepted: 12/02/2021] [Indexed: 06/14/2023]
Abstract
BACKGROUND Chemotherapy-induced thrombocytopenia (CIT) is a severe adverse drug reaction, and the main reason for CIT is the destruction of megakaryocytes (MKs, precursor cells of platelet) in bone marrow by chemotherapy. Peanut skin, the seed coat of Arachis hypogaea L., is a traditional Chinese medicine commonly used to treat thrombocytopenia. However, its active compounds and the mechanisms remain unclear. PURPOSE This study aims to clarify the active compounds of peanut skin to exhibit thrombogenic effects against CIT and their underlying mechanisms in vitro and in vivo. STUDY DESIGN The bioassay-guided isolation based on the proliferation of MKs was used to explore the possible platelet-enhancing ingredients in peanut skin. HSCCC technique coupled with preparative HPLC was used to separate the active compounds. Dami cells and carboplatin-treated mice model were used to evaluate the thrombogenic effects of PS-1. Network pharmacology, molecular docking, dynamics simulation studies, kinase activity, surface plasmon resonance (SPR), cellular thermal shift assay (CETSA), isothermal dose-response fingerprint (ITDRFCETSA) and western blot analysis were performed to investigate the mechanisms of PS-1. RESULTS Proanthocyanidin A1 (PS-1) and its stereoisomers (PS-2-4) were demonstrated to promote the proliferation of MKs (Dami cells), especially PS-1 (EC50 = 8.58 μM). Further studies demonstrated that PS-1 could induce the differentiation of Dami cells in dose/time-dependent manner. Biological target analysis showed that PS-1 directly bound to JAK2 (KD = 2.06 μM) to exert potent activating effect (EC50 = 0.66 μM). Oral administration of PS-1 (25 or 50 mg/kg) significantly improved CIT, but this effect was confirmed to be inhibited by JAK2 inhibitor AG490, indicating that PS-1 exerted its efficacy through JAK2 in vivo. CONCLUSION Proanthocyanins (PS-1-4) derived from peanut skin were first clarified as platelet-enhancing ingredients to improve CIT. The underlying mechanism of PS-1 was proved to promote the proliferation and differentiation of MKs via JAK2/STAT3 pathway both in vitro and in vivo.
Collapse
Affiliation(s)
- Rong Wang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Xiaolong Hu
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Jingjin Wang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Lina Zhou
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Yu Hong
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Yuanhao Zhang
- Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Suzhou 215028, People's Republic of China
| | - Fei Xiong
- State Key Laboratory of Bioelectronics, Jiangsu Laboratory for Biomaterials and Devices, Southeast University, Nanjing 210009, People's Republic of China
| | - Xiaoqi Zhang
- Institute of Traditional Chinese Medicine & Natural Products, Jinan University, Guangzhou 510632, People's Republic of China
| | - Wen-Cai Ye
- Institute of Traditional Chinese Medicine & Natural Products, Jinan University, Guangzhou 510632, People's Republic of China
| | - Hao Wang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China.
| |
Collapse
|
7
|
Voeltzel T, Fossard G, Degaud M, Geistlich K, Gadot N, Jeanpierre S, Mikaelian I, Brevet M, Anginot A, Le Bousse-Kerdilès MC, Trichet V, Lefort S, Maguer-Satta V. A minimal standardized human bone marrow microphysiological system to assess resident cell behavior during normal and pathological processes. Biomater Sci 2021; 10:485-498. [PMID: 34904143 DOI: 10.1039/d1bm01098k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Bone marrow is a complex and dynamic microenvironment that provides essential cues to resident cells. We developed a standardized three-dimensional (3D) model to decipher mechanisms that control human cells during hematological and non-hematological processes. Our simple 3D-model is constituted of a biphasic calcium phosphate-based scaffold and human cell lines to ensure a high reproducibility. We obtained a minimal well-organized bone marrow-like structure in which various cell types and secreted extracellular matrix can be observed and characterized by in situ imaging or following viable cell retrieval. The complexity of the system can be increased and customized, with each cellular component being independently modulated according to the issue investigated. Introduction of pathological elements in this 3D-system accurately reproduced changes observed in patient bone marrow. Hence, we have developed a handy and flexible standardized microphysiological system that mimics human bone marrow, allowing histological analysis and functional assays on collected cells.
Collapse
Affiliation(s)
- Thibault Voeltzel
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France.,Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France.,Université de Lyon, 69000, Lyon, France.,Department of Cancer Initiation and Tumor cell Identity and Lyon, France.,CNRS GDR 3697 MicroNiT, Tours, France.
| | - Gaëlle Fossard
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France.,Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France.,Université de Lyon, 69000, Lyon, France.,Department of Cancer Initiation and Tumor cell Identity and Lyon, France.,Hospices Civils de Lyon, Hematology Department, Centre Hospitalier Lyon Sud, F-69495 Pierre Bénite, France
| | - Michaël Degaud
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France.,Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France.,Université de Lyon, 69000, Lyon, France.,Department of Cancer Initiation and Tumor cell Identity and Lyon, France.,Hospices Civils de Lyon, Hematology Department, Centre Hospitalier Lyon Sud, F-69495 Pierre Bénite, France
| | - Kevin Geistlich
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France.,Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France.,Université de Lyon, 69000, Lyon, France.,Department of Cancer Initiation and Tumor cell Identity and Lyon, France.,Centre Léon Bérard, Lyon, France
| | - Nicolas Gadot
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France.,Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France.,Université de Lyon, 69000, Lyon, France.,Research Pathology Platform, Department of Translational Research and Innovation, Centre Léon Bérard, Lyon, France
| | - Sandrine Jeanpierre
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France.,Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France.,Université de Lyon, 69000, Lyon, France.,Department of Cancer Initiation and Tumor cell Identity and Lyon, France.,Centre Léon Bérard, Lyon, France
| | - Ivan Mikaelian
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France.,Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France.,Université de Lyon, 69000, Lyon, France.,Department of Cancer Initiation and Tumor cell Identity and Lyon, France
| | - Marie Brevet
- Pathology Department, Hospices Civils de Lyon, Bron F-69500, France
| | - Adrienne Anginot
- UMR1197, Université Paris-Saclay, 94800 Villejuif, France.,CNRS GDR 3697 MicroNiT, Tours, France.
| | | | - Valérie Trichet
- INSERM, UMR 1238, PHYOS, Faculty of Medicine, University of Nantes, Nantes, France.,CNRS GDR 3697 MicroNiT, Tours, France.
| | - Sylvain Lefort
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France.,Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France.,Université de Lyon, 69000, Lyon, France.,Department of Cancer Initiation and Tumor cell Identity and Lyon, France.,CNRS GDR 3697 MicroNiT, Tours, France.
| | - Véronique Maguer-Satta
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France.,Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France.,Université de Lyon, 69000, Lyon, France.,Department of Cancer Initiation and Tumor cell Identity and Lyon, France.,CNRS GDR 3697 MicroNiT, Tours, France. .,Centre Léon Bérard, Lyon, France
| |
Collapse
|
8
|
Warsi S, Blank U, Dahl M, Hooi Min Grahn T, Schmiderer L, Andradottir S, Karlsson S. BMP signaling is required for postnatal murine hematopoietic stem cell self-renewal. Haematologica 2021; 106:2203-2214. [PMID: 32675226 PMCID: PMC8327730 DOI: 10.3324/haematol.2019.236125] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Indexed: 12/12/2022] Open
Abstract
Life-long production of blood from hematopoietic stem cells (HSC) is a process of strict modulation. Intrinsic and extrinsic signals govern fate options like self-renewal – a cardinal feature of HSC. Bone morphogenetic proteins (BMP) have an established role in embryonic hematopoiesis, but less is known about its functions in adulthood. Previously, SMAD-mediated BMP signaling has been proven dispensable for HSC. However, the BMP type-II receptor (BMPR-II) is highly expressed in HSC, leaving the possibility that BMP function via alternative pathways. Here, we establish that BMP signaling is required for selfrenewal of adult HSC. Through conditional knockout we show that BMPR-II deficient HSC have impaired self-renewal and regenerative capacity. BMPR-II deficient cells have reduced p38 activation, implying that non-SMAD pathways operate downstream of BMP in HSC. Indeed, a majority of primitive hematopoietic cells do not engage in SMADmediated responses downstream of BMP in vivo. Furthermore, deficiency of BMPR-II results in increased expression of TJP1, a known regulator of self-renewal in other stem cells, and knockdown of TJP1 in primitive hematopoietic cells partly rescues the BMPR-II null phenotype. This suggests TJP1 may be a universal stem cell regulator. In conclusion, BMP signaling, in part mediated through TJP1, is required endogenously by adult HSC to maintain self-renewal capacity and proper resilience of the hematopoietic system during regeneration.
Collapse
Affiliation(s)
- Sarah Warsi
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Ulrika Blank
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Maria Dahl
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Tan Hooi Min Grahn
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Ludwig Schmiderer
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | | | - Stefan Karlsson
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| |
Collapse
|
9
|
Jeanpierre S, Arizkane K, Thongjuea S, Grockowiak E, Geistlich K, Barral L, Voeltzel T, Guillemin A, Gonin-Giraud S, Gandrillon O, Nicolini FE, Mead AJ, Maguer-Satta V, Lefort S. The quiescent fraction of chronic myeloid leukemic stem cells depends on BMPR1B, Stat3 and BMP4-niche signals to persist in patients in remission. Haematologica 2021; 106:111-122. [PMID: 32001529 PMCID: PMC7776261 DOI: 10.3324/haematol.2019.232793] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 01/27/2020] [Indexed: 12/16/2022] Open
Abstract
Chronic myeloid leukemia arises from the transformation of hematopoietic stem cells by the BCR-ABL oncogene. Though transformed cells are predominantly BCR-ABL-dependent and sensitive to tyrosine kinase inhibitor treatment, some BMPR1B+ leukemic stem cells are treatment-insensitive and rely, among others, on the bone morphogenetic protein (BMP) pathway for their survival via a BMP4 autocrine loop. Here, we further studied the involvement of BMP signaling in favoring residual leukemic stem cell persistence in the BM of patients having achieved remission under treatment. We demonstrate by single-cell RNASequencing analysis that a sub-fraction of surviving BMPR1B+ leukemic stem cells are co-enriched in BMP signaling, quiescence and stem cell signatures, without modulation of the canonical BMP target genes, but enrichment in actors of the Jak2/Stat3 signaling pathway. Indeed, based on a new model of persisting CD34+CD38– leukemic stem cells, we show that BMPR1B+ cells display co-activated Smad1/5/8 and Stat3 pathways. Interestingly, we reveal that only the BMPR1B+ cells adhering to stromal cells display a quiescent status. Surprisingly, this quiescence is induced by treatment, while non-adherent BMPR1B+ cells treated with tyrosine kinase inhibitors continued to proliferate. The subsequent targeting of BMPR1B and Jak2 pathways decreased quiescent leukemic stem cells by promoting their cell cycle re-entry and differentiation. Moreover, while Jak2-inhibitors alone increased BMP4 production by mesenchymal cells, the addition of the newly described BMPR1B inhibitor (E6201) impaired BMP4-mediated production by stromal cells. Altogether, our data demonstrate that targeting both BMPR1B and Jak2/Stat3 efficiently impacts persisting and dormant leukemic stem cells hidden in their BM microenvironment.
Collapse
Affiliation(s)
| | | | | | | | | | - Lea Barral
- Centre de Recherche en Cancérologie de Lyon
| | | | - Anissa Guillemin
- Laboratoire de biologie et modélisation de la cellule. LBMC - Ecole Normale Superieure - Lyon
| | - Sandrine Gonin-Giraud
- Laboratoire de biologie et modélisation de la cellule. LBMC - Ecole Normale Superieure - Lyon
| | - Olivier Gandrillon
- Laboratoire de biologie et modélisation de la cellule. LBMC - Ecole Normale Superieure - Lyon
| | | | | | | | | |
Collapse
|
10
|
Targeting BMP signaling in the bone marrow microenvironment of myeloid leukemia. Biochem Soc Trans 2020; 48:411-418. [DOI: 10.1042/bst20190223] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 02/18/2020] [Accepted: 02/18/2020] [Indexed: 12/29/2022]
Abstract
The bone morphogenetic protein (BMP) pathway regulates the fate and proliferation of normal hematopoietic stem cells (HSC) as well as interactions with their niche. While BMP2 and BMP4 promote HSC differentiation, only BMP4 maintains HSC pool and favors interactions with their niche. In myeloid leukemia, we have identified intrinsic and extrinsic dysregulations of the BMP pathway in Chronic Myeloid Leukemia (CML) and Acute Myeloid leukemia (AML) responsible for leukemic stem cells (LSC) survival. In AML, BMP pathway alterations sustain and promote resistant immature-like leukemic cells by activating a new signaling cascade. Binding of BMP4 to BMPR1A leads to ΔNp73 expression, which in turn induces NANOG, altogether associated with a poor patient's prognosis. Despite efficient targeted therapies, like Tyrosine Kinase Inhibitors (TKI) in CML, many patients retain LSCs. Our laboratory demonstrated that the BMP pathway sustains a permanent pool of LSCs expressing high levels of BMPR1B receptor, that evolve upon treatment to progressively implement a BMP4 autocrine loop, leading to TKI-resistant cells. Single cell RNA-Seq analysis of TKI-persisting LSCs showed a co-enrichment of BMP with Jak2-signaling, quiescence and stem cell (SC) signatures. Using a new model of persisting LSCs, we recently demonstrated that BMPR1B+ cells display co-activated Smad1/5/8 and Stat3 pathways and could be targeted by blocking BMPR1B/Jak2 signal. Lastly, a specific BMPR1B inhibitor impaired BMP4-mediated LSC protection against TKIs. Altogether, data based on various studies including ours, indicate that BMP targeting could eliminate leukemic cells within a protective bone marrow microenvironment to efficiently impact residual resistance or persistence of LSCs in myeloid leukemia.
Collapse
|
11
|
Bone marrow niche crosses paths with BMPs: a road to protection and persistence in CML. Biochem Soc Trans 2020; 47:1307-1325. [PMID: 31551354 DOI: 10.1042/bst20190221] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 08/23/2019] [Accepted: 08/29/2019] [Indexed: 12/21/2022]
Abstract
Chronic myeloid leukaemia (CML) is a paradigm of precision medicine, being one of the first cancers to be treated with targeted therapy. This has revolutionised CML therapy and patient outcome, with high survival rates. However, this now means an ever-increasing number of patients are living with the disease on life-long tyrosine kinase inhibitor (TKI) therapy, with most patients anticipated to have near normal life expectancy. Unfortunately, in a significant number of patients, TKIs are not curative. This low-level disease persistence suggests that despite a molecularly targeted therapeutic approach, there are BCR-ABL1-independent mechanisms exploited to sustain the survival of a small cell population of leukaemic stem cells (LSCs). In CML, LSCs display many features akin to haemopoietic stem cells, namely quiescence, self-renewal and the ability to produce mature progeny, this all occurs through intrinsic and extrinsic signals within the specialised microenvironment of the bone marrow (BM) niche. One important avenue of investigation in CML is how the disease highjacks the BM, thereby remodelling this microenvironment to create a niche, which enables LSC persistence and resistance to TKI treatment. In this review, we explore how changes in growth factor levels, in particular, the bone morphogenetic proteins (BMPs) and pro-inflammatory cytokines, impact on cell behaviour, extracellular matrix deposition and bone remodelling in CML. We also discuss the challenges in targeting LSCs and the potential of dual targeting using combination therapies against BMP receptors and BCR-ABL1.
Collapse
|
12
|
Voeltzel T, Flores-Violante M, Zylbersztejn F, Lefort S, Billandon M, Jeanpierre S, Joly S, Fossard G, Milenkov M, Mazurier F, Nehme A, Belhabri A, Paubelle E, Thomas X, Michallet M, Louache F, Nicolini FE, Caron de Fromentel C, Maguer-Satta V. A new signaling cascade linking BMP4, BMPR1A, ΔNp73 and NANOG impacts on stem-like human cell properties and patient outcome. Cell Death Dis 2018; 9:1011. [PMID: 30262802 PMCID: PMC6160490 DOI: 10.1038/s41419-018-1042-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 07/20/2018] [Accepted: 08/20/2018] [Indexed: 12/23/2022]
Abstract
In a significant number of cases cancer therapy is followed by a resurgence of more aggressive tumors derived from immature cells. One example is acute myeloid leukemia (AML), where an accumulation of immature cells is responsible for relapse following treatment. We previously demonstrated in chronic myeloid leukemia that the bone morphogenetic proteins (BMP) pathway is involved in stem cell fate and contributes to transformation, expansion, and persistence of leukemic stem cells. Here, we have identified intrinsic and extrinsic dysregulations of the BMP pathway in AML patients at diagnosis. BMP2 and BMP4 protein concentrations are elevated within patients’ bone marrow with a BMP4-dominant availability. This overproduction likely depends on the bone marrow microenvironment, since MNCs do not overexpress BMP4 transcripts. Intrinsically, the receptor BMPR1A transcript is increased in leukemic samples with more cells presenting this receptor at the membrane. This high expression of BMPR1A is further increased upon BMP4 exposure, specifically in AML cells. Downstream analysis demonstrated that BMP4 controls the expression of the survival factor ΔNp73 through its binding to BMPR1A. At the functional level, this results in the direct induction of NANOG expression and an increase of stem-like features in leukemic cells, as shown by ALDH and functional assays. In addition, we identified for the first time a strong correlation between ΔNp73, BMPR1A and NANOG expression with patient outcome. These results highlight a new signaling cascade initiated by tumor environment alterations leading to stem-cell features and poor patients’ outcome.
Collapse
Affiliation(s)
- Thibault Voeltzel
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 69000, Lyon, France. .,Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69008, Lyon, France. .,Université de Lyon, 69000, Lyon, France. .,Department of Tumor Escape Signaling, INSERM U1052, CNRS UMR5286, 69000, Lyon, France. .,Université de Lyon 1, 69000, Lyon, France.
| | - Mario Flores-Violante
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 69000, Lyon, France.,Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69008, Lyon, France.,Université de Lyon, 69000, Lyon, France.,Department of Tumor Escape Signaling, INSERM U1052, CNRS UMR5286, 69000, Lyon, France.,Université de Lyon 1, 69000, Lyon, France
| | - Florence Zylbersztejn
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 69000, Lyon, France.,Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69008, Lyon, France.,Université de Lyon, 69000, Lyon, France.,Department of Tumor Escape Signaling, INSERM U1052, CNRS UMR5286, 69000, Lyon, France.,Université de Lyon 1, 69000, Lyon, France
| | - Sylvain Lefort
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 69000, Lyon, France.,Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69008, Lyon, France.,Université de Lyon, 69000, Lyon, France.,Department of Tumor Escape Signaling, INSERM U1052, CNRS UMR5286, 69000, Lyon, France.,Université de Lyon 1, 69000, Lyon, France
| | - Marion Billandon
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 69000, Lyon, France.,Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69008, Lyon, France.,Université de Lyon, 69000, Lyon, France.,Department of Tumor Escape Signaling, INSERM U1052, CNRS UMR5286, 69000, Lyon, France.,Université de Lyon 1, 69000, Lyon, France
| | - Sandrine Jeanpierre
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 69000, Lyon, France.,Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69008, Lyon, France.,Université de Lyon, 69000, Lyon, France.,Department of Tumor Escape Signaling, INSERM U1052, CNRS UMR5286, 69000, Lyon, France.,Université de Lyon 1, 69000, Lyon, France.,Centre Léon Bérard, 69000, Lyon, France
| | - Stéphane Joly
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 69000, Lyon, France.,Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69008, Lyon, France.,Université de Lyon, 69000, Lyon, France.,Department of Tumor Escape Signaling, INSERM U1052, CNRS UMR5286, 69000, Lyon, France.,Université de Lyon 1, 69000, Lyon, France
| | - Gaelle Fossard
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 69000, Lyon, France.,Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69008, Lyon, France.,Université de Lyon, 69000, Lyon, France.,Department of Tumor Escape Signaling, INSERM U1052, CNRS UMR5286, 69000, Lyon, France.,Université de Lyon 1, 69000, Lyon, France.,Hospices Civils de Lyon, Hematology Department, Centre Hospitalier Lyon Sud, 69495, Pierre Bénite, France
| | - Milen Milenkov
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 69000, Lyon, France.,Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69008, Lyon, France.,Université de Lyon, 69000, Lyon, France.,Department of Tumor Escape Signaling, INSERM U1052, CNRS UMR5286, 69000, Lyon, France.,Université de Lyon 1, 69000, Lyon, France
| | - Frédéric Mazurier
- CNRS ERL 7001, 37032, Tours, France.,CNRS GDR 3697 MicroNiT, Tours, France
| | | | - Amine Belhabri
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 69000, Lyon, France.,Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69008, Lyon, France.,Université de Lyon, 69000, Lyon, France.,Department of Tumor Escape Signaling, INSERM U1052, CNRS UMR5286, 69000, Lyon, France.,Université de Lyon 1, 69000, Lyon, France.,Centre Léon Bérard, 69000, Lyon, France
| | - Etienne Paubelle
- Hospices Civils de Lyon, Hematology Department, Centre Hospitalier Lyon Sud, 69495, Pierre Bénite, France
| | - Xavier Thomas
- Hospices Civils de Lyon, Hematology Department, Centre Hospitalier Lyon Sud, 69495, Pierre Bénite, France
| | - Mauricette Michallet
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 69000, Lyon, France.,Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69008, Lyon, France.,Université de Lyon, 69000, Lyon, France.,Department of Tumor Escape Signaling, INSERM U1052, CNRS UMR5286, 69000, Lyon, France.,Université de Lyon 1, 69000, Lyon, France.,Centre Léon Bérard, 69000, Lyon, France
| | - Fawzia Louache
- CNRS GDR 3697 MicroNiT, Tours, France.,Inserm, UMR1170, 94000, Villejuif, France
| | - Franck-Emmanuel Nicolini
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 69000, Lyon, France.,Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69008, Lyon, France.,Université de Lyon, 69000, Lyon, France.,Department of Tumor Escape Signaling, INSERM U1052, CNRS UMR5286, 69000, Lyon, France.,Université de Lyon 1, 69000, Lyon, France.,Centre Léon Bérard, 69000, Lyon, France
| | - Claude Caron de Fromentel
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 69000, Lyon, France.,Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69008, Lyon, France.,Université de Lyon, 69000, Lyon, France.,Université de Lyon 1, 69000, Lyon, France
| | - Véronique Maguer-Satta
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 69000, Lyon, France. .,Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69008, Lyon, France. .,Université de Lyon, 69000, Lyon, France. .,Department of Tumor Escape Signaling, INSERM U1052, CNRS UMR5286, 69000, Lyon, France. .,Université de Lyon 1, 69000, Lyon, France. .,CNRS GDR 3697 MicroNiT, Tours, France.
| |
Collapse
|
13
|
Bhat FA, Advani J, Khan AA, Mohan S, Pal A, Gowda H, Chakrabarti P, Keshava Prasad TS, Chatterjee A. A network map of thrombopoietin signaling. J Cell Commun Signal 2018; 12:737-743. [PMID: 30039510 DOI: 10.1007/s12079-018-0480-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 07/05/2018] [Indexed: 10/28/2022] Open
Abstract
Thrombopoietin (THPO), also known as megakaryocyte growth and development factor (MGDF), is a cytokine involved in the production of platelets. THPO is a glycoprotein produced by liver and kidney. It regulates the production of platelets by stimulating the differentiation and maturation of megakaryocyte progenitors. It acts as a ligand for MPL receptor, a member of the hematopoietic cytokine receptor superfamily and is essential for megakaryocyte maturation. THPO binding induces homodimerization of the receptor which results in activation of JAKSTAT and MAPK signaling cascades that subsequently control cellular proliferation, differentiation and other signaling events. Despite the importance of THPO signaling in various diseases and biological processes, a detailed signaling network of THPO is not available in any publicly available database. Therefore, in this study, we present a resource of signaling events induced by THPO that was manually curated from published literature on THPO. Our manual curation of thrombopoietin pathway resulted in identification of 48 molecular associations, 66 catalytic reactions, 100 gene regulation events, 19 protein translocation events and 43 activation/inhibition reactions that occur upon activation of thrombopoietin receptor by THPO. THPO signaling pathway is made available on NetPath, a freely available human signaling pathway resource developed previously by our group. We believe this resource will provide a platform for scientific community to accelerate further research in this area on potential therapeutic interventions.
Collapse
Affiliation(s)
- Firdous A Bhat
- Institute of Bioinformatics, International Technology Park, Bangalore, 560 066, India.,School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, 690525, India
| | - Jayshree Advani
- Institute of Bioinformatics, International Technology Park, Bangalore, 560 066, India.,Manipal Academy of Higher Education, Manipal, 576104, India
| | - Aafaque Ahmad Khan
- Institute of Bioinformatics, International Technology Park, Bangalore, 560 066, India.,School of Biotechnology, Kalinga Institute of Industrial Technology, Bhubaneswar, 751024, India
| | - Sonali Mohan
- Institute of Bioinformatics, International Technology Park, Bangalore, 560 066, India
| | - Arnab Pal
- Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Harsha Gowda
- Institute of Bioinformatics, International Technology Park, Bangalore, 560 066, India
| | - Prantar Chakrabarti
- Department of Haematology, Nil Ratan Sircar Medical College and Hospital, Kolkata, 700014, India
| | - T S Keshava Prasad
- Institute of Bioinformatics, International Technology Park, Bangalore, 560 066, India. .,Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), University Road, Mangalore, 575018, India.
| | - Aditi Chatterjee
- Institute of Bioinformatics, International Technology Park, Bangalore, 560 066, India.
| |
Collapse
|
14
|
Zylbersztejn F, Flores-Violante M, Voeltzel T, Nicolini FE, Lefort S, Maguer-Satta V. The BMP pathway: A unique tool to decode the origin and progression of leukemia. Exp Hematol 2018; 61:36-44. [PMID: 29477370 DOI: 10.1016/j.exphem.2018.02.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 02/08/2018] [Accepted: 02/13/2018] [Indexed: 12/25/2022]
Abstract
The microenvironment (niche) governs the fate of stem cells (SCs) by balancing self-renewal and differentiation. Increasing evidence indicates that the tumor niche plays an active role in cancer, but its important properties for tumor initiation progression and resistance remain to be identified. Clinical data show that leukemic stem cell (LSC) survival is responsible for disease persistence and drug resistance, probably due to their sustained interactions with the tumor niche. Bone morphogenetic protein (BMP) signaling is a key pathway controlling stem cells and their niche. BMP2 and BMP4 are important in both the normal and the cancer context. Several studies have revealed profound alterations of the BMP signaling in cancer SCs, with major deregulations of the BMP receptors and their downstream signaling elements. This was illustrated in the hematopoietic system by pioneer studies in chronic myelogenous leukemia that may now be expanded to acute myeloid leukemia and lymphoid leukemia, as reviewed here. At diagnosis, cells from the leukemic microenvironment are the major providers of soluble BMPs. Conversely, LSCs display altered receptors and downstream BMP signaling elements accompanied by altered functional responses to BMPs. These studies reveal the role of BMPs in tumor initiation, in addition to their known effects in later stages of transformation and progression. They also reveal the importance of BMPs in fueling cell transformation and expansion by overamplifying a natural SC response. This mechanism may explain the survival of LSCs independently of the initial oncogenic event and therefore may be involved in resistance processes.
Collapse
Affiliation(s)
- Florence Zylbersztejn
- Centre National de la Recherche Scientifique Unité Mixte de Recherche 5286, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Université de Lyon, 69000, Lyon, France; Department of Signaling of Tumor Escape, Lyon, France
| | - Mario Flores-Violante
- Centre National de la Recherche Scientifique Unité Mixte de Recherche 5286, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Université de Lyon, 69000, Lyon, France; Department of Signaling of Tumor Escape, Lyon, France
| | - Thibault Voeltzel
- Centre National de la Recherche Scientifique Unité Mixte de Recherche 5286, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Université de Lyon, 69000, Lyon, France; Department of Signaling of Tumor Escape, Lyon, France
| | - Franck-Emmanuel Nicolini
- Centre National de la Recherche Scientifique Unité Mixte de Recherche 5286, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Université de Lyon, 69000, Lyon, France; Department of Signaling of Tumor Escape, Lyon, France; Centre Léon Bérard, 69000 Lyon, France
| | - Sylvain Lefort
- Centre National de la Recherche Scientifique Unité Mixte de Recherche 5286, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Université de Lyon, 69000, Lyon, France; Department of Signaling of Tumor Escape, Lyon, France
| | - Véronique Maguer-Satta
- Centre National de la Recherche Scientifique Unité Mixte de Recherche 5286, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Université de Lyon, 69000, Lyon, France; Department of Signaling of Tumor Escape, Lyon, France.
| |
Collapse
|
15
|
Calpe S, Correia ACP, Sancho-Serra MDC, Krishnadath KK. Comparison of newly developed anti-bone morphogenetic protein 4 llama-derived antibodies with commercially available BMP4 inhibitors. MAbs 2017; 8:678-88. [PMID: 26967714 PMCID: PMC4966848 DOI: 10.1080/19420862.2016.1158380] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Due to improved understanding of the role of bone morphogenetic protein 4 (BMP4) in an increasing number of diseases, the development of selective inhibitors of BMP4 is an attractive therapeutic option. The currently available BMP4 inhibitors are not suitable as therapeutics because of their low specificity and low effectiveness. Here, we compared newly generated anti-BMP4 llama-derived antibodies (VHHs) with 3 different types of commercially available BMP4 inhibitors, natural antagonists, small molecule BMPR inhibitors and conventional anti-BMP4 monoclonal antibodies. We found that the anti-BMP4 VHHs were as effective as the natural antagonist or small molecule inhibitors, but had higher specificity. We also showed that commercial anti-BMP4 antibodies were inferior in terms of both specificity and effectiveness. These findings might result from the fact that the VHHs C4C4 and C8C8 target a small region within the BMPR1 epitope of BMP4, whereas the commercial antibodies target other areas of the BMP4 molecule. Our results show that the newly developed anti-BMP4 VHHs are promising antibodies with better specificity and effectivity for inhibition of BMP4, making them an attractive tool for research and for therapeutic applications.
Collapse
Affiliation(s)
- Silvia Calpe
- a Center for Experimental & Molecular Medicine , Academic Medical Center , Meibergdreef , Amsterdam , The Netherlands
| | - Ana C P Correia
- a Center for Experimental & Molecular Medicine , Academic Medical Center , Meibergdreef , Amsterdam , The Netherlands
| | - Maria Del Carmen Sancho-Serra
- a Center for Experimental & Molecular Medicine , Academic Medical Center , Meibergdreef , Amsterdam , The Netherlands
| | - Kausilia K Krishnadath
- a Center for Experimental & Molecular Medicine , Academic Medical Center , Meibergdreef , Amsterdam , The Netherlands.,b Department of Gastroenterology & Hepatology , Academic Medical Center , Meibergdreef, Amsterdam , The Netherlands
| |
Collapse
|
16
|
Immature CML cells implement a BMP autocrine loop to escape TKI treatment. Blood 2017; 130:2860-2871. [PMID: 29138221 DOI: 10.1182/blood-2017-08-801019] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 11/03/2017] [Indexed: 12/14/2022] Open
Abstract
The BCR-ABL specific tyrosine kinase inhibitors (TKI) changed the outcome of chronic myeloid leukemia (CML), turning a life-threatening disease into a chronic illness. However, TKI are not yet curative, because most patients retain leukemic stem cells (LSC) and their progenitors in bone marrow and relapse following treatment cessation. At diagnosis, deregulation of the bone morphogenetic protein (BMP) pathway is involved in LSC and progenitor expansion. Here, we report that BMP pathway alterations persist in TKI-resistant patients. In comparison with patients in complete cytogenetic remission, TKI-resistant LSC and progenitors display high levels of BMPR1b expression and alterations of its cellular localization. In vitro treatment of immature chronic phase CML cells with TKI alone, or in combination with interferon-α, results in the preferential survival of BMPR1b+ cells. We demonstrated persistent and increasing BMP4 production by patients' mesenchymal cells with resistance. Patient follow-up revealed an increase of BMPR1b expression and in BMP4 expression in LSC from TKI-resistant patients in comparison with diagnosis, while remaining unchanged in sensitive patients. Both leukemic and nonleukemic cells exhibit higher BMP4 levels in the bone marrow of TKI-resistant patients. Exposure to BMP2/BMP4 does not alter BCR-ABL transcript expression but is accompanied by the overexpression of TWIST-1, a transcription factor highly expressed in resistant LSC. By modulating BMP4 or BMPR1b expression, we show that these elements are involved in TKI resistance. In summary, we reveal that persistence of BMP alterations and existence of an autocrine loop promote CML-primitive cells' TKI resistance.
Collapse
|
17
|
Emerging roles of the bone morphogenetic protein pathway in cancer: potential therapeutic target for kinase inhibition. Biochem Soc Trans 2017; 44:1117-34. [PMID: 27528760 DOI: 10.1042/bst20160069] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Indexed: 12/15/2022]
Abstract
Bone morphogenetic proteins (BMPs) belong to the transforming growth factor-β (TGF-β) family signalling pathway. Similar to TGF-β, the complex roles of BMPs in development and disease are demonstrated by their dichotomous roles in various cancers and cancer stages. Although early studies implicated BMP signalling in tumour suppressive phenotypes, the results of more recent experiments recognize BMPs as potent tumour promoters. Many of these complexities are becoming illuminated by understanding the role of BMPs in their contextual role in unique cell types of cancer and the impact of their surrounding tumour microenvironment. Here we review the emerging roles of BMP signalling in cancer, with a focus on the molecular underpinnings of BMP signalling in individual cancers as a valid therapeutic target for cancer prevention and treatment.
Collapse
|
18
|
Wang Y, Jiang L, Mo X, Lan Y, Yang X, Liu X, Zhang J, Zhu L, Liu J, Wu X. Megakaryocytic Smad4 Regulates Platelet Function through Syk and ROCK2 Expression. Mol Pharmacol 2017; 92:285-296. [PMID: 28663280 DOI: 10.1124/mol.116.107417] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 06/21/2017] [Indexed: 01/03/2023] Open
Abstract
Smad4, a key transcription factor in the transforming growth factor-β signaling pathway, is involved in a variety of cell physiologic and pathologic processes. Here, we characterized megakaryocyte/platelet-specific Smad4 deficiency in mice to elucidate its effect on platelet function. We found that megakaryocyte/platelet-specific loss of Smad4 caused mild thrombocytopenia and significantly extended first occlusion time and tail bleeding time in mice. Smad4-deficient platelets showed reduced agonist-induced platelet aggregation. Further studies showed that a severe defect was seen in integrin αIIbβ3-mediated bidirectional (inside-out and outside-in) signaling in Smad4-deficient platelets, as evidenced by reduced fibrinogen binding and α-granule secretion, suppressed platelet spreading and clot retraction. Microarray analysis showed that the expression levels of multiple genes were altered in Smad4-deficient platelets. Among these genes, spleen tyrosine kinase (Syk) and Rho-associated coiled-coil containing protein kinase 2 (ROCK2) were downregulated several times as confirmed by quantitative reverse-transcription polymerase chain reaction and immunoblotting. Further research showed that Smad4 directly regulates ROCK2 transcription but indirectly regulates Syk. Megakaryocyte/platelet-specific Smad4 deficiency caused decreased expression levels of Syk and ROCK2 in platelets. These results suggest potential links among Smad4 deficiency, attenuated Syk, and ROCK2 expression and defective platelet activation.
Collapse
Affiliation(s)
- Yanhua Wang
- Department of Laboratory Medicine, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, People's Republic of China (Y.W.); Institute for Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai, People's Republic of China (L.J., X.M.); State Key Laboratory of Proteomics, Genetic Laboratory of Development and Diseases, Institute of Biotechnology, Beijing, People's Republic of China (Y.L., X.Y.); Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China (X.L., J.Z.); Cyrus Tang Hematology Center, Soochow University, Suzhou, People's Republic of China (L.Z.); Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China (J.L.); and The Central Laboratory of The Eighth People's Hospital of Shanghai, Shanghai, People's Republic of China (X.W.)
| | - Lirong Jiang
- Department of Laboratory Medicine, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, People's Republic of China (Y.W.); Institute for Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai, People's Republic of China (L.J., X.M.); State Key Laboratory of Proteomics, Genetic Laboratory of Development and Diseases, Institute of Biotechnology, Beijing, People's Republic of China (Y.L., X.Y.); Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China (X.L., J.Z.); Cyrus Tang Hematology Center, Soochow University, Suzhou, People's Republic of China (L.Z.); Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China (J.L.); and The Central Laboratory of The Eighth People's Hospital of Shanghai, Shanghai, People's Republic of China (X.W.)
| | - Xi Mo
- Department of Laboratory Medicine, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, People's Republic of China (Y.W.); Institute for Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai, People's Republic of China (L.J., X.M.); State Key Laboratory of Proteomics, Genetic Laboratory of Development and Diseases, Institute of Biotechnology, Beijing, People's Republic of China (Y.L., X.Y.); Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China (X.L., J.Z.); Cyrus Tang Hematology Center, Soochow University, Suzhou, People's Republic of China (L.Z.); Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China (J.L.); and The Central Laboratory of The Eighth People's Hospital of Shanghai, Shanghai, People's Republic of China (X.W.)
| | - Yu Lan
- Department of Laboratory Medicine, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, People's Republic of China (Y.W.); Institute for Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai, People's Republic of China (L.J., X.M.); State Key Laboratory of Proteomics, Genetic Laboratory of Development and Diseases, Institute of Biotechnology, Beijing, People's Republic of China (Y.L., X.Y.); Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China (X.L., J.Z.); Cyrus Tang Hematology Center, Soochow University, Suzhou, People's Republic of China (L.Z.); Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China (J.L.); and The Central Laboratory of The Eighth People's Hospital of Shanghai, Shanghai, People's Republic of China (X.W.)
| | - Xiao Yang
- Department of Laboratory Medicine, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, People's Republic of China (Y.W.); Institute for Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai, People's Republic of China (L.J., X.M.); State Key Laboratory of Proteomics, Genetic Laboratory of Development and Diseases, Institute of Biotechnology, Beijing, People's Republic of China (Y.L., X.Y.); Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China (X.L., J.Z.); Cyrus Tang Hematology Center, Soochow University, Suzhou, People's Republic of China (L.Z.); Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China (J.L.); and The Central Laboratory of The Eighth People's Hospital of Shanghai, Shanghai, People's Republic of China (X.W.)
| | - Xinyi Liu
- Department of Laboratory Medicine, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, People's Republic of China (Y.W.); Institute for Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai, People's Republic of China (L.J., X.M.); State Key Laboratory of Proteomics, Genetic Laboratory of Development and Diseases, Institute of Biotechnology, Beijing, People's Republic of China (Y.L., X.Y.); Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China (X.L., J.Z.); Cyrus Tang Hematology Center, Soochow University, Suzhou, People's Republic of China (L.Z.); Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China (J.L.); and The Central Laboratory of The Eighth People's Hospital of Shanghai, Shanghai, People's Republic of China (X.W.)
| | - Jian Zhang
- Department of Laboratory Medicine, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, People's Republic of China (Y.W.); Institute for Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai, People's Republic of China (L.J., X.M.); State Key Laboratory of Proteomics, Genetic Laboratory of Development and Diseases, Institute of Biotechnology, Beijing, People's Republic of China (Y.L., X.Y.); Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China (X.L., J.Z.); Cyrus Tang Hematology Center, Soochow University, Suzhou, People's Republic of China (L.Z.); Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China (J.L.); and The Central Laboratory of The Eighth People's Hospital of Shanghai, Shanghai, People's Republic of China (X.W.)
| | - Li Zhu
- Department of Laboratory Medicine, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, People's Republic of China (Y.W.); Institute for Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai, People's Republic of China (L.J., X.M.); State Key Laboratory of Proteomics, Genetic Laboratory of Development and Diseases, Institute of Biotechnology, Beijing, People's Republic of China (Y.L., X.Y.); Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China (X.L., J.Z.); Cyrus Tang Hematology Center, Soochow University, Suzhou, People's Republic of China (L.Z.); Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China (J.L.); and The Central Laboratory of The Eighth People's Hospital of Shanghai, Shanghai, People's Republic of China (X.W.)
| | - Junling Liu
- Department of Laboratory Medicine, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, People's Republic of China (Y.W.); Institute for Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai, People's Republic of China (L.J., X.M.); State Key Laboratory of Proteomics, Genetic Laboratory of Development and Diseases, Institute of Biotechnology, Beijing, People's Republic of China (Y.L., X.Y.); Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China (X.L., J.Z.); Cyrus Tang Hematology Center, Soochow University, Suzhou, People's Republic of China (L.Z.); Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China (J.L.); and The Central Laboratory of The Eighth People's Hospital of Shanghai, Shanghai, People's Republic of China (X.W.)
| | - Xiaolin Wu
- Department of Laboratory Medicine, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, People's Republic of China (Y.W.); Institute for Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai, People's Republic of China (L.J., X.M.); State Key Laboratory of Proteomics, Genetic Laboratory of Development and Diseases, Institute of Biotechnology, Beijing, People's Republic of China (Y.L., X.Y.); Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China (X.L., J.Z.); Cyrus Tang Hematology Center, Soochow University, Suzhou, People's Republic of China (L.Z.); Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China (J.L.); and The Central Laboratory of The Eighth People's Hospital of Shanghai, Shanghai, People's Republic of China (X.W.)
| |
Collapse
|
19
|
Clément F, Xu X, Donini CF, Clément A, Omarjee S, Delay E, Treilleux I, Fervers B, Le Romancer M, Cohen PA, Maguer-Satta V. Long-term exposure to bisphenol A or benzo(a)pyrene alters the fate of human mammary epithelial stem cells in response to BMP2 and BMP4, by pre-activating BMP signaling. Cell Death Differ 2016; 24:155-166. [PMID: 27740625 PMCID: PMC5260492 DOI: 10.1038/cdd.2016.107] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 08/31/2016] [Accepted: 08/31/2016] [Indexed: 12/11/2022] Open
Abstract
Bone morphogenetic protein 2 (BMP2) and BMP4 are key regulators of the fate and differentiation of human mammary epithelial stem cells (SCs), as well as of their niches, and are involved in breast cancer development. We established that MCF10A immature mammary epithelial cells reliably reproduce the BMP response that we previously identified in human primary epithelial SCs. In this model, we observed that BMP2 promotes luminal progenitor commitment and expansion, whereas BMP4 prevents lineage differentiation. Environmental pollutants are known to promote cancer development, possibly by providing cells with stem-like features and by modifying their niches. Bisphenols, in particular, were shown to increase the risk of developing breast cancer. Here, we demonstrate that chronic exposure to low doses of bisphenol A (BPA) or benzo(a)pyrene (B(a)P) alone has little effect on SCs properties of MCF10A cells. Conversely, we show that this exposure affects the response of immature epithelial cells to BMP2 and BMP4. Furthermore, the modifications triggered in MCF10A cells on exposure to pollutants appeared to be predominantly mediated by altering the expression and localization of type-1 receptors and by pre-activating BMP signaling, through the phosphorylation of small mothers against decapentaplegic 1/5/8 (SMAD1/5/8). By analyzing stem and progenitor properties, we reveal that BPA prevents the maintenance of SC features prompted by BMP4, whereas promoting cell differentiation towards a myoepithelial phenotype. Inversely, B(a)P prevents BMP2-mediated luminal progenitor commitment and expansion, leading to the retention of stem-like properties. Overall, our data indicate that BPA and B(a)P distinctly alter the fate and differentiation potential of mammary epithelial SCs by modulating BMP signaling.
Collapse
Affiliation(s)
- Flora Clément
- Univ Lyon, Université Claude Bernard Lyon 1, Lyon, F-69008, France.,Univ Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon Cedex 08, F-69008, France.,Department of Tumor Escape Signaling, Centre Léon Bérard, Lyon, France
| | - Xinyi Xu
- Univ Lyon, Université Claude Bernard Lyon 1, Lyon, F-69008, France.,Univ Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon Cedex 08, F-69008, France.,Department of Tumor Escape Signaling, Centre Léon Bérard, Lyon, France
| | - Caterina F Donini
- Univ Lyon, Université Claude Bernard Lyon 1, Lyon, F-69008, France.,Univ Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon Cedex 08, F-69008, France.,Department of Cancer and Environnement, Centre Léon Bérard, Lyon, France
| | - Alice Clément
- Univ Lyon, Université Claude Bernard Lyon 1, Lyon, F-69008, France.,Univ Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon Cedex 08, F-69008, France.,Department of Tumor Escape Signaling, Centre Léon Bérard, Lyon, France
| | - Soleilmane Omarjee
- Univ Lyon, Université Claude Bernard Lyon 1, Lyon, F-69008, France.,Univ Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon Cedex 08, F-69008, France.,Department of Cancer Cell Plasticity, Centre Léon Bérard, Lyon, France
| | - Emmanuel Delay
- Univ Lyon, Université Claude Bernard Lyon 1, Lyon, F-69008, France.,Univ Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon Cedex 08, F-69008, France.,Department of Tumor Escape Signaling, Centre Léon Bérard, Lyon, France.,Centre Léon Bérard, Lyon, France
| | - Isabelle Treilleux
- Univ Lyon, Université Claude Bernard Lyon 1, Lyon, F-69008, France.,Univ Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon Cedex 08, F-69008, France.,Department of Cancer Cell Plasticity, Centre Léon Bérard, Lyon, France.,Centre Léon Bérard, Lyon, France
| | - Béatrice Fervers
- Univ Lyon, Université Claude Bernard Lyon 1, Lyon, F-69008, France.,Univ Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon Cedex 08, F-69008, France.,Department of Cancer and Environnement, Centre Léon Bérard, Lyon, France
| | - Muriel Le Romancer
- Univ Lyon, Université Claude Bernard Lyon 1, Lyon, F-69008, France.,Univ Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon Cedex 08, F-69008, France.,Department of Cancer Cell Plasticity, Centre Léon Bérard, Lyon, France
| | - Pascale A Cohen
- Univ Lyon, Université Claude Bernard Lyon 1, Lyon, F-69008, France.,Univ Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon Cedex 08, F-69008, France.,Department of Cancer and Environnement, Centre Léon Bérard, Lyon, France
| | - Véronique Maguer-Satta
- Univ Lyon, Université Claude Bernard Lyon 1, Lyon, F-69008, France.,Univ Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon Cedex 08, F-69008, France.,Department of Tumor Escape Signaling, Centre Léon Bérard, Lyon, France.,CNRS GDR 3697 Micronit, Tours, France
| |
Collapse
|
20
|
Chapellier M, Maguer-Satta V. BMP2, a key to uncover luminal breast cancer origin linked to pollutant effects on epithelial stem cells niche. Mol Cell Oncol 2015; 3:e1026527. [PMID: 27314065 PMCID: PMC4909443 DOI: 10.1080/23723556.2015.1026527] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 03/02/2015] [Accepted: 03/02/2015] [Indexed: 11/15/2022]
Abstract
Chronic exposure of epithelial cells to high levels of bone morphogenetic protein 2 (BMP2) has recently been demonstrated to initiate stem cell transformation toward a luminal tumor-like phenotype through a BMP2–BMPR1B-dependent mechanism. Carcinogen-driven deregulation of the stem cell niche could therefore represent a driving force to promote transformation and dictate the ultimate breast tumor subtype.
Collapse
Affiliation(s)
| | - Véronique Maguer-Satta
- CNRS UMR5286; Center de Recherche en Cancérologie de Lyon; Lyon, France; Inserm U1052; Center de Recherche en Cancérologie de Lyon; Lyon, France; Université de Lyon;, Lyon, France; Department of Immunity, Virus and Microenvironment; Lyon, France
| |
Collapse
|
21
|
Chapellier M, Bachelard-Cascales E, Schmidt X, Clément F, Treilleux I, Delay E, Jammot A, Ménétrier-Caux C, Pochon G, Besançon R, Voeltzel T, Caron de Fromentel C, Caux C, Blay JY, Iggo R, Maguer-Satta V. Disequilibrium of BMP2 levels in the breast stem cell niche launches epithelial transformation by overamplifying BMPR1B cell response. Stem Cell Reports 2015; 4:239-54. [PMID: 25601208 PMCID: PMC4325271 DOI: 10.1016/j.stemcr.2014.12.007] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 12/09/2014] [Accepted: 12/09/2014] [Indexed: 01/22/2023] Open
Abstract
Understanding the mechanisms of cancer initiation will help to prevent and manage the disease. At present, the role of the breast microenvironment in transformation remains unknown. As BMP2 and BMP4 are important regulators of stem cells and their niches in many tissues, we investigated their function in early phases of breast cancer. BMP2 production by tumor microenvironment appeared to be specifically upregulated in luminal tumors. Chronic exposure of immature human mammary epithelial cells to high BMP2 levels initiated transformation toward a luminal tumor-like phenotype, mediated by the receptor BMPR1B. Under physiological conditions, BMP2 controlled the maintenance and differentiation of early luminal progenitors, while BMP4 acted on stem cells/myoepithelial progenitors. Our data also suggest that microenvironment-induced overexpression of BMP2 may result from carcinogenic exposure. We reveal a role for BMP2 and the breast microenvironment in the initiation of stem cell transformation, thus providing insight into the etiology of luminal breast cancer. High BMP2 levels are provided by endothelial and stroma cells in luminal tumors Chronic exposure to high BMP2 levels initiate mammary epithelial transformation Luminal tumors likely arise from an amplified BMP2/BMPR1B-mediated normal response Radiation and bisphenols perturbed BMP2 production by the mammary niche stroma
Collapse
Affiliation(s)
- Marion Chapellier
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Université de Lyon, 69000 Lyon, France; Department of Immunity, Virus, and Microenvironment, 69000 Lyon, France; Université de Lyon 1, ISPB, 69000 Lyon, France
| | | | - Xenia Schmidt
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Université de Lyon, 69000 Lyon, France; Department of Immunity, Virus, and Microenvironment, 69000 Lyon, France; Université de Lyon 1, ISPB, 69000 Lyon, France
| | - Flora Clément
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Université de Lyon, 69000 Lyon, France; Department of Immunity, Virus, and Microenvironment, 69000 Lyon, France; Université de Lyon 1, ISPB, 69000 Lyon, France
| | - Isabelle Treilleux
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Université de Lyon, 69000 Lyon, France; Department of Immunity, Virus, and Microenvironment, 69000 Lyon, France; Université de Lyon 1, ISPB, 69000 Lyon, France; Centre Léon Bérard, 69000 Lyon, France
| | - Emmanuel Delay
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Université de Lyon, 69000 Lyon, France; Department of Immunity, Virus, and Microenvironment, 69000 Lyon, France; Université de Lyon 1, ISPB, 69000 Lyon, France; Centre Léon Bérard, 69000 Lyon, France
| | - Alexandre Jammot
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Université de Lyon, 69000 Lyon, France; Department of Immunity, Virus, and Microenvironment, 69000 Lyon, France; Université de Lyon 1, ISPB, 69000 Lyon, France
| | - Christine Ménétrier-Caux
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Université de Lyon, 69000 Lyon, France; Department of Immunity, Virus, and Microenvironment, 69000 Lyon, France; Université de Lyon 1, ISPB, 69000 Lyon, France; Centre Léon Bérard, 69000 Lyon, France
| | - Gaëtan Pochon
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Université de Lyon, 69000 Lyon, France; Department of Immunity, Virus, and Microenvironment, 69000 Lyon, France; Université de Lyon 1, ISPB, 69000 Lyon, France
| | - Roger Besançon
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Université de Lyon, 69000 Lyon, France; Department of Immunity, Virus, and Microenvironment, 69000 Lyon, France; Université de Lyon 1, ISPB, 69000 Lyon, France
| | - Thibault Voeltzel
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Université de Lyon, 69000 Lyon, France; Department of Immunity, Virus, and Microenvironment, 69000 Lyon, France; Université de Lyon 1, ISPB, 69000 Lyon, France
| | - Claude Caron de Fromentel
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Université de Lyon, 69000 Lyon, France; Department of Immunity, Virus, and Microenvironment, 69000 Lyon, France; Université de Lyon 1, ISPB, 69000 Lyon, France
| | - Christophe Caux
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Université de Lyon, 69000 Lyon, France; Department of Immunity, Virus, and Microenvironment, 69000 Lyon, France; Université de Lyon 1, ISPB, 69000 Lyon, France; Centre Léon Bérard, 69000 Lyon, France
| | - Jean-Yves Blay
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Université de Lyon, 69000 Lyon, France; Department of Immunity, Virus, and Microenvironment, 69000 Lyon, France; Université de Lyon 1, ISPB, 69000 Lyon, France; Centre Léon Bérard, 69000 Lyon, France
| | - Richard Iggo
- Inserm U916, Institut Bergonié, University of Bordeaux, 33076 Bordeaux, France
| | - Véronique Maguer-Satta
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Université de Lyon, 69000 Lyon, France; Department of Immunity, Virus, and Microenvironment, 69000 Lyon, France; Université de Lyon 1, ISPB, 69000 Lyon, France.
| |
Collapse
|
22
|
Emmrich S, Rasche M, Schöning J, Reimer C, Keihani S, Maroz A, Xie Y, Li Z, Schambach A, Reinhardt D, Klusmann JH. miR-99a/100~125b tricistrons regulate hematopoietic stem and progenitor cell homeostasis by shifting the balance between TGFβ and Wnt signaling. Genes Dev 2014; 28:858-74. [PMID: 24736844 PMCID: PMC4003278 DOI: 10.1101/gad.233791.113] [Citation(s) in RCA: 126] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
miR-99a/100, let-7, and miR-125b paralogs are encoded in two tricistrons and are highly expressed in hematopoietic stem cells (HSCs). Emmrich et al. demonstrate that miR-99a/100∼125b tricistrons are transcribed as a polycistronic message and functionally converge at the combinatorial block of the TGFβ pathway. Down-regulating tumor suppressor genes APC/APC2 stabilized active β-catenin and enhanced Wnt signaling. These tricistronic miRNAs promoted sustained expansion of murine and human HSCs by switching the balance between Wnt and TGFβ signaling. Although regulation of stem cell homeostasis by microRNAs (miRNAs) is well studied, it is unclear how individual miRNAs genomically encoded within an organized polycistron can interact to induce an integrated phenotype. miR-99a/100, let-7, and miR-125b paralogs are encoded in two tricistrons on human chromosomes 11 and 21. They are highly expressed in hematopoietic stem cells (HSCs) and acute megakaryoblastic leukemia (AMKL), an aggressive form of leukemia with poor prognosis. Here, we show that miR-99a/100∼125b tricistrons are transcribed as a polycistronic message transactivated by the homeobox transcription factor HOXA10. Integrative analysis of global gene expression profiling, miRNA target prediction, and pathway architecture revealed that miR-99a/100, let-7, and miR-125b functionally converge at the combinatorial block of the transforming growth factor β (TGFβ) pathway by targeting four receptor subunits and two SMAD signaling transducers. In addition, down-regulation of tumor suppressor genes adenomatous polyposis coli (APC)/APC2 stabilizes active β-catenin and enhances Wnt signaling. By switching the balance between Wnt and TGFβ signaling, the concerted action of these tricistronic miRNAs promoted sustained expansion of murine and human HSCs in vitro or in vivo while favoring megakaryocytic differentiation. Hence, our study explains the high phylogenetic conservation of the miR-99a/100∼125b tricistrons controlling stem cell homeostasis, the deregulation of which contributes to the development of AMKL.
Collapse
Affiliation(s)
- Stephan Emmrich
- Pediatric Hematology and Oncology, Hannover Medical School, Hannover 30625, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Primitive CML cell expansion relies on abnormal levels of BMPs provided by the niche and on BMPRIb overexpression. Blood 2013; 122:3767-77. [PMID: 24100446 DOI: 10.1182/blood-2013-05-501460] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Leukemic stem cells in chronic phase chronic myelogenous leukemia (CP-CML) are responsible for disease persistence and eventual drug resistance, most likely because they survive, expand, and are sustained through interactions with their microenvironment. Bone morphogenetic proteins 2 (BMP2) and 4 (BMP4) regulate the fate and proliferation of normal hematopoietic stem cells, as well as interactions with their niche. We show here that the intrinsic expression of members of the BMP response pathway are deregulated in CML cells with differences exhibited in mature (CD34(-)) and immature (CD34(+)) compartments. These changes are accompanied by altered functional responses of primitive leukemic cells to BMP2 and BMP4 and strong increases in soluble BMP2 and BMP4 in the CML bone marrow. Using primary cells and a cell line mimicking CP-CML, we found that myeloid progenitor expansion is driven by the exposure of immature cells overexpressing BMP receptor Ib to BMP2 and BMP4. In summary, we demonstrate that deregulation of intracellular BMP signaling in primary CP-CML samples corrupts and amplifies their response to exogenous BMP2 and BMP4, which are abnormally abundant within the tumor microenvironment. These results provide new insights with regard to leukemic stem cell biology and suggest possibilities for the development of novel therapeutic tools specifically targeting the CML niche.
Collapse
|
24
|
Ali A, Bluteau O, Messaoudi K, Palazzo A, Boukour S, Lordier L, Lecluse Y, Rameau P, Kraus-Berthier L, Jacquet-Bescond A, Lelièvre H, Depil S, Dessen P, Solary E, Raslova H, Vainchenker W, Plo I, Debili N. Thrombocytopenia induced by the histone deacetylase inhibitor abexinostat involves p53-dependent and -independent mechanisms. Cell Death Dis 2013; 4:e738. [PMID: 23887629 PMCID: PMC3730430 DOI: 10.1038/cddis.2013.260] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Revised: 06/12/2013] [Accepted: 06/17/2013] [Indexed: 12/02/2022]
Abstract
Abexinostat is a pan histone deacetylase inhibitor (HDACi) that demonstrates efficacy in malignancy treatment. Like other HDACi, this drug induces a profound thrombocytopenia whose mechanism is only partially understood. We have analyzed its effect at doses reached in patient plasma on in vitro megakaryopoiesis derived from human CD34+ cells. When added at day 0 in culture, abexinostat inhibited CFU-MK growth, megakaryocyte (MK) proliferation and differentiation. These effects required only a short incubation period. Decreased proliferation was due to induction of apoptosis and was not related to a defect in TPO/MPL/JAK2/STAT signaling. When added later (day 8), the compound induced a dose-dependent decrease (up to 10-fold) in proplatelet (PPT) formation. Gene profiling from MK revealed a silencing in the expression of DNA repair genes with a marked RAD51 decrease at protein level. DNA double-strand breaks were increased as attested by elevated γH2AX phosphorylation level. Moreover, ATM was phosphorylated leading to p53 stabilization and increased BAX and p21 expression. The use of a p53 shRNA rescued apoptosis, and only partially the defect in PPT formation. These results suggest that HDACi induces a thrombocytopenia by a p53-dependent mechanism along MK differentiation and a p53-dependent and -independent mechanism for PPT formation.
Collapse
Affiliation(s)
- A Ali
- Institut National de la Santé et de la Recherche Médicale, UMR 1009, Equipe labellisée Ligue Contre le Cancer, Laboratoire d'Excellence GR-Ex.114 rue Edouard Vaillant, 94805 Villejuif, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Schneider L, Pellegatta S, Favaro R, Pisati F, Roncaglia P, Testa G, Nicolis SK, Finocchiaro G, d'Adda di Fagagna F. DNA damage in mammalian neural stem cells leads to astrocytic differentiation mediated by BMP2 signaling through JAK-STAT. Stem Cell Reports 2013; 1:123-38. [PMID: 24052948 PMCID: PMC3757751 DOI: 10.1016/j.stemcr.2013.06.004] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Revised: 06/11/2013] [Accepted: 06/12/2013] [Indexed: 01/17/2023] Open
Abstract
The consequences of DNA damage generation in mammalian somatic stem cells, including neural stem cells (NSCs), are poorly understood despite their potential relevance for tissue homeostasis. Here, we show that, following ionizing radiation-induced DNA damage, NSCs enter irreversible proliferative arrest with features of cellular senescence. This is characterized by increased cytokine secretion, loss of stem cell markers, and astrocytic differentiation. We demonstrate that BMP2 is necessary to induce expression of the astrocyte marker GFAP in irradiated NSCs via a noncanonical signaling pathway engaging JAK-STAT. This is promoted by ATM and antagonized by p53. Using a SOX2-Cre reporter mouse model for cell-lineage tracing, we demonstrate irradiation-induced NSC differentiation in vivo. Furthermore, glioblastoma assays reveal that irradiation therapy affects the tumorigenic potential of cancer stem cells by ablating self-renewal and inducing astroglial differentiation.
Collapse
Affiliation(s)
- Leonid Schneider
- IFOM Foundation-The FIRC Institute of Molecular Oncology Foundation, Via Adamello 16, 20139 Milan, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Su YC, Li SC, Peng HY, Ho YH, Chen LJ, Liao HF. RAD001-mediated STAT3 upregulation and megakaryocytic differentiation. Thromb Haemost 2013; 109:540-9. [PMID: 23329056 DOI: 10.1160/th12-10-0734] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Accepted: 12/11/2012] [Indexed: 01/15/2023]
Abstract
RAD001 is currently used as an immunosuppressant and anticancer drug. Megakaryocyte (MK) differentiation includes development from pluripotent stem cells to proliferation and differentiation toward MK formation and platelet maturation. Our preliminary assay showed that RAD001 might stimulate MK differentiation; however, the exact regulatory mechanisms needed to be elucidated. By the ex vivo assay, RAD001 induced MK differentiation in human haematopoietic stem cells, with both the stimulation of CFU-GM colony formation and CD61 surface marker expression. Then, BALB/c mice were orally administrated with or without agrylin and/or RAD001 for 15 days. The platelet count and bone marrow CFU-MK colony formation were eliminated by agrylin, but unchanged in RAD001 and RAD001 plus agrylin mice. An ex vivo assay of bone marrow-derived stem cells demonstrated that RAD001 increased the number of CFU-MK colonies. The MK count in bone section indicated the decreased effect by agrylin and then recovered by RAD001. The level of plasma thrombopoietin was also enhanced in RAD001-treated mice. The effect of RAD001 on human leukaemic K562 and HEL cells showed the growth inhibition and MK differentiation activities; including morphological observation, CD41 and CD61 expression, and platelet factor 4 secretion. In RAD001-treated HEL cells, p-STAT3 expression, STAT3 translocation, and STAT3-DNA binding activity were up-regulated. Furthermore, STAT3 siRNA decreased the p-STAT3 and CD61 expression, as well as the CD61 fluorescence intensity, indicating that STAT3 may be critical in RAD001-mediated MK differentiation. Conclusion, the present study demonstrated that RAD001 might have the capacity to induce MK differentiation through the up-regulation of STAT3 signalling.
Collapse
Affiliation(s)
- Yu-Chieh Su
- Department of Internal Medicine, Buddhist Dalin Tzu Chi General Hospital, and Department of Biochemical Science and Technology, National Chiayi University, 300 University Road, Chiayi 600, Taiwan
| | | | | | | | | | | |
Collapse
|
27
|
Crispino JD, Le Beau MM. BMP meets AML: induction of BMP signaling by a novel fusion gene promotes pediatric acute leukemia. Cancer Cell 2012; 22:567-8. [PMID: 23153530 PMCID: PMC3501979 DOI: 10.1016/j.ccr.2012.10.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
In this issue of Cancer Cell, Gruber et al. report that a significant proportion of children with acute megakaryoblastic leukemia acquire a translocation that confers enhanced BMP signaling and promotes self-renewal of hematopoietic progenitors. This study presents novel therapeutic targets that may lead to improved therapies for this aggressive leukemia.
Collapse
Affiliation(s)
- John D Crispino
- Division of Hematology/Oncology, Northwestern University, University of Chicago, Chicago, IL 60637, USA.
| | | |
Collapse
|
28
|
Gruber TA, Gedman AL, Zhang J, Koss CS, Marada S, Ta HQ, Chen SC, Su X, Ogden SK, Dang J, Wu G, Gupta V, Andersson AK, Pounds S, Shi L, Easton J, Barbato MI, Mulder HL, Manne J, Wang J, Rusch M, Ranade S, Ganti R, Parker M, Ma J, Radtke I, Ding L, Cazzaniga G, Biondi A, Kornblau SM, Ravandi F, Kantarjian H, Nimer SD, Döhner K, Döhner H, Ley TJ, Ballerini P, Shurtleff S, Tomizawa D, Adachi S, Hayashi Y, Tawa A, Shih LY, Liang DC, Rubnitz JE, Pui CH, Mardis ER, Wilson RK, Downing JR. An Inv(16)(p13.3q24.3)-encoded CBFA2T3-GLIS2 fusion protein defines an aggressive subtype of pediatric acute megakaryoblastic leukemia. Cancer Cell 2012; 22:683-97. [PMID: 23153540 PMCID: PMC3547667 DOI: 10.1016/j.ccr.2012.10.007] [Citation(s) in RCA: 192] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Revised: 09/05/2012] [Accepted: 10/17/2012] [Indexed: 01/12/2023]
Abstract
To define the mutation spectrum in non-Down syndrome acute megakaryoblastic leukemia (non-DS-AMKL), we performed transcriptome sequencing on diagnostic blasts from 14 pediatric patients and validated our findings in a recurrency/validation cohort consisting of 34 pediatric and 28 adult AMKL samples. Our analysis identified a cryptic chromosome 16 inversion (inv(16)(p13.3q24.3)) in 27% of pediatric cases, which encodes a CBFA2T3-GLIS2 fusion protein. Expression of CBFA2T3-GLIS2 in Drosophila and murine hematopoietic cells induced bone morphogenic protein (BMP) signaling and resulted in a marked increase in the self-renewal capacity of hematopoietic progenitors. These data suggest that expression of CBFA2T3-GLIS2 directly contributes to leukemogenesis.
Collapse
MESH Headings
- Animals
- Bone Morphogenetic Proteins/metabolism
- Child
- Chromosome Inversion
- Chromosomes, Human, Pair 16
- Drosophila/genetics
- Drosophila/growth & development
- Gene Expression Profiling
- Humans
- Kruppel-Like Transcription Factors/genetics
- Leukemia, Megakaryoblastic, Acute/classification
- Leukemia, Megakaryoblastic, Acute/diagnosis
- Leukemia, Megakaryoblastic, Acute/genetics
- Mice
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/metabolism
- Oncogene Proteins, Fusion/physiology
- Prognosis
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/metabolism
- Recombinant Fusion Proteins/physiology
- Repressor Proteins/genetics
- Sequence Analysis, RNA
- Signal Transduction
- Tumor Suppressor Proteins/genetics
Collapse
Affiliation(s)
- Tanja A. Gruber
- St. Jude Children's Research Hospital – Washington University Pediatric Cancer Genome Project, Memphis, TN, USA and St. Louis, MO, USA
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Amanda Larson Gedman
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jinghui Zhang
- St. Jude Children's Research Hospital – Washington University Pediatric Cancer Genome Project, Memphis, TN, USA and St. Louis, MO, USA
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Cary S. Koss
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Suresh Marada
- Department of Biochemistry, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Huy Q. Ta
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Shann-Ching Chen
- Hartwell Center for Biotechnology and Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Xiaoping Su
- Department of Bioinformatics and Computational Biology, University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - Stacey K. Ogden
- Department of Biochemistry, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jinjun Dang
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Gang Wu
- St. Jude Children's Research Hospital – Washington University Pediatric Cancer Genome Project, Memphis, TN, USA and St. Louis, MO, USA
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Vedant Gupta
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Anna K. Andersson
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Stanley Pounds
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Lei Shi
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - John Easton
- St. Jude Children's Research Hospital – Washington University Pediatric Cancer Genome Project, Memphis, TN, USA and St. Louis, MO, USA
- Pediatric Cancer Genome Project, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Michael I. Barbato
- St. Jude Children's Research Hospital – Washington University Pediatric Cancer Genome Project, Memphis, TN, USA and St. Louis, MO, USA
- Pediatric Cancer Genome Project, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Heather L. Mulder
- St. Jude Children's Research Hospital – Washington University Pediatric Cancer Genome Project, Memphis, TN, USA and St. Louis, MO, USA
- Pediatric Cancer Genome Project, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jayanthi Manne
- St. Jude Children's Research Hospital – Washington University Pediatric Cancer Genome Project, Memphis, TN, USA and St. Louis, MO, USA
- Pediatric Cancer Genome Project, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jianmin Wang
- St. Jude Children's Research Hospital – Washington University Pediatric Cancer Genome Project, Memphis, TN, USA and St. Louis, MO, USA
- Information Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Michael Rusch
- St. Jude Children's Research Hospital – Washington University Pediatric Cancer Genome Project, Memphis, TN, USA and St. Louis, MO, USA
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | | | - Ramapriya Ganti
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Matthew Parker
- St. Jude Children's Research Hospital – Washington University Pediatric Cancer Genome Project, Memphis, TN, USA and St. Louis, MO, USA
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jing Ma
- St. Jude Children's Research Hospital – Washington University Pediatric Cancer Genome Project, Memphis, TN, USA and St. Louis, MO, USA
- Hartwell Center for Biotechnology and Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Ina Radtke
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Li Ding
- St. Jude Children's Research Hospital – Washington University Pediatric Cancer Genome Project, Memphis, TN, USA and St. Louis, MO, USA
- Washington University School of Medicine, Siteman Cancer Center, St. Louis, MO, USA, The Genome Institute at Washington University, St Louis, MO, USA
| | - Giovanni Cazzaniga
- Centro Ricerca Tettamanti, Pediatric Clinic, Univ. Milan Bicocca, Monza, Italy
| | - Andrea Biondi
- Pediatric Unit, University of Milan-Bicocca, San Gerardo Hospital, Monza, Italy
| | - Steven M. Kornblau
- Department of Blood and Marrow Transplantation, University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - Farhad Ravandi
- Department of Leukemia, University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - Hagop Kantarjian
- Department of Leukemia, University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - Stephen D. Nimer
- Molecular Pharmacology and Chemistry Program, Sloan Kettering Institute , New York, NY, USA
| | - Konstanze Döhner
- Department of Internal Medicine III, University of Ulm, Ulm, Germany
| | - Hartmut Döhner
- Department of Internal Medicine III, University of Ulm, Ulm, Germany
| | - Timothy J. Ley
- St. Jude Children's Research Hospital – Washington University Pediatric Cancer Genome Project, Memphis, TN, USA and St. Louis, MO, USA
- Washington University School of Medicine, Siteman Cancer Center, St. Louis, MO, USA, The Genome Institute at Washington University, St Louis, MO, USA
| | - Paola Ballerini
- Laboratoire d'Hématologie, Hôpital A. Trousseau, Paris, France
| | - Sheila Shurtleff
- St. Jude Children's Research Hospital – Washington University Pediatric Cancer Genome Project, Memphis, TN, USA and St. Louis, MO, USA
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Daisuke Tomizawa
- Department of Pediatrics, Tokyo Medical and Dental University, Tokyo, Japan
| | - Souichi Adachi
- Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yasuhide Hayashi
- Department of Haematology/Oncology, Gunma Children's Medical Center, Shibukawa, Japan
| | - Akio Tawa
- Dept. of Pediatrics, National Hospital Organization Osaka National Hospital, Osaka, Japan
| | - Lee-Yung Shih
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, Taipei, Taiwan
| | - Der-Cherng Liang
- Division of Pediatric Hematology Oncology, Mackay Memorial Hospital, Taipei Taiwan
| | - Jeffrey E. Rubnitz
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Ching-Hon Pui
- St. Jude Children's Research Hospital – Washington University Pediatric Cancer Genome Project, Memphis, TN, USA and St. Louis, MO, USA
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Elaine R Mardis
- St. Jude Children's Research Hospital – Washington University Pediatric Cancer Genome Project, Memphis, TN, USA and St. Louis, MO, USA
- Washington University School of Medicine, Siteman Cancer Center, St. Louis, MO, USA, The Genome Institute at Washington University, St Louis, MO, USA
| | - Richard K Wilson
- St. Jude Children's Research Hospital – Washington University Pediatric Cancer Genome Project, Memphis, TN, USA and St. Louis, MO, USA
- Washington University School of Medicine, Siteman Cancer Center, St. Louis, MO, USA, The Genome Institute at Washington University, St Louis, MO, USA
| | - James R. Downing
- St. Jude Children's Research Hospital – Washington University Pediatric Cancer Genome Project, Memphis, TN, USA and St. Louis, MO, USA
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| |
Collapse
|
29
|
Thiollier C, Lopez CK, Gerby B, Ignacimouttou C, Poglio S, Duffourd Y, Guégan J, Rivera-Munoz P, Bluteau O, Mabialah V, Diop M, Wen Q, Petit A, Bauchet AL, Reinhardt D, Bornhauser B, Gautheret D, Lecluse Y, Landman-Parker J, Radford I, Vainchenker W, Dastugue N, de Botton S, Dessen P, Bourquin JP, Crispino JD, Ballerini P, Bernard OA, Pflumio F, Mercher T. Characterization of novel genomic alterations and therapeutic approaches using acute megakaryoblastic leukemia xenograft models. ACTA ACUST UNITED AC 2012; 209:2017-31. [PMID: 23045605 PMCID: PMC3478932 DOI: 10.1084/jem.20121343] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Acute megakaryoblastic leukemia (AMKL) is a heterogeneous disease generally associated with poor prognosis. Gene expression profiles indicate the existence of distinct molecular subgroups, and several genetic alterations have been characterized in the past years, including the t(1;22)(p13;q13) and the trisomy 21 associated with GATA1 mutations. However, the majority of patients do not present with known mutations, and the limited access to primary patient leukemic cells impedes the efficient development of novel therapeutic strategies. In this study, using a xenotransplantation approach, we have modeled human pediatric AMKL in immunodeficient mice. Analysis of high-throughput RNA sequencing identified recurrent fusion genes defining new molecular subgroups. One subgroup of patients presented with MLL or NUP98 fusion genes leading to up-regulation of the HOX A cluster genes. A novel CBFA2T3-GLIS2 fusion gene resulting from a cryptic inversion of chromosome 16 was identified in another subgroup of 31% of non-Down syndrome AMKL and strongly associated with a gene expression signature of Hedgehog pathway activation. These molecular data provide useful markers for the diagnosis and follow up of patients. Finally, we show that AMKL xenograft models constitute a relevant in vivo preclinical screening platform to validate the efficacy of novel therapies such as Aurora A kinase inhibitors.
Collapse
Affiliation(s)
- Clarisse Thiollier
- Institut National de la Santé et de la Recherche Médicale (INSERM) Unité 985, 94805 Villejuif, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Tamura S, Nagasawa A, Masuda Y, Tsunematsu T, Hayasaka K, Matsuno K, Shimizu C, Ozaki Y, Moriyama T. BDNF, produced by a TPO-stimulated megakaryocytic cell line, regulates autocrine proliferation. Biochem Biophys Res Commun 2012; 427:542-6. [PMID: 23022197 DOI: 10.1016/j.bbrc.2012.09.093] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Accepted: 09/14/2012] [Indexed: 12/30/2022]
Abstract
While human platelets release endogenous brain-derived neurotrophic factor (BDNF) upon activation, a previous report on MEG-01, a megakaryocytic cell line, found no trace of BDNF production, and the pathophysiological function of platelet BDNF has remained elusive. In the present study, we demonstrate that MEG-01 produces BDNF in the presence of TPO and that this serves to potentiate cell proliferation. Our in vitro findings suggest that BDNF regulates MEG-01 proliferation in an autocrine manner, and we suggest that BDNF may be a physiological autocrine regulator of megakaryocyte progenitors.
Collapse
Affiliation(s)
- Shogo Tamura
- Graduate School of Health Sciences, Hokkaido University, Sapporo, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Sagorny K, Chapellier M, Laperrousaz B, Maguer-Satta V. [BMP and cancer: the Yin and Yang of stem cells]. Med Sci (Paris) 2012; 28:416-22. [PMID: 22549870 DOI: 10.1051/medsci/2012284020] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
In a normal context, bone morphogenetic proteins (BMPs), members of the TGFβ superfamily, are key players in adult stem cell biology. They are involved in the control of the overall functional and phenotypic properties of the stem cell population (self-renewal, proliferation, differentiation, apoptosis, quiescence, etc.). They can act directly on the stem cell or through its microenvironment, contributing to the tight balance of this system. In the tumorigenic context, alterations of the BMP signalling are involved in the deregulation of the interaction between stem cells and their microenvironment and, as such, participate to the different steps of the transformation process.
Collapse
|
32
|
Lineage regulators direct BMP and Wnt pathways to cell-specific programs during differentiation and regeneration. Cell 2011; 147:577-89. [PMID: 22036566 DOI: 10.1016/j.cell.2011.09.044] [Citation(s) in RCA: 242] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Revised: 08/01/2011] [Accepted: 09/23/2011] [Indexed: 01/08/2023]
Abstract
BMP and Wnt signaling pathways control essential cellular responses through activation of the transcription factors SMAD (BMP) and TCF (Wnt). Here, we show that regeneration of hematopoietic lineages following acute injury depends on the activation of each of these signaling pathways to induce expression of key blood genes. Both SMAD1 and TCF7L2 co-occupy sites with master regulators adjacent to hematopoietic genes. In addition, both SMAD1 and TCF7L2 follow the binding of the predominant lineage regulator during differentiation from multipotent hematopoietic progenitor cells to erythroid cells. Furthermore, induction of the myeloid lineage regulator C/EBPα in erythroid cells shifts binding of SMAD1 to sites newly occupied by C/EBPα, whereas expression of the erythroid regulator GATA1 directs SMAD1 loss on nonerythroid targets. We conclude that the regenerative response mediated by BMP and Wnt signaling pathways is coupled with the lineage master regulators to control the gene programs defining cellular identity.
Collapse
|
33
|
Liu S, Hu P, Hou Y, Li P, Li X, Tian Q. The Additive Effect of Mesenchymal Stem Cells and Bone Morphogenetic Protein 2 on γ-Irradiated Bone Marrow in Mice. Cell Biochem Biophys 2011; 61:539-50. [DOI: 10.1007/s12013-011-9236-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
34
|
Sensitivity and gene expression profile of fresh human acute myeloid leukemia cells exposed ex vivo to AS602868. Cancer Chemother Pharmacol 2010; 68:97-105. [PMID: 20844879 DOI: 10.1007/s00280-010-1458-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2010] [Accepted: 09/02/2010] [Indexed: 10/19/2022]
Abstract
PURPOSE The need for new treatment options for acute myeloid leukemia (AML) is increasing. AS602868 is a novel investigational drug with reported activity on AML cells. METHODS We studied gene expression profiles in AML blasts exposed to AS602868 in order to better understand its mechanism of action. We analyzed the in vitro cytotoxicity of AS602868 alone or in combination with daunorubicin, etoposide or cytarabine. To document AS602868-induced IKK2 inhibition in fresh AML cells, a flow cytometry analysis of IκB was performed. Finally, the effect of AS602868 on gene expression in fresh AML cells was analyzed. RESULTS The results show that AML cells are globally as sensitive to AS602868 as they are to cytarabine, with large interindividual variations. Combinations with conventional antileukemic agents showed enhanced cytotoxic activity in subsets of patients. IKK2 appeared to be effectively inhibited by 100 μM AS602868 in fresh leukemic cells. Gene expression profiling and gene ontology analyses identified several groups of genes induced/inhibited by exposure to AS602868 and/or exhibiting a correlation with sensitivity to this agent in vitro. Of note, the expression of several genes related to immune function was found to be significantly altered after exposure to AS602868. CONCLUSION These data suggest that AS602868 is cytotoxic against fresh human AML blasts and provide insights regarding the mechanisms of cytotoxicity.
Collapse
|
35
|
Martelli AM, Evangelisti C, Chiarini F, Grimaldi C, Cappellini A, Ognibene A, McCubrey JA. The emerging role of the phosphatidylinositol 3-kinase/Akt/mammalian target of rapamycin signaling network in normal myelopoiesis and leukemogenesis. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2010; 1803:991-1002. [DOI: 10.1016/j.bbamcr.2010.04.005] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2010] [Revised: 04/06/2010] [Accepted: 04/06/2010] [Indexed: 12/19/2022]
|
36
|
Inducible Fli-1 gene deletion in adult mice modifies several myeloid lineage commitment decisions and accelerates proliferation arrest and terminal erythrocytic differentiation. Blood 2010; 116:4795-805. [PMID: 20733157 DOI: 10.1182/blood-2010-02-270405] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
This study investigated the role of the ETS transcription factor Fli-1 in adult myelopoiesis using new transgenic mice allowing inducible Fli-1 gene deletion. Fli-1 deletion in adult induced mild thrombocytopenia associated with a drastic decrease in large mature megakaryocytes number. Bone marrow bipotent megakaryocytic-erythrocytic progenitors (MEPs) increased by 50% without increase in erythrocytic and megakaryocytic common myeloid progenitor progeny, suggesting increased production from upstream stem cells. These MEPs were almost unable to generate pure colonies containing large mature megakaryocytes, but generated the same total number of colonies mainly identifiable as erythroid colonies containing a reduced number of more differentiated cells. Cytological and fluorescence-activated cell sorting analyses of MEP progeny in semisolid and liquid cultures confirmed the drastic decrease in large mature megakaryocytes but revealed a surprisingly modest (50%) reduction of CD41-positive cells indicating the persistence of a megakaryocytic commitment potential. Symmetrical increase and decrease of monocytic and granulocytic progenitors were also observed in the progeny of purified granulocytic-monocytic progenitors and common myeloid progenitors. In summary, this study indicates that Fli-1 controls several lineages commitment decisions at the stem cell, MEP, and granulocytic-monocytic progenitor levels, stimulates the proliferation of committed erythrocytic progenitors at the expense of their differentiation, and is a major regulator of late stages of megakaryocytic differentiation.
Collapse
|
37
|
Nørgaard NN, Holien T, Jönsson S, Hella H, Espevik T, Sundan A, Standal T. CpG-oligodeoxynucleotide inhibits Smad-dependent bone morphogenetic protein signaling: effects on myeloma cell apoptosis and in vitro osteoblastogenesis. THE JOURNAL OF IMMUNOLOGY 2010; 185:3131-9. [PMID: 20702733 DOI: 10.4049/jimmunol.0903605] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The TLR9 agonist CpG-oligodeoxynucleotide (CpG-ODN) with a phosphorothioate backbone (PTO-CpG-ODN) is evaluated in clinical trials as a vaccine adjuvant or as treatment of cancers. Bone morphogenetic proteins (BMPs) regulate growth and differentiation of several cell types, and also induce apoptosis of cancer cells. Cross-talk between BMP- and TLR-signaling has been reported, and we aimed to investigate whether CpG-ODN influenced BMP-induced osteoblast differentiation or BMP-induced apoptosis of malignant plasma cells. We found that PTO-CpG-ODN inhibited BMP-2-induced osteoblast differentiation from human mesenchymal stem cells. Further, PTO-CpG-ODN counteracted BMP-2- and BMP-6-induced apoptosis of the human myeloma cell lines IH-1 and INA-6, respectively. In contrast, PTO-CpG-ODN did not antagonize the antiproliferative effect of BMP-2 on hMSCs or IH-1 cells. Inhibition of Smad-signaling and p38 MAPK-signaling indicated that apoptosis of IH-1 cells is dependent on Smad-signaling downstream of BMP, whereas the antiproliferative effect of BMP-2 on IH-1 cells also involves p38 MAPK-signaling. Together, the data suggested a specific inhibition by PTO-CpG-ODN on BMP-Smad-signaling. Supporting this we found that PTO-CpG-ODN inhibited BMP-induced phosphorylation of receptor-Smads in human mesenchymal stem cells and myeloma cell lines. This effect appeared to be independent of TLR9 because GpC-ODN and other ODNs with the ability to form multimeric structures inhibited Smad-signaling as efficiently as PTO-CpG-ODNs, and because knockdown of TLR9 by small interfering RNA in INA-6 cells did not blunt the effect of PTO-CpG-ODN. In conclusion, our results demonstrate that PTO-CpG-ODN inhibits BMP-signaling, and thus might provoke unwanted TLR9-independent side effects in patients.
Collapse
Affiliation(s)
- Nikolai N Nørgaard
- Department of Cancer Research and Molecular Medicine, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | | | | | | | | | | | | |
Collapse
|
38
|
Levy O, Ruvinov E, Reem T, Granot Y, Cohen S. Highly efficient osteogenic differentiation of human mesenchymal stem cells by eradication of STAT3 signaling. Int J Biochem Cell Biol 2010; 42:1823-30. [PMID: 20691278 DOI: 10.1016/j.biocel.2010.07.017] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2010] [Revised: 07/11/2010] [Accepted: 07/27/2010] [Indexed: 11/16/2022]
Abstract
Human bone marrow-derived mesenchymal stem cells (hMSCs) are promising candidates for cellular therapy owing to their multipotency to differentiate into several cell lineages. Elucidating the signaling events involved in the response of hMSCs to diverse stimulants affecting their differentiation may considerably promote their clinical use. In this study, we attempted to illuminate the molecular signaling networks involved in bone morphogenetic protein (BMP)-stimulated hMSC osteogenic differentiation. We demonstrate that eradication of signal transducers and activators of transcription (STAT) signaling considerably enhances BMP-induced osteogenic differentiation of hMSCs. BMP 2 and 4 are shown for the first time to activate the Janus-activated kinase (JAK)-STAT pathway in hMSC. Specifically, we reveal that JAK2 mediates STAT3 tyrosine phosphorylation in response to the two BMPs, whereas BMP2- and BMP4-induced STAT3 serine phosphorylation involves two divergent cascades, namely the mTOR and ERK1/2 cascades, respectively. Furthermore, elimination of the STAT3 signaling pathway by the inhibitors, AG490 or STAT3 siRNA, results in the acceleration and augmentation of BMPs-induced osteogenic differentiation, thus proposing a role for JAK-STAT signaling as a negative regulator of this process in MSCs. We believe that the findings presented in this study may be the basis for the development of a useful strategy to better control stem cell fate through intervention in molecular signaling networks. Hopefully, such a strategy will include the development of more efficient and controllable protocols for hMSC differentiation and facilitate their use in regenerative medicine.
Collapse
Affiliation(s)
- Oren Levy
- The Department of Life Sciences, Ben Gurion University of the Negev, Beer Sheva, Israel.
| | | | | | | | | |
Collapse
|
39
|
Abstract
Bone morphogenetic protein 4 (BMP4) is required for mesoderm commitment to the hematopoietic lineage during early embryogenesis. However, deletion of BMP4 is early embryonically lethal and its functional role in definitive hematopoiesis is unknown. Consequently, we used a BMP4 hypomorph to investigate the role of BMP4 in regulating hematopoietic stem cell (HSC) function and maintaining steady-state hematopoiesis in the adult. Reporter gene expression shows that Bmp4 is expressed in cells associated with the hematopoietic microenvironment including osteoblasts, endothelial cells, and megakaryocytes. Although resting hematopoiesis is normal in a BMP4-deficient background, the number of c-Kit+, Sca-1+, Lineage- cells is significantly reduced. Serial transplantation studies reveal that BMP4-deficient recipients have a microenvironmental defect that reduces the repopulating activity of wild-type HSCs. This defect is even more pronounced in a parabiosis model that demonstrates a profound reduction in wild-type hematopoietic cells within the bone marrow of BMP4-deficient recipients. Furthermore, wild-type HSCs that successfully engraft into the BMP4-deficient bone marrow show a marked decrease in functional stem cell activity when tested in a competitive repopulation assay. Taken together, these findings indicate BMP4 is a critical component of the hematopoietic microenvironment that regulates both HSC number and function.
Collapse
|
40
|
Bluteau D, Lordier L, Di Stefano A, Chang Y, Raslova H, Debili N, Vainchenker W. Regulation of megakaryocyte maturation and platelet formation. J Thromb Haemost 2009; 7 Suppl 1:227-34. [PMID: 19630806 DOI: 10.1111/j.1538-7836.2009.03398.x] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Each day in every human, approximately 1 x 10(11) platelets are produced by the cytoplasmic fragmentation of megakaryocytes (MK), their marrow precursor cells. Platelets are the predominating factor in the process of hemostasis and thrombosis. Recent studies have shown that platelets also play a hitherto unsuspected role in several other processes such as inflammation, innate immunity, neoangiogenesis and tumor metastasis. The late phases of MK differentiation identified by polyploidization, maturation and organized fragmentation of the cytoplasm leading to the release of platelets in the blood stream represent a unique model of differentiation. The molecular and cellular mechanisms regulating platelet biogenesis are better understood and may explain several platelet disorders. This review focuses on MK polyploidization, and platelet formation, and discusses their alteration in some platelet disorders.
Collapse
Affiliation(s)
- D Bluteau
- INSERM, U790, 39 rue Camille Desmoulins, Villejuif, France
| | | | | | | | | | | | | |
Collapse
|
41
|
Cailleteau C, Liagre B, Beneytout JL. A proteomic approach to the identification of molecular targets in subsequent apoptosis of HEL cells after diosgenin-induced megakaryocytic differentiation. J Cell Biochem 2009; 107:785-96. [DOI: 10.1002/jcb.22176] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|