1
|
Dietsche L, Stirm K, Lysenko V, Schneidawind C, Tzankov A, Müller A, Theocharides APA. Loss of SMAD1 in acute myeloid leukemia with KMT2A::AFF1 and KMT2A::MLLT3 fusion genes. Front Oncol 2025; 14:1481713. [PMID: 39834944 PMCID: PMC11743462 DOI: 10.3389/fonc.2024.1481713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 12/10/2024] [Indexed: 01/22/2025] Open
Abstract
Introduction KMT2A-rearrangements define a subclass of acute leukemias characterized by a distinct gene expression signature linked to the dysfunctional oncogenic fusion proteins arising from various chromosomal translocations involving the KMT2A (also known as MLL1) gene. Research on the disease pathomechanism in KMT2A-rearranged acute leukemias has mainly focused on the upregulation of the stemness-related genes of the HOX-family and their co-factor MEIS1. Results Here we report the KMT2A::AFF1 and KMT2A::MLLT3 fusion gene-dependent downregulation of SMAD1, a TGF-β signaling axis transcription factor. SMAD1 expression is lost in the majority of AML patient samples and cell lines containing the two fusion genes KMT2A::AFF1 and KMT2A::MLLT3 compared to non-rearranged controls. Loss of SMAD1 expression is inducible by introducing the respective two KMT2A fusion genes into hematopoietic stem and progenitor cells. The loss of SMAD1 correlated with a markedly reduced amount of H3K4me3 levels at the SMAD1 promoter in tested cells with KMT2A::AFF1 and KMT2A::MLLT3. The expression of SMAD1 in cells with KMT2A::AFF1 fusion genes impacted the growth of cells in vitro and influenced engraftment of the KMT2A::AFF1 cell line MV4-11 in vivo. In MV4-11 cells SMAD1 expression caused a downregulation of HOXA9 and MEIS1, which was reinforced by TGF-β stimulation. Moreover, in MV4-11 cells SMAD1 presence sensitized cells for TGF-β mediated G1-arrest. Conclusion Overall, our data contributes to the understanding of the role of TGF-β signaling in acute myeloid leukemia with KMT2A::AFF1 by showing that SMAD1 loss can influence the growth dynamics and contribute to the pathogenic expression of disease driving factors.
Collapse
Affiliation(s)
- Lisa Dietsche
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital Zurich, Zurich, Switzerland
| | - Kristin Stirm
- Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland
| | - Veronika Lysenko
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital Zurich, Zurich, Switzerland
| | - Corina Schneidawind
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital Zurich, Zurich, Switzerland
| | - Alexandar Tzankov
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Anne Müller
- Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland
| | - Alexandre P. A. Theocharides
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
2
|
Leask A, Fadl A, Naik A. A modest proposal: targeting αv integrin-mediated activation of latent TGFbeta as a novel therapeutic approach to treat scleroderma fibrosis. Expert Opin Investig Drugs 2024; 33:279-285. [PMID: 38393748 DOI: 10.1080/13543784.2024.2323528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 02/22/2024] [Indexed: 02/25/2024]
Abstract
INTRODUCTION The potent profibrotic cytokine transforming growth factor-β (TGF-β) has been associated with the onset and progression of the fibrosis seen in the autoimmune connective tissue disease scleroderma (systemic sclerosis, SSc). AREA COVERED This review explores the data supporting the notion that TGF-β contributes to SSc fibrosis and examines why initiating clinical trials in SSc aimed at targeting integrin-mediated latent TGF-β activation is timely. EXPERT OPINION Targeting TGF-β directly has not been proven to be clinically effective in this disease. Conversely, targeting matrix stiffness, which perpetuates fibrosis, may have more promise. Intriguingly, targeting integrin-mediated activation of latent TGF-β, which bridges these concepts, may have therapeutic value.
Collapse
Affiliation(s)
- Andrew Leask
- College of Dentistry, University of Saskatchewan, Saskatoon, SK, Canada
| | - Asmaa Fadl
- College of Dentistry, University of Saskatchewan, Saskatoon, SK, Canada
| | - Angha Naik
- College of Dentistry, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
3
|
Sistigu A, Musella M, Galassi C, Vitale I, De Maria R. Tuning Cancer Fate: Tumor Microenvironment's Role in Cancer Stem Cell Quiescence and Reawakening. Front Immunol 2020; 11:2166. [PMID: 33193295 PMCID: PMC7609361 DOI: 10.3389/fimmu.2020.02166] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 08/10/2020] [Indexed: 12/12/2022] Open
Abstract
Cancer cell dormancy is a common feature of human tumors and represents a major clinical barrier to the long-term efficacy of anticancer therapies. Dormant cancer cells, either in primary tumors or disseminated in secondary organs, may reawaken and relapse into a more aggressive disease. The mechanisms underpinning dormancy entry and exit strongly resemble those governing cancer cell stemness and include intrinsic and contextual cues. Cellular and molecular components of the tumor microenvironment persistently interact with cancer cells. This dialog is highly dynamic, as it evolves over time and space, strongly cooperates with intrinsic cell nets, and governs cancer cell features (like quiescence and stemness) and fate (survival and outgrowth). Therefore, there is a need for deeper insight into the biology of dormant cancer (stem) cells and the mechanisms regulating the equilibrium quiescence-versus-proliferation are vital in our pursuit of new therapeutic opportunities to prevent cancer from recurring. Here, we review and discuss microenvironmental regulations of cancer dormancy and its parallels with cancer stemness, and offer insights into the therapeutic strategies adopted to prevent a lethal recurrence, by either eradicating resident dormant cancer (stem) cells or maintaining them in a dormant state.
Collapse
Affiliation(s)
- Antonella Sistigu
- Istituto di Patologia Generale, Università Cattolica del Sacro Cuore, Rome, Italy.,Tumor Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Martina Musella
- Istituto di Patologia Generale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Claudia Galassi
- Istituto di Patologia Generale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Ilio Vitale
- IIGM - Italian Institute for Genomic Medicine, c/o IRCSS Candiolo (TO), Candiolo, Italy.,Candiolo Cancer Institute, FPO - IRCCS, Candiolo, Italy
| | - Ruggero De Maria
- Istituto di Patologia Generale, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario "A. Gemelli" - IRCCS, Rome, Italy
| |
Collapse
|
4
|
Watanabe S, Sakamoto R, Yamamoto H, Imaya M, Yamashita T, Anann T, Nakamura K. Pediatric Pure Red Cell Aplasia Caused by Tacrolimus After Living-Donor Liver Transplant. EXP CLIN TRANSPLANT 2019; 18:838-841. [PMID: 31724928 DOI: 10.6002/ect.2019.0075] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Pure red cell aplasia is a relatively rare disease characterized by selective suppression of erythroid precursors in the bone marrow. This disease can also develop secondary to several other diseases and as a side effect of certain drugs. Tacrolimus, a potent immunosuppressant, is widely used in organ transplant. Several cases of pure red cell aplasia due to tacrolimus administration in organ transplant recipients have been reported.Here, we report a case of reversible pure red cell aplasia that developed during tacrolimus therapy following living-donor liver transplant. The patient, a 1-year-old girl diagnosed with progressive familial intrahepatic cholestasis type II, underwent living-donor liver transplant when she was 10 months old. She was started on 3 immunosuppressants posttransplant: tacrolimus (0.1 mg/kg/day twice daily), mycophenolate mofetil, and prednisolone (0.2 mg/kg/day). One year after transplant, she developed severe progressive anemia. Her hemoglobin concentration was extremely low (5.4 g/dL). A bone marrow biopsy revealed severe hypoplasia of the erythroblasts with no abnormality of other myelocytes. These findings were suggestive of pure red cell aplasia; we suspected that tacrolimus had caused this based on similar previous cases of tacrolimus-associated pure red cell aplasia. Accordingly, tacrolimus was switched to cyclosporine after this diagnosis. One week after this switch, the patient's red blood cell counts, reticulocytes, and hemoglobin concentration increased. Although tacrolimus is considered to have no significant potential for myelosuppression, cases of tacrolimus-related pure red cell aplasia have occurred. In patients who develop pure red cell aplasia during tacrolimus treatment following living-donor liver transplant, clinicians should consider switching from tacrolimus to another immunosuppressant.
Collapse
Affiliation(s)
- Suguru Watanabe
- From the Department of Pediatrics, Kumamoto University, Kumamoto, Japan
| | | | | | | | | | | | | |
Collapse
|
5
|
Binder S, Luciano M, Horejs-Hoeck J. The cytokine network in acute myeloid leukemia (AML): A focus on pro- and anti-inflammatory mediators. Cytokine Growth Factor Rev 2018; 43:8-15. [PMID: 30181021 DOI: 10.1016/j.cytogfr.2018.08.004] [Citation(s) in RCA: 133] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 08/22/2018] [Indexed: 12/15/2022]
Abstract
Cytokines exert profound effects on the progression of hematopoietic malignancies such as acute myeloid leukemia (AML). Critical roles of cytokines in the context of inflammation have gained special interest. While pro-inflammatory mediators such as IL-1β, TNF-α and IL-6 tend to increase AML aggressiveness, anti-inflammatory mediators such as TGF-β and IL-10 appear to impede AML progression. Dysregulation of the complex interactions between pro- and anti-inflammatory cytokines in AML may create a pro-tumorigenic microenvironment with effects on leukemic cell proliferation, survival and drug-resistance. This article summarizes current knowledge about the functions of pro- and anti-inflammatory cytokines in AML, their modes of action, and therapeutic interventions with potential to improve clinical outcomes for AML patients.
Collapse
Affiliation(s)
- Stephanie Binder
- University of Salzburg, Department of Biosciences, Hellbrunner Str. 34, Salzburg, Austria
| | - Michela Luciano
- University of Salzburg, Department of Biosciences, Hellbrunner Str. 34, Salzburg, Austria
| | - Jutta Horejs-Hoeck
- University of Salzburg, Department of Biosciences, Hellbrunner Str. 34, Salzburg, Austria.
| |
Collapse
|
6
|
|
7
|
Mullen AC, Wrana JL. TGF-β Family Signaling in Embryonic and Somatic Stem-Cell Renewal and Differentiation. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a022186. [PMID: 28108485 DOI: 10.1101/cshperspect.a022186] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Soon after the discovery of transforming growth factor-β (TGF-β), seminal work in vertebrate and invertebrate models revealed the TGF-β family to be central regulators of tissue morphogenesis. Members of the TGF-β family direct some of the earliest cell-fate decisions in animal development, coordinate complex organogenesis, and contribute to tissue homeostasis in the adult. Here, we focus on the role of the TGF-β family in mammalian stem-cell biology and discuss its wide and varied activities both in the regulation of pluripotency and in cell-fate commitment.
Collapse
Affiliation(s)
- Alan C Mullen
- Gastrointestinal Unit, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114.,Harvard Stem Cell Institute, Cambridge, Massachusetts 02138
| | - Jeffrey L Wrana
- Lunenfeld-Tanenbam Research Institute, Mount Sinai Hospital and Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1X5, Canada
| |
Collapse
|
8
|
Vaidya A, Kale VP. TGF-β signaling and its role in the regulation of hematopoietic stem cells. SYSTEMS AND SYNTHETIC BIOLOGY 2015; 9:1-10. [PMID: 25972984 DOI: 10.1007/s11693-015-9161-2] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 01/16/2015] [Accepted: 01/21/2015] [Indexed: 01/15/2023]
Abstract
Transforming growth factor-betas (TGF-βs) and their family members that include bone morphogenic proteins and activins have been implicated in the regulation of proliferation, hibernation, quiescence and differentiation of hematopoietic stem cells (HSCs). Increasing evidence suggests that the superfamily of TGF-βs play an integral role in the intercellular cross-talk between the stem cells and their microenvironment as well as within the cells at an intracellular level. Active sites of hematopoiesis, such as fetal liver and bone marrow are known to have abundant presence of TGF-β indicating their importance in the maintenance and regulation of hematopoiesis. One of the striking features of TGF-β superfamily is the variety of effects they evoke, contingent on the developing history of the responding cells. In the present review, we discuss the Smad-dependent and Smad-independent TGF-β signaling pathways in order to understand and underscore their role in the regulation of HSCs.
Collapse
Affiliation(s)
- Anuradha Vaidya
- Symbiosis School of Biomedical Sciences (SSBS), Symbiosis International University (SIU), Symbiosis Knowledge Village, Lavale, Mulshi, Pune, 412115 Maharashtra India
| | - Vaijayanti P Kale
- National Centre for Cell Science (NCCS), NCCS Complex, Pune University Campus, Ganeshkhind, Pune, 411007 Maharashtra India
| |
Collapse
|
9
|
Glass C, Wilson M, Gonzalez R, Zhang Y, Perkins AS. The role of EVI1 in myeloid malignancies. Blood Cells Mol Dis 2014; 53:67-76. [PMID: 24495476 DOI: 10.1016/j.bcmd.2014.01.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 12/26/2013] [Indexed: 01/01/2023]
Abstract
The EVI1 oncogene at human chr 3q26 is rearranged and/or overexpressed in a subset of acute myeloid leukemias and myelodysplasias. The EVI1 protein is a 135 kDa transcriptional regulator with DNA-binding zinc finger domains. Here we provide a critical review of the current state of research into the molecular mechanisms by which this gene plays a role in myeloid malignancies. The major pertinent cellular effects are blocking myeloid differentiation and preventing cellular apoptosis, and several potential mechanisms for these phenomena have been identified. Evidence supports a role for EVI1 in inducing cellular quiescence, and this may contribute to the resistance to chemotherapy seen in patients with neoplasms that overexpress EVI1. Another isoform, MDS1-EVI1 (or PRDM3), encoded by the same locus as EVI1, harbors an N-terminal histone methyltransferase(HMT) domain; experimental findings indicate that this protein and its HMT activity are critical for the progression of a subset of AMLs, and this provides a potential target for therapeutic intervention.
Collapse
Affiliation(s)
- Carolyn Glass
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine, Rochester, NY 14642, USA
| | - Michael Wilson
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine, Rochester, NY 14642, USA
| | - Ruby Gonzalez
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine, Rochester, NY 14642, USA
| | - Yi Zhang
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine, Rochester, NY 14642, USA
| | - Archibald S Perkins
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine, Rochester, NY 14642, USA.
| |
Collapse
|
10
|
Zuo W, Huang F, Chiang YJ, Li M, Du J, Ding Y, Zhang T, Lee HW, Jeong LS, Chen Y, Deng H, Feng XH, Luo S, Gao C, Chen YG. c-Cbl-mediated neddylation antagonizes ubiquitination and degradation of the TGF-β type II receptor. Mol Cell 2013; 49:499-510. [PMID: 23290524 DOI: 10.1016/j.molcel.2012.12.002] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2012] [Revised: 10/23/2012] [Accepted: 11/29/2012] [Indexed: 02/07/2023]
Abstract
Transforming growth factor β (TGF-β) is a potent antiproliferative factor in multiple types of cells. Deregulation of TGF-β signaling is associated with the development of many cancers, including leukemia, though the molecular mechanisms are largely unclear. Here, we show that Casitas B-lineage lymphoma (c-Cbl), a known proto-oncogene encoding an ubiquitin E3 ligase, promotes TGF-β signaling by neddylating and stabilizing the type II receptor (TβRII). Knockout of c-Cbl decreases the TβRII protein level and desensitizes hematopoietic stem or progenitor cells to TGF-β stimulation, while c-Cbl overexpression stabilizes TβRII and sensitizes leukemia cells to TGF-β. c-Cbl conjugates neural precursor cell-expressed, developmentally downregulated 8 (NEDD8), a ubiquitin-like protein, to TβRII at Lys556 and Lys567. Neddylation of TβRII promotes its endocytosis to EEA1-positive early endosomes while preventing its endocytosis to caveolin-positive compartments, therefore inhibiting TβRII ubiquitination and degradation. We have also identified a neddylation-activity-defective c-Cbl mutation from leukemia patients, implying a link between aberrant TβRII neddylation and leukemia development.
Collapse
Affiliation(s)
- Wei Zuo
- The State Key Laboratory of Biomembrane and Membrane Biotechnology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Abstract
Chronic myeloid leukemia (CML) has long served as a paradigm for generating new insights into the cellular origin, pathogenesis and improved approaches to treating many types of human cancer. Early studies of the cellular phenotypes and genotypes represented in leukemic populations obtained from CML patients established the concept of an evolving clonal disorder originating in and initially sustained by a rare, multipotent, self-maintaining hematopoietic stem cell (HSC). More recent investigations continue to support this model, while also revealing new insights into the cellular and molecular mechanisms that explain how knowledge of CML stem cells and their early differentiating progeny can predict the differing and variable features of chronic phase and blast crisis. In particular, these emphasize the need for new agents that effectively and specifically target CML stem cells to produce non-toxic, but curative therapies that do not require lifelong treatments.
Collapse
|
12
|
Bhatwadekar AD, Guerin E, Jarajapu YP, Caballero S, Sheridan C, Kent D, Kennedy L, Lansang MC, Ruscetti FW, Pepine CJ, Higgins PJ, Bartelmez SH, Grant MB. Transient inhibition of transforming growth factor-beta1 in human diabetic CD34+ cells enhances vascular reparative functions. Diabetes 2010; 59:2010-9. [PMID: 20460428 PMCID: PMC2911069 DOI: 10.2337/db10-0287] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE Peripheral blood CD34(+) cells from diabetic patients demonstrate reduced vascular reparative function due to decreased proliferation and diminished migratory prowess, largely resulting from decreased nitric oxide (NO) bioavailability. The level of TGF-beta, a key factor that modulates stem cell quiescence, is increased in the serum of type 2 diabetic patients. We asked whether transient TGF-beta1 inhibition in CD34(+) cells would improve their reparative ability. RESEARCH DESIGN AND METHODS To inhibit TGF-beta1 protein expression, CD34(+) cells were treated ex vivo with antisense phosphorodiamidate morpholino oligomers (TGF-beta1-PMOs) and analyzed for cell surface CXCR4 expression, cell survival in the absence of added growth factors, SDF-1-induced migration, NO release, and in vivo retinal vascular reparative ability. RESULTS TGF-beta1-PMO treatment of diabetic CD34(+) cells resulted in increased expression of CXCR4, enhanced survival in the absence of growth factors, and increased migration and NO release as compared with cells treated with control PMO. Using a retinal ischemia reperfusion injury model in mice, we observed that recruitment of diabetic CD34(+) cells to injured acellular retinal capillaries was greater after TGF-beta1-PMO treatment compared with control PMO-treated cells. CONCLUSIONS Transient inhibition of TGF-beta1 may represent a promising therapeutic strategy for restoring the reparative capacity of dysfunctional diabetic CD34(+) cells.
Collapse
Affiliation(s)
| | - E.P. Guerin
- Pharmacology and Therapeutics, University of Florida, Gainesville, Florida
- The Vision Clinic, Circular Road, Kilkenny, Ireland
| | | | - Sergio Caballero
- Pharmacology and Therapeutics, University of Florida, Gainesville, Florida
| | - Carl Sheridan
- School of Clinical Sciences University of Liverpool, Liverpool, U.K
| | - David Kent
- The Vision Clinic, Circular Road, Kilkenny, Ireland
| | - Laurence Kennedy
- Division of Endocrinology, Diabetes, and Metabolism, University of Florida, Gainesville, Florida
| | - M. Cecilia Lansang
- Division of Endocrinology, Diabetes, and Metabolism, University of Florida, Gainesville, Florida
| | - Frank W. Ruscetti
- Laboratory of Experimental Immunology, Center for Cancer Research, National Cancer Institute–Frederick, Frederick, Maryland
| | - Carl J. Pepine
- Division of Cardiology, University of Florida, Gainesville, Florida
| | - Paul J. Higgins
- Center for Cell Biology & Cancer Research Albany Medical College, Albany, New York; and
| | - Stephen H. Bartelmez
- BetaStem Therapeutics Inc., San Francisco, California
- Corresponding authors: Maria B. Grant, , and Stephen Bartelmez,
| | - Maria B. Grant
- Pharmacology and Therapeutics, University of Florida, Gainesville, Florida
- Corresponding authors: Maria B. Grant, , and Stephen Bartelmez,
| |
Collapse
|
13
|
Insights into signaling and function of hematopoietic stem cells at the single-cell level. Curr Opin Hematol 2009; 16:255-8. [PMID: 19465850 DOI: 10.1097/moh.0b013e32832c6705] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
PURPOSE OF REVIEW Development of a technique prospectively to isolate hematopoietic stem cells (HSCs) to near homogeneity has enabled clonal analysis and thus converted our understanding of HSCs from conceptual and qualitative to realistic and quantitative. Recent studies have revealed that despite their high proliferation potential, most HSCs are in G0 and enter cell cycle only after a long interval. This dormancy of HSCs, which seems to be important for long-term maintenance of 'stemness', appears to be regulated by the exchange of signals between HSCs and the bone marrow niche. Analysis of intersignaling and intrasignaling events in HSCs in and out of the bone marrow niche has begun. RECENT FINDINGS With the help of advances in confocal microscopy, laser scanning microscopy, and personal computer computational power over the last decade, it has become evident that thrombopoietin/c-Mpl signaling plays a role in HSC self-renewal and AKT-forkhead box O signaling in HSC dormancy. Furthermore, transforming growth factor-beta has been indicated as a candidate niche signal to induce hibernation in HSCs. SUMMARY Understanding of the signaling events between HSCs and niche is critical not only for stem cell biology in general and for transplantation medicine but also for the development of novel cancer therapy.
Collapse
|
14
|
|
15
|
Métais JY, Dunbar CE. The MDS1-EVI1 gene complex as a retrovirus integration site: impact on behavior of hematopoietic cells and implications for gene therapy. Mol Ther 2008; 16:439-49. [PMID: 18227842 DOI: 10.1038/sj.mt.6300372] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Gene therapy trials have been performed with virus-based vectors that have the ability to integrate permanently into genomic DNA and thus allow prolonged expression of corrective genes after transduction of hematopoietic stem and progenitor cells. Adverse events observed during the X-linked severe combined immunodeficiency gene therapy trial revealed a significant risk of genotoxicity related to retrovirus vector integration and activation of adjacent proto-oncogenes, with several cases of T-cell leukemia linked to vector activation of the LMO2 gene. In patients with chronic granulomatous disease (CGD), rhesus macaques, and mice receiving hematopoietic stem and progenitor cells transduced with retrovirus vectors, a highly non-random pattern of vector integration has been reported. The most striking finding has been overrepresentation of integrations in one specific genomic locus, a complex containing the MDS1 and the EVI1 genes. Most evidence suggests that this overrepresentation is primarily due to a modification of primitive myeloid cell behavior by overexpression of EVI1 or MDS1-EVI1, as opposed to a specific predilection for integration at this site. Three different proteins can be produced from this complex locus: MDS1, MDS1-EVI1, and EVI1. This review will summarize current knowledge regarding this locus and its gene products, with specific focus on issues with relevance to gene therapy, leukemogenesis, and hematopoiesis. Insights into the mechanisms that result in altered hematopoiesis and leukemogenesis when this locus is dysregulated could improve the safety of gene therapy in the future.
Collapse
Affiliation(s)
- Jean-Yves Métais
- Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | |
Collapse
|
16
|
Ruscetti FW, Dubois C, Falk LA, Jacobsen SE, Sing G, Longo DL, Wiltrout RH, Keller JR. In vivo and in vitro effects of TGF-beta 1 on normal and neoplastic haemopoiesis. CIBA FOUNDATION SYMPOSIUM 2007; 157:212-27; discussion 227-31. [PMID: 1649034 DOI: 10.1002/9780470514061.ch14] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
TGF-beta 1 and TGF-beta 2 are equipotent selective inhibitors of murine and human haemopoiesis in vitro. Primitive haemopoietic cells such as the high proliferative potential progenitor cell and the colony-forming unit (CFU)-GEMM are directly inhibited by TGF-beta whereas the more differentiated CFU-G, CFU-M and CFU-E are not. Recombinant TGF-beta 1 administered intraperitoneally or intravenously to mice selectively inhibits haemopoietic colony formation in a time- and dose-dependent manner to the same extent as seen in vitro. The progenitors are reversibly prevented from entering the cell cycle. This inhibitory action of TGF-beta functions on at least two levels: (1) down-modulation of the cell surface expression of receptors for growth stimulatory molecules and (2) interference with the intracellular signalling pathways of these molecules. In addition, expression of TGF-beta receptors is regulated during cytokine stimulation of haemopoiesis. Neoplastic B lymphocytes can proliferate by escaping from a TGF-beta-mediated autocrine inhibitory loop. Activation signals (e.g. phorbol esters) inhibit tumour cell growth by stimulating active TGF-beta production and inducing cell surface expression of TGF-beta receptors. These results indicate that TGF-beta may be useful as a bone marrow protective and/or an antitumour agent.
Collapse
Affiliation(s)
- F W Ruscetti
- Biological Response Modifiers Program, National Cancer Institute-Frederick Cancer Research and Development Center, MD 21701-1201
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Moody JL, Singbrant S, Karlsson G, Blank U, Aspling M, Flygare J, Bryder D, Karlsson S. Endoglin is not critical for hematopoietic stem cell engraftment and reconstitution but regulates adult erythroid development. Stem Cells 2007; 25:2809-19. [PMID: 17673527 DOI: 10.1634/stemcells.2006-0602] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Endoglin is a transforming growth factor-beta (TGF-beta) accessory receptor recently identified as being highly expressed on long-term repopulating hematopoietic stem cells (HSC). However, little is known regarding its function in these cells. We have used two complementary approaches toward understanding endoglin's role in HSC biology: one that efficiently knocks down expression via lentiviral-driven short hairpin RNA and another that uses retroviral-mediated overexpression. Altering endoglin expression had functional consequences for hematopoietic progenitors in vitro such that endoglin-suppressed myeloid progenitors (colony-forming unit-granulocyte macrophage) displayed a higher degree of sensitivity to TGF-beta-mediated growth inhibition, whereas endoglin-overexpressing cells were partially resistant. However, transplantation of transduced bone marrow enriched in primitive hematopoietic stem and progenitor cells revealed that neither endoglin suppression nor endoglin overexpression affected the ability of stem cells to short-term or long-term repopulate recipient marrow. Furthermore, transplantation of cells altered in endoglin expression yielded normal white blood cell proportions and peripheral blood platelets. Interestingly, decreasing endoglin expression increased the clonogenic capacity of early blast-forming unit-erythroid progenitors, whereas overexpression compromised erythroid differentiation at the basophilic erythroblast phase, suggesting a pivotal role for endoglin at key stages of adult erythropoietic development.
Collapse
Affiliation(s)
- Jennifer L Moody
- Molecular Medicine and Gene Therapy, Institute of Laboratory Medicine, Lund Strategic Research Center for Stem Cell Biology and Cell Therapy, Lund University Hospital, Lund, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Isufi I, Seetharam M, Zhou L, Sohal D, Opalinska J, Pahanish P, Verma A. Transforming growth factor-beta signaling in normal and malignant hematopoiesis. J Interferon Cytokine Res 2007; 27:543-52. [PMID: 17651015 DOI: 10.1089/jir.2007.0009] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Transforming growth factor-beta (TGF-beta) is an important physiologic regulator of cell growth and differentiation. TGF-beta has been shown to inhibit the proliferation of quiescent hematopoietic stem cells and stimulate the differentiation of late progenitors to erythroid and myeloid cells. Insensitivity to TGF-beta is implicated in the pathogenesis of many myeloid and lymphoid neoplasms. Loss of extracellular TGF receptors and disruption of intracellular TGF-beta signaling by oncogenes is seen in a variety of malignant and premalignant states. TGF-beta can also affect tumor growth and survival by influencing the secretion of other growth factors and manipulation of the tumor microenvironment. Recent development of small molecule inhibitors of TGF-beta receptors and other signaling intermediaries may allow us to modulate TGF signaling for future therapeutic interventions in cancer.
Collapse
Affiliation(s)
- Iris Isufi
- Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | | | | | | | | | | |
Collapse
|
19
|
Blank U, Karlsson G, Moody JL, Utsugisawa T, Magnusson M, Singbrant S, Larsson J, Karlsson S. Smad7 promotes self-renewal of hematopoietic stem cells. Blood 2006; 108:4246-54. [PMID: 16917010 DOI: 10.1182/blood-2006-02-005611] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The Smad-signaling pathway downstream of the transforming growth factor-beta superfamily of ligands is an evolutionarily conserved signaling circuitry with critical functions in a wide variety of biologic processes. To investigate the role of this pathway in the regulation of hematopoietic stem cells (HSCs), we have blocked Smad signaling by retroviral gene transfer of the inhibitory Smad7 to murine HSCs. We report here that the self-renewal capacity of HSCs is promoted in vivo upon blocking of the entire Smad pathway, as shown by both primary and secondary bone marrow (BM) transplantations. Importantly, HSCs overexpressing Smad7 have an unperturbed differentiation capacity as evidenced by normal contribution to both lymphoid and myeloid cell lineages, suggesting that the Smad pathway regulates self-renewal independently of differentiation. Moreover, phosphorylation of Smads was inhibited in response to ligand stimulation in BM cells, thus verifying impairment of the Smad-signaling cascade in Smad7-overexpressing cells. Taken together, these data reveal an important and previously unappreciated role for the Smad-signaling pathway in the regulation of self-renewal of HSCs in vivo.
Collapse
Affiliation(s)
- Ulrika Blank
- Molecular Medicine and Gene Therapy, Institute of Laboratory Medicine, Lund, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Ruscetti FW, Akel S, Bartelmez SH. Autocrine transforming growth factor-beta regulation of hematopoiesis: many outcomes that depend on the context. Oncogene 2005; 24:5751-63. [PMID: 16123808 DOI: 10.1038/sj.onc.1208921] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Transforming growth factor-beta (TGF-beta) is a pleiotropic regulator of all stages of hematopoieis. The three mammalian isoforms (TGF-beta1, 2 and 3) have distinct but overlapping effects on hematopoiesis. Depending on the differentiation stage of the target cell, the local environment and the concentration and isoform of TGF-beta, in vivo or in vitro, TGF-beta can be pro- or antiproliferative, pro- or antiapoptotic, pro- or antidifferentiative and can inhibit or increase terminally differentiated cell function. TGF-beta is a major regulator of stem cell quiescence, at least in vitro. TGF-beta can act directly or indirectly through effects on the bone marrow microenvironment. In addition, paracrine and autocrine actions of TGF-beta have overlapping but distinct regulatory effects on hematopoietic stem/progenitor cells. Since TGF-beta can act in numerous steps in the hematopoietic cascade, loss of function mutations in hematopoeitic stem cells (HSC) have different effects on hematopoiesis than transient blockade of autocrine TGF-beta1. Transient neutralization of autocrine TGF-beta in HSC has therapeutic potential. In myeloid and erythroid leukemic cells, autocrine TGF-beta1 and/or its Smad signals controls the ability of these cells to respond to various differentiation inducers, suggesting that this pathway plays a role in determining the cell fate of leukemic cells.
Collapse
Affiliation(s)
- Francis W Ruscetti
- Laboratory of Experimental Immunology, Center for Cancer Research, National Cancer Institute-Frederick, Frederick, MD 21702-1201, USA.
| | | | | |
Collapse
|
21
|
Abstract
The TGF-beta family of ligands, including TGF-beta, bone morphogenetic protein (BMP) and activin, signal through Smad pathways to regulate the fate of hematopoietic progenitor and stem cells during development and postnatally. BMP regulates hematopoietic stem cell (HSC) specification during development, while TGF-beta1, 2 and 3 are not essential for the generation of HSCs. BMP4 can increase proliferation of human hematopoietic progenitors, while TGF-beta acts as a negative regulator of hematopoietic progenitor and stem cells in vitro. In contrast, TGF-beta signaling deficiency in vivo does not affect proliferation of HSCs and does not affect lineage choice either. Therefore, the outcome of Smad signaling is very context dependent in hematopoiesis and regulation of hematopoietic stem and progenitor cells is more complicated in the bone marrow microenvironment in vivo than is seen in liquid cultures ex vivo. Smad signaling regulates hematopoiesis by crosstalk with other regulatory signals and future research will define in more detail how the various pathways interact and how the knowledge obtained can be used to develop advanced cell therapies.
Collapse
Affiliation(s)
- Jonas Larsson
- Molecular Medicine and Gene Therapy, Institute of Laboratory Medicine, The Lund Strategic Research Center for Stem Cell Biology and Cell Therapy, Lund University, BMC A12, Lund 221 84, Sweden
| | | |
Collapse
|
22
|
Yatsula B, Lin S, Read AJ, Poholek A, Yates K, Yue D, Hui P, Perkins AS. Identification of binding sites of EVI1 in mammalian cells. J Biol Chem 2005; 280:30712-22. [PMID: 16006653 DOI: 10.1074/jbc.m504293200] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The leukemia-associated protein EVI1 possesses seven zinc fingers within an N-terminal domain (amino acids 1-250) that binds to GACAAGATA. Single amino acid missense mutants of EVI1 were developed that failed to bind DNA either in vitro, as assessed by gel shift assay, or in vivo, as shown by transactivation studies. Specifically, mutation R205N lacks high affinity binding to the GACAAGATA motif. Putative EVI1 target genes were identified by using an EVI1-(1-250)-VP16 fusion protein that acts as a transcriptional activator with the binding specificity of EVI1. Sixteen genes induced in NIH 3T3 cells by wild type EVI1-VP16 but not by mutant forms were identified. Sequence analysis revealed evolutionarily conserved GACAAGATA-like motifs within 10 kb of their transcription start sites, and by chromatin immunoprecipitation in fibroblasts, we showed occupancy of many of these sites by EVI1-VP16. To assess whether native EVI1 binds to these sites in EVI1-transformed myeloid cells, we performed chromatin immunoprecipitation in 32Dcl3 and NFS58 cells, using anti-EVI1 antisera, and we showed that the majority of these sites is bound by wild type EVI1. These putative target genes include Gadd45g, Gata2, Zfpm2/Fog2, Skil (SnoN), Klf5 (BTEB2), Dcn, and Map3k14 (Nik). In this study we demonstrated for the first time that the N-terminal DNA binding domain of EVI1 has the capacity to bind to endogenous genes. We hypothesized that these genes play a critical role in EVI1-induced transformation.
Collapse
Affiliation(s)
- Bogdan Yatsula
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut 06510-8023, USA
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Tse KF, Inayat MS, Morrow JK, Hughes NK, Oakley OR, Gallicchio VS. Reconstitution of erythroid, megakaryocyte and myeloid hematopoietic support function with neutralizing antibodies against IL-4 and TGFbeta1 in long-term bone marrow cultures infected with LP-BM5 murine leukemia virus. Virus Res 2005; 113:1-15. [PMID: 15869820 DOI: 10.1016/j.virusres.2005.03.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2004] [Revised: 03/25/2005] [Accepted: 03/25/2005] [Indexed: 11/25/2022]
Abstract
Murine acquired immunodeficiency syndrome (MAIDS) induced by a defective LP-BM5 murine leukemia virus (MuLV) produces hematopoietic cytopenias similar to HIV in patients with AIDS. The pathogenesis of MAIDS induced cytopenias remains obscure; however, direct retroviral infection of bone marrow stroma has been implicated to play a role. To evaluate the consequential effect of viral infection, primary stromal cell cultures were transiently incubated in vitro with LP-BM5 MuLV viral supernatant. Reverse transcription polymerase chain reaction (RT-PCR) and Southern blot hybridization revealed that defective LP-BM5 MuLV infection resulted in elevated levels of IL-4 and TGFbeta1 transcript expression in infected stromal cells. The increased expression of both IL-4 and TGFbeta1 transcripts was associated with enhanced production of corresponding proteins as determined by quantitative western blot analyses. Hematopoietic reconstitution assays revealed that the hematopoietic support function of stromal cells was significantly reduced following transient exposure to LP-BM5 MuLV. The production of nonadherent mononuclear cells and the growth of myeloid, megakaryocyte and erythroid lineages were all suppressed in infected cultures. Culture supernatant conditioned by infected stromal cells demonstrated growth-inhibitory activity for hematopoietic progenitor colony formation. This growth-inhibitory activity could be significantly abolished by addition of anti-IL-4 and/or anti-TGFbeta1 neutralizing antibodies to the culture supernatant or directly to the stromal cell cultures. This study demonstrates LP-BM5 MuLV increases two known cytokines to suppress hematopoiesis implicating viral infection can directly suppress hematopoiesis mediated by inhibitors released from marrow stroma.
Collapse
Affiliation(s)
- Kam-Fai Tse
- Laboratory of Experimental Immunohematopoiesis and Developmental Therapeutics, Hematology/Oncology Division, University of Kentucky, Lexington, KY 40536-0084, USA
| | | | | | | | | | | |
Collapse
|
24
|
Larsson J, Blank U, Klintman J, Magnusson M, Karlsson S. Quiescence of hematopoietic stem cells and maintenance of the stem cell pool is not dependent on TGF-β signaling in vivo. Exp Hematol 2005; 33:592-6. [PMID: 15850837 DOI: 10.1016/j.exphem.2005.02.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2004] [Revised: 02/08/2005] [Accepted: 02/09/2005] [Indexed: 11/15/2022]
Abstract
OBJECTIVE Maintained quiescence of hematopoietic stem cells (HSCs) is of critical importance to prevent premature exhaustion of the stem cell pool under conditions of hematopoietic stress. The growth inhibitory cytokine transforming growth factor beta (TGF-beta) has been shown to play a critical role in maintaining quiescence of HSCs in vitro. Here, we have used conditional knockout mice for the TGF-beta type I receptor (TbetaRI) to ask whether the naturally quiescent state of HSCs in vivo is dependent on TGF-beta signaling and thus whether TGF-beta serves as a protective factor for the stem cell pool during conditions of stress. METHODS TbetaRI null and control bone marrow chimeras were subjected to repeated treatments with the cell cycle-specific cytotoxic drug 5-fluorouracil (5-FU) and surviving HSCs were assayed by competitive transplantation experiments. Exhaustion of stem cells was provoked by serially transplanting TGF-beta signaling-deficient as well as normal BM cells into lethally irradiated recipients, which were monitored for survival. RESULTS Surprisingly, we found that TGF-beta receptor-deficient HSCs have similar susceptibility, compared to controls, to repeated 5-FU treatments, indicative of normally maintained quiescence in these cells. Likewise, hematopoietic failure occurred at similar stages in serially transplanted recipients of TbetaRI null and control BM, respectively, demonstrating normal consumption of the stem cell pool during hematopoietic stress. CONCLUSIONS These findings clearly demonstrate that, despite a key role in vitro, TGF-beta does not provide the necessary signal that induces the quiescent state of HSCs and maintains the stem cell pool in vivo.
Collapse
Affiliation(s)
- Jonas Larsson
- Department of Molecular Medicine and Gene Therapy, Institute of Laboratory Medicine, Lund University Hospital, Lund, Sweden
| | | | | | | | | |
Collapse
|
25
|
Kale VP. Differential activation of MAPK signaling pathways by TGF-beta1 forms the molecular mechanism behind its dose-dependent bidirectional effects on hematopoiesis. Stem Cells Dev 2004; 13:27-38. [PMID: 15068691 DOI: 10.1089/154732804773099236] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
We have earlier reported that transforming growth factor-beta1 (TGF-beta1), a well-known inhibitor of hematopoiesis, stimulated colony formation from adult human bone marrow mononuclear cells (BM MNC) when used at low concentrations. We examined the possible molecular mechanism behind this bidirectional effect using CD34+ cells isolated from human BM for clonal assays and the KG1a cell line as a model system for analysis of proteins for signaling pathways by immunoblotting. We found that TGF-beta1 at low doses (picogram levels) stimulated the colony formation from CD34+ cells, indicating that these progenitors form the direct target of stimulatory action of TGF-beta1. CD34+ cells were found to be more sensitive to the TGF-beta1 concentration than the total MNC. We used the KG1a cell line as a model system for identification of mitogen-activated protein kinase (MAPK) and AKT signaling pathways involved in the process. Low doses strongly induced p44/42 MAPK phosphorylation, whereas high doses induced p38 activation. Use of specific p44/42 MAPK inhibitor PD 98059 in the colony assay abrogated the stimulatory effect of low TGF-beta1. On the other hand, use of p38 MAPK inhibitor SB 203580 along with low TGF-beta1 concentrations had a synergistic effect on stimulation of colony formation. Treatment of BM MNC with Anisomycin, which activates stress kinases, resulted in a dose-dependent inhibition of colony formation. This inhibition could not be rescued by stimulatory doses of TGF-beta1. Phosphorylation of AKT was found to occur in a dose-dependent way but declined slightly at the highest concentration used (10 ng/ml). Inhibition of the AKT pathway by LY 294002 strongly suppressed colony formation. These data indicate clearly that sustained activation of p44/42 MAPK perhaps forms the stimulatory signal induced by low TGF-beta1, whereas activation of p38 forms the inhibitory pathway.
Collapse
Affiliation(s)
- V P Kale
- National Center for Cell Science (NCCS), Ganeshkhind, Pune 411007, Maharashtra, India.
| |
Collapse
|
26
|
Akel S, Petrow-Sadowski C, Laughlin MJ, Ruscetti FW. Neutralization of autocrine transforming growth factor-beta in human cord blood CD34(+)CD38(-)Lin(-) cells promotes stem-cell-factor-mediated erythropoietin-independent early erythroid progenitor development and reduces terminal differentiation. Stem Cells 2004; 21:557-67. [PMID: 12968110 DOI: 10.1634/stemcells.21-5-557] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Transforming growth factor (TGF)-beta1 exerts autocrine and paracrine effects on hematopoiesis. Here, we have attempted to evaluate the effect of endogenous TGF-beta1 on early erythroid development from primitive human hematopoietic stem cells (HSCs) and to assess the effects of TGF-beta1 on different phases of erythropoiesis. Cord blood CD34(+)CD38(-) lineage-marker-negative (Lin(-)) cells were cultured in serum-free conditions using various combinations of stem cell factor (SCF), erythropoietin (Epo), and TGF-beta-neutralizing antibody. Generation of erythroid progenitors was assessed using colony assay and flow cytometry. Terminal erythroid differentiation was examined when SCF/Epo-stimulated cells were recultured in the presence of Epo with and without TGF-beta1. Anti-TGF-beta augmented the proliferation of CD34(+)CD38(-)Lin(-) cells (day 21) in SCF-stimulated (6.4-fold +/- 1.5-fold) and SCF/Epo-stimulated (2.9-fold +/- 1.2-fold) cultures. Cells stimulated by SCF/Epo underwent similar levels of erythroid differentiation with and without anti-TGF-beta. While SCF alone stimulated the production of tryptase-positive mast cells, cells stimulated by SCF/anti-TGF-beta were predominantly erythroid (CD36(+)CD14(-) and glycophorin A positive). A distinct expansion of erythroid progenitors (CD34(+)CD36(+)CD14(-)) with the potential to form erythroid colonies was seen, revealing early Epo-independent erythroid development. In contrast, the kinetics of erythroid progenitor generation from primitive HSCs indicate that TGF-beta1 is not inhibitory in late erythropoiesis, but it accelerated the conversion of large BFU-E into colony-forming units-erythroid. Finally, TGF-beta1 accelerated Epo-induced terminal erythroid differentiation and resulted in a greater level of enucleation (22% +/- 6% versus 7% +/- 3%) in serum-free conditions. Serum addition stimulated enucleation (54% +/- 18%), which was lower (26% +/- 14%) with anti-TGF-beta, suggesting that optimal erythroid enucleation is Epo dependent, requiring serum factors including TGF-beta1.
Collapse
Affiliation(s)
- Salem Akel
- Leukocyte Biology Section, Basic Research Laboratory, Center for Cancer Research, NCI-Frederick, Frederick, Maryland 21702-1201, USA
| | | | | | | |
Collapse
|
27
|
Larsson J, Blank U, Helgadottir H, Björnsson JM, Ehinger M, Goumans MJ, Fan X, Levéen P, Karlsson S. TGF-beta signaling-deficient hematopoietic stem cells have normal self-renewal and regenerative ability in vivo despite increased proliferative capacity in vitro. Blood 2003; 102:3129-35. [PMID: 12842983 DOI: 10.1182/blood-2003-04-1300] [Citation(s) in RCA: 115] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Studies in vitro implicate transforming growth factor beta (TGF-beta) as a key regulator of hematopoiesis with potent inhibitory effects on progenitor and stem cell proliferation. In vivo studies have been hampered by early lethality of knock-out mice for TGF-beta isoforms and the receptors. To directly assess the role of TGF-beta signaling for hematopoiesis and hematopoietic stem cell (HSC) function in vivo, we generated a conditional knock-out model in which a disruption of the TGF-beta type I receptor (T beta RI) gene was induced in adult mice. HSCs from induced mice showed increased proliferation recruitment when cultured as single cells under low stimulatory conditions in vitro, consistent with an inhibitory role of TGF-beta in HSC proliferation. However, induced T beta RI null mice show normal in vivo hematopoiesis with normal numbers and differentiation ability of hematopoietic progenitor cells. Furthermore HSCs from T beta RI null mice exhibit a normal cell cycle distribution and do not differ in their ability long term to repopulate primary and secondary recipient mice following bone marrow transplantation. These findings challenge the classical view that TGF-beta is an essential negative regulator of hematopoietic stem cells under physiologic conditions in vivo.
Collapse
Affiliation(s)
- Jonas Larsson
- Department of Molecular Medicine and Gene Therapy, Institute of Laboratory Medicine, Lund University Hospital, BMC A12, 221 84 Lund, Sweden.
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Duman M, Irken G, Oren H, Uçar C, Atabay B, Yilmaz S, Yüksel F, Ozkan H. Plasma transforming growth factor beta1 levels in thrombocytopenic and nonthrombocytopenic neonates. Neonatology 2003; 84:206-13. [PMID: 14504444 DOI: 10.1159/000072304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In this study, we determined the plasma TGF-beta1 levels in healthy and thrombocytopenic and nonthrombocytopenic neonates who had perinatal risk factors and examined the association between plasma TGF-beta1 levels and platelet counts in these newborns to investigate the role of TGF-beta1 in the pathogenesis of neonatal thrombocytopenia. Three groups were defined in this prospective study: group 1, thrombocytopenic neonates (n=22) who had perinatal risk factors; group 2, nonthrombocytopenic neonates who had similar perinatal risk factors for thrombocytopenia (n=20); group 3, healthy and nonthrombocytopenic neonates without any risk factors (n=20). Plasma TGF-beta1 levels were measured with ELISA. Plasma TGF-beta1 levels of the thrombocytopenic neonates were significantly lower than those of healthy nonthrombocytopenic neonates but did not differ significantly from nonthrombocytopenic neonates who had similar perinatal risk factors for thrombocytopenia. There was a significant positive correlation between plasma TGF-beta1 levels and platelet counts. Further studies are needed to determine the cause of low plasma TGF-beta1 levels in thrombocytopenic neonates and to investigate the role of plasma TGF-beta1 levels in the pathogenesis of neonatal thrombocytopenia.
Collapse
Affiliation(s)
- Murat Duman
- Departments of Hematology and Neonatology, Dokuz Eylül University Faculty of Medicine, Izmir, Turkey
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Suzuki M, Harashima A, Okochi A, Yamamoto M, Matsuo Y, Motoda R, Yoshioka T, Orita K. Transforming growth factor-beta(1) augments granulocyte-macrophage colony-stimulating factor-induced proliferation of umbilical cord blood CD34(+) cells with an associated tyrosine phosphorylation of STAT5. Exp Hematol 2002; 30:1132-8. [PMID: 12384143 DOI: 10.1016/s0301-472x(02)00902-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
OBJECTIVE Several investigators have reported that transforming growth factor (TGF)-beta(1) and granulocyte-macrophage colony-stimulating factor (GM-CSF) synergistically support cell proliferation. However, the mechanisms involved have not been elucidated. To clarify the mechanisms of the synergistic action of TGF-beta(1) and GM-CSF, we compared the activation states of STAT5 and mitogen-activated protein kinase in CD34(+) cells and in GM-CSF-dependent hematopoietic cell lines. MATERIALS AND METHODS Human CD34(+) cells and GM-CSF-dependent cell lines (FKH-1, YNH-1, and M-07e) were stimulated with 1.25 ng/mL GM-CSF and/or 0.25 ng/mL TGF-beta(1), and 1.25 ng/mL GM-CSF and/or 0.25 ng/mL, 0.025 ng/mL TGF-beta(1), respectively, and cell proliferation was analyzed by [3H]thymidine uptake. Expression of signal transduction proteins and their phosphorylation states were determined by Western blotting. RESULTS TGF-beta(1) synergistically enhanced the GM-CSF-augmented growth of CD34(+) cells and FKH-1 cells, but inhibited the growth of YNH-1 and M-07e cells. Tyrosine phosphorylation of STAT5 induced by GM-CSF was enhanced by stimulation with the combination of TGF-beta(1) and GM-CSF (TGF-beta(1)/GM-CSF) compared with that induced by GM-CSF alone in CD34(+) cells and FKH-1 cells. However, combinations of TGF-beta(1)/GM-CSF caused inhibition of GM-CSF-induced tyrosine phosphorylation in M-07e cells. No significant difference was observed in mitogen-activated protein kinase activation between CD34(+) cells and FKH-1 cells stimulated with GM-CSF/TGF-beta(1) or GM-CSF alone. CONCLUSIONS Results suggest that TGF-beta(1) may augment GM-CSF-induced proliferation of CD34(+) cells in association with enhanced tyrosine phosphorylation of STAT5. Our data suggest a novel mechanism for the synergistic enhancement of cellular growth induced by the combination of TGF-beta(1) and GM-CSF.
Collapse
Affiliation(s)
- Motoyuki Suzuki
- Fujisaki Cell Center, Hayashibara Biochemical Laboratories, Inc., Okayama, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Cheng T, Scadden DT. Cell cycle entry of hematopoietic stem and progenitor cells controlled by distinct cyclin-dependent kinase inhibitors. Int J Hematol 2002; 75:460-5. [PMID: 12095144 DOI: 10.1007/bf02982107] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The therapeutic promise of hematopoietic stem cells in medicine has been expanded as broader differentiation potential of the cells has gained experimental support. However, hurdles for stem cell manipulation in vitro and tissue regeneration in vivo remain because of lack of the molecular biology of the stem cells. In particular, elucidating the molecular control of cell cycle entry is necessary for rational stem cell expansion strategies. Understanding how the stem and progenitor cell populations are controlled by negative regulators of cell cycle entry may provide one basis for manipulating these cells. In this mini-review, we focus on the rationale of targeting the cyclin-dependent kinase inhibitors (CKIs) in stem cell biology. Two CKI members, p21(Cip1/Waf1) (p21) and p27kip1 (p27), have been shown to govern the pool sizes of hematopoietic stem and progenitor cells, respectively. Of note, their inhibitory roles in primitive hematopoietic cells are distinct from the action of the inhibitory cytokine, transforming growth factor-beta1 (TGF-beta1). Therefore, the distinct roles of p21, p27, and TGF-beta1 in hematopoietic cells offer attractive targets for specific manipulation of the stem or progenitor cell populations in therapeutic strategies.
Collapse
Affiliation(s)
- Tao Cheng
- Department of Radiation Oncology and University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pennsylvania 15213, USA.
| | | |
Collapse
|
31
|
Wright SH, Brown J, Knight PA, Thornton EM, Kilshaw PJ, Miller HRP. Transforming growth factor-beta1 mediates coexpression of the integrin subunit alphaE and the chymase mouse mast cell protease-1 during the early differentiation of bone marrow-derived mucosal mast cell homologues. Clin Exp Allergy 2002; 32:315-24. [PMID: 11929499 DOI: 10.1046/j.1365-2222.2002.01233.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND Mucosal mast cells (MMC) play a central role in gut hypersensitivities and inflammation. They are morphologically, biochemically and functionally distinct from their connective tissue counterparts. Massive hyperplasia of MMC occurs 7-10 days after intestinal infection with nematodes but it has never been possible to replicate this phenomenon in vitro. OBJECTIVE (1) To determine whether mouse bone marrow-derived mast cells (mBMMC) grown in the presence of transforming growth factor (TGF)-beta1 could develop over the same time frame (7-10 days) as MMC in parasitized mice. (2) To compare the early expression of surface receptors (integrins alphaE and beta7, c-kit and FcepsilonR) with that of the MMC-specific granule chymase mouse mast cell protease-1 (mMCP-1). METHODS Mouse bone marrow cells were cultured in the presence of IL-9, IL-3 and Stem Cell Factor (SCF) with or without TGF-beta1. mBMMC were quantified after toluidine blue or Leishmans' staining. Expression of MMC-specific mouse mast cell proteases was analysed by ELISA, immunohistochemistry and RT-PCR. Surface antigen expression was characterized by flow cytometry and confocal microscopy. RESULTS TGF-beta1 promotes the development of abundant MMC-like mBMMC from bone marrow progenitor cells with kinetics, which closely parallel that seen in vivo. mRNA transcripts encoding mMCP-1 and -2 are readily detectable by day 4 ex vivo in cultures grown in the presence of TGF-beta1. Between 30 and 40% and 75-90% of the cells in these cultures on days 4 and 7, respectively, have typical mast cell morphology, are c-kit+, FcepsilonR+, integrin alphaEbeta7+, and express and secrete abundant mMCP-1. The integrin alphaE subunit is coexpressed with mMCP-1. CONCLUSION The kinetics of mMCP-1+/alphaE+ mBMMC development, regulated by TGF-beta1, are consistent with that seen in vivo in the parasitized intestine. The normally down-regulatory functions of TGF-beta1 in haematopoiesis are superseded in this culture system by its ability to promote the early expression of alphaE and mMCP-1.
Collapse
Affiliation(s)
- S H Wright
- Department of Veterinary Clinical Studies, Wellcome Trust Centre For Research in Comparative Respiratory Medicine, Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Easter Bush, Roslin, Midlothian EH25 9RG, Scotland, UK
| | | | | | | | | | | |
Collapse
|
32
|
Pinto do O P, Wandzioch E, Kolterud A, Carlsson L. Multipotent hematopoietic progenitor cells immortalized by Lhx2 self-renew by a cell nonautonomous mechanism. Exp Hematol 2001; 29:1019-28. [PMID: 11495708 DOI: 10.1016/s0301-472x(01)00666-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
OBJECTIVE Direct molecular and cellular studies of hematopoietic stem cells (HSCs) are hampered by the low levels of HSCs in hematopoietic tissues. To address these issues, we generated immortalized multipotent hematopoietic precursor cell (HPC) lines by expressing the LIM-homeobox gene Lhx2 (previously LH2) in hematopoietic progenitors derived from embryonic stem cells differentiated in vitro. MATERIALS AND METHODS To validate further the relevance of the HPC lines as a model for normal HSCs, we analyzed in detail the growth requirements of HPC lines in vitro. RESULTS Lhx2 immortalized the HPC lines by a putatively novel and cell nonautonomous mechanism. Self-renewal of the HPC lines is dependent on functional Lhx2 expression. Most early-acting hematopoiesis-related growth factors show synergistic effects on the HPC lines, whereas late-acting factors do not induce differentiation by themselves. Transforming growth factor-beta(1) is a potent inhibitor of proliferation of the HPC lines. HPC lines form cobblestone areas with high efficiency when seeded onto stromal cell lines, and the cobblestone area-forming cell can be maintained in these cultures for several months. CONCLUSIONS Our data show that, in many respects, HPC lines are similar to normal hematopoietic progenitor/stem cells on the cellular level, in contrast to most previously described multipotent hematopoietic cell lines. The cell nonautonomous mechanism for immortalization of the HPC lines suggests that Lhx2 regulates, directly or indirectly, soluble mediators involved in self-renewal of the HPC lines.
Collapse
Affiliation(s)
- P Pinto do O
- Department of Microbiology, University of Umeå, Umeå, Sweden
| | | | | | | |
Collapse
|
33
|
Ruscetti FW, Bartelmez SH. Transforming growth factor beta, pleiotropic regulator of hematopoietic stem cells: potential physiological and clinical relevance. Int J Hematol 2001; 74:18-25. [PMID: 11530800 DOI: 10.1007/bf02982545] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Transforming growth factor beta (TGF-beta) is a pleiotropic regulator of all stages of hematopoieis. Depending on the differentiation stage of the target cell, the local environment, and the concentration of TGF-beta, TGF-beta can be proproliferative or antiproliferative, proapoptotic or antiapoptotic, and/or prodifferentiative or antidifferentiative. TGF-beta is the major regulator of stem cell quiescence and can act directly or indirectly through effects on the marrow microenvironment. In addition, paracrine and autocrine actions of TGF-beta have overlapping but distinct regulatory effects on hematopoietic stem/progenitor cells. Neutralization of autocrine TGF-beta has therapeutic potential.
Collapse
Affiliation(s)
- F W Ruscetti
- The Basic Research Laboratory, Center for Cancer Research, National Cancer Institute-Frederick, Maryland 21702-1201, USA.
| | | |
Collapse
|
34
|
Ohno N, Izawa A, Hattori M, Kageyama R, Sudo T. dlk inhibits stem cell factor-induced colony formation of murine hematopoietic progenitors: Hes-1-independent effect. Stem Cells 2001; 19:71-9. [PMID: 11209092 DOI: 10.1634/stemcells.19-1-71] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Delta-like (dlk) is a family of transmembrane proteins containing epidermal growth factor-like repeat motifs homologous to the notch/delta/serrate family. Recent studies suggest that dlk is a negative regulator of adipocyte differentiation, a promoting factor of cobblestone area colony formation, and a molecule which influences stromal cell-pre-B cell interactions and augments cellularity of developing thymocytes. However, the role of dlk in regulating the growth and differentiation of hematopoietic progenitors remains unclear. In the present study, we examined the effect of dlk on the proliferation of murine hematopoietic progenitors by hematopoietic growth factors. Soluble dlk-IgG Fc chimeric protein completely inhibited the colony formation of lineage-marker negative (Lin-) bone marrow cells by GM-CSF, G-CSF, or macrophage-CSF (M-CSF) in the presence of stem cell factor (SCF). However, dlk failed to inhibit the colony formation of Lin- bone marrow cells by CSF, as described above, or M-CSF plus interleukin 3. Furthermore, dlk failed to inhibit the colony formation of Hes-1-null fetal liver cells by M-CSF in the presence of SCF. These findings suggest that dlk is an important regulator of hematopoietic progenitor proliferation. Depending on the presence of SCF, dlk may act as a growth inhibitor, although dlk signaling does not mediate Hes-1 transcription factor.
Collapse
Affiliation(s)
- N Ohno
- Pharmaceutical Research Laboratories, Toray Industries, Inc., Tebiro, Kamakura, Japan
| | | | | | | | | |
Collapse
|
35
|
Kouro T, Medina KL, Oritani K, Kincade PW. Characteristics of early murine B-lymphocyte precursors and their direct sensitivity to negative regulators. Blood 2001; 97:2708-15. [PMID: 11313262 DOI: 10.1182/blood.v97.9.2708] [Citation(s) in RCA: 85] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Recently, a collection of surface markers was exploited to isolate viable Lin(-) TdT(+) cells from murine bone marrow. These early pro-B cells were enriched for B-lineage lymphocyte precursor activity measured by short-term culture and had little responsiveness to myeloid growth factors. Early precursors can be propagated with remarkably high cloning frequencies in stromal cell-free, serum-free cultures, permitting this analysis of direct regulatory factors. Expression of the interleukin-7 receptor (IL-7Ralpha) chain marks functional precursors and IL-7 is necessary for progression beyond the CD45RA(+) CD19(-) stage. Efficient survival and differentiation were only observed when stem cell factor and Flt-3 ligand were also present. IL-7-responsive CD19(+) precursors are estrogen resistant. However, B-lineage differentiation was selectively abrogated when highly purified Lin(-) precursors were treated with hormone in the absence of stromal cells. In addition, early stages of B lymphopoiesis were arrested by limitin, a new interferon (IFN)-like cytokine as well as IFN-alpha, IFN-gamma, or transforming growth factor beta (TGF-beta), but not by epidermal growth factor (EGF). Lin(-) TdT(+) early pro-B cells are shown here to be CD27(+) AA4.1(+/-)Ki-67(+) Ly-6C(-) Ly-6A/Sca-1(Lo/-)Thy-1(-)CD43(+) CD4(+/-)CD16/32(Lo/-)CD44(Hi) and similar in some respects to the "common lymphoid progenitors" (CLP) identified by others. Although early pro-B cells have lost myeloid differentiation potential, transplantation experiments described here reveal that at least some can generate T lymphocytes. Of particular importance is the demonstration that a pivotal early stage of lymphopoiesis is directly sensitive to negative regulation by hormones and cytokines.
Collapse
Affiliation(s)
- T Kouro
- Immunobiology and Cancer Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | | | | | | |
Collapse
|
36
|
Francis JM, Heyworth CM, Spooncer E, Pierce A, Dexter TM, Whetton AD. Transforming growth factor-beta 1 induces apoptosis independently of p53 and selectively reduces expression of Bcl-2 in multipotent hematopoietic cells. J Biol Chem 2000; 275:39137-45. [PMID: 10993901 DOI: 10.1074/jbc.m007212200] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Transforming growth factor-beta1 (TGF-beta1) can inhibit cell proliferation or induce apoptosis in multipotent hematopoietic cells. To study the mechanisms of TGF-beta1 action on primitive hematopoietic cells, we used the interleukin-3 (IL-3)-dependent, multipotent FDCP-Mix cell line. TGF-beta1-mediated growth inhibition was observed in high concentrations of IL-3, while at lower IL-3 concentrations TGF-beta1 induced apoptosis. The proapoptotic effects of TGF-beta1 occur via a p53-independent pathway, since p53(null) FDCP-Mix demonstrated the same responses to TGF-beta1. IL-3 has been suggested to enhance survival via an increase in (antiapoptotic) Bcl-x(L) expression. In FDCP-Mix cells, neither IL-3 nor TGF-beta1 induced any change in Bcl-x(L) protein levels or the proapoptotic proteins Bad or Bax. However, TGF-beta1 had a major effect on Bcl-2 levels, reducing them in the presence of high and low concentrations of IL-3. Overexpression of Bcl-2 in FDCP-Mix cells rescued them from TGF-beta1-induced apoptosis but was incapable of inhibiting TGF-beta1-mediated growth arrest. We conclude that TGF-beta1-induced cell death is independent of p53 and inhibited by Bcl-2, with no effect on Bcl-x(L). The significance of these results for stem cell survival in bone marrow are discussed.
Collapse
Affiliation(s)
- J M Francis
- Leukaemia Research Fund Cellular Development Unit, Department of Biomolecular Sciences, UMIST, Sackville St., Manchester, M60 1QD, United Kingdom
| | | | | | | | | | | |
Collapse
|
37
|
Katayama N, Mahmud N, Nishii K, Ohishi K, Masuya M, Mitani H, Araki H, Suzuki H, Kobayashi K, Minami N, Shiku H. Bcl-2 in cell-cycle regulation of hematopoietic cells by transforming growth factor-beta1. Leuk Lymphoma 2000; 39:601-5. [PMID: 11342343 DOI: 10.3109/10428190009113390] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
We reported that several growth factors regulate the doubling time of hematopoietic progenitor cells by modulating the time required to pass through the G1 phase. As recent studies revealed the link between cell death and cell-cycle progression, we asked if cell death regulators such as Bcl-2 play a role in regulating the cell-cycle of hematopoietic cells by growth factors. Among growth factors, transforming growth factor-beta1 (TGF-beta1), a negative regulator of hematopoiesis, was chosen. When a large number of cells was required for analysis, we used IL-3-dependent Ba/F3 cells instead of primary hematopoietic progenitor cells because the response of Ba/F3 cells to TGF-beta1 was similar to that of primary hematopoietic progenitor cells. TGF-beta1 decelerated the cell-cycling of hematopoietic cells by inducing a delay in G1 to S phase transition, an event associated with increase in the level of Bcl-2 as well as p27, a cyclin/cyclin-dependent kinase inhibitor. In experiments using Ba/F3 cells with the potential to produce Bcl-2 in an inducible manner, Bcl-2 apparently functions upstream of p27. The effects of TGF-beta1 on Bcl-2 and p27 expression as well as cell growth were abrogated by c-kit ligand. These findings suggest that Bcl-2 plays a crucial role in regulating the cell-cycle of hematopoietic progenitor cells.
Collapse
Affiliation(s)
- N Katayama
- The Second Department of Internal Medicine, Mie University School of Medicine, Tsu, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
AbstractHematopoiesis is a remarkable cell-renewal process that leads to the continuous generation of large numbers of multiple mature cell types, starting from a relatively small stem cell compartment. A highly complex but efficient regulatory network is necessary to tightly control this production and to maintain the hematopoietic tissue in homeostasis. During the last 3 decades, constantly growing numbers of molecules involved in this regulation have been identified. They include soluble cytokines and growth factors, cell–cell interaction molecules, and extracellular matrix components, which provide a multifunctional scaffolding specific for each tissue. The cloning of numerous growth factors and their mass production have led to their possible use for both fundamental research and clinical application.
Collapse
|
39
|
Abstract
Hematopoiesis is a remarkable cell-renewal process that leads to the continuous generation of large numbers of multiple mature cell types, starting from a relatively small stem cell compartment. A highly complex but efficient regulatory network is necessary to tightly control this production and to maintain the hematopoietic tissue in homeostasis. During the last 3 decades, constantly growing numbers of molecules involved in this regulation have been identified. They include soluble cytokines and growth factors, cell–cell interaction molecules, and extracellular matrix components, which provide a multifunctional scaffolding specific for each tissue. The cloning of numerous growth factors and their mass production have led to their possible use for both fundamental research and clinical application.
Collapse
|
40
|
de Vries P, Singer JW. Lisofylline suppresses ex vivo release by murine spleen cells of hematopoietic inhibitors induced by cancer chemotherapeutic agents. Exp Hematol 2000; 28:916-23. [PMID: 10989192 DOI: 10.1016/s0301-472x(00)00493-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Many cytotoxic cancer therapeutic drugs activate stress response signaling pathways that transcriptionally activate a variety of genes. We decided to determine if cytotoxic therapies induce inflammatory cytokines with inhibitory effects on hematopoiesis and if lisofylline (LSF), a novel antiinflammatory compound, suppresses this induction. Mice were treated with cytosine beta-d-arabinofuranoside (AraC), cis-platinum(II)diammine-dichloride (CisP), etoposide (VP-16), or melphalan at clinically relevant doses, with or without LSF. Spleen cell conditioned media (CM) derived from mice treated with cytotoxic agents, but not from control or LSF treated mice, reduced colony formation by murine bone marrow progenitors belonging to the myeloid, erythroid, megakaryocytic, and B-lymphoid lineages. LSF (100 mg/kg), administered either simultaneously with or up to 48 hours before the cytotoxic agents, suppressed the release of this inhibitory activity. Treatment of inhibitory CM with neutralizing antibodies against known growth inhibitory cytokines, including tumor necrosis factor alpha, transforming growth factor beta, and macrophage inflammatory protein-1alpha, resulted in enhanced colony growth. We conclude that treatment of mice with chemotherapeutic drugs induces the ex vivo production of multilineage hematopoietic inhibitors and that induction of these inhibitors could be abrogated by treatment with LSF. These findings suggest a mechanism whereby LSF can accelerate recovery of hematopoiesis following cytotoxic therapies.
Collapse
Affiliation(s)
- P de Vries
- Cell Therapeutics, Inc., Seattle, WA 98119, USA.
| | | |
Collapse
|
41
|
Matsumura T, Whelan MC, Li XQ, Trippel SB. Regulation by IGF-I and TGF-beta1 of Swarm-rat chondrosarcoma chondrocytes. J Orthop Res 2000; 18:351-5. [PMID: 10937620 DOI: 10.1002/jor.1100180305] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The growth factors transforming growth factor-beta 1 and insulin-like growth factor-I influence a wide range of cellular actions, including the growth of several neoplastic cell types. Their role in the regulation of neoplastic chondrocytes remains unclear. We tested the hypotheses that transforming growth factor-beta 1 and insulin-like growth factor-I differentially regulate neoplastic chondrocytes and interact to modulate the mitotic and matrix synthetic activities of neoplastic chondrocytes. We used Swarm-rat chondrosarcoma chondrocytes to investigate the effect of each factor individually and of both factors in combination on [(3)H]thymidine incorporation into DNA and on [(35)S]sulfate incorporation into glycosaminoglycans. Each factor increased [(3)H]thymidine incorporation 2.7-fold: transforming growth factor-beta 1 achieved this effect at a 20-fold lower concentration than insulin-like growth factor-I. In contrast, insulin-like growth factor-I stimulated [(35)S]sulfate incorporation 3.5-fold; this was twice the maximal effect of transforming growth factor-beta 1. Transforming growth factor-beta 1 and insulin-like growth factor-I each decreased the proportion of newly synthesized glycosaminoglycans that were retained in the cells and pericellular matrix, indicating that the anabolic effect of these factors is only partly directed toward cell-associated matrix production. The mitogenic and matrix synthetic actions of insulin-like growth factor-I and transforming growth factor-beta 1 were synergistic. In concert, they increased [(3)H]thymidine incorporation approximately 12-fold, an effect three times greater than the sum of the maximal stimulation achieved by each factor individually. Similarly, transforming growth factor-beta 1 and insulin-like growth factor-I together increased glycosaminoglycan synthesis approximately two times more than the sum of their maximal individual effects. Taken together, these data indicate that these chondrosarcoma chondrocytes are positively regulated by insulin-like growth factor-I and transforming growth factor-beta 1 and that these growth factors interact to augment the mitotic and matrix synthetic actions of the chondrocytes. If supported in human models, the sensitivity to growth factors of these cells suggests that interventions directed toward growth factor inhibition may be of therapeutic value.
Collapse
Affiliation(s)
- T Matsumura
- Department of Orthopaedic Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, USA
| | | | | | | |
Collapse
|
42
|
Letterio JJ. Murine models define the role of TGF-beta as a master regulator of immune cell function. Cytokine Growth Factor Rev 2000; 11:81-7. [PMID: 10708955 DOI: 10.1016/s1359-6101(99)00031-3] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Many members of transforming growth factor-beta (TGF-beta) superfamily, including not only TGF-beta, but also the activins, and bone morphogenetic proteins (BMPs), have been demonstrated to affect the development and function of immune cells. From the proliferation and differentiation of pluripotent stem cells, to the activation and migration of mature lymphoid and myeloid lineages, the TGF-betas have been recognized for their ability to modulate the manner in which such cells respond to stimuli in their environment. Recent studies involving disruption of this pathway in genetically engineered mice now emphasize the importance of this activity and validate functional models predicted by in vitro studies. Phenotypic differences between mice harboring mutations in the TGF-beta1 ligand and the TGF-beta receptor-activated signaling intermediate Smad3 are presented and serve to highlight the valuable role of these in vivo genetic tests of function.
Collapse
Affiliation(s)
- J J Letterio
- Laboratory of Cell Regulation and Carcinogenesis, The National Cancer Institute, NIH, Bethesda, MD 20892-5055, USA.
| |
Collapse
|
43
|
Batard P, Monier MN, Fortunel N, Ducos K, Sansilvestri-Morel P, Phan T, Hatzfeld A, Hatzfeld JA. TGF-(beta)1 maintains hematopoietic immaturity by a reversible negative control of cell cycle and induces CD34 antigen up-modulation. J Cell Sci 2000; 113 ( Pt 3):383-90. [PMID: 10639326 DOI: 10.1242/jcs.113.3.383] [Citation(s) in RCA: 111] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Somatic stem cells are largely quiescent in spite of their considerable proliferative potential. Transforming growth factor-(beta)1 (TGF-(beta)1) appears to be a good candidate for controlling this quiescence. Indeed, various mutations in the TGF-beta signalling pathway are responsible for neoplasic proliferation of primitive stem/progenitor cells in human tissues of various origins. In hemopoietic single cell culture assays, blocking autocrine and endogeneous TGF-(beta)1 triggers the cell cycling of high proliferative potential undifferenciated stem/progenitor cells. However, it has never been demonstrated whether TGF-(beta)1 has an apoptotic effect or a differentiating effect on these primitive cells, as already described for more mature cells. Using single cell experiments both in liquid or semi-solid culture assays and dye tracking experiments by flow cytometry, we demonstrate that low, physiological concentrations of TGF-(beta)1, which specifically maintain primitive human hemopoietic stem/progenitor cells in quiescence, have a reversible effect and do not induce apoptosis. We moreover demonstrate that these low concentrations prevent the rapid loss of the mucin-like protein CD34, a most common marker of immature hematopoietic stem/progenitor cells, which is progressively lost during differentiation. TGF-(beta)1 not only up-modulated the CD34 antigen before S phase entry but also maintained a high level of CD34 expression on cells which had escaped cell cycle inhibition, suggesting that proliferation inhibition and differentiation control by TGF-(beta)1 may be independent. These data provide additional evidence that TGF-(beta)1 acts as a key physiological factor ensuring the maintenance of a stem cell reserve.
Collapse
Affiliation(s)
- P Batard
- Laboratoire de Biologie Cellulaire et Moléculaire des Facteurs de Croissance, UPR 1983 du Centre National de la Recherche Scientifique, IFC1, rue Guy Moquet, France
| | | | | | | | | | | | | | | |
Collapse
|
44
|
All-Trans Retinoic Acid Delays the Differentiation of Primitive Hematopoietic Precursors (lin−c-kit+Sca-1+) While Enhancing the Terminal Maturation of Committed Granulocyte/Monocyte Progenitors. Blood 1999. [DOI: 10.1182/blood.v94.2.483] [Citation(s) in RCA: 90] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
All-trans retinoic acid (ATRA) is a potent inducer of terminal differentiation of malignant promyelocytes, but its effects on more primitive hematopoietic progenitors and stem cells are less clear. In this study, we investigated the effect of ATRA on highly enriched murine hematopoietic precursor cells (lin−c-kit+Sca-1+) grown in liquid suspension culture for 28 days. ATRA initially slowed the growth of these hematopoietic precursors but prolonged and markedly enhanced their colony-forming cell production compared with the hematopoietic precursors cultured in its absence. At 7 and 14 days of culture, a substantially greater percentage of cells cultured with ATRA did not express lineage-associated antigens (55.4% at day 7 and 68.6% at day 14) and retained expression of Sca-1 (44.7% at day 7 and 79.9% at day 14) compared with cells grown in its absence (lin−cells: 31.5% at day 7 and 4% at day 14; Sca-1+: 10.4% at day 7 and 0.7% at day 14). Moreover, a marked inhibition of granulocyte production was observed in cultures continuously incubated with ATRA. Significantly, ATRA markedly prolonged and enhanced the production of transplantable colony-forming unit-spleen (CFU-S) during 14 days of liquid suspension culture. In contrast with its effects on primitive lin−c-kit+Sca-1+hematopoietic precursors, ATRA did not exert the same effects on the more committed lin−c-kit+Sca-1−progenitor cells. Moreover, the late addition of ATRA (7 days post-culture initiation) to cultures of primitive hematopoietic precursors resulted in a marked decrease in colony-forming cell production in these cultures, which was associated with enhanced granulocyte differentiation. These observations indicate that ATRA has different effects on hematopoietic cells depending on their maturational state, preventing and/or delaying the differentiation of primitive hematopoietic precursors while enhancing the terminal differentiation of committed granulocyte/monocyte progenitors.
Collapse
|
45
|
All-Trans Retinoic Acid Delays the Differentiation of Primitive Hematopoietic Precursors (lin−c-kit+Sca-1+) While Enhancing the Terminal Maturation of Committed Granulocyte/Monocyte Progenitors. Blood 1999. [DOI: 10.1182/blood.v94.2.483.414k12_483_495] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
All-trans retinoic acid (ATRA) is a potent inducer of terminal differentiation of malignant promyelocytes, but its effects on more primitive hematopoietic progenitors and stem cells are less clear. In this study, we investigated the effect of ATRA on highly enriched murine hematopoietic precursor cells (lin−c-kit+Sca-1+) grown in liquid suspension culture for 28 days. ATRA initially slowed the growth of these hematopoietic precursors but prolonged and markedly enhanced their colony-forming cell production compared with the hematopoietic precursors cultured in its absence. At 7 and 14 days of culture, a substantially greater percentage of cells cultured with ATRA did not express lineage-associated antigens (55.4% at day 7 and 68.6% at day 14) and retained expression of Sca-1 (44.7% at day 7 and 79.9% at day 14) compared with cells grown in its absence (lin−cells: 31.5% at day 7 and 4% at day 14; Sca-1+: 10.4% at day 7 and 0.7% at day 14). Moreover, a marked inhibition of granulocyte production was observed in cultures continuously incubated with ATRA. Significantly, ATRA markedly prolonged and enhanced the production of transplantable colony-forming unit-spleen (CFU-S) during 14 days of liquid suspension culture. In contrast with its effects on primitive lin−c-kit+Sca-1+hematopoietic precursors, ATRA did not exert the same effects on the more committed lin−c-kit+Sca-1−progenitor cells. Moreover, the late addition of ATRA (7 days post-culture initiation) to cultures of primitive hematopoietic precursors resulted in a marked decrease in colony-forming cell production in these cultures, which was associated with enhanced granulocyte differentiation. These observations indicate that ATRA has different effects on hematopoietic cells depending on their maturational state, preventing and/or delaying the differentiation of primitive hematopoietic precursors while enhancing the terminal differentiation of committed granulocyte/monocyte progenitors.
Collapse
|
46
|
A Novel Function for Transforming Growth Factor-β1: Upregulation of the Expression and the IgE-Independent Extracellular Release of a Mucosal Mast Cell Granule-Specific β-Chymase, Mouse Mast Cell Protease-1. Blood 1999. [DOI: 10.1182/blood.v93.10.3473.410k01_3473_3486] [Citation(s) in RCA: 118] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Intestinal mucosal mast cells (IMMC) express granule neutral proteases that are regulated by T-cell–derived cytokines, including interleukin-3 (IL-3) and IL-9, and by stem cell factor (SCF). The IMMC-specific chymase, mouse mast cell protease-1 (mMCP-1), is released in substantial quantities into the blood stream during gastrointestinal allergic responses. We used cultured bone marrow-derived mast cells (mBMMC) to identify cytokines that regulate the expression and extracellular release of mMCP-1. When grown in IL-3–rich WEHI (15% vol/vol) and 50 ng/mL recombinant rat SCF (rrSCF) bone marrow cells supplemented with IL-9 (5 ng/mL) differentiated into mBMMC that expressed a maximum of less than 250 ng mMCP-1/106 cells and 189 ng mMCP-1/mL of culture supernatant. Supplementation of the same three cytokines with transforming growth factor-β1(TGF-β1; 1 ng/mL) resulted in substantially enhanced expression (6 μg/106 mBMMC) and extracellular release (2 μg/mL of culture supernatant) of mMCP-1. The response to TGF-β1 was dose-dependent, with maximal effect at 1 ng/mL, and was associated with immunohistochemical and ultrastructural changes in the secretory granules. IL-9–induced expression of mMCP-1 may be due to endogenously expressed TGF-β1, because it was blocked by anti–TGF-β antibodies. In conclusion, the expression and extracellular release of the IMMC-specific chymase, mMCP-1, is strictly regulated by TGF-β1.
Collapse
|
47
|
Mahmud N, Katayama N, Nishii K, Sugawara T, Komada Y, Mitani H, Araki H, Ohishi K, Watanabe M, Masuya M, Nishikawa M, Minami N, Ohashi H, Shiku H. Possible involvement of bcl-2 in regulation of cell-cycle progression of haemopoietic cells by transforming growth factor-beta1. Br J Haematol 1999. [DOI: 10.1111/j.1365-2141.1999.01324.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
48
|
Kale VP, Limaye LS. Stimulation of adult human bone marrow by factors secreted by fetal liver hematopoietic cells: in vitro evaluation using semisolid clonal assay system. Stem Cells 1999; 17:107-16. [PMID: 10195571 DOI: 10.1002/stem.170107] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Fetal liver infusion (FLI) therapy has been used in various disorders, such as aplastic anemia, leukemia, metabolic disorders, etc., and has been shown to result in stimulation of autologous hematopoiesis in many cases. The aim of the present study was to elucidate the mechanism of stimulation of adult hematopoiesis by fetal liver hematopoietic cells (FLHC) and to identify the factors involved in the process using a clonal assay system in vitro. The effect of FLHC on the clonal growth of bone marrow cells was studied using a co-culture system consisting of mitomycin C-treated FLHC with 2 x 10(5) bone marrow (BM) mononuclear cells. It was observed that FLHC induced a two- to four-fold increase in the BM colony formation. A further increase in the number of FLHC did not, however, result in an equivalent fold increase in the colony formation, indicating that the number of cells in the BM population responsive to FLHC was perhaps the limiting factor. When the effect of fetal liver cell conditioned medium (FLCM) was examined in a similar fashion, it was observed that the FLCM showed a 1.5- to 4-fold increase in the colony formation when used at 1%-5% along with limiting amounts of growth factors. Higher concentrations of conditioned medium resulted in inhibitory responses. One of the principal factors responsible for the stimulatory activity of FLCM was shown to be transforming growth factor-beta1 (TGF-beta1), by a variety of experiments such as its quantitation in FLCM by enzyme-linked immunosorbent assay, antibody neutralization, and reconstruction experiments using purified TGF-beta1 and normal medium. In these reconstitution experiments, TGF-beta1 stimulated the colony formation when it was applied at 1-50 pg/ml, but at higher concentration it induced an inhibitory effect, mimicking the behavior earlier seen with FLCM. Our data strongly suggest that one of the mechanisms in stimulation of a recipient's hematopoiesis could be mediated by the action of TGF-beta1 secreted by infused FLHC and could provide a rational framework on which FLI therapy can be further evaluated.
Collapse
Affiliation(s)
- V P Kale
- National Centre for Cell Science, Ganeshkhind, Pune, India
| | | |
Collapse
|
49
|
Zhang XJ, Tsung HC, Caen JP, Li XL, Yao Z, Han ZC. Vasculogenesis from embryonic bodies of murine embryonic stem cells transfected by Tgf-beta1 gene. ENDOTHELIUM : JOURNAL OF ENDOTHELIAL CELL RESEARCH 1999; 6:95-106. [PMID: 9930643 DOI: 10.3109/10623329809072196] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Mouse embryonic stem (ES) cells transfected with a 1.7 kb cDNA of porcine transforming growth factor type beta1 (TGFbeta1), known as ES-T cells, were found to be able to differentiate in vitro into cystic embryonic bodies (EBs) with outspread tubular structures. Morphological analysis using light, phase-contrast and electron microscopes revealed that in culture, the EBs of ES-T cells initially developed some flat endothelial-like cells which further proliferated and migrated to form thread structures. At 8-10 days after EB formation, these thread structures further developed into net-like and tubular structures connecting directly to EBs. Immunofluorescent assays using antibodies against Flk-1 and von Willebrand factor (vWF) indicated that these net-like and tubular structures of ES-T cells consisted of vascular endothelial cells. Further analysis by RT-PCR revealed that the EBs with tubular structures expressed the mRNA of other markers of vascular endothelial cells, including VE-cadherin and platelet-endothelial cell adhesion molecule (PECAM). Cells of hematopoietic origin were not detected on the outside of EBs by immunostaining using several antibodies specific for granulocytes, macrophages and lymphocytes as well as by benzidine staining for erythroid cells on the outside of EBs. Our data demonstrates that the transfer of TGFbeta1 into ES cells results in a significant vasculogenesis without concomitant hematopoiesis. ES-T cells could therefore provide an excellent model for studying blood vessel formation and vasculogenic and hematopoietic interactions.
Collapse
Affiliation(s)
- X J Zhang
- Shanghai Institute of Cell Biology, Chinese Academy of Sciences
| | | | | | | | | | | |
Collapse
|
50
|
Blank M, George J, Barak V, Tincani A, Koike T, Shoenfeld Y. Oral Tolerance to Low Dose β2-Glycoprotein I: Immunomodulation of Experimental Antiphospholipid Syndrome. THE JOURNAL OF IMMUNOLOGY 1998. [DOI: 10.4049/jimmunol.161.10.5303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Abstract
Oral tolerance was induced in BALB/c mice by feeding low dose β2-glycoprotein I (β2GPI). The β2GPI-fed mice did not develop serologic and clinical markers of experimental antiphospholipid syndrome (APS) upon immunization with the autoantigen. The treated group was characterized by low titers of serum anti-β2GPI and anticardiolipin Abs in the serum, lack of fetal resorptions, low incidence of thrombocytopenia, and normal aPTT (activated partial thromboplastin time) values. β2GPI given orally before priming with β2GPI resulted in complete prevention of experimental APS development; β2GPI given at an early stage of the disease reduced clinical manifestations. However, administration of β2GPI 70 days postimmunization had a less significant effect on disease expression. Tolerized mice exhibited a diminished T lymphocyte proliferation response to β2GPI in comparison with β2GPI-immunized mice fed with OVA. When nontolerant β2GPI-primed T lymphocytes were mixed with T lymphocytes derived from tolerized mice, a significant inhibition of proliferation upon exposure to β2GPI was observed. The induction of suppression was β2GPI specific and driven, as well as TGF-β mediated. The β2GPI-specific response of T lymphocytes from the β2GPI-fed mice was reversed by anti-TGF-β Abs. The tolerance was adoptively transferred by CD8+ T cells from the tolerized mice into naive mice. Those CD8+ cells were MHC class I restricted, found to secrete TGF-β, and had no cytolytic activity. Oral administration of β2GPI suppressed priming of CTLs in the recipient mice. In sum, β2GPI-induced oral tolerance has an immunomodulatory effect in experimental APS, demonstrating the importance of β2GPI in the pathogenesis of the disease.
Collapse
Affiliation(s)
- Miri Blank
- *Research Unit of Autoimmune Diseases, Department of Medicine B, Sheba Medical Center, Tel-Hashomer, and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Jacob George
- *Research Unit of Autoimmune Diseases, Department of Medicine B, Sheba Medical Center, Tel-Hashomer, and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Vivian Barak
- †Immunology Laboratory for Diagnosis, Oncology Department, Hadassah Medical Center, Jerusalem, Israel
| | - Angela Tincani
- ‡Clinical Immunology Unit, Spedali Civili, Brescia, Italy; and
| | - Takao Koike
- §Department of Medicine II, Hokkaido University School of Medicine, Sapporo, Japan
| | - Yehuda Shoenfeld
- *Research Unit of Autoimmune Diseases, Department of Medicine B, Sheba Medical Center, Tel-Hashomer, and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|