151
|
Johnson K, Shapiro-Shelef M, Tunyaplin C, Calame K. Regulatory events in early and late B-cell differentiation. Mol Immunol 2005; 42:749-61. [PMID: 15829263 DOI: 10.1016/j.molimm.2004.06.039] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2004] [Accepted: 06/18/2004] [Indexed: 01/29/2023]
Abstract
We are studying transcriptional control of critical developmental decision points in B lymphocytes. Commitment to the B-lymphocyte lineage is dependent on the transcriptional regulator Pax5 and committed B lymphocytes represent the first developmental stage when V(H)-to-DJ recombination occurs in the immunoglobulin (Ig) heavy chain locus. We summarize our recent studies showing that methylation of histone H3 lysine 9, a heterochromatic chromatin modification, is present in the Ig V(H) region in hematopoietic progenitors and in non-B lineage hematopoietic cells. Pax5 is both necessary and sufficient to remove this heterochromatic mark in B cells. Using genetically altered mice, we have shown that terminal differentiation of B cells to memory and Ig-secreting plasma cells depends on the transcriptional repressor Blimp-1. Recent studies demonstrating a requirement for Blimp-1 in the formation of pre-plasma memory B cells, Ig-secreting plasma cells as well as preliminary data suggesting a requirement for Blimp-1 in the maintenance of long-lived plasma cells are summarized. We also summarize our recent studies on the regulation of Blimp-1, showing direct repression by Bcl-6 and providing evidence for activation by NF-kappaB following toll-like receptor signaling.
Collapse
Affiliation(s)
- Kristen Johnson
- Department of Microbiology, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | | | | | | |
Collapse
|
152
|
Dias S, Silva H, Cumano A, Vieira P. Interleukin-7 is necessary to maintain the B cell potential in common lymphoid progenitors. ACTA ACUST UNITED AC 2005; 201:971-9. [PMID: 15767371 PMCID: PMC2213099 DOI: 10.1084/jem.20042393] [Citation(s) in RCA: 172] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Interleukin-7 (IL-7) promotes survival and expansion of lymphoid precursors. We show here that, in addition, IL-7 has a fundamental role, as early as the stage of the multipotent (B/T/NK) common lymphoid progenitor (CLP), in maintaining the B cell differentiation program open. CLPs generated in the absence of IL-7 have normal T/NK differentiation potential, but severely impaired B potential. Accordingly, CLPs from IL-7–deficient mice express lower amounts of early B cell factor (EBF) and Pax5 than wild-type CLPs, but similar amounts of GATA-3. Importantly, induced overexpression of EBF is sufficient to restore the B potential in these cells. These results indicate that IL-7 directs commitment of CLPs by modulating EBF expression. This is the first example of a cytokine influencing lymphoid lineage commitment in multipotent progenitors and highlights the relevance of the expression of a functional IL-7 receptor at the CLP stage.
Collapse
Affiliation(s)
- Sheila Dias
- Unité du Développement des Lymphocytes, Institute National de la Santé et de la Recherche Medicale U668, Institut Pasteur, 75724 Paris, France
| | | | | | | |
Collapse
|
153
|
Ardouin L, Rolink AG, Mura AM, Gommeaux J, Melchers F, Busslinger M, Malissen M, Malissen B. Rapidin vivo analysis of mutant forms of the LAT adaptor usingPax5-Lat double-deficient pro-B?cells. Eur J Immunol 2005; 35:977-86. [PMID: 15719364 DOI: 10.1002/eji.200425836] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Following injection into recombinase-activating gene-deficient (Rag1(-/-)) mice, pro-B cells lacking the Pax5 transcription factor (Pax5(-/-)) develop into most major hematopoietic lineages, with the notable exception of B cells. We assessed whether Pax5(-/-) pro-B cells that were also rendered deficient for the linker for activation of T cells (LAT), an adaptor essential for T cell receptor signaling, can be used for the rapid in vivo analysis of mutant forms of LAT. We showed that Pax5(-/-) Lat(-/-) pro-B cell lines can be infected with recombinant retroviruses expressing a LAT cDNA and sorted for the expression of LAT. When injected into Rag1(-/-) mice, they restore normal intrathymic T cell development and give rise to functional peripheral T cells. Considering that the handling of Pax5(-/-) pro-B cell lines is easier than that of bone marrow hematopoietic precursors, we used them for the rapid functional analysis of a novel Lat allelic series. When compared to knock-in and transgenic approaches, a major advantage of our Pax5(-/-) pro-B cell-based experimental approach consists in the production of mice bearing a given mutation within 2-3 months. Therefore, it constitutes a powerful first-line screen for mutations worth fastidious knock-in approaches.
Collapse
Affiliation(s)
- Laurence Ardouin
- Centre d'Immunologie de Marseille-Luminy, INSERM, CNRS, Université de la Méditerranée, Marseille, France
| | | | | | | | | | | | | | | |
Collapse
|
154
|
Abstract
PURPOSE OF REVIEW Theoretic and, in particular, mathematic models can help biologists to select and design experiments, to highlight general principles, to discriminate similar and to link different phenomena, and to predict novel features. Specifically, they contribute to an understanding of latent mechanisms and crucial parameters of biologic processes. The following review gives an overview of recent developments in the field of hematopoietic tissue stem cell modeling. RECENT FINDINGS A number of experimental findings on heterogeneity, flexibility, and plasticity of hematopoietic and other tissue stem cells are challenging the classic stem cell concept of a predefined intrinsic stem cell program. Self-organizing systems provide a more elegant and comprehensive alternative to explain experimental data. SUMMARY Within the last few decades, modeling approaches in stem cell biology have evolved and now encompass a broad spectrum of phenomena, ranging from the cellular level to the tissue level. The application of theoretic models is currently suggesting that we abandon the classic assumption of a strict developmental hierarchy and understand stem cell organization as a dynamic, functional process. Such a perspective has implications for a prospective characterization of tissue stem cells (eg, regarding gene expression profiles and genetic regulation patterns).
Collapse
Affiliation(s)
- Markus Loeffler
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Germany.
| | | |
Collapse
|
155
|
Affiliation(s)
- Barbara A Osborne
- Department of Veterinary & Animal Sciences, University of Massachusetts, Amherst, Massachusetts 01003, USA.
| | | |
Collapse
|
156
|
Neuroblastoma and pre-B lymphoma cells share expression of key transcription factors but display tissue restricted target gene expression. BMC Cancer 2004; 4:80. [PMID: 15544702 PMCID: PMC544580 DOI: 10.1186/1471-2407-4-80] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2004] [Accepted: 11/15/2004] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Transcription factors are frequently involved in the process of cellular transformation, and many malignancies are characterized by a distinct genetic event affecting a specific transcription factor. This probably reflects a tissue specific ability of transcription factors to contribute to the generation of cancer but very little is known about the precise mechanisms that governs these restricted effects. METHODS To investigate this selectivity in target gene activation we compared the overall gene expression patterns by micro-array analysis and expression of target genes for the transcription factor EBF in lymphoma and neuroblastoma cells by RT-PCR. The presence of transcription factors in the different model cell lines was further investigated by EMSA analysis. RESULTS In pre-B cells mb-1 and CD19 are regulate by EBF-1 in collaboration with Pax-5 and E-proteins. We here show that neuroblastoma cells express these three, for B cell development crucial transcription factors, but nevertheless fail to express detectable levels of their known target genes. Expression of mb-1 could, however, be induced in neuroblastoma cells after disruption of the chromatin structure by treatment with 5-azacytidine and Trichostatin A. CONCLUSION These data suggest that transcription factors are able to selectively activate target genes in different tissues and that chromatin structure plays a key role in the regulation of this activity.
Collapse
|
157
|
Bellantuono I. Haemopoietic stem cells. Int J Biochem Cell Biol 2004; 36:607-20. [PMID: 15010327 DOI: 10.1016/j.biocel.2003.10.008] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2003] [Revised: 08/14/2003] [Accepted: 10/14/2003] [Indexed: 12/14/2022]
Abstract
Considerable effort has been made in recent years in understanding the mechanisms that govern stem cell generation, proliferation, self-renewal, commitment and lately plasticity. In the development of the haemopoietic system during embryonic and fetal life the notion of different pools of stem cells arising from the endothelium is gaining consensus. Gene expression profiling of populations of stem cells is bringing to light categories of genes important for self-renewal or commitment. Besides the role of transcription factors in lineage decision, the role of soluble factors and transmembrane proteins, very active at the time of embryo development, are taking central stage in the maintenance and in vitro expansion of haemopoietic stem cells (HSCs). The hierarchical model of haemopoietic development is being questioned with reports of lineage switching and plasticity of haemopoietic stem cells to non-haemopoietic cells. Yet the understanding of the overall process is still very fragmented and hypothetical. This is mainly due to the absence of appropriate markers to enable selection of homogeneous stem cell populations and the need to rely on retrospective functional assays, able only to determine the overall behaviour of a population of cells. This review is intended to be an overview of the haemopoietic system and a critical re-visitation of issues such as plasticity and self-renewal important for therapeutic applications of haemopoietic stem cells.
Collapse
Affiliation(s)
- Ilaria Bellantuono
- Stem Cell Research Group, Giving for Living Research Centre, Royal Manchester Children Hospital, Hospital Road, Manchester M27 4HA, UK.
| |
Collapse
|
158
|
Höflinger S, Kesavan K, Fuxa M, Hutter C, Heavey B, Radtke F, Busslinger M. Analysis of Notch1 Function by In Vitro T Cell Differentiation of Pax5 Mutant Lymphoid Progenitors. THE JOURNAL OF IMMUNOLOGY 2004; 173:3935-44. [PMID: 15356142 DOI: 10.4049/jimmunol.173.6.3935] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Signaling through the Notch1 receptor is essential for T cell development in the thymus. Stromal OP9 cells ectopically expressing the Notch ligand Delta-like1 mimic the thymic environment by inducing hemopoietic stem cells to undergo in vitro T cell development. Notch1 is also expressed on Pax5-/- pro-B cells, which are clonable lymphoid progenitors with a latent myeloid potential. In this study, we demonstrate that Pax5-/- progenitors efficiently differentiate in vitro into CD4+CD8+ alphabeta and gammadelta T cells upon coculture with OP9-Delta-like1 cells. In vitro T cell development of Pax5-/- progenitors strictly depends on Notch1 function and progresses through normal developmental stages by expressing T cell markers and rearranging TCRbeta, gamma, and delta loci in the correct temporal sequence. Notch-stimulated Pax5-/- progenitors efficiently down-regulate the expression of B cell-specific genes, consistent with a role of Notch1 in preventing B lymphopoiesis in the thymus. At the same time, Notch signaling rapidly induces cell surface expression of the c-Kit receptor and transcription of the target genes Deltex1 and pre-Talpha concomitant with the activation of TCR Vbeta germline transcription and the regulatory genes GATA3 and Tcf1. These data suggest that Notch1 acts upstream of GATA3 and Tcf1 in early T cell development and regulates Vbeta-DJbeta rearrangements by controlling the chromatin accessibility of Vbeta genes at the TCRbeta locus.
Collapse
MESH Headings
- Animals
- B-Lymphocyte Subsets/cytology
- B-Lymphocyte Subsets/metabolism
- B-Lymphocyte Subsets/physiology
- Cell Differentiation/genetics
- Cell Differentiation/immunology
- Cell Line
- Cell Lineage/genetics
- Cell Lineage/immunology
- Clone Cells
- Coculture Techniques
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/physiology
- Down-Regulation/genetics
- Down-Regulation/immunology
- Gene Expression Regulation/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mutation
- PAX5 Transcription Factor
- Receptor, Notch1
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, gamma-delta/genetics
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/physiology
- Signal Transduction/genetics
- Signal Transduction/immunology
- Stem Cells/cytology
- Stem Cells/metabolism
- Stem Cells/physiology
- Stromal Cells/physiology
- T-Lymphocyte Subsets/cytology
- T-Lymphocyte Subsets/metabolism
- T-Lymphocyte Subsets/physiology
- Transcription Factors/genetics
- Transcription Factors/physiology
Collapse
Affiliation(s)
- Sonja Höflinger
- Research Institute of Molecular Pathology, Vienna Biocenter, Vienna, Austria
| | | | | | | | | | | | | |
Collapse
|
159
|
Seet CS, Brumbaugh RL, Kee BL. Early B cell factor promotes B lymphopoiesis with reduced interleukin 7 responsiveness in the absence of E2A. ACTA ACUST UNITED AC 2004; 199:1689-700. [PMID: 15210745 PMCID: PMC2212815 DOI: 10.1084/jem.20032202] [Citation(s) in RCA: 128] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The basic helix-loop-helix transcription factors encoded by the E2A gene function at the apex of a transcriptional hierarchy involving E2A, early B cell factor (EBF), and Pax5, which is essential for B lymphopoiesis. In committed B lineage progenitors, E2A proteins have also been shown to regulate many lineage-associated genes. Herein, we demonstrate that the block in B lymphopoiesis imposed by the absence of E2A can be overcome by expression of EBF, but not Pax5, indicating that EBF is the essential target of E2A required for development of B lineage progenitors. Our data demonstrate that EBF, in synergy with low levels of alternative E2A-related proteins (E proteins), is sufficient to promote expression of most B lineage genes. Remarkably, however, we find that E2A proteins are required for interleukin 7-dependent proliferation due, in part, to a role for E2A in optimal expression of N-myc. Therefore, high levels of E protein activity are essential for the activation of EBF and N-myc, whereas lower levels of E protein activity, in synergy with other B lineage transcription factors, are sufficient for expression of most B lineage genes.
Collapse
Affiliation(s)
- Christopher S Seet
- Department of Pathology, University of Chicago, 5841 S. Maryland Avenue, MC 1089, Chicago, IL 60637, USA
| | | | | |
Collapse
|
160
|
Abstract
The generation of B-lymphocytes from hematopoietic stem cells is controlled by multiple transcription factors regulating distinct developmental aspects. Ikaros and PU.1 act in parallel pathways to control the development of lymphoid progenitors in part by regulating the expression of essential signaling receptors (Flt3, c-Kit, and IL-7R alpha). The generation of the earliest B cell progenitors depends on E2A and EBF, which coordinately activate the B cell gene expression program and immunoglobulin heavy-chain gene rearrangements at the onset of B-lymphopoiesis. Pax5 restricts the developmental options of lymphoid progenitors to the B cell lineage by repressing the transcription of lineage-inappropriate genes and simultaneously activating the expression of B-lymphoid signaling molecules. LEF1 and Sox4 contribute to the survival and proliferation of pro-B cells in response to extracellular signals. Finally, IRF4 and IRF8 together control the termination of pre-B cell receptor signaling and thus promote differentiation to small pre-B cells undergoing light-chain gene rearrangements.
Collapse
Affiliation(s)
- Meinrad Busslinger
- Research Institute of Molecular Pathology, Vienna Biocenter, A-1030 Vienna, Austria.
| |
Collapse
|
161
|
Crozatier M, Ubeda JM, Vincent A, Meister M. Cellular immune response to parasitization in Drosophila requires the EBF orthologue collier. PLoS Biol 2004; 2:E196. [PMID: 15314643 PMCID: PMC509289 DOI: 10.1371/journal.pbio.0020196] [Citation(s) in RCA: 172] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2004] [Accepted: 04/22/2004] [Indexed: 11/21/2022] Open
Abstract
Drosophila immune response involves three types of hemocytes ('blood cells'). One cell type, the lamellocyte, is induced to differentiate only under particular conditions, such as parasitization by wasps. Here, we have investigated the mechanisms underlying the specification of lamellocytes. We first show that collier (col), the Drosophila orthologue of the vertebrate gene encoding early B-cell factor (EBF), is expressed very early during ontogeny of the lymph gland, the larval hematopoietic organ. In this organ, Col expression prefigures a specific posterior region recently proposed to act as a signalling centre, the posterior signalling centre (PSC). The complete lack of lamellocytes in parasitized col mutant larvae revealed the critical requirement for Col activity in specification of this cell type. In wild-type larvae, Col expression remains restricted to the PSC following parasitization, despite the massive production of lamellocytes. We therefore propose that Col endows PSC cells with the capacity to relay an instructive signal that orients hematopoietic precursors towards the lamellocyte fate in response to parasitization. Considered together with the role of EBF in lymphopoiesis, these findings suggest new parallels in cellular immunity between Drosophila and vertebrates. Further investigations on Col/EBF expression and function in other phyla should provide fresh insight into the evolutionary origin of lymphoid cells.
Collapse
Affiliation(s)
- Michèle Crozatier
- 1Centre de Biologie du Développement, Centre National de la Recherche Scientifique and Université Paul SabatierToulouse, France
| | - Jean-Michel Ubeda
- 2Institut de Biologie Moléculaire et CellulaireCentre National de la Recherche Scientifique, StrasbourgFrance
| | - Alain Vincent
- 1Centre de Biologie du Développement, Centre National de la Recherche Scientifique and Université Paul SabatierToulouse, France
| | - Marie Meister
- 2Institut de Biologie Moléculaire et CellulaireCentre National de la Recherche Scientifique, StrasbourgFrance
| |
Collapse
|
162
|
Johnson K, Pflugh DL, Yu D, Hesslein DGT, Lin KI, Bothwell ALM, Thomas-Tikhonenko A, Schatz DG, Calame K. B cell-specific loss of histone 3 lysine 9 methylation in the V(H) locus depends on Pax5. Nat Immunol 2004; 5:853-61. [PMID: 15258579 PMCID: PMC1635547 DOI: 10.1038/ni1099] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2004] [Accepted: 06/23/2004] [Indexed: 12/28/2022]
Abstract
Immunoglobulin heavy chain rearrangement (V(H)-to-DJ(H)) occurs only in B cells, suggesting it is inhibited in other lineages. Here we found that in the mouse V(H) locus, methylation of lysine 9 on histone H3 (H3-K9), a mark of inactive chromatin, was present in non-B lineage cells but was absent in B cells. As others have shown that H3-K9 methylation can inhibit V(D)J recombination on engineered substrates, our data support the idea that H3-K9 methylation inhibits endogenous V(H)-to-DJ(H) recombination. We also show that Pax5, a transcription factor required for B cell commitment, is necessary and sufficient for the removal of H3-K9 methylation in the V(H) locus and provide evidence that one function of Pax5 is to remove this inhibitory modification by a mechanism of histone exchange, thus allowing B cell-specific V(H)-to-DJ(H) recombination.
Collapse
Affiliation(s)
- Kristen Johnson
- Department of Microbiology, Columbia University College of Physicians and Surgeons, New York, New York 10032, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
163
|
Tokoyoda K, Egawa T, Sugiyama T, Choi BI, Nagasawa T. Cellular niches controlling B lymphocyte behavior within bone marrow during development. Immunity 2004; 20:707-18. [PMID: 15189736 DOI: 10.1016/j.immuni.2004.05.001] [Citation(s) in RCA: 582] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2003] [Revised: 03/02/2004] [Accepted: 03/31/2004] [Indexed: 01/01/2023]
Abstract
In bone marrow, hematopoiesis is thought to depend on special microenvironments known as niches that maintain blood cells. However, the identity of niches and interaction of blood cells with niches remain poorly understood. Here we identify stage-specific cellular niches for B lymphopoiesis. The earliest precursors, pre-pro-B cells and end-stage B cells, plasma cells require CXC chemokine ligand (CXCL)12. CXCL12-expressing cells are a small population of stromal cells, scattered throughout bone marrow and located some distance from the cells expressing interleukin (IL)-7. Multipotent hematopoietic progenitors are attached to the processes of CXCL12-expressing cells and pre-pro-B cells adjoin their cell bodies. Maturer pro-B cells that require IL-7 have moved away and adjoin the IL-7-expressing cells. Plasma cells again seed CXCL12-expressing cells. We demonstrate the B lymphocyte characteristic location and movement between specific niches within bone marrow during development and suggest that CXCL12 maintains the cells in the niche.
Collapse
Affiliation(s)
- Koji Tokoyoda
- Department of Medical Systems Control, Institute for Frontier Medical Sciences, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | | | | | | | | |
Collapse
|
164
|
Abstract
It is generally assumed that once a cell commits to a certain lineage it no longer can change its fate. A new study in this issue of Cell provides compelling evidence that committed mature B lineage cells can be reprogrammed to become macrophages.
Collapse
Affiliation(s)
- Cornelis Murre
- Divison of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
165
|
Abstract
Sepsis is a syndrome of significant morbidity and mortality. Unlike the advances made in other diseases processes, improvements in outcome from sepsis, severe sepsis, and septic shock have been modest. Current research has altered our understanding of sepsis pathogenesis such that present models and definitions are still evolving. One relatively novel cell type, the dendritic cell, is the subject of much current investigation in sepsis. Although our present understanding of dendritic cell biology is incomplete, growing evidence supports the importance of this antigen-presenting cell in the normal and maladaptive responses to microbial invasion and tissue injury. A better understanding of this cell's basic biology as well as its potential as a therapeutic target will undoubtedly play increasing roles in the development of new strategies for the treatment of the septic patient.
Collapse
Affiliation(s)
- Philip Efron
- Department of Surgery, University of Florida College of Medicine, Gainesville 32610, USA
| | | |
Collapse
|
166
|
Abstract
The foundations of experimental hematology were laid by histologists, and while their contributions were enormous, they were limited in their interpretation of very dynamic processes by the static nature of the methodology. The middle of the twentieth century saw the introduction of techniques for hematopoietic cell marking and development of in vitro and in vivo assays for primitive hematopoietic cells, allowing dynamic studies of hematopoiesis. Paralleling this was an understanding of cellular immunology with the discovery of the role of the thymus and the identification of T and B lymphocyte lineages. In the 1960s a series of ontogenetic studies in birds and subsequently in mice revealed that hematopoietic and lymphoid development involved migration streams of primitive cells that colonized developing primary lymphoid organs as well as spleen, marrow, and liver. The yolk sac was proposed as the ultimate origin of these lympho-hematopoietic precursors. Subsequent studies identified a region associated with the dorsal aorta as the primary site of "definitive" stem cells. These opposing views are currently achieving a compromise that recognizes that both sites contribute stem cells involved in seeding the developing tissues. The clear distinction between the local origin of the inducing microenvironment provided by the endoderm or by stroma derived from mesenchymal stem cells of mesodermal origin, and the immigrant origin of the hematopoietic stem cells and progenitors, raises intriguing questions in the current climate of stem cell plasticity, cell fusion, and discovery of stem cells in adult marrow with the capacity to generate hematopoiesis as well as other mesodermal, ectodermal, and endodermal lineages.
Collapse
Affiliation(s)
- Malcolm A S Moore
- Cell Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA.
| |
Collapse
|
167
|
Johnson K, Calame K. Transcription factors controlling the beginning and end of B-cell differentiation. Curr Opin Genet Dev 2004; 13:522-8. [PMID: 14550419 DOI: 10.1016/j.gde.2003.08.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Powerful techniques--including gene targeting, genomic and proteomic analyses--are allowing a more complete understanding of the transcription factors that drive B-cell development. Regulatory cascades involving multiple steps have been identified at many decision points. In addition, feedback loops and transcriptional inhibition of alternative fates have been found to be important both for commitment to the B lymphocyte lineage and for terminal B-cell differentiation.
Collapse
Affiliation(s)
- Kristen Johnson
- Department of Microbiology, Columbia University College of Physicians and Surgeons, New York, New York 10032, USA
| | | |
Collapse
|
168
|
Fuxa M, Skok J, Souabni A, Salvagiotto G, Roldan E, Busslinger M. Pax5 induces V-to-DJ rearrangements and locus contraction of the immunoglobulin heavy-chain gene. Genes Dev 2004; 18:411-22. [PMID: 15004008 PMCID: PMC359395 DOI: 10.1101/gad.291504] [Citation(s) in RCA: 318] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The subnuclear location and chromatin state of the immunoglobulin heavy-chain (IgH) locus have been implicated in the control of VDJ recombination. VH-to-DJH rearrangement of distal, but not proximal V(H) genes, furthermore, depends on the B-lineage commitment factor Pax5 (BSAP). He e we demonstrate that ectopic Pax5 expression from the Ikaros promote induces proximal rather than distal VH-DJH rearrangements in Ik(Pax5/+) thymocytes, thus recapitulating the loss-of-function phenotype of Pax5-/- pro-B cells. The phenotypic similarities of both cell types include (1) chromatin accessibility of distal VH genes in the absence of VH-DJH rearrangements, (2) expression of the B-cell-specific regulator EBF, (3) central location of IgH alleles within the nucleus, and (4) physical separation of distal VH genes from proximal segments in an extended IgH locus. Reconstitution of Pax5 expression in Pax5-/- pro-B cells induced large-scale contraction and distal VH-DJH rearrangements of the IgH locus. Hence, VH-DJH recombination is regulated in two steps during early B-lymphopoiesis. The IgH locus is first repositioned from its default location at the nuclear periphery toward the center of the nucleus, which facilitates proximal VH-DJH recombination. Pax5 subsequently activates locus contraction and distal VH-DJH rearrangements in collaboration with an unknown factor that is present in pro-B cells, but absent in thymocytes.
Collapse
Affiliation(s)
- Martin Fuxa
- Research Institute of Molecular Pathology, Vienna Biocente, A-1030 Vienna, Austria
| | | | | | | | | | | |
Collapse
|
169
|
Sato H, Saito-Ohara F, Inazawa J, Kudo A. Pax-5 Is Essential for κ Sterile Transcription during Igκ Chain Gene Rearrangement. THE JOURNAL OF IMMUNOLOGY 2004; 172:4858-65. [PMID: 15067064 DOI: 10.4049/jimmunol.172.8.4858] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Pax-5 is the key regulator in B cell development. Pax-5-deficient mice show defects in B cell commitment and recombination of IgH chain gene rearrangement from DJ to VDJ. Previously, we found that Pax-5 bound to KI and KII sites, which play a crucial role in kappa-chain gene rearrangement. However, the function of Pax-5 in Ig kappa chain gene rearrangement has not been investigated. To address this issue, we newly established pre-BI cell lines expressing the pre-B cell receptor from Pax-5-deficient mice and used them in an in vitro culture system, in which kappa-chain gene rearrangement is induced by removing IL-7. By examining the Pax-5-deficient pre-BI (knockout (KO)) cells, we show in this study that, despite recombination-activating gene 1 and 2 expression, these KO cells did not rearrange the kappa-chain gene following the absence of kappa sterile transcription. Consistent with these data, fluorescent in situ hybridization analyses revealed that the J(kappa) locus in KO cells was located at the nuclear periphery as a repressive compartment. Transfection of KO cells with Pax-5 constructs indicated that the transactivation domain of Pax-5 was required for kappa sterile transcription and kappa-chain gene rearrangement. Moreover, the hormone-inducible system in KO cells demonstrated that Pax-5 directly functioned in kappa sterile transcription. These results indicate that Pax-5 is necessary for kappa sterile transcription during Ig kappa chain gene rearrangement.
Collapse
Affiliation(s)
- Hiromu Sato
- Department of Life Science, Tokyo Institute of Technology, Yokohama, Japan
| | | | | | | |
Collapse
|
170
|
Ikawa T, Kawamoto H, Wright LYT, Murre C. Long-Term Cultured E2A-Deficient Hematopoietic Progenitor Cells Are Pluripotent. Immunity 2004; 20:349-60. [PMID: 15030778 DOI: 10.1016/s1074-7613(04)00049-4] [Citation(s) in RCA: 112] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2003] [Revised: 01/19/2004] [Accepted: 02/11/2004] [Indexed: 11/23/2022]
Abstract
E2A proteins are essential for the development of B cells beyond the progenitor cell stage. Here we have isolated E2A-deficient bone marrow-derived cells that have the ability to grow long-term in vitro and coexpress, at low levels, regulators of different hematopoietic cell lineages. When transferred into lethally irradiated hosts, E2A-deficient hematopoietic progenitor cells reconstitute the T, NK, myeloid, dendritic, and erythroid lineages but fail to develop into mature B lineage cells. Enforced expression of E47 in E2A-deficient hematopoietic progenitor cells directly activates the transcription of a subset of B lineage-specific genes, including lambda5, mb-1, and Pax5. In contrast, E47 inhibits the expression of regulators of other hematopoietic lineages, including TCF-1 and GATA-1. These observations indicate that E2A-deficient hematopoietic progenitor cells remain pluripotent after long-term culture in vitro and that E2A proteins play a critical role in B cell commitment.
Collapse
Affiliation(s)
- Tomokatsu Ikawa
- Division of Biological Sciences, 0377 University of California, San Diego, La Jolla, CA 92093, USA
| | | | | | | |
Collapse
|
171
|
Smith E, Sigvardsson M. The roles of transcription factors in B lymphocyte commitment, development, and transformation. J Leukoc Biol 2004; 75:973-81. [PMID: 14982952 DOI: 10.1189/jlb.1103554] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Studies of normal blood cell development and malignant transformation of hematopoietic cells have shown that the correctly regulated expression of stage- and lineage-specific genes is a key issue in hematopoiesis. Experiments in transgenic mice have defined a number of transcription factors such as SCL/Tal, core-binding factor/acute myeloid leukemia, and c-myb, all crucial for the establishment of definitive hematopoiesis and development of all blood cell lineages. Other regulators such as IKAROS, E47/E2A, early B cell factor, Sox-4, and B cell-specific activator protein (Pax-5) appear crucial, more or less selectively, for B lymphopoiesis, allowing for detailed analysis of the development of this lineage. In addition, several of these transcription factors are found translocated in human tumors, often resulting in aberrant gene expression or production of modified proteins. This article concerns the role of transcription factors in B lymphoid development with special focus on lineage initiation and commitment events but also to some extent on the roles of transcription factors in human B lymphoid malignancies.
Collapse
Affiliation(s)
- Emma Smith
- Stemcell Center, Lund University, BMC B12, 22184 Lund, Sweden
| | | |
Collapse
|
172
|
Lalonde JP, Lim R, Ingley E, Tilbrook PA, Thompson MJ, McCulloch R, Beaumont JG, Wicking C, Eyre HJ, Sutherland GR, Howe K, Solomon E, Williams JH, Klinken SP. HLS5, a Novel RBCC (Ring Finger, B Box, Coiled-coil) Family Member Isolated from a Hemopoietic Lineage Switch, Is a Candidate Tumor Suppressor. J Biol Chem 2004; 279:8181-9. [PMID: 14662771 DOI: 10.1074/jbc.m306751200] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Hemopoietic cells, apparently committed to one lineage, can be reprogrammed to display the phenotype of another lineage. The J2E erythroleukemic cell line has on rare occasions developed the features of monocytic cells. Subtractive hybridization was used in an attempt to identify genes that were up-regulated during this erythroid to myeloid transition. We report here on the isolation of hemopoietic lineage switch 5 (Hls5), a gene expressed by the monocytoid variant cells, but not the parental J2E cells. Hls5 is a novel member of the RBCC (Ring finger, B box, coiled-coil) family of genes, which includes Pml, Herf1, Tif-1alpha, and Rfp. Hls5 was expressed in a wide range of adult tissues; however, at different stages during embryogenesis, Hls5 was detected in the branchial arches, spinal cord, dorsal root ganglia, limb buds, and brain. The protein was present in cytoplasmic granules and punctate nuclear bodies. Isolation of the human cDNA and genomic DNA revealed that the gene was located on chromosome 8p21, a region implicated in numerous leukemias and solid tumors. Enforced expression of Hls5 in HeLa cells inhibited cell growth, clonogenicity, and tumorigenicity. It is conceivable that HLS5 is one of the tumor suppressor genes thought to reside at the 8p21 locus.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Apoptosis
- Apoptosis Regulatory Proteins
- Base Sequence
- Brain/embryology
- Brain Chemistry
- Branchial Region/chemistry
- Branchial Region/embryology
- Carrier Proteins/chemistry
- Carrier Proteins/genetics
- Carrier Proteins/physiology
- Cell Cycle
- Cell Differentiation
- Cell Line, Tumor
- Cell Nucleus/chemistry
- Chromosomes, Human, Pair 8
- Cytoplasmic Granules/chemistry
- DNA/analysis
- DNA, Complementary/chemistry
- DNA, Complementary/isolation & purification
- Embryonic and Fetal Development
- Extremities/embryology
- Ganglia, Spinal/chemistry
- Ganglia, Spinal/embryology
- Genes, Tumor Suppressor
- HeLa Cells
- Hematopoietic Stem Cells/cytology
- Humans
- Leukemia, Erythroblastic, Acute
- Mice
- Microscopy, Fluorescence
- Molecular Sequence Data
- Open Reading Frames
- Spinal Cord/chemistry
- Spinal Cord/embryology
- Transfection
Collapse
Affiliation(s)
- Jean-Philippe Lalonde
- Laboratory for Cancer Medicine, Western Australian Institute for Medical Research, Royal Perth Hospital and the Center for Medical Research, The University of Western Australia, Perth, Western Australia 6000, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
173
|
Traver D, Akashi K. Lineage commitment and developmental plasticity in early lymphoid progenitor subsets. Adv Immunol 2004; 83:1-54. [PMID: 15135627 DOI: 10.1016/s0065-2776(04)83001-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- David Traver
- Dana-Farber Cancer Institute, Boston Massachusetts 02115, USA
| | | |
Collapse
|
174
|
Perry SS, Wang H, Pierce LJ, Yang AM, Tsai S, Spangrude GJ. L-selectin defines a bone marrow analog to the thymic early T-lineage progenitor. Blood 2003; 103:2990-6. [PMID: 15070675 DOI: 10.1182/blood-2003-09-3030] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The recent description of an early T-lineage progenitor (ETP) population in adult mouse thymus implies the presence of a bone marrow predecessor that has not yet been identified. Here we describe a Lin(Neg) Sca-1(Pos) c-kit(Hi) Thy-1.1(Neg) L-selectin(Pos) adult mouse bone marrow population that resembles the thymic ETP in both antigen expression phenotype and posttransplantation lineage potential. These cells produce wavelike kinetics of thymic seeding and reconstitute the irradiated thymus with kinetics comparable to a thymocyte graft after intravenous transplantation. Transient B-lineage reconstitution is also observed, but little myeloid potential can be detected in transplant experiments. A second subset of progenitors is L-selectin(Neg) and is highly enriched for rapid and persistent T- and B-lineage potential, as well as some myeloid potential. L-selectin (CD62L) is therefore an effective marker for separating lymphoid progenitors from myeloid progenitors and hematopoietic stem cells in mouse bone marrow.
Collapse
Affiliation(s)
- S Scott Perry
- Department of Medicine, University of Utah School of Medicine, Salt Lake City 84132, USA
| | | | | | | | | | | |
Collapse
|
175
|
Araki H, Katayama N, Yamashita Y, Mano H, Fujieda A, Usui E, Mitani H, Ohishi K, Nishii K, Masuya M, Minami N, Nobori T, Shiku H. Reprogramming of human postmitotic neutrophils into macrophages by growth factors. Blood 2003; 103:2973-80. [PMID: 15070673 DOI: 10.1182/blood-2003-08-2742] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
It is generally recognized that postmitotic neutrophils give rise to polymorphonuclear neutrophils alone. We obtained evidence for a lineage switch of human postmitotic neutrophils into macrophages in culture. When the CD15+CD14- cell population, which predominantly consists of band neutrophils, was cultured with granulocyte macrophage-colony-stimulating factor, tumor necrosis factor-alpha, interferon-gamma, and interleukin-4, and subsequently with macrophage colony-stimulating factor alone, the resultant cells had morphologic, cytochemical, and phenotypic features of macrophages. In contrast to the starting population, they were negative for myeloperoxidase, specific esterase, and lactoferrin, and they up-regulated nonspecific esterase activity and the expression of macrophage colony-stimulating factor receptor, mannose receptor, and HLA-DR. CD15+CD14- cells proceeded to macrophages through the CD15-CD14- cell population. Microarray analysis of gene expression also disclosed the lineage conversion from neutrophils to macrophages. Macrophages derived from CD15+CD14- neutrophils had phagocytic function. Data obtained using 3 different techniques, including Ki-67 staining, bromodeoxyuridine incorporation, and cytoplasmic dye labeling, together with the yield of cells, indicated that the generation of macrophages from CD15+CD14- neutrophils did not result from a contamination of progenitors for macrophages. Our data show that in response to cytokines, postmitotic neutrophils can become macrophages. This may represent another differentiation pathway toward macrophages in human postnatal hematopoiesis.
Collapse
Affiliation(s)
- Hiroto Araki
- Second Department of Internal Medicine, Mie University School of Medicine, Tsu, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
176
|
Kozmik Z, Daube M, Frei E, Norman B, Kos L, Dishaw LJ, Noll M, Piatigorsky J. Role of Pax genes in eye evolution: a cnidarian PaxB gene uniting Pax2 and Pax6 functions. Dev Cell 2003; 5:773-85. [PMID: 14602077 DOI: 10.1016/s1534-5807(03)00325-3] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
PaxB from Tripedalia cystophora, a cubomedusan jellyfish possessing complex eyes (ocelli), was characterized. PaxB, the only Pax gene found in this cnidarian, is expressed in the larva, retina, lens, and statocyst. PaxB contains a Pax2/5/8-type paired domain and octapeptide, but a Pax6 prd-type homeodomain. Pax2/5/8-like properties of PaxB include a DNA binding specificity of the paired domain, activation and inhibitory domains, and the ability to rescue spa(pol), a Drosophila Pax2 eye mutant. Like Pax6, PaxB activates jellyfish crystallin and Drosophila rhodopsin rh6 promoters and induces small ectopic eyes in Drosophila. Pax6 has been considered a "master" control gene for eye development. Our data suggest that the ancestor of jellyfish PaxB, a PaxB-like protein, was the primordial Pax protein in eye evolution and that Pax6-like genes evolved in triploblasts after separation from Cnidaria, raising the possibility that cnidarian and sophisticated triploblastic eyes arose independently.
Collapse
Affiliation(s)
- Zbynek Kozmik
- Laboratory of Molecular and Developmental Biology, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | |
Collapse
|
177
|
Dazert S, Aletsee C, Brors D, Sudhoff H, Ryan AF, Müller AM. Regeneration of Inner Ear Cells from Stem Cell Precursors—A Future Concept of Hearing Rehabilitation? DNA Cell Biol 2003; 22:565-70. [PMID: 14577909 DOI: 10.1089/104454903322405455] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The use of stem cells offers new and powerful strategies for future tissue development and engineering. Common features of stem cells are both their capacity for self-renewal and the ability to differentiate into mature effector cells. Since the establishment of embryonic stem cells from early human embryos, research on and clinical application of human ES cells belong to the most controversial topics in our society. Great hopes are based upon the remarkable observation that human ES cells can be greatly expanded in vitro, and that they can differentiate into various clinically important cell types. Recent advances in the cloning of mammals by nuclear transplantation provide new concepts for autologous replacement of damaged and degenerated tissues. In contrast, somatic stem cells of the adult organism were considered to be more restricted in their developmental potential. However, recent investigations suggest that somatic stem cells may have a wider differentiation potential than previously thought. In otology, initial experiments have revealed neural stem cell survival in cochlear cell cultures and under neurotrophin influence, neural stem cells seemed to develop into a neuronal phenotype. Further studies have to be carried out to investigate the full potential of stem cells as well as the molecular mechanisms that are involved in regulating cellular identity and plasticity. Clinically, advances in stem cell biology may provide a permanent source of replacement cells for treating human diseases and could open the development of new concepts for cell and tissue regeneration for a causal treatment of chronic degenerative diseases.
Collapse
Affiliation(s)
- S Dazert
- Hals-Nasen-Ohrenklinik der Ruhr-Universität Bochum, St. Elisabeth Hospital, Bochum, Germany.
| | | | | | | | | | | |
Collapse
|
178
|
Iwasaki H, Mizuno SI, Wells RA, Cantor AB, Watanabe S, Akashi K. GATA-1 Converts Lymphoid and Myelomonocytic Progenitors into the Megakaryocyte/Erythrocyte Lineages. Immunity 2003; 19:451-62. [PMID: 14499119 DOI: 10.1016/s1074-7613(03)00242-5] [Citation(s) in RCA: 146] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
GATA-1 is an essential transcription factor for megakaryocyte and erythrocyte (MegE) development. Here we show that hematopoietic progenitors can be reprogrammed by the instructive action of GATA-1. Enforced expression of GATA-1 in hematopoietic stem cells led to loss of self-renewal activity and the exclusive generation of MegE lineages. Strikingly, ectopic GATA-1 reprogrammed common lymphoid progenitors as well as granulocyte/monocyte (GM) progenitors to differentiate into MegE lineages, while inhibiting normal lymphoid or GM differentiation. GATA-1 upregulated critical MegE-related transcription factors such as FOG-1 and GATA-2 in lymphoid and GM progenitors, and their MegE development did not require "permissive" erythropoietin signals. Furthermore, GATA-1 induced apoptosis of proB and myelomonocytic cells, which could not be prevented by enforced permissive Bcl-2 or myeloid cytokine signals. Thus, GATA-1 specifically instructs MegE commitment while excluding other fate outcomes in stem and progenitor cells, suggesting that regulation of GATA-1 is critical in maintaining multilineage homeostasis.
Collapse
Affiliation(s)
- Hiromi Iwasaki
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | |
Collapse
|
179
|
Abstract
Methods for directly turning a somatic cell type into another type (a process referred to as transdifferentiation) would be beneficial for producing replacement cells for therapeutic applications. Adult stem cells have been shown to display a broader differentiation potential than anticipated and may contribute to tissues other than those in which they reside. In addition, novel transdifferentiation strategies are being developed. I report recent results on the functional reprogramming of a somatic cell using a nuclear and cytoplasmic extract derived from another somatic cell type. The reprogramming of 293T fibroblasts in an extract from T cells is evidenced by nuclear uptake and the assembly of transcription factors, induction of activity of a chromatin remodelling complex, changes in chromatin composition and activation of lymphoid cell-specific genes. The reprogrammed cells express T-cell-specific surface molecules and a complex regulatory function. Reprogramming cells in cell-free extracts may create possibilities for producing replacement cells for therapeutic applications. The system may also constitute a powerful tool to examine the mechanisms of nuclear reprogramming, at least as they occur in vitro.
Collapse
Affiliation(s)
- Philippe Collas
- Institute of Medical Biochemistry, University of Oslo, PO Box 1112 Blindern, 0317 Oslo, Norway.
| |
Collapse
|
180
|
Heavey B, Charalambous C, Cobaleda C, Busslinger M. Myeloid lineage switch of Pax5 mutant but not wild-type B cell progenitors by C/EBPalpha and GATA factors. EMBO J 2003; 22:3887-97. [PMID: 12881423 PMCID: PMC169053 DOI: 10.1093/emboj/cdg380] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The developmental potential of hematopoietic progenitors is restricted early on to either the erythromyeloid or lymphoid lineages. The broad developmental potential of Pax5(-/-) pro-B cells is in apparent conflict with such a strict separation, although these progenitors realize the myeloid and erythroid potential with lower efficiency compared to the lymphoid cell fates. Here we demonstrate that ectopic expression of the transcription factors C/EBPalpha, GATA1, GATA2 and GATA3 strongly promoted in vitro macrophage differentiation and myeloid colony formation of Pax5(-/-) pro-B cells. GATA2 and GATA3 expression also resulted in efficient engraftment and myeloid development of Pax5(-/-) pro-B cells in vivo. The myeloid transdifferentiation of Pax5(-/-) pro-B cells was accompanied by the rapid activation of myeloid genes and concomitant repression of B-lymphoid genes by C/EBPalpha and GATA factors. These data identify the Pax5(-/-) pro-B cells as lymphoid progenitors with a latent myeloid potential that can be efficiently activated by myeloid transcription factors. The same regulators were unable to induce a myeloid lineage switch in Pax5(+/+) pro-B cells, indicating that Pax5 dominates over myeloid transcription factors in B-lymphocytes.
Collapse
Affiliation(s)
- Barry Heavey
- Research Institute of Molecular Pathology, Vienna Biocenter,Dr. Bohr-Gasse 7, A-1030 Vienna, Austria
| | | | | | | |
Collapse
|
181
|
Reynaud D, Lefort N, Manie E, Coulombel L, Levy Y. In vitro identification of human pro-B cells that give rise to macrophages, natural killer cells, and T cells. Blood 2003; 101:4313-21. [PMID: 12560235 DOI: 10.1182/blood-2002-07-2085] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
In this study we report the molecular and functional characterization of very early interleukin 7 receptor alpha (IL-7Ralpha)+-CD79a+CD19- B-cell progenitors, produced by human CD34+CD19-CD10- cord blood cells grown in the presence of stromal cells and cytokines. Purified IL-7Ralpha+CD79a+CD19- cells transcribed the B-lymphoid specific genes E2A, EBF, TdT, Rag-1, had initiated DJH rearrangements, but almost lacked Pax-5 mRNA. When exposed to appropriate environmental conditions, these cells repressed B-cell genes and completely differentiated into CD14+ macrophages, CD56+ natural killer cells, and CD4high T cells. Retention of the DJH rearranged genes in both CD14+ and CD56+ cells unambiguously demonstrates that early B-cell genes, expressed prior to Pax-5, can be activated in a multipotent human progenitor cell whose final fate, including in non-B lineages, is determined by external signals.
Collapse
Affiliation(s)
- Damien Reynaud
- Institut national de la santé et de la recherche médicale (INSERM), Paris, France
| | | | | | | | | |
Collapse
|
182
|
Cotta CV, Zhang Z, Kim HG, Klug CA. Pax5 determines B- versus T-cell fate and does not block early myeloid-lineage development. Blood 2003; 101:4342-6. [PMID: 12560221 DOI: 10.1182/blood-2002-10-3139] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Progenitor B cells deficient in Pax5 are developmentally multipotent, suggesting that Pax5 is necessary to maintain commitment to the B-cell lineage. Commitment may be mediated, in part, by Pax5 repression of myeloid-specific genes. To determine whether Pax5 expression in multipotential cells is sufficient to restrict development to the B-cell lineage in vivo, we enforced expression of Pax5 in hematopoietic stem cells using a retroviral vector. Peripheral blood analysis of all animals reconstituted with Pax5-expressing cells indicated that more than 90% of Pax5-expressing cells were B220+ mature B cells that were not malignant. Further analysis showed that Pax5 completely blocked T-lineage development in the thymus but did not inhibit myelopoiesis or natural killer (NK) cell development in bone marrow. These results implicate Pax5 as a critical regulator of B- versus T-cell developmental fate and suggest that Pax5 may promote commitment to the B-cell lineage by mechanisms that are independent of myeloid gene repression.
Collapse
Affiliation(s)
- Claudiu V Cotta
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294-3300, USA
| | | | | | | |
Collapse
|
183
|
Liu P, Keller JR, Ortiz M, Tessarollo L, Rachel RA, Nakamura T, Jenkins NA, Copeland NG. Bcl11a is essential for normal lymphoid development. Nat Immunol 2003; 4:525-32. [PMID: 12717432 DOI: 10.1038/ni925] [Citation(s) in RCA: 258] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2002] [Accepted: 03/11/2003] [Indexed: 12/11/2022]
Abstract
Bcl11a (also called Evi9) functions as a myeloid or B cell proto-oncogene in mice and humans, respectively. Here we show that Bcl11a is essential for postnatal development and normal lymphopoiesis. Bcl11a mutant embryos lack B cells and have alterations in several types of T cells. Phenotypic and expression studies show that Bcl11a functions upstream of the transcription factors Ebf1 and Pax5 in the B cell pathway. Transplantation studies show that these defects in Bcl11a mutant mice are intrinsic to fetal liver precursor cells. Mice transplanted with Bcl11a-deficient cells died from T cell leukemia derived from the host. Thus, Bcl11a may also function as a non-autonomous T cell tumor suppressor gene.
Collapse
Affiliation(s)
- Pentao Liu
- Mouse Cancer Genetics Program, National Cancer Institute-Frederick, Maryland 21702, USA
| | | | | | | | | | | | | | | |
Collapse
|
184
|
Iwasaki-Arai J, Iwasaki H, Miyamoto T, Watanabe S, Akashi K. Enforced granulocyte/macrophage colony-stimulating factor signals do not support lymphopoiesis, but instruct lymphoid to myelomonocytic lineage conversion. J Exp Med 2003; 197:1311-22. [PMID: 12756267 PMCID: PMC2193786 DOI: 10.1084/jem.20021843] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
We evaluated the effects of ectopic granulocyte/macrophage colony-stimulating factor (GM-CSF) signals on hematopoietic commitment and differentiation. Lineage-restricted progenitors purified from mice with the ubiquitous transgenic human GM-CSF receptor (hGM-CSFR) were used for the analysis. In cultures with hGM-CSF alone, hGM-CSFR-expressing (hGM-CSFR+) granulocyte/monocyte progenitors (GMPs) and megakaryocyte/erythrocyte progenitors (MEPs) exclusively gave rise to granulocyte/monocyte (GM) and megakaryocyte/erythroid (MegE) colonies, respectively, providing formal proof that GM-CSF signals support the GM and MegE lineage differentiation without affecting the physiological myeloid fate. hGM-CSFR transgenic mice were crossed with mice deficient in interleukin (IL)-7, an essential cytokine for T and B cell development. Administration of hGM-CSF in these mice could not restore T or B lymphopoiesis, indicating that enforced GM-CSF signals cannot substitute for IL-7 to promote lymphopoiesis. Strikingly, >50% hGM-CSFR+ common lymphoid progenitors (CLPs) and >20% hGM-CSFR+ pro-T cells gave rise to granulocyte, monocyte, and/or myeloid dendritic cells, but not MegE lineage cells in the presence of hGM-CSF. Injection of hGM-CSF into mice transplanted with hGM-CSFR+ CLPs blocked their lymphoid differentiation, but induced development of GM cells in vivo. Thus, hGM-CSF transduces permissive signals for myeloerythroid differentiation, whereas it transmits potent instructive signals for the GM differentiation to CLPs and early T cell progenitors. These data suggest that a majority of CLPs and a fraction of pro-T cells possess plasticity for myelomonocytic differentiation that can be activated by ectopic GM-CSF signals, supporting the hypothesis that the down-regulation of GM-CSFR is a critical event in producing cells with a lymphoid-restricted lineage potential.
Collapse
Affiliation(s)
- Junko Iwasaki-Arai
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, 44 Binney Street, Boston, MA 02115, USA
| | | | | | | | | |
Collapse
|
185
|
Hozumi K, Abe N, Chiba S, Hirai H, Habu S. Active form of Notch members can enforce T lymphopoiesis on lymphoid progenitors in the monolayer culture specific for B cell development. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 170:4973-9. [PMID: 12734340 DOI: 10.4049/jimmunol.170.10.4973] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The in vitro induction of T lymphopoiesis needs the precise stereoscopic structure of thymus tissues as seen in fetal thymus organ culture. In this study, we demonstrated for the first time that the introduction of the intracellular region of Notch1 can induce T cells expressing TCR without any thymic environment. In the coculture on the monolayer of OP-9, which was originally known to support B cell specific development, hemopoietic progenitors developed into Thy-1(+)CD25(+) T lineage cells if the progenitor cells were infected with the retrovirus containing Notch1 intracellular domains. The Thy-1(+) cells progressed to a further developmental stage, CD4 and CD8 double-positive cells expressing TCR on the cell surface, if they were further cultured on OP-9 or in the thymus. However, T cell induction by intracellular Notch1 failed unless both OP-9 and IL-7 were present. It is notable that Notch2 and Notch3 showed an effect on T lymphopoiesis similar to that of Notch1. These results indicate that in vitro T lymphopoiesis is inducible by signaling via Notch family members in a lineage-specific manner but shares other stroma-derived factors including IL-7 with B lymphopoiesis.
Collapse
Affiliation(s)
- Katsuto Hozumi
- Department of Immunology, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | | | | | | | | |
Collapse
|
186
|
Abstract
Considerable progress has been made over the past few years in determining the key molecular players that regulate the commitment and development of early B-lineage cells in mouse bone marrow (transcription factors, cytokines and their receptors). However, confusion remains as to the precise stages, surface phenotypes and lineage restrictions that are observed early in B-cell development. Increasingly powerful flow cytometry analysis of genetically altered animals (knockouts and transgenics) combined with sensitive functional assays may provide an answer, a "scorecard" for the cell stages.
Collapse
Affiliation(s)
- Richard R Hardy
- Institute for Cancer Research, Fox Chase Cancer Center, 7701 Burholme Avenue, Philadelphia, PA 19111, USA.
| |
Collapse
|
187
|
Yu D, Allman D, Goldschmidt MH, Atchison ML, Monroe JG, Thomas-Tikhonenko A. Oscillation between B-lymphoid and myeloid lineages in Myc-induced hematopoietic tumors following spontaneous silencing/reactivation of the EBF/Pax5 pathway. Blood 2003; 101:1950-5. [PMID: 12406913 PMCID: PMC4547547 DOI: 10.1182/blood-2002-06-1797] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
B lymphomagenesis is an uncontrolled expansion of immature precursors that fail to complete their differentiation program. This failure could be at least partly explained by inappropriate expression of several oncogenic transcription factors, such as Pax5 and Myc. Both Pax5 and c-Myc are implicated in the pathogenesis of non-Hodgkin lymphomas. To address their role in lymphomagenesis, we analyzed B-cell lymphomas derived from p53-null bone marrow progenitors infected in vivo by a Myc-encoding retrovirus. All Myc-induced lymphomas invariably maintained expression of Pax5, which is thought to be incompatible with terminal differentiation. However, upon culturing in vitro, several cell lines spontaneously down-regulated Pax5 and its target genes CD19, N-Myc, and MB1. Unexpectedly, other B-cell markers (eg, CD45R) were also down-regulated, and markers of myeloid lineage (CD11b and F4/80 antigen) were acquired instead. Moreover, cells assumed the morphology reminiscent of myeloid cells. A pool of F4/80-positive cells as well as several single-cell clones were obtained and reinjected into syngeneic mice. Remarkably, pooled cells rapidly re-expressed Pax5 and formed tumors of relatively mature lymphoid phenotype, with surface immunoglobulins being abundantly expressed. Approximately half of tumorigenic single-cell clones also abandoned myeloid differentiation and gave rise to B lymphomas. However, when secondary lymphoma cells were returned to in vitro conditions, they once again switched to myeloid differentiation. This process could be curbed via enforced expression of retrovirally encoded Pax5. Our data demonstrate that some Myc target cells are bipotent B-lymphoid/myeloid progenitors with the astonishing capacity to undergo successive rounds of lineage switching.
Collapse
MESH Headings
- Animals
- Antigens, CD19/biosynthesis
- Antigens, CD19/genetics
- Antigens, Differentiation/biosynthesis
- Antigens, Differentiation/genetics
- Antigens, Differentiation, B-Lymphocyte/biosynthesis
- Antigens, Differentiation, B-Lymphocyte/genetics
- CD11b Antigen/biosynthesis
- CD11b Antigen/genetics
- Cell Adhesion
- Cell Differentiation
- Cell Lineage/genetics
- Cell Size
- Cell Transformation, Neoplastic/genetics
- Clone Cells/transplantation
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/physiology
- Gene Expression Regulation, Neoplastic
- Gene Silencing
- Genes, myc
- Leukocyte Common Antigens/biosynthesis
- Leukocyte Common Antigens/genetics
- Lymphocytes/pathology
- Lymphoma, B-Cell/genetics
- Lymphoma, B-Cell/pathology
- Mice
- Mice, Inbred C57BL
- Myeloid Cells/pathology
- Neoplasm Proteins/biosynthesis
- Neoplasm Proteins/genetics
- Neoplasm Transplantation
- PAX5 Transcription Factor
- Receptors, Antigen, B-Cell/biosynthesis
- Receptors, Antigen, B-Cell/genetics
- Recombinant Fusion Proteins/physiology
- Trans-Activators/physiology
- Transcription Factors/genetics
- Transcription Factors/physiology
- Tumor Cells, Cultured/metabolism
- Tumor Cells, Cultured/pathology
Collapse
Affiliation(s)
- Duonan Yu
- Departments of Pathobiology, Pathology and Laboratory Medicine, and Animal Biology, University of Pennsylvania, Philadelphia, PA 19104-6051, USA
| | | | | | | | | | | |
Collapse
|
188
|
Perry SS, Pierce LJ, Slayton WB, Spangrude GJ. Characterization of thymic progenitors in adult mouse bone marrow. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 170:1877-86. [PMID: 12574354 DOI: 10.4049/jimmunol.170.4.1877] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Thymic cellularity is maintained throughout life by progenitor cells originating in the bone marrow. In this study, we describe adult mouse bone cells that exhibit several features characteristic of prothymocytes. These include 1) rapid thymic engraftment kinetics following i.v. transplantation, 2) dramatic expansion of thymic progeny, and 3) limited production of hemopoietic progeny other than thymocytes. The adult mouse bone marrow population that is depleted of cells expressing any of a panel of lineage-specific Ags, stem cell Ag-1 positive, and not expressing the Thy1.1 Ag (Thy1.1(-)) (Thy1.1(-) progenitors) can repopulate the thymus 9 days more rapidly than can hemopoietic stem cells, a rate of thymic repopulation approaching that observed with transplanted thymocytes. Additionally, Thy1.1(-) progenitors expand prolifically to generate thymocyte progeny comparable in absolute numbers to those observed from parallel hemopoietic stem cell transplants, and provide a source of progenitors that spans multiple waves of thymic seeding. Nevertheless, the Thy1.1(-) population yields relatively few B cells and rare myeloid progeny posttransplant. These observations describe the phenotype of an adult mouse bone marrow population highly enriched for rapidly engrafting, long-term thymocyte progenitors. Furthermore, they note disparity in B and T cell expansion from this lymphoid progenitor population and suggest that it contains the progenitor primarily responsible for seeding the thymus throughout life.
Collapse
Affiliation(s)
- S Scott Perry
- Department of Pathology, University of Utah, 30 North 1900 East, Salt Lake City, UT 84132, USA
| | | | | | | |
Collapse
|
189
|
Hesslein DGT, Pflugh DL, Chowdhury D, Bothwell ALM, Sen R, Schatz DG. Pax5 is required for recombination of transcribed, acetylated, 5' IgH V gene segments. Genes Dev 2003; 17:37-42. [PMID: 12514097 PMCID: PMC195966 DOI: 10.1101/gad.1031403] [Citation(s) in RCA: 127] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2002] [Accepted: 11/04/2002] [Indexed: 01/31/2023]
Abstract
Pax5-deficient progenitor B (pro-B) cells are thought to be severely defective for recombination of all immunoglobulin heavy chain (IgH) V gene segments, but the mechanism by which Pax5 regulates this process has not been defined. To address this issue, we have examined the assembly of the IgH locus in Pax5-deficient pro-B cells and find, unexpectedly, that 3' IgH V gene segments, which lie closest to the D-J-Cmu region, recombine efficiently, but progressively more distal V gene segments recombine progressively less efficiently. Histone acetylation and germ-line transcription correlate strongly with an open or an accessible chromatin structure thought to be permissive for V(D)J recombination, and defects in recombination are typically accompanied by deficits in these processes. We were therefore surprised to observe that distal V(H) gene segments in Pax5-/- pro-B cells exhibit no defect in these measures of accessibility. The finding of transcribed, histone acetylated gene segments that fail to recombine suggests that a Pax5-dependent regulatory mechanism is required in addition to standard constraints of accessibility to control V(H) gene recombination.
Collapse
Affiliation(s)
- David G T Hesslein
- Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut 06520-8011, USA
| | | | | | | | | | | |
Collapse
|
190
|
Abstract
T lymphocytes originate from pluripotent precursors and undergo lasting commitment to the T cell developmental fate during their processing in the thymus. Commitment includes both the acquisition of essential T cell characteristics and the foreclosing of other developmental options. Gain of T cell characteristics is probably mediated by separate mechanisms, at least in detail, from loss of alternative developmental potentials. Programmed shifts in survival requirements make changes irreversible. Here we review the current evidence identifying the regulatory components of this commitment pathway, and the first hints of how they work together. Roles for PU.1, GATA-3, and their target genes are highlighted.
Collapse
Affiliation(s)
- Ellen V Rothenberg
- Division of Biology 156-29, California Institute of Technology, Pasadena, CA 91125, USA.
| |
Collapse
|
191
|
Souabni A, Cobaleda C, Schebesta M, Busslinger M. Pax5 promotes B lymphopoiesis and blocks T cell development by repressing Notch1. Immunity 2002; 17:781-93. [PMID: 12479824 DOI: 10.1016/s1074-7613(02)00472-7] [Citation(s) in RCA: 178] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The B lineage commitment factor Pax5 (BSAP) is exclusively expressed in B lymphocytes of the blood system. To study the effect of Pax5 on the development of other hematopoietic lineages, we generated a heterozygous knockin mouse carrying a Pax5 minigene under the control of the Ikaros locus. Conditional and constitutive activation of the Ik(Pax5) allele demonstrated that precocious Pax5 expression in hematopoietic stem cells and progenitors failed to interfere with myeloid development and only weakly affected erythroblast formation. Instead, pan-hematopoietic Pax5 expression strongly promoted B cell development at the expense of T lymphopoiesis. Pax5 thereby interfered with T lineage commitment and early thymocyte development by repressing the transcription of the T cell specification gene Notch1.
Collapse
Affiliation(s)
- Abdallah Souabni
- Research Institute of Molecular Pathology, Vienna Biocenter, Dr. Bohr-Gasse 7, A-1030 Vienna, Austria
| | | | | | | |
Collapse
|
192
|
de Andrés B, Gonzalo P, Minguet S, Martínez-Marin JA, Soro PG, Marcos MAR, Gaspar ML. The first 3 days of B-cell development in the mouse embryo. Blood 2002; 100:4074-81. [PMID: 12393735 DOI: 10.1182/blood-2002-03-0809] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
B-lineage-committed cells are believed to arise in the liver of mouse embryos at 14 days after coitus (dpc). However, pre-B-specific gene transcripts and DJH gene rearrangements have been detected in earlier, midgestation embryos. We describe here a population of c-kit(+)AA4.1(+)CD19(+)Pax5(+) cells present in the aorta-gonad-mesonephros (AGM) area and in the livers of 11-dpc mouse embryos. In contrast to multipotent c-kit(+)AA4.1(+)CD19(-) hematopoietic stem cells (HSCs), these c-kit(+)AA4.1(+)CD19(+) progenitors differentiated only to B-lineage cells in vitro. We propose that mouse embryonic B lymphopoiesis starts earlier than previously thought, at 10 to 11 dpc, both in liver and extra-liver hematopoietic sites. The B-cell differentiation program is not delayed with respect to the emerging lymphohematopoiesis events in the midgestation mouse embryo (8-9 dpc).
Collapse
Affiliation(s)
- Belen de Andrés
- Centro Nacional de Microbiologia, Instituto de Salud Carlos III (ISCIII), Majadahonda, Spain
| | | | | | | | | | | | | |
Collapse
|
193
|
Abstract
B lymphocyte development is regulated by the nuclear proteins Early B cell factor (EBF) and Pax-5. EBF and Pax-5 work separately and in concert to activate genes required for B cell differentiation. Recent studies have defined mechanisms by which these two factors control transcription, including chromatin remodeling activities and recruitment of partner proteins. This review addresses the structures, functions, and roles of these proteins in early B cell commitment and development, as well as in later stages of B cell differentiation.
Collapse
Affiliation(s)
- Holly Maier
- Integrated Department of Immunology, National Jewish Medical and Research Center, 1400 Jackson Street K516B, Denver, CO 80206, USA
| | | |
Collapse
|
194
|
Abstract
In some respects, our understanding of the cellular and molecular aspects of early T-cell differentiation is lagging behind that of B cells. Papers describing gene-knockout and reporter-transgenic mice in which thymocyte development is affected are often difficult to interpret. Progress in this field will be hampered unless a more detailed phenotypic and molecular analysis of progenitor thymocytes at the single-cell level is carried out.
Collapse
Affiliation(s)
- Rod Ceredig
- U548 INSERM, CEA-G, 17 rue des Martyrs, F-38054 Grenoble, France.
| | | |
Collapse
|
195
|
Miller JP, Izon D, DeMuth W, Gerstein R, Bhandoola A, Allman D. The earliest step in B lineage differentiation from common lymphoid progenitors is critically dependent upon interleukin 7. J Exp Med 2002; 196:705-11. [PMID: 12208884 PMCID: PMC2193997 DOI: 10.1084/jem.20020784] [Citation(s) in RCA: 160] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Little is known about the signals that promote early B lineage differentiation from common lymphoid progenitors (CLPs). Using a stromal-free culture system, we show that interleukin (IL)-7 is sufficient to promote the in vitro differentiation of CLPs into B220(+) CD19(+) B lineage progenitors. Consistent with current models of early B cell development, surface expression of B220 was initiated before CD19 and was accompanied by the loss of T lineage potential. To address whether IL-7 receptor (R) activity is essential for early B lineage development in vivo, we examined the frequencies of CLPs and downstream pre-pro- and pro-B cells in adult mice lacking either the alpha chain or the common gamma chain (gamma(c)) of the IL-7R. The data indicate that although gamma(c)(-/-) mice have normal frequencies of CLPs, both gamma(c)(-/-) and IL-7R(alpha)(-/-) mice lack detectable numbers of all downstream early B lineage precursors, including pre-pro-B cells. These findings challenge previous notions regarding the point in B cell development affected by the loss of IL-7R signaling and suggest that IL-7 plays a key and requisite role during the earliest phases of B cell development.
Collapse
Affiliation(s)
- Juli P Miller
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | | | | | | | | | |
Collapse
|
196
|
Avots A, Harder F, Schmittwolf C, Petrovic S, Müller AM. Plasticity of hematopoietic stem cells and cellular memory. Immunol Rev 2002; 187:9-21. [PMID: 12366679 DOI: 10.1034/j.1600-065x.2002.18702.x] [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: 01/18/2023]
Abstract
Stem cell systems represent an effective and powerful approach for tissue development and regeneration of diverse tissue types. Common and defining features of these exceptional cells are the capacity for self-renewal and the potential for differentiation into multiple mature cell types. Recently, surprising new observations have indicated that stem cells isolated from one adult tissue can also give rise to mature cells of other cell lineages, irrespective of classical germ layer designations. This discovery has resulted in quantum leaps in both scientific knowledge and the potential applications of stem cells. The new findings contradict central dogmas of commitment and differentiation of stem and progenitor cells. However, the true potential of somatic stem cells is just emerging and the new findings have to be defined more fully and integrated into a unifying model of stem cell potential and behavior. Here we analyze the developmental potential of hematopoietic stem cells of mouse and man following their injection into the murine preimplantation blastocyst, an environment that allows the development of all cell lineages. In addition, we discuss the emerging lines of evidence of the developmental plasticity of hematopoietic and other somatic stem cells and consider how cellular memory of transcriptional states is established and may be potentially involved in this phenomenon.
Collapse
Affiliation(s)
- Andris Avots
- Institut für Medizinische Strahlenkunde und Zellforschung, Universität Würzburg, 97078 Würzburg, Germany
| | | | | | | | | |
Collapse
|
197
|
Spangrude GJ. Divergent models of lymphoid lineage specification: do clonal assays provide all the answers? Immunol Rev 2002; 187:40-7. [PMID: 12366681 DOI: 10.1034/j.1600-065x.2002.18704.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Hematopoietic stem cells that drive blood development in mouse and man have been well characterized in recent years. In contrast, detailed analysis of the next stages of development, the progenitor cells that have begun to differentiate along specific hematopoietic lineages, is now only in its infancy. The process of myeloid differentiation has been relatively accessible to experimental manipulation due to the availability of culture systems able to support the progenitors for myeloid lineages, and the identification of cytokines capable of driving myeloid differentiation. Studies of early lymphoid differentiation, however, have lagged behind. In particular, the characterization of the first progenitors for the lymphoid lineages is far from complete, due mainly to inefficient assay systems for growing these cell lineages in vitro. Two laboratories have published conflicting data regarding the specification of lymphoid lineages in the mouse. Both groups of investigators utilize elegant clonal approaches to characterize progenitor cell subsets. While these experiments define lineage potential in the most rigorous manner possible, the divergent results suggest that clonal assays must be supplemented with more physiologic studies in order to define the actual differentiation pathways that function in vivo.
Collapse
|
198
|
Abstract
The earliest stages of intrathymic T-cell development include not only the acquisition of T-cell characteristics but also programmed loss of potentials for B, natural killer, and dendritic cell development. Evidence from genetics and cell-transfer studies suggests an order and some components of the mechanisms involved in loss of these options, but some of the interpretations conflict. The conflicts can be resolved by a view that postulates overlapping windows of developmental opportunity and individual mechanisms regulating progression along each pathway. This view is consistent with molecular evidence for the expression patterns of positive regulators of non-T developmental pathways, SCL, PU.1 and Id2, in early thymocytes. To some extent, overexpression of such regulators redirects thymocyte development in vitro. Specific commitment functions may normally terminate this developmental plasticity. Both PU.1 overexpression and stimulation of ectopically expressed growth factor receptors can perturb T- and myeloid/dendritic-cell divergence, but only in permissive stages. A cell-line system that approximates DN3-stage thymocytes reveals that PU.1 can alter specification even in a homogeneous population. However, the response of the population to PU.1 is sharply discontinuous. These studies show a critical role for regulatory context in restricting plasticity, which is probably maintained by interacting transcription factor networks.
Collapse
Affiliation(s)
- Ellen V Rothenberg
- Division of Biology, 156-29, California Institute of Technology, Pasadena, CA 91125, USA.
| | | |
Collapse
|
199
|
Dzierzak E. Hematopoietic stem cells and their precursors: developmental diversity and lineage relationships. Immunol Rev 2002; 187:126-38. [PMID: 12366688 DOI: 10.1034/j.1600-065x.2002.18711.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Within the context of the developing embryo, restrictions in cell lineage potential occur through cell-cell interactions and signaling molecules, leading to changes in genetic programs and to the emergence of disparate tissues containing functionally distinct cell types including somatic stem cells. Tissue maintenance in the adult is thought to occur through specific stem cells, and in the case of the hematopoietic system, through hematopoietic stem cells (HSCs). These cells arise in midgestation within the region of the embryo containing the dorsal aorta, gonads, and mesonephros (AGM) and are thought to maintain a distinct hematopoietic lineage-restricted fate. However, recent transplantation experiments suggest that within the adult, HSCs previously thought to be restricted can, under certain circumstances, display unexpected lineage potentials. With these surprising and controversial results, it is becoming apparent that a better understanding of the developmental processes, molecular programs and lineage relationships leading to the emergence of adult stem cells will provide insight into the incremental steps involved in lineage determination, and perhaps possibilities for the manipulated differentiation of stem cells. The most widely studied, accessible stem cell and cellular differentiation hierarchy is that of the hematopoietic system. With the issue of stem cell potential in the forefront, the focus of this review is on the development of the hematopoietic system: how HSCs arise in the embryo, the lineage relationships of hematopoietic cells as they are generated, and the identification of precursor cells fated to the hematopoietic lineage throughout ontogeny.
Collapse
Affiliation(s)
- Elaine Dzierzak
- Erasmus University Medical Center, Department of Cell Biology and Genetics, PO Box 1738, 3000 DR Rotterdam, The Netherlands.
| |
Collapse
|
200
|
Rolink AG, Schaniel C, Melchers F. Stability and plasticity of wild-type and Pax5-deficient precursor B cells. Immunol Rev 2002; 187:87-95. [PMID: 12366685 DOI: 10.1034/j.1600-065x.2002.18708.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
In Pax5-deficient mice, B cell development is blocked at the pre-BI cell stage. Like wild-type, Pax5-/- pre-BI cells can be grown long-term in vitro in the presence of stromal cells and IL-7. However, unlike their wild-type in vitro-grown counterparts, Pax5-/- pre-BI cells possess an extraordinary developmental plasticity showing hematopoietic stem cell features such as multipotency and self renewing capacity. Here we review and discuss this in vitro and in vivo plasticity of Pax5-/- pre-BI cells.
Collapse
Affiliation(s)
- Antonius G Rolink
- Division of Immunology, Pharmazentrum, University of Basel, Klingelbergstrasse 50, CH-4056 Basel/Switzerland.
| | | | | |
Collapse
|