1
|
Kao CY, Mills JA, Burke CJ, Morse B, Marques BF. Role of Cytokines and Growth Factors in the Manufacturing of iPSC-Derived Allogeneic Cell Therapy Products. BIOLOGY 2023; 12:biology12050677. [PMID: 37237491 DOI: 10.3390/biology12050677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 04/23/2023] [Accepted: 04/27/2023] [Indexed: 05/28/2023]
Abstract
Cytokines and other growth factors are essential for cell expansion, health, function, and immune stimulation. Stem cells have the additional reliance on these factors to direct differentiation to the appropriate terminal cell type. Successful manufacturing of allogeneic cell therapies from induced pluripotent stem cells (iPSCs) requires close attention to the selection and control of cytokines and factors used throughout the manufacturing process, as well as after administration to the patient. This paper employs iPSC-derived natural killer cell/T cell therapeutics to illustrate the use of cytokines, growth factors, and transcription factors at different stages of the manufacturing process, ranging from the generation of iPSCs to controlling of iPSC differentiation into immune-effector cells through the support of cell therapy after patient administration.
Collapse
Affiliation(s)
- Chen-Yuan Kao
- Process and Product Development, Century Therapeutics, Philadelphia, PA 19104, USA
| | - Jason A Mills
- Process and Product Development, Century Therapeutics, Philadelphia, PA 19104, USA
| | - Carl J Burke
- Process and Product Development, Century Therapeutics, Philadelphia, PA 19104, USA
| | - Barry Morse
- Research and Development, Century Therapeutics, Philadelphia, PA 19104, USA
| | - Bruno F Marques
- Process and Product Development, Century Therapeutics, Philadelphia, PA 19104, USA
| |
Collapse
|
2
|
NKT Cells in Mice Originate from Cytoplasmic CD3-Positive, CD4 -CD8 - Double-Negative Thymocytes that Express CD44 and IL-7Rα. Sci Rep 2019; 9:1874. [PMID: 30755654 PMCID: PMC6372634 DOI: 10.1038/s41598-018-37811-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 12/11/2018] [Indexed: 12/16/2022] Open
Abstract
Although natural killer T cells (NKT cells) are thought to be generated from CD4+CD8+ (DP) thymocytes, the developmental origin of CD4−CD8− (DN) NKT cells has remained unclear. In this study, we found the level of NK1.1 expression was highest in DN cells, followed by CD4 and CD8 (SP) and DP cells. The level of NK1.1 expression was highest in CD44+CD25− (DN1) cells, after that CD44+CD25+ (DN2), finally, CD44−CD25− (DN3) and CD44− CD25+ (DN4) cells. Unexpectedly, cytoplasmic CD3 was not only expressed in SP and DP thymocytes but also in most DN thymocytes at various stages. The mean fluorescence of cytoplasmic and surface CD3 in DN cells was significantly lower than in mature (SP) T and NKT cells in the thymus and spleen. Interestingly, there were more NKT cells in DN-cytoplasmic CD3 expression cells was higher than in DN-surface CD3 expression cells. There were more CD3-NKT cells in DN1 thymocytes than in TCR-β-NKT cells. NKT cells expressed higher levels of IL-7Rα which was correlated with CD44 expression in the thymus. Our data suggest that T cells and NKT cells follow similar patterns of expression with respect to cytoplasmic and surface CD3. Cytoplasmic CD3 could be used as a marker for early stage T cells. Both cytoplasmic CD3 and surface CD3 were expressed in mature T cells and immature T cells, including the immature cytoplasmic CD3+ surface CD3− and surface CD3+TCR-β− cells in DN1-NKT thymocytes. CD44 could be used as an additional marker of NKT cells which may originate from cytoplasmic CD3-positive DN thymocytes that express CD44 and IL-7Rα in mice.
Collapse
|
3
|
Chaves P, Zriwil A, Wittmann L, Boukarabila H, Peitzsch C, Jacobsen SEW, Sitnicka E. Loss of Canonical Notch Signaling Affects Multiple Steps in NK Cell Development in Mice. THE JOURNAL OF IMMUNOLOGY 2018; 201:3307-3319. [PMID: 30366956 DOI: 10.4049/jimmunol.1701675] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 09/27/2018] [Indexed: 11/19/2022]
Abstract
Within the hematopoietic system, the Notch pathway is critical for promoting thymic T cell development and suppressing the B and myeloid lineage fates; however, its impact on NK lymphopoiesis is less understood. To study the role of Notch during NK cell development in vivo, we investigated different NK cell compartments and function in Rbp-Jkfl/flVav-Cretg/+ mice, in which Rbp-Jk, the major transcriptional effector of canonical Notch signaling, was specifically deleted in all hematopoietic cells. Peripheral conventional cytotoxic NK cells in Rbp-Jk-deleted mice were significantly reduced and had an activated phenotype. Furthermore, the pool of early NK cell progenitors in the bone marrow was decreased, whereas immature NK cells were increased, leading to a block in NK cell maturation. These changes were cell intrinsic as the hematopoietic chimeras generated after transplantation of Rbp-Jk-deficient bone marrow cells had the same NK cell phenotype as the Rbp-Jk-deleted donor mice, whereas the wild-type competitors did not. The expression of several crucial NK cell regulatory pathways was significantly altered after Rbp-Jk deletion. Together, these results demonstrate the involvement of canonical Notch signaling in regulation of multiple stages of NK cell development.
Collapse
Affiliation(s)
- Patricia Chaves
- Lund Research Center for Stem Cell Biology and Cell Therapy, Lund University, 221 84 Lund, Sweden.,Division of Molecular Hematology, Department of Laboratory Medicine, Lund University, 221 84 Lund, Sweden
| | - Alya Zriwil
- Lund Research Center for Stem Cell Biology and Cell Therapy, Lund University, 221 84 Lund, Sweden.,Division of Molecular Hematology, Department of Laboratory Medicine, Lund University, 221 84 Lund, Sweden
| | - Lilian Wittmann
- Lund Research Center for Stem Cell Biology and Cell Therapy, Lund University, 221 84 Lund, Sweden.,Division of Molecular Hematology, Department of Laboratory Medicine, Lund University, 221 84 Lund, Sweden
| | - Hanane Boukarabila
- Haematopoietic Stem Cell Laboratory, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DS, United Kingdom
| | - Claudia Peitzsch
- Lund Research Center for Stem Cell Biology and Cell Therapy, Lund University, 221 84 Lund, Sweden
| | - Sten Eirik W Jacobsen
- Haematopoietic Stem Cell Laboratory, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DS, United Kingdom.,MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DS, United Kingdom.,Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, 141 86 Stockholm, Sweden; and.,Department of Cell and Molecular Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Ewa Sitnicka
- Lund Research Center for Stem Cell Biology and Cell Therapy, Lund University, 221 84 Lund, Sweden; .,Division of Molecular Hematology, Department of Laboratory Medicine, Lund University, 221 84 Lund, Sweden
| |
Collapse
|
4
|
Croy BA, Ashkar AA, Minhas K, Greenwood JD. Can Murine Uterine Natural Killer Cells Give Insights Into the Pathogenesis of Preeclampsia? ACTA ACUST UNITED AC 2016. [DOI: 10.1177/107155760000700104] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- B. Anne Croy
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | | | | | - James D. Greenwood
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
5
|
Cheng K, Ibrahim A, Hensley MT, Shen D, Sun B, Middleton R, Liu W, Smith RR, Marbán E. Relative roles of CD90 and c-kit to the regenerative efficacy of cardiosphere-derived cells in humans and in a mouse model of myocardial infarction. J Am Heart Assoc 2014; 3:e001260. [PMID: 25300435 PMCID: PMC4323830 DOI: 10.1161/jaha.114.001260] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Background The regenerative potential of cardiosphere‐derived cells (CDCs) for ischemic heart disease has been demonstrated in mice, rats, pigs, and a recently completed clinical trial (CADUCEUS). CDCs are CD105+ stromal cells of intrinsic cardiac origin, but the antigenic characteristics of the active fraction remain to be defined. CDCs contain a small minority of c‐kit+ cells, which have been argued to be cardiac progenitors, and a variable fraction of CD90+ cells whose bioactivity is unclear. Methods We performed a retrospective analysis of data from the CADUCEUS trial and a prospective mouse study to elucidate the roles of c‐kit+ and CD90+ cells in human CDCs. Here, we show, surprisingly, that c‐kit expression has no relationship to CDCs' therapeutic efficacy in humans, and depletion of c‐kit+ cells does not undermine the structural and functional benefits of CDCs in a mouse model of myocardial infarction (MI). In contrast, CD90 expression negatively correlates with the therapeutic benefit of CDCs in humans (ie, higher CD90 expression associated with lower efficacy). Depletion of CD90+ cells augments the functional potency of CDCs in murine MI. CD90− CDCs secrete lower levels of inflammatory cytokines and can differentiate into cardiomyocytes in vitro and in vivo. Conclusion The majority population of CDCs (CD105+/CD90−/c‐kit−) constitutes the active fraction, both in terms of therapeutic efficacy and in the ability to undergo cardiomyogenic differentiation. The c‐kit+ fraction is neither necessary for, nor contributory to, the regenerative efficacy of CDCs.
Collapse
Affiliation(s)
- Ke Cheng
- Cedars-Sinai Heart Institute, Los Angeles, CA (K.C., A.I., B.S., R.M., W.L., R.R.S., E.M.) Department of Molecular Biomedical Sciences and Center for Comparative Medicine and Translational Research, College of Veterinary Medicine, North Carolina State University, Raleigh, NC (K.C., T.H.) Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University Raleigh, NC (K.C.)
| | - Ahmed Ibrahim
- Cedars-Sinai Heart Institute, Los Angeles, CA (K.C., A.I., B.S., R.M., W.L., R.R.S., E.M.)
| | - M Taylor Hensley
- Department of Molecular Biomedical Sciences and Center for Comparative Medicine and Translational Research, College of Veterinary Medicine, North Carolina State University, Raleigh, NC (K.C., T.H.)
| | - Deliang Shen
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (D.S.)
| | - Baiming Sun
- Cedars-Sinai Heart Institute, Los Angeles, CA (K.C., A.I., B.S., R.M., W.L., R.R.S., E.M.)
| | - Ryan Middleton
- Cedars-Sinai Heart Institute, Los Angeles, CA (K.C., A.I., B.S., R.M., W.L., R.R.S., E.M.)
| | - Weixin Liu
- Cedars-Sinai Heart Institute, Los Angeles, CA (K.C., A.I., B.S., R.M., W.L., R.R.S., E.M.)
| | - Rachel R Smith
- Cedars-Sinai Heart Institute, Los Angeles, CA (K.C., A.I., B.S., R.M., W.L., R.R.S., E.M.) Capricor Therapeutics Inc, Beverly Hills, CA (R.R.S.)
| | - Eduardo Marbán
- Cedars-Sinai Heart Institute, Los Angeles, CA (K.C., A.I., B.S., R.M., W.L., R.R.S., E.M.)
| |
Collapse
|
6
|
Kirkham CL, Carlyle JR. Complexity and Diversity of the NKR-P1:Clr (Klrb1:Clec2) Recognition Systems. Front Immunol 2014; 5:214. [PMID: 24917862 PMCID: PMC4041007 DOI: 10.3389/fimmu.2014.00214] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 04/28/2014] [Indexed: 11/26/2022] Open
Abstract
The NKR-P1 receptors were identified as prototypical natural killer (NK) cell surface antigens and later shown to be conserved from rodents to humans on NK cells and subsets of T cells. C-type lectin-like in nature, they were originally shown to be capable of activating NK cell function and to recognize ligands on tumor cells. However, certain family members have subsequently been shown to be capable of inhibiting NK cell activity, and to recognize proteins encoded by a family of genetically linked C-type lectin-related ligands. Some of these ligands are expressed by normal, healthy cells, and modulated during transformation, infection, and cellular stress, while other ligands are upregulated during the immune response and during pathological circumstances. Here, we discuss historical and recent developments in NKR-P1 biology that demonstrate this NK receptor–ligand system to be far more complex and diverse than originally anticipated.
Collapse
Affiliation(s)
- Christina L Kirkham
- Department of Immunology, University of Toronto, Sunnybrook Research Institute , Toronto, ON , Canada
| | - James R Carlyle
- Department of Immunology, University of Toronto, Sunnybrook Research Institute , Toronto, ON , Canada
| |
Collapse
|
7
|
Li TS, Cheng K, Malliaras K, Smith RR, Zhang Y, Sun B, Matsushita N, Blusztajn A, Terrovitis J, Kusuoka H, Marbán L, Marbán E. Direct comparison of different stem cell types and subpopulations reveals superior paracrine potency and myocardial repair efficacy with cardiosphere-derived cells. J Am Coll Cardiol 2012; 59:942-53. [PMID: 22381431 DOI: 10.1016/j.jacc.2011.11.029] [Citation(s) in RCA: 350] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2011] [Revised: 10/19/2011] [Accepted: 11/22/2011] [Indexed: 10/28/2022]
Abstract
OBJECTIVES The goal of this study was to conduct a direct head-to-head comparison of different stem cell types in vitro for various assays of potency and in vivo for functional myocardial repair in the same mouse model of myocardial infarction. BACKGROUND Adult stem cells of diverse origins (e.g., bone marrow, fat, heart) and antigenic identity have been studied for repair of the damaged heart, but the relative utility of the various cell types remains unclear. METHODS Human cardiosphere-derived cells (CDCs), bone marrow-derived mesenchymal stem cells, adipose tissue-derived mesenchymal stem cells, and bone marrow mononuclear cells were compared. RESULTS CDCs revealed a distinctive phenotype with uniform expression of CD105, partial expression of c-kit and CD90, and negligible expression of hematopoietic markers. In vitro, CDCs showed the greatest myogenic differentiation potency, highest angiogenic potential, and relatively high production of various angiogenic and antiapoptotic-secreted factors. In vivo, injection of CDCs into the infarcted mouse hearts resulted in superior improvement of cardiac function, the highest cell engraftment and myogenic differentiation rates, and the least-abnormal heart morphology 3 weeks after treatment. CDC-treated hearts also exhibited the lowest number of apoptotic cells. The c-kit(+) subpopulation purified from CDCs produced lower levels of paracrine factors and inferior functional benefit when compared with unsorted CDCs. To validate the comparison of cells from various human donors, selected results were confirmed in cells of different types derived from individual rats. CONCLUSIONS CDCs exhibited a balanced profile of paracrine factor production and, among various comparator cell types/subpopulations, provided the greatest functional benefit in experimental myocardial infarction.
Collapse
Affiliation(s)
- Tao-Sheng Li
- Cedars-Sinai Heart Institute, Los Angeles, California, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
HEB in the spotlight: Transcriptional regulation of T-cell specification, commitment, and developmental plasticity. Clin Dev Immunol 2012; 2012:678705. [PMID: 22577461 PMCID: PMC3346973 DOI: 10.1155/2012/678705] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2011] [Accepted: 12/12/2011] [Indexed: 12/02/2022]
Abstract
The development of T cells from multipotent progenitors in the thymus occurs by cascades of interactions between signaling molecules and transcription factors, resulting in the loss of alternative lineage potential and the acquisition of the T-cell functional identity. These processes require Notch signaling and the activity of GATA3, TCF1, Bcl11b, and the E-proteins HEB and E2A. We have shown that HEB factors are required to inhibit the thymic NK cell fate and that HEBAlt allows the passage of T-cell precursors from the DN to DP stage but is insufficient for suppression of the NK cell lineage choice. HEB factors are also required to enforce the death of cells that have not rearranged their TCR genes. The synergistic interactions between Notch1, HEBAlt, HEBCan, GATA3, and TCF1 are presented in a gene network model, and the influence of thymic stromal architecture on lineage choice in the thymus is discussed.
Collapse
|
9
|
Thompson PK, Zúñiga-Pflücker JC. On becoming a T cell, a convergence of factors kick it up a Notch along the way. Semin Immunol 2011; 23:350-9. [PMID: 21981947 DOI: 10.1016/j.smim.2011.08.007] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2010] [Accepted: 08/19/2011] [Indexed: 12/18/2022]
Abstract
The thymus is seeded by bone marrow-derived progenitors, which undergo a series of differentiation and proliferation events in order to generate functional T lymphocytes. The Notch signaling pathway, together with multiple transcription factors, act in concert to commit progenitors to a T-lineage fate, extinguishing non-T cell potential, inducing thymocyte differentiation and supporting proliferation and survival along the way to becoming a mature T cell. This review focuses on recent evidence regarding the complex interplay between the Notch pathway and other key transcription factors at specific lineage-decision points during the program of T cell development.
Collapse
Affiliation(s)
- Patrycja K Thompson
- Department of Immunology, University of Toronto, Sunnybrook Research Institute, Toronto, Ontario M4N 3M5, Canada
| | | |
Collapse
|
10
|
HEB-deficient T-cell precursors lose T-cell potential and adopt an alternative pathway of differentiation. Mol Cell Biol 2010; 31:971-82. [PMID: 21189289 DOI: 10.1128/mcb.01034-10] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Early thymocytes possess multilineage potential, which is progressively restricted as cells transit through the double-negative stages of T-cell development. DN1 cells retain the ability to become natural killer cells, dendritic cells, B cells, and myeloid cells as well as T cells, but these options are lost by the DN3 stage. The Notch1 signaling pathway is indispensable for initiation of the T-cell lineage and inhibitory for the B-cell lineage, but the regulatory mechanisms by which the T-cell fate is locked in are largely undefined. Previously, we discovered that the E-protein transcription factor HEBAlt promoted T-cell specification. Here, we report that HEB(-/-) T-cell precursors have compromised Notch1 function and lose T-cell potential. Moreover, reconstituting HEB(-/-) precursors with Notch1 activity enforced fidelity to the T-cell fate. However, instead of becoming B cells, HEB(-/-) DN3 cells adopted a DN1-like phenotype and could be induced to differentiate into thymic NK cells. HEB(-/-) DN1-like cells retained GATA3 and Id2 expression but had lower levels of the Bcl11b gene, a Notch target gene. Therefore, our studies have revealed a new set of interactions between HEB, Notch1, and GATA3 that regulate the T-cell fate choice in developing thymocytes.
Collapse
|
11
|
Blossom SJ, Doss JC. Trichloroethylene alters central and peripheral immune function in autoimmune-prone MRL(+/+) mice following continuous developmental and early life exposure. J Immunotoxicol 2009; 4:129-41. [PMID: 18958721 DOI: 10.1080/15476910701337035] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Trichloroethylene (TCE) is a widespread environmental toxicant known to promote CD4(+) T-lymphocyte activation, IFNgamma production, and autoimmunity in adult MRL(+/+) mice. Because developing tissues may be more sensitive to toxicant exposure, it was hypothesized that continuous TCE exposure beginning at conception might induce even more pronounced CD4(+) T-lymphocyte effects and exacerbate the development of autoimmunity in MRL(+/+) mice. In the current study, MRL(+/+) mice were exposed to occupationally-relevant doses of TCE from conception until adulthood (i.e., 7-8 wk-of-age). The CD4(+) T-lymphocyte effects in the thymus and periphery were evaluated, as well as serum antibody levels. TCE exposure altered the number of thymocyte subsets, and reduced the capacity of the most immature CD4-/CD8- thymocytes to undergo apoptosis in vitro. In the periphery, T-lymphocyte IFN(gamma) production was monitored in the blood prior to sacrifice by intracellular cytokine staining and flow cytometry. TCE induced a dose-dependent increase in T-lymphocyte IFN(gamma) as early as 4-5-week-of-age. However, these effects were transient, and not observed in splenic T-lymphocytes in 7-8-week-old mice. In contrast, the serum levels of anti-histone autoantibodies and total IgG(2a) were significantly elevated in the TCE-exposed offspring. The data illustrated that occupationally-relevant doses of TCE administered throughout development until adulthood affected central and peripheral immune function in association with early signs of autoimmunity. Future studies will address the possibility that early-life exposure to TCE may alter some aspect of self tolerance in the thymus, leading to autoimmune disease later in life.
Collapse
Affiliation(s)
- Sarah J Blossom
- Department of Pediatrics, Arkansas Children's Hospital Research Institute, University of Arkansas for Medical Sciences College of Medicine, Little Rock, Arkansas 72202, USA.
| | | |
Collapse
|
12
|
Extrinsic and intrinsic regulation of early natural killer cell development. Immunol Res 2008; 40:193-207. [PMID: 18266115 DOI: 10.1007/s12026-007-8006-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Natural killer (NK) cells are lymphocytes that play a critical role in both adaptive and innate immune responses. These cells develop from multipotent progenitors in the embryonic thymus and neonatal or adult bone marrow and recent evidence suggests that a subset of these cells may develop in the thymus. Thymus- and bone marrow-derived NK cells have unique phenotypes and functional abilities supporting the hypothesis that the microenvironment dictates the outcome of NK cell development. A detailed understanding of the mechanisms controlling this developmental program will be required to determine how alterations in NK cell development lead to disease and to determine how to harness this developmental program for therapeutic purposes. In this review, we discuss some of the known extrinsic stromal-cell derived factors and cell intrinsic transcription factors that function in guiding NK cell development.
Collapse
|
13
|
Abstract
Natural killer (NK) cells have been thought to develop from committed progenitors in the bone marrow. However, a novel pathway of thymus-dependent NK-cell development that produces a unique subset of NK cells expressing CD127 has recently been reported. We now have identified 2 populations of NK progenitors, one in the thymus and the other in the lymph node (LN). Immature double-negative 2 (CD4(-)CD8(-)CD44(+)CD25(+)) thymocytes have potential to produce NK cells with rearranged T-cell receptor gamma genes (Tcrgamma(+)) in vitro. Tcrgamma(+) NK cells are rare in spleen but relatively abundant in the thymus and LN. Approximately 20% of LN NK cells are Tcrgamma(+), and they are found at similar levels in both CD127(+) and CD127(-) subsets. Moreover, a subpopulation of LN cells resembling immature thymocytes differentiates into Tcrgamma(+) NK cells in vitro and also repopulates the NK compartment in lymphopenic mice. Athymic mice lack the LN NK progenitors expressing CD127 as well as Tcrgamma(+) NK cells. These results suggest that Tcrgamma(+) NK cells may be generated from unique progenitors in the thymus as well as in the LN.
Collapse
|
14
|
de Andrés B, Cortegano I, Serrano N, del Rio B, Martín P, Gonzalo P, Marcos MAR, Gaspar ML. A population of CD19highCD45R-/lowCD21low B lymphocytes poised for spontaneous secretion of IgG and IgA antibodies. THE JOURNAL OF IMMUNOLOGY 2007; 179:5326-34. [PMID: 17911619 DOI: 10.4049/jimmunol.179.8.5326] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Ab responses to selected Ags are produced by discrete B cell populations whose presence and functional relevance vary along the ontogeny. The earliest B lineage-restricted precursors in gestational day 11 mouse embryos display the CD19(+)CD45R/B220(-) phenotype. Phenotypically identical cells persist throughout gestation and in postnatal life, in parallel to the later-arising, CD19(+)CD45R(+) B cells. Very early after birth, the CD19(+)CD45R(-) B cell subset included high frequencies of spontaneously Ig-secreting cells. In the adult spleen, a small subset of CD19(high)CD45R(-/low)IgM(+/-)IgD(-)CD21/Cr2(-/low) cells, which was detected in perifollicular areas, displayed genetic and phenotypical traits of highly differentiated B cells, and was enriched in IgG- and IgA-secreting plasma cells. In vitro differentiation and in vivo adoptive transfer experiments of multipotent hemopoietic progenitors revealed that these CD19(high)CD45R(-/low) B cells were preferentially regenerated by embryo-, but not by adult bone marrow-, derived progenitors, except when the latter were inoculated into newborn mice. Both the early ontogenical emergence and the natural production of serum Igs, are shared features of this CD19(high)CD45R(-/low) B cell population with innate-like B lymphocytes such as B1 and marginal zone B cells, and suggest that the new population might be related to that category.
Collapse
Affiliation(s)
- Belén de Andrés
- Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain.
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Boos MD, Yokota Y, Eberl G, Kee BL. Mature natural killer cell and lymphoid tissue-inducing cell development requires Id2-mediated suppression of E protein activity. ACTA ACUST UNITED AC 2007; 204:1119-30. [PMID: 17452521 PMCID: PMC2118569 DOI: 10.1084/jem.20061959] [Citation(s) in RCA: 301] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The Id2 transcriptional repressor is essential for development of natural killer (NK) cells, lymphoid tissue–inducing (LTi) cells, and secondary lymphoid tissues. Id2 was proposed to regulate NK and LTi lineage specification from multipotent progenitors through suppression of E proteins. We report that NK cell progenitors are not reduced in the bone marrow (BM) of Id2−/− mice, demonstrating that Id2 is not essential for NK lineage specification. Rather, Id2 is required for development of mature (m) NK cells. We define the mechanism by which Id2 functions by showing that a reduction in E protein activity, through deletion of E2A, overcomes the need for Id2 in development of BM mNK cells, LTi cells, and secondary lymphoid tissues. However, mNK cells are not restored in the blood or spleen of Id2−/−E2A−/− mice, suggesting a role for Id2 in suppression of alternative E proteins after maturation. Interestingly, the few splenic mNK cells in Id2−/− and Id2−/−E2A−/− mice have characteristics of thymus-derived NK cells, which develop in the absence of Id2, implying a differential requirement for Id2 in BM and thymic mNK development. Our findings redefine the essential functions of Id2 in lymphoid development and provide insight into the dynamic regulation of E and Id proteins during this process.
Collapse
Affiliation(s)
- Markus D Boos
- Committee on Immunology, University of Chicago, Chicago, IL 60657, USA
| | | | | | | |
Collapse
|
16
|
Mesci A, Ljutic B, Makrigiannis AP, Carlyle JR. NKR-P1 biology: from prototype to missing self. Immunol Res 2006; 35:13-26. [PMID: 17003506 DOI: 10.1385/ir:35:1:13] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 11/30/1999] [Indexed: 11/11/2022]
Abstract
Natural killer (NK) cells represent lymphocytes of the innate immune system capable of recognizing and destroying a broad array of target cells, including tumors, virus-infected cells, antibodycoated cells, foreign transplants, and "stressed" cells. NK cells eliminate their targets through two main effector mechanisms, cytokine secretion and cell-mediated cytotoxicity, which in turn depend on detection of target cells through a complex integration of stimulatory and inhibitory receptor-ligand interactions. The NKR-P1 molecules were the first family of NK cell receptors identified, yet they have remained enigmatic in their contribution to self-nonself discrimination until recently. Here, we outline a brief history of the NKR-P1 receptor family, then examine recent data providing insight into their genetic regulation, signaling function, cognate ligands, and gene organization and diversity.
Collapse
Affiliation(s)
- Aruz Mesci
- Department of Immunology, University of Toronto, Sunnybrook & Women's Research Institute, 2075 Bayview Avenue, Toronto, ON, M4N 3M5, Canada
| | | | | | | |
Collapse
|
17
|
Abstract
Transcriptional regulation of T-cell development involves successive interactions between complexes of transcriptional regulators and their binding sites within the regulatory regions of each gene. The regulatory modules that control expression of T-lineage genes frequently include binding sites for a core set of regulators that set the T-cell-specific background for signal-dependent control, including GATA-3, Notch/CSL, c-myb, TCF-1, Ikaros, HEB/E2A, Ets, and Runx factors. Additional regulators in early thymocytes include PU.1, Id-2, SCL, Spi-B, Erg, Gfi-1, and Gli. Many of these factors are involved in simultaneous regulation of non-T-lineage genes, T-lineage genes, and genes involved in cell cycle control, apoptosis, or survival. Potential and known interactions between early thymic transcription factors such as GATA-3, SCL, PU.1, Erg, and Spi-B are explored. Regulatory modules involved in the expression of several critical T-lineage genes are described, and models are presented for shifting occupancy of the DNA-binding sites in the regulatory modules of pre-Talpha, T-cell receptor beta (TCRbeta), recombinase activating genes 1 and 2 (Rag-1/2), and CD4 during T-cell development. Finally, evidence is presented that c-kit, Erg, Hes-1, and HEBAlt are expressed differently in Rag-2(-/-) thymocytes versus normal early thymocytes, which provide insight into potential regulatory interactions that occur during normal T-cell development.
Collapse
Affiliation(s)
- Michele K Anderson
- Sunnybrook and Women's College Health Sciences Center, Division of Molecular and Cell Biology, University of Toronto, Department of Immunology, Toronto, ON, Canada.
| |
Collapse
|
18
|
Xu H, Williams MS, Spain LM. Patterns of expression, membrane localization, and effects of ectopic expression suggest a function for MS4a4B, a CD20 homolog in Th1 T cells. Blood 2005; 107:2400-8. [PMID: 16293604 PMCID: PMC1895730 DOI: 10.1182/blood-2005-08-3340] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The membrane-spanning 4A (MS4A) family of proteins includes CD20, Fc epsilonRIbeta, and HTm4, whose genes are grouped in a chromosomal location that is associated with increased susceptibility to allergy and atopic asthma. One family member, Chandra/MS4a4B, was reported to be expressed in T helper 1 (Th1) T cells but not Th2 T cells. In the present study, Ms4a4b was isolated in a screen of genes differentially expressed during thymocyte development. MS4a4B was detected in immature CD4- CD8- CD44+ CD25- thymocytes, turned off during further stages of thymocyte development and reexpressed in mature single-positive thymocytes. MS4a4B expression was found in naive CD8+ and CD4+ peripheral T cells and natural killer (NK) cells but not in B cells. MS4a4B is expressed at the cell surface with its C-terminus located in the cytoplasm. When expressed in a T-cell hybridoma by retroviral vector, MS4a4B protein constitutively associated with lipid raft microdomains, whereas in primary T cells endogenous MS4a4B protein became enriched in rafts after T-cell activation. Overexpression of MS4a4B in primary CD4+ T-cell blasts enhanced T-cell receptor (TCR)-induced Th1 cytokine production. These results suggest that MS4a4B expression is tightly regulated during T-cell development and that MS4a4B expression promotes Th1 function and/or differentiation.
Collapse
Affiliation(s)
- Hui Xu
- Department of Microbiology and Immunology, Center for Vascular and Inflammatory Diseases, The University of Maryland School of Medicine, 800 W Baltimore St, Baltimore, MD 21201, USA
| | | | | |
Collapse
|
19
|
Sambandam A, Maillard I, Zediak VP, Xu L, Gerstein RM, Aster JC, Pear WS, Bhandoola A. Notch signaling controls the generation and differentiation of early T lineage progenitors. Nat Immunol 2005; 6:663-70. [PMID: 15951813 DOI: 10.1038/ni1216] [Citation(s) in RCA: 291] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2005] [Accepted: 05/17/2005] [Indexed: 12/29/2022]
Abstract
Signaling by the transmembrane receptor Notch is critical for T lineage development, but progenitor subsets that first receive Notch signals have not been defined. Here we identify an immature subset of early T lineage progenitors (ETPs) in the thymus that expressed the tyrosine kinase receptor Flt3 and had preserved B lineage potential at low progenitor frequency. Notch signaling was active in ETPs and was required for generation of the ETP population. Additionally, Notch signals contributed to the subsequent differentiation of ETPs. In contrast, multipotent hematopoietic progenitors circulated in the blood even in the absence of Notch signaling, suggesting that critical Notch signals during early T lineage development are delivered early after thymic entry.
Collapse
Affiliation(s)
- Arivazhagan Sambandam
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Ljutic B, Carlyle JR, Filipp D, Nakagawa R, Julius M, Zúñiga-Pflücker JC. Functional requirements for signaling through the stimulatory and inhibitory mouse NKR-P1 (CD161) NK cell receptors. THE JOURNAL OF IMMUNOLOGY 2005; 174:4789-96. [PMID: 15814704 DOI: 10.4049/jimmunol.174.8.4789] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The NK cell receptor protein 1 (NKR-P1) (CD161) molecules represent a family of type II transmembrane C-type lectin-like receptors expressed predominantly by NK cells. Despite sharing a common NK1.1 epitope, the mouse NKR-P1B and NKR-P1C receptors possess opposing functions in NK cell signaling. Engagement of NKR-P1C stimulates cytotoxicity of target cells, Ca2+ flux, phosphatidylinositol turnover, kinase activity, and cytokine production. In contrast, NKR-P1B engagement inhibits NK cell cytotoxicity. Nonetheless, it remains unclear how different signaling outcomes are mediated at the molecular level. Here, we demonstrate that both NKR-P1B and NKR-P1C associate with the tyrosine kinase, p56(lck). The interaction is mediated through the di-cysteine CxCP motif in the cytoplasmic domains of NKR-P1B/C. Disrupting this motif leads to abrogation of both stimulatory and inhibitory NKR-P1 signals. In addition, mutation of the consensus ITIM (LxYxxL) in NKR-P1B abolishes both its Src homology 2-containing protein tyrosine phosphatase-1 recruitment and inhibitory function. Strikingly, engagement of NKR-P1C on NK cells obtained from Lck-deficient mice failed to induce NK cytotoxicity. These results reveal a role for Lck in the initiation of NKR-P1 signals, and demonstrate a requirement for the ITIM in NKR-P1-mediated inhibition.
Collapse
MESH Headings
- Amino Acid Motifs
- Amino Acid Sequence
- Animals
- Antigens, Ly
- Antigens, Surface/chemistry
- Antigens, Surface/genetics
- Antigens, Surface/metabolism
- Calcium Signaling
- Cell Line
- Humans
- Jurkat Cells
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Lectins, C-Type/chemistry
- Lectins, C-Type/genetics
- Lectins, C-Type/metabolism
- Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Models, Immunological
- Molecular Sequence Data
- Mutagenesis, Site-Directed
- NK Cell Lectin-Like Receptor Subfamily B
- Sequence Homology, Amino Acid
- Signal Transduction
Collapse
Affiliation(s)
- Belma Ljutic
- Department of Immunology, University of Toronto, Sunnybrook and Women's College Health Sciences Centre, Toronto, Ontario, Canada
| | | | | | | | | | | |
Collapse
|
21
|
Marshall D, Bagley J, Le P, Hogquist K, Cyr S, Von Schild E, Pykett M, Rosenzweig M. T cell generation including positive and negative selection ex vivo in a three-dimensional matrix. ACTA ACUST UNITED AC 2004; 12:565-74. [PMID: 14594513 DOI: 10.1089/152581603322448277] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The prevailing paradigm is that T cell differentiation is dependent on interactions between stem cells and neuroectodermal thymic cells in the context of a three-dimensional environment. We evaluated the utility of a three-dimensional matrix, the Cytomatrix, to facilitate T cell differentiation. Thymus stroma grown on the Cytomatrix and seeded with hematopoietic progenitors was observed to support the development of both CD4(+) and CD8(+) T cells. Murine transgenic models used to address T cell selection demonstrated that both positive and negative selection was maintained in the context of MHC Class I. These data demonstrate that this in vitro system using neuroectoderm tissue is capable of the efficient production of T cells from hematopoietic progenitors and presents the possibility of generating and adoptively transferring immune cells to patients.
Collapse
|
22
|
Carlyle JR, Jamieson AM, Gasser S, Clingan CS, Arase H, Raulet DH. Missing self-recognition of Ocil/Clr-b by inhibitory NKR-P1 natural killer cell receptors. Proc Natl Acad Sci U S A 2004; 101:3527-32. [PMID: 14990792 PMCID: PMC373496 DOI: 10.1073/pnas.0308304101] [Citation(s) in RCA: 166] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The NKR-P1 family of C-type lectin-like receptors are expressed on natural killer (NK) cells and NKT cells. We report the cloning and characterization of a cognate ligand for the inhibitory mouse NK receptors (NKR)-P1B and NKR-P1D (CD161b/d). The NKR-P1B/D ligand is osteoclast inhibitory lectin (Ocil), also known as Clr-b, a member of a previously cloned group of C-type lectin-related (Clr) proteins linked to the NKR-P1 receptors in the mouse NK gene complex (NKC). Expression of Ocil/Clr-b on mouse tumor cell lines inhibits NK cell-mediated killing. Inhibition is blocked with a new mAb (4A6) specific for Ocil/Clr-b. By using 4A6 mAb, we demonstrate that Ocil/Clr-b is displayed at high levels on nearly all hematopoietic cells, with the exception of erythrocytes, in a pattern that is similar to that of class I MHC molecules. Remarkably, Ocil/Clr-b is frequently down-regulated on mouse tumor cell lines, indicating a role for this receptor-ligand system in a new form of "missing self-recognition" of tumor cells.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal
- Antigens, Surface/genetics
- Antigens, Surface/metabolism
- Base Sequence
- Cell Line, Tumor
- Cloning, Molecular
- DNA, Complementary/genetics
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Lectins, C-Type/genetics
- Lectins, C-Type/metabolism
- Ligands
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Molecular Sequence Data
- NK Cell Lectin-Like Receptor Subfamily B
- Receptors, Immunologic/genetics
- Receptors, Immunologic/metabolism
Collapse
Affiliation(s)
- James R Carlyle
- Department of Molecular and Cell Biology and Cancer Research Laboratory, University of California, 485 Life Sciences Addition, Berkeley, CA 94720, USA.
| | | | | | | | | | | |
Collapse
|
23
|
Ljutic B, Carlyle JR, Zúñiga-Pflücker JC. Identification of upstream cis-acting regulatory elements controlling lineage-specific expression of the mouse NK cell activation receptor, NKR-P1C. J Biol Chem 2003; 278:31909-17. [PMID: 12813047 DOI: 10.1074/jbc.m212869200] [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: 01/08/2023] Open
Abstract
Mouse NKR-P1C (NK1.1) is a homodimeric type II transmembrane protein expressed on natural killer (NK) cells, NKT cells, and on CD117+ progenitor thymocytes capable of giving rise to cells of the T and NK lineages. Although its physiological ligands remain unknown, NKR-P1C engagement with a monoclonal antibody (mAb) leads to interferon-gamma (IFN-gamma) production and the directed release of cytotoxic granules from NK cells. We have cloned and sequenced a approximately 10-kb genomic fragment corresponding to the 5'-flanking region of the C57Bl/6 mouse NKR-P1C gene. A transcriptional initiation site has been mapped in NK cells and an NK1.1+ T cell line by primer extension and rapid amplification of 5'-cDNA ends (5'-RACE) techniques. Although the 5'-flanking region of NKR-P1C is TATA-less, we have identified an initiator region and a downstream promoter element, which together constitute the principal minimal functional promoter. Computational analysis of the 10-kb 5'-flanking region revealed potential regulatory factor binding sites. DNaseI hypersensitivity assays identified a single hypersensitive site (HS) about a 9-kb upstream of the transcriptional initiation site. This site, termed HS1, was able to act as a transcriptional enhancer element in an NK cell line, while minimally affecting transcription in non-NK cell lines. Moreover, the HS1 element was shown to function as a promoter, with a transcript detected only in fetal NK1.1+ cells. An additional promoter and two non-coding exons were also characterized. These results identify the minimal upstream cis-acting elements, and point to a complex regulatory mechanism involved in the lineage-specific control of NKR-P1C expression in NK lymphocytes.
Collapse
Affiliation(s)
- Belma Ljutic
- Department of Immunology, University of Toronto, Sunnybrook and Women's Health Sciences Centre, Toronto, Ontario M4N 3M5, Canada
| | | | | |
Collapse
|
24
|
Lin CW, Yang JY, Chuang YC, Chen YH, Albitar M, Hsu SM. Presence of restricted killer immunoglobulin-like receptor repertoire and monoclonal T-cell receptor gamma rearrangement as evidence of mixed NK/T-cell differentiation in a subset of sinonasal lymphomas. J Transl Med 2003; 83:55-64. [PMID: 12533686 DOI: 10.1097/01.lab.0000047491.62596.a3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Most sinonasal lymphomas have a restricted killer immunoglobulin-like receptor (KIR) repertoire without a monoclonal T-cell receptor-gamma (TCR-gamma) rearrangement, implying an NK lineage. However, the lineage assignment of sinonasal lymphoma with a monoclonal TCR-gamma rearrangement is unclear because of its mixed NK/T phenotype. The possibility of a mixed NK/T lineage arises with the discovery of T cells with NK features, such as KIR(+) T cells or Valpha24(+) NKT cells. The former might transform into a T-cell lymphoma with both a monoclonal TCR-gamma rearrangement and a restricted KIR repertoire; the latter might give rise to a T-cell lymphoma with a monoclonal Valpha24 rearrangement and possibly a restricted KIR repertoire. To identify such mixed-lineage lymphomas, we undertook a survey of 15 consecutive sinonasal lymphomas and found six with both a restricted KIR repertoire and a monoclonal TCR-gamma rearrangement, consistent with KIR(+) T-cell lymphomas. Among these six cases, four female CD56(-)/CD44(-)/CD8(-)/CD45RO(+)/CD45RA(-) cases constituted a distinct group with a better prognosis than the rest of the male cases of sinonasal lymphomas. None of the six cases had a monoclonal Valpha24 repertoire, thus excluding a derivation from NKT cells. The predominance of KIR(+) T cells that normally function in chronic viral infections over Valpha24(+) NKT cells that typically recognize glycolipid antigens is consistent with the known association of Epstein-Barr virus infection with sinonasal lymphoma. The demonstration of mixed lineage in a mature lymphoid neoplasm is unusual and echoes the World Health Organization classification that placed NK-cell and T-cell lymphomas in a mixed group.
Collapse
Affiliation(s)
- Chung-Wu Lin
- Department of Pathology, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | | | | | | | | | | |
Collapse
|
25
|
Abstract
Molecules that are essential to natural killer (NK) cell development have been identified mostly through characterizing knock-out mice that exhibit NK deficiencies. Such studies have shown that the interaction of membrane lymphotoxin (LT) on NK cells with its receptor on stromal elements is necessary for inducing a permissive microenvironment for NK development. Also, transcription factors such as Id2, interferon regulatory factors-1 (IRF-1), IRF-2, and Ets-1 are indispensable while PU.1 has a somewhat selective role. In addition, recent studies have identified T/NK progenitors (T/NKPs) in the fetal liver that precede migration to the fetal thymus as well as the earliest committed NK precursors in the bone marrow.
Collapse
Affiliation(s)
- Rebecca H Lian
- Department of Pathology, University of Chicago, 5841 S. Maryland Avenue, Chicago, IL 60637, USA.
| | | |
Collapse
|
26
|
Abstract
Lymphocytes arise from hematopoietic stem cells through the coordinated action of transcription factors. The E proteins (E12, E47, HEB and E2-2) have emerged as key regulators of both B and T lymphocyte differentiation. This review summarizes the current data and examines the various functions of E proteins and their antagonists, Id2 and Id3, throughout lymphoid maturation. Beyond an established role in B and T lineage commitment, E proteins continue to be essential at subsequent stages of development. E protein activity regulates the expression of surrogate and antigen receptor genes, promotes Ig and TCR rearrangements, and coordinates cell survival and proliferation with developmental progression in response to TCR signaling. Finally, this review also discusses the role of E47 as a tumor suppressor.
Collapse
Affiliation(s)
- Melanie W Quong
- Division of Biology, University of California, San Diego, 9500 Gilman Drive, MC 0366, La Jolla, California 92093-0366, USA.
| | | | | |
Collapse
|
27
|
Shimozato O, Ortaldo JR, Komschlies KL, Young HA. Impaired NK cell development in an IFN-gamma transgenic mouse: aberrantly expressed IFN-gamma enhances hematopoietic stem cell apoptosis and affects NK cell differentiation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 168:1746-52. [PMID: 11823506 DOI: 10.4049/jimmunol.168.4.1746] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Aberrant expression of IFN-gamma has been demonstrated to cause a wide variety of alterations in cell function and development. Previously we reported that constitutive expression of IFN-gamma in bone marrow (BM) and thymus results in a total absence of B cells and a substantial decrease in the number of hematopoietic progenitor cells. In this study, we demonstrate a severe deficiency of NK1.1(+)CD3(-) cells in this transgenic mouse model. Compared with normal control littermates, we found a pronounced reduction of NK cells in IFN-gamma transgenic mouse spleen and liver despite maintenance of normal function. In addition, we observed a reduced number of BM cells in the IFN-gamma transgenic mouse despite normal expression of hematopoietic growth factors in the BM. Interestingly, these cells were less responsive to stem cell factor (SCF) despite c-kit expression on hematopoietic stem cells (HSCs). We observed that addition of exogenous IFN-gamma inhibited proliferation of HSCs and differentiation of NK precursors from HSCs in normal mice in response to SCF, IL-7, fms-like tyrosine kinase 3 ligand, and IL-15. Furthermore, we found that HSCs express the IFN-gammaRalpha subunit and undergo apoptosis in response to exogenous IFN-gamma. Thus, we have demonstrated the occurrence of a severe deficiency of NK cells and lower numbers of BM cells in an IFN-gamma transgenic mouse model. Furthermore, because exogenous IFN-gamma affects the responsiveness to hematopoietic growth factors such as SCF in vitro, our results indicate that chronic expression of IFN-gamma in vivo leads to widespread immune system defects, including alterations in NK cell differentiation.
Collapse
Affiliation(s)
- Osamu Shimozato
- Laboratory of Experimental Immunology, Center for Cancer Research, and Intramural Research Support Program, Science Applications International Corp.-Frederick, National Cancer Institute, Frederick, MD 21702, USA
| | | | | | | |
Collapse
|
28
|
Abstract
During development, it is obvious that enormous multiplication and diversification of cells is required to build a body plan from a single fertilized egg and that these two processes, proliferation and differentiation, must be coordinated properly. Id proteins, negative regulators of basic helix-loop-helix transcription factors, possess the ability to inhibit differentiation and to stimulate proliferation, and are useful molecules for investigating the mechanisms regulating development. In the past few years, our understanding of the roles of Id proteins has been substantially enhanced by the detailed investigation of genetically modified animals. The data have indicated that the functions of Id proteins in vivo are functionally related to those revealed by earlier work in cell culture systems. However, unexpected organs and cell types have also been found to require Id proteins for their normal development. This review looks at the advances made in our understanding of the in vivo functions of Id proteins. The topics discussed include neurogenesis, natural killer cell development, lymphoid organogenesis, mammary gland development and spermatogenesis.
Collapse
Affiliation(s)
- Y Yokota
- Department of Biochemistry, Fukui Medical University, Shimoaizuki 23-3, Matsuoka, Fukui 910-1193, Japan.
| |
Collapse
|
29
|
Abstract
The generation of a peripheral T-cell pool is essential for normal immune system function. CD4+ and CD8+ T cells are produced most efficiently in the thymus, which provides a complexity of discrete cellular microenvironments. Specialized stromal cells, that make up such microenvironments, influence each stage in the maturation programme of immature T-cell precursors. Progress has recently been made in elucidating events that regulate the development of intrathymic microenvironments, as well as mechanisms of thymocyte differentiation. It is becoming increasingly clear that the generation and maintenance of thymic environments that are capable of supporting efficient T-cell development, requires complex interplay between lymphoid and stromal compartments of the thymus.
Collapse
Affiliation(s)
- G Anderson
- Department of Anatomy, MRC Centre for Immune Regulation, University of Birmingham, Edgbaston, UK.
| | | |
Collapse
|
30
|
Alonso-C LM, Muñoz JJ, Zapata AG. Delineation of intrathymic T, NK, and dendritic cell (DC) progenitors in fetal and adult rats: demonstration of a bipotent T/DC intermediate precursor. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:3635-41. [PMID: 11564776 DOI: 10.4049/jimmunol.167.7.3635] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We previously published study results stating that the early rat fetal liver contains a high frequency of T/dendritic cells (DCs), but rarely T/NK bipotent common progenitors. Now, by using xenogenic rat/SCID mouse fetal thymic organ cultures, we extend these observations to the thymus, in which conflicting data have been published in human and mouse. On the one hand, enriched adult intrathymic CD45+CD2- triple negative for CD8, CD4, and CD3 Ag cell progenitors, which contained both rearranged TCRbeta chain and pre-Talpha chain transcripts, completely lacked NKR-P1A expressing cells, and upon limiting dilution conditions, generated T- and T/DC-containing lobes, but no T/NK or NK ones were found. On the other hand, the CD45+CD2- triple negative for CD8, CD4, and CD3 Ags cell population obtained from 15- and 16-day-old fetal rat thymus can be divided into NKR-P1A- and NKR-P1A(low) cell subpopulations that differ in several aspects. Both cell subsets expressed pre-TCRalpha chain transcripts, but only the former contained fully rearranged TCRbeta chain transcripts. Upon limiting dilution, T cell-committed progenitors were only found in the NKR-P1A- cell population, whereas NK-committed progenitors were present in the NKR-P1A(low) population. More importantly, bipotential T/NK progenitors were very rare and were found only in the NKR-P1A(low) cell population, whereas bipotential T/DC progenitors, only previously suggested in the adult mouse thymus, were observed frequently in the NKR-P1A-CD2- cell subpopulation. Our results demonstrate, therefore, that a common intrathymic T/DC intermediate represents the main T cell developmental pathway in rat thymus.
Collapse
MESH Headings
- Animals
- Antigens, Surface/analysis
- CD2 Antigens/analysis
- Cell Lineage
- Cells, Cultured
- Dendritic Cells/immunology
- Immunophenotyping
- Killer Cells, Natural/immunology
- Lectins, C-Type
- Leukocyte Common Antigens/analysis
- Mice
- Mice, SCID
- NK Cell Lectin-Like Receptor Subfamily B
- RNA, Messenger/biosynthesis
- Rats
- Rats, Wistar
- Receptors, Antigen, T-Cell, alpha-beta/biosynthesis
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Stem Cells/classification
- Stem Cells/immunology
- T-Lymphocytes/immunology
- Thymus Gland/embryology
- Thymus Gland/immunology
Collapse
Affiliation(s)
- L M Alonso-C
- Servicio Común de Investigación and Departamento de Biología Celular, Faculty of Biology, Complutense University of Madrid, 28040 Madrid, Spain
| | | | | |
Collapse
|
31
|
Yoshida H, Kawamoto H, Santee SM, Hashi H, Honda K, Nishikawa S, Ware CF, Katsura Y, Nishikawa SI. Expression of alpha(4)beta(7) integrin defines a distinct pathway of lymphoid progenitors committed to T cells, fetal intestinal lymphotoxin producer, NK, and dendritic cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:2511-21. [PMID: 11509590 DOI: 10.4049/jimmunol.167.5.2511] [Citation(s) in RCA: 122] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
During embryogenesis, the Peyer's patch anlagen are induced by a cell population that produces lymphotoxin (LT) alpha(1)beta(2) following stimulation of IL-7Ralpha. In this study, we show that the LT-producing cell is localized within the IL-7Ralpha(+) and integrin alpha(4)beta(7) (alpha(4)beta(7))(+) population in the embryonic intestine. Lineage commitment to the LT producer phenotype in the fetal liver coincides with expression of alpha(4)beta(7). Before expression of alpha(4)beta(7), the potential of IL-7Ralpha(+) population to generate B cells is lost. However, the progenitors for T cells and LT producer cells reside in the IL-7Ralpha(+)alpha(4)beta(7)(+) cells, but during subsequent differentiation, the potential to give rise to T cells is lost. This IL-7Ralpha(+)alpha(4)beta(7)(+) population migrates to the intestine, where it induces the Peyer's patch anlagen. When stimulated with IL-15 or IL-3 and TNF, the intestinal IL-7Ralpha(+)alpha(4)beta(7)(+) population can differentiate into fully competent NK1.1(+) NK cells or CD11c(+) APCs. Expression of alpha(4)beta(7) is lost during differentiation of both lineages; IL-7Ralpha expression is lost during NK1.1(+) cells differentiation. A newly discovered lineage(-)IL-7Ralpha(+)c-Kit(+)alpha(4)beta(7)(+) population in the fetal liver is committed to T, NK, dendritic, and fetal intestinal LT producer lineage, the latter being an intermediate stage during differentiation of NK and dendritic cells.
Collapse
Affiliation(s)
- H Yoshida
- Department of Molecular Genetics, Graduate School of Medicine, and Department of Immunology, Institute for Frontier Medical Sciences, Kyoto University, Sakyo, Kyoto, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Dejbakhsh-Jones S, Garcia-Ojeda ME, Chatterjea-Matthes D, Zeng D, Strober S. Clonable progenitors committed to the T lymphocyte lineage in the mouse bone marrow; use of an extrathymic pathway. Proc Natl Acad Sci U S A 2001; 98:7455-60. [PMID: 11390986 PMCID: PMC34690 DOI: 10.1073/pnas.131559798] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2000] [Indexed: 11/18/2022] Open
Abstract
We searched for clonable committed T cell progenitors in the adult mouse bone marrow and isolated rare (approximately 0.05%) cells with the Thy-1hiCD2-CD16+CD44hiCD25-Lin- phenotype. In vivo experiments showed that these cells were progenitors committed only to reconstituting the T cell lineage of irradiated Ly5 congenic hosts. Reconstitution of the thymus was minimal compared with that of the bone marrow, spleen, and lymph nodes. At limiting dilutions, donor T cell reconstitution of the spleen frequently occurred without detectable donor cells in the thymus. Progenitors were capable of rapidly reconstituting athymic hosts. In conclusion, the clonable bone marrow progenitors were capable of T cell reconstitution predominantly by means of an extrathymic pathway.
Collapse
Affiliation(s)
- S Dejbakhsh-Jones
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | | | | | |
Collapse
|
33
|
Rosmaraki EE, Douagi I, Roth C, Colucci F, Cumano A, Di Santo JP. Identification of committed NK cell progenitors in adult murine bone marrow. Eur J Immunol 2001; 31:1900-9. [PMID: 11433387 DOI: 10.1002/1521-4141(200106)31:6<1900::aid-immu1900>3.0.co;2-m] [Citation(s) in RCA: 268] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Natural killer (NK) cells play important roles in innate immunity by lysing tumor and virally infected cells and by producing cytokines including interferon-gamma. While NK cell progenitors have been described in the fetal thymus, NK cell generation from hematopoietic stem cells (HSC) in the bone marrow (BM) occurs throughout life, and in athymic mice and humans. Interleukin (IL)-15 promotes NK development in vitro and is essential for the generation of normal numbers of NK cells in vivo. By characterizing BM cells expressing IL-15 receptor components, we found marked heterogeneity within the IL-2 receptor beta chain(+) (CD122(+)) subset, which included cells uniquely committed to the NK lineage. These CD122(+) NK cell precursors (NKP) are negative for markers used to identify mature NK cells, including NK1.1, DX5 and members of Ly-49 family, and fail to demonstrate natural cytotoxicity against susceptible target cells. In vitro culture of NKP generates mature lytic NK1.1(+) cells at high frequencies, while they do not give rise to T, B, myeloid or erythroid cells under appropriate conditions. NKP lack transcripts associated with early B and T cell differentiation (pTalpha, lambda5 and CD3epsilon), but express a group of genes (IL-15Ralpha, Id2, GATA-3 and Ets-1) and the 2B4 marker, which may define NK cell commitment. We propose that NKP represent the earliest adult BM precursor uniquely restricted to the NK cell lineage.
Collapse
Affiliation(s)
- E E Rosmaraki
- Unité des Cytokines et Développement Lymphoïde, Institut Pasteur, Paris, France
| | | | | | | | | | | |
Collapse
|
34
|
Ikawa T, Fujimoto S, Kawamoto H, Katsura Y, Yokota Y. Commitment to natural killer cells requires the helix-loop-helix inhibitor Id2. Proc Natl Acad Sci U S A 2001; 98:5164-9. [PMID: 11296270 PMCID: PMC33181 DOI: 10.1073/pnas.091537598] [Citation(s) in RCA: 130] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
We have previously described how T and natural killer (NK) lineage commitment proceeds from common T/NK progenitors (p-T/NK) in the murine fetal thymus (FT), with the use of a clonal assay system capable of discriminating p-T/NK from unipotent T or NK lineage-committed progenitors (p-T and p-NK, respectively). The molecular mechanisms controlling the commitment processes, however, are yet to be defined. In this study, we investigated the progenitor activity of FT cells from Id2-/- mice that exhibit defective NK cell development. In the Id2-/- FT, NK cells were greatly reduced, and a cell population that exclusively contains p-NK in the wild-type thymus was completely missing. Id2-/- FT progenitors were unable to differentiate into NK cells in IL-2-supplemented-FT organ culture. Single progenitor analysis demonstrated that all Id2-/- fetal thymic progenitors are destined for the T cell lineage, whereas progenitors for T/NK, T, and NK cell lineages were found in the control. Interestingly, the total progenitor number was similar between Id2-/- and Id2+/+ embryos analyzed. Expression of Id2 was correlated with p-NK activity. Our results suggest that Id2 is indispensable in thymic NK cell development, where it most probably restricts bipotent T/NK progenitors to the NK cell lineage.
Collapse
MESH Headings
- Animals
- Cell Differentiation/drug effects
- Cell Lineage/drug effects
- Cells, Cultured
- DNA-Binding Proteins/chemistry
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Flow Cytometry
- Gene Deletion
- Helix-Loop-Helix Motifs
- Hyaluronan Receptors/metabolism
- Inhibitor of Differentiation Protein 2
- Interleukin-2/pharmacology
- Killer Cells, Natural/cytology
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/metabolism
- Lymphocyte Count
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Models, Immunological
- Organ Culture Techniques
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Interleukin-2/analysis
- Receptors, Interleukin-2/metabolism
- Repressor Proteins
- Reverse Transcriptase Polymerase Chain Reaction
- Stem Cells/cytology
- Stem Cells/drug effects
- Stem Cells/metabolism
- Thymus Gland/cytology
- Thymus Gland/drug effects
- Thymus Gland/embryology
- Thymus Gland/metabolism
- Transcription Factors
Collapse
Affiliation(s)
- T Ikawa
- Department of Immunology, Institute for Frontier Medical Sciences, Kyoto University, Shogoin Kawahara-cho 53, Sakyo-ku, 606-8507 Kyoto, Japan
| | | | | | | | | |
Collapse
|
35
|
Trop S, De Sepulveda P, Zúñiga-Pflücker JC, Rottapel R. Overexpression of suppressor of cytokine signaling-1 impairs pre-T-cell receptor-induced proliferation but not differentiation of immature thymocytes. Blood 2001; 97:2269-77. [PMID: 11290587 DOI: 10.1182/blood.v97.8.2269] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Cytokines play an essential role during early T-cell development. However, the mechanisms controlling cytokine signaling in developing thymocytes have not been elucidated. Cytokine receptor signaling can be modulated by suppressor of cytokine signaling-1 (SOCS-1), which acts as a negative regulator of Janus kinases. SOCS-1 is normally expressed throughout thymocyte development; however, retroviral-mediated overexpression of SOCS-1 in fetal liver-derived hematopoietic progenitors prevented their progression beyond the earliest stage of T-cell development. Further analysis revealed that SOCS-1 expression is transiently suppressed following pre-T-cell receptor (TCR) signaling. Moreover, constitutive expression of SOCS-1 abrogated pre-TCR- mediated expansion of immature thymocytes but did not interfere with differentiation. These findings reveal that SOCS-1 serves to regulate cytokine signaling at critical checkpoints during early T-cell development.
Collapse
Affiliation(s)
- S Trop
- Departments of Immunology, Medicine, and Medical Biophysics, University of Toronto, Ontario, Canada
| | | | | | | |
Collapse
|
36
|
Iwabuchi K, Iwabuchi C, Tone S, Itoh D, Tosa N, Negishi I, Ogasawara K, Uede T, Onoé K. Defective development of NK1.1+ T-cell antigen receptor alphabeta+ cells in zeta-associated protein 70 null mice with an accumulation of NK1.1+ CD3- NK-like cells in the thymus. Blood 2001; 97:1765-75. [PMID: 11238119 DOI: 10.1182/blood.v97.6.1765] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Development of natural killer 1.1+ (NK1.1+) CD3+ (NK1.1+ T) cells was analyzed in zeta-associated protein 70 (ZAP-70) null ((-/-)) mice. Both NK1.1+ TCRalphabeta+ and NK1.1+ TCRgammadelta+ cell populations were absent in the thymus and spleen. By contrast, the number of NK1.1+ CD3- cells was increased in these tissues. The NK1.1+ CD3- thymocytes in ZAP-70(-/-) mice had surface phenotypes in common with NK or NK1.1+ T cells. However, some of them were discordant either with NK cells or with NK1.1+ T cells. The NK1.1+ CD3- cells produced interferon-gamma upon stimulation with NK1.1 cross-linking in the presence of interleukin-2 and exhibited a substantial cytotoxicity against YAC-1 cells. Moreover, the generation of NK1.1+ T cells with invariant Valpha14Jalpha281 chains was induced from the NK1.1+ CD3- thymocytes following stimulation with phorbol myristate acetate and ionomycin in a neonatal thymic organ culture. An introduction of TCRalpha and beta transgenes to the ZAP-70(-/-) mice resulted in generation of an NK1.1+ TCRalphabeta(dim) population, whereas no substantial CD4+ CD8- or CD4- CD8+ population that expressed the introduced TCRalphabeta was generated in the mainstream T lineage. These findings demonstrate that ZAP-70 kinase is indispensable for the development of NK1.1+ T cells and that the unique NK1.1+ CD3- thymocytes in ZAP-70(-/-) mice contain immediate precursors of NK1.1+ T cells.
Collapse
MESH Headings
- Animals
- Antigens/metabolism
- Antigens, Ly
- Antigens, Surface
- CD3 Complex/genetics
- CD3 Complex/metabolism
- Cell Differentiation/drug effects
- Gene Rearrangement/drug effects
- Immunophenotyping
- Ionomycin/pharmacology
- Killer Cells, Natural/cytology
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/immunology
- Lectins, C-Type
- Mice
- Mice, Knockout
- Mice, Transgenic
- NK Cell Lectin-Like Receptor Subfamily B
- Protein-Tyrosine Kinases/genetics
- Protein-Tyrosine Kinases/pharmacology
- Protein-Tyrosine Kinases/physiology
- Proteins/metabolism
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Spleen/cytology
- Spleen/immunology
- T-Lymphocytes/cytology
- T-Lymphocytes/drug effects
- T-Lymphocytes/immunology
- Tetradecanoylphorbol Acetate/pharmacology
- Thymus Gland/cytology
- Thymus Gland/immunology
- ZAP-70 Protein-Tyrosine Kinase
Collapse
Affiliation(s)
- K Iwabuchi
- Division of Immunobiology and Molecular Immunology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
Natural killer (NK) cells are cytotoxic cells that play a critical role in the innate immune response against infections and tumors. Recent studies on NK cell biology have demonstrated that besides their cytotoxic function, NK cells express cytokine and chemokine receptors and also that they secrete other immunoregulatory cytokines and chemokines, supporting their relevance in the regulation of the immune response by promoting downstream adaptive, Th1 mediated, responses against infections. Immunosenescence is the deterioration of the immune response associated with aging. It is characterized mainly by a defective T cell response, but includes changes in the number and function of other cells of the innate immune system. Age-associated alterations in the number and function of NK cells have been reported. There is a general consensus that a progressive increase in the percentage of NK cells with a mature phenotype occurs in elderly donors associated with an impairment of their cytotoxic capacity when considered on a "per cell" basis. The response of NK cells from elderly individuals to IL-2 or other cytokines is also decreased in terms of proliferation, expression of CD69 and killing of NK-resistant cell lines. Furthermore early IFN-gamma and chemokine production in response to IL-2 or IL-12 is also decreased. However aging does not significantly alter other NK cell functions such as TNF-alpha production or perforin induction in response to IL-2. The percentage of T cells that co-express NK cell markers is also increased in aging. These results indicate that the increase in the number of "classical" mature NK and NK/T cells in aging is associated with a defective functional capacity of NK cells. Low NK cell number or function in elderly individuals is associated with increased mortality risk and increased incidence of severe infections, supporting the role of NK cells in the defense against infections in the elderly.
Collapse
Affiliation(s)
- R Solana
- Department of Immunology, Faculty of Medicine, "Reina Sofia" University Hospital, University of Córdoba, Av. Menendez Pidal s/n, E-14004, Córdoba, Spain.
| | | |
Collapse
|
38
|
Ikawa T, Kawamoto H, Fujimoto S, Katsura Y. Commitment of common T/Natural killer (NK) progenitors to unipotent T and NK progenitors in the murine fetal thymus revealed by a single progenitor assay. J Exp Med 1999; 190:1617-26. [PMID: 10587352 PMCID: PMC2195728 DOI: 10.1084/jem.190.11.1617] [Citation(s) in RCA: 130] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
We have established a new clonal assay system that can evenly support the development of T and natural killer (NK) cells. With this system, we show that all T cell progenitors in the earliest CD44(+)CD25(-)FcgammaRII/III(-) fetal thymus (FT) cell population retain NK potential, and that the NK lineage-committed progenitors (p-NK) also exist in this population. T cell lineage-committed progenitors (p-T), which are unable to generate NK cells, first appear at the CD44(+)CD25(-) FcgammaRII/III(+) stage in day 12 FT. The proportion of p-T markedly increases during the transition from the CD44(+)CD25(-) stage to the CD44(+)CD25(+) stage in day 14 FT. On the other hand, p-NK preferentially increase in number at the CD44(+)CD25(-) stage between days 12 and 14 of gestation. The production of p-NK continues up to the CD44(+)CD25(+) stage, but ceases before the rearrangement of T cell receptor beta chain genes. It was further shown that the CD44(+)CD25(-) CD122(+) population of day 14 FT exclusively contains p-NK. These results indicate that the earliest T cell progenitor migrating into the FT is T/NK bipotent, and strongly suggest that the bipotent progenitor continuously produces p-NK and p-T until the CD44(+)CD25(+) stage.
Collapse
Affiliation(s)
- Tomokatsu Ikawa
- Department of Immunology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Hiroshi Kawamoto
- Department of Immunology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Shinji Fujimoto
- Department of Immunology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Yoshimoto Katsura
- Department of Immunology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| |
Collapse
|
39
|
Spain LM, Guerriero A, Kunjibettu S, Scott EW. T Cell Development in PU.1-Deficient Mice. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.163.5.2681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
These studies address the role of PU.1 in T cell development through the analysis of PU.1−/− mice. We show that the majority of PU.1−/− thymocytes are blocked in differentiation prior to T cell commitment, and contain a population of thymocyte progenitors with the cell surface phenotype of CD44+, HSAbright, c-kitint, Thy-1−, CD25−, Sca-1−, CD4−, and CD8−. These cells correspond in both number and cell surface phenotype with uncommitted thymocyte progenitors found in wild-type fetal thymus. RT-PCR analysis demonstrated that PU.1 is normally expressed in this early progenitor population, but is down-regulated during T cell commitment. Rare PU.1−/− thymi, however, contained small numbers of thymocytes expressing markers of T cell commitment. Furthermore, almost 40% of PU.1−/− thymi placed in fetal thymic organ culture are capable of T cell development. Mature PU.1−/− thymocytes generated during organ culture proliferated and produced IL-2 in response to stimulation through the TCR. These data demonstrate that PU.1 is not absolutely required for T cell development, but does play a role in efficient commitment and/or early differentiation of most T progenitors.
Collapse
Affiliation(s)
| | - Anastasia Guerriero
- †Institute for Human Gene Therapy, University of Pennsylvania Medical School, Philadelphia, PA 19104
| | | | - Edward W. Scott
- †Institute for Human Gene Therapy, University of Pennsylvania Medical School, Philadelphia, PA 19104
| |
Collapse
|
40
|
Abstract
Very small placentae and absence of uterine natural killer (uNK) cells are amongst the reproductive deficits found in the natural killer (NK) cell and thymus-derived (T) cell immunodeficient mouse tgepsilon26. These defects can be reversed by grafting of adult tgepsilon26 females with bone marrow from T and B cell immunodeficient scid/scid donors. We report here that a second protocol, grafting of neonatal tgepsilon26 females with immunocompetent bone marrow pretreated with antibody to Thy-1, successfully established the uNK cell lineage and ameliorated the phenotype. Further, comparisons of mid-gestation (days 10-16) placental area measurements from tgepsilon26 and seven other immunodeficient strains to time-matched tissues from four strains of immunocompetent mice indicate that lymphocytes of the NK but not the T or B cell lineages are able to influence placental size during normal gestation and that this action is independent of interleukin 2. Area measurements of placentae produced in manipulated tgepsilon26 pregnancies (maternal bone marrow engraftment, outcrossing to immunocompetent males and reciprocal embryo transfers with an immunocompetent strain) suggest that NK cell competence is required in each of the maternal and fetal compartments to optimize placental growth.
Collapse
Affiliation(s)
- M Guimond
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ontario, N1G 2W1, Canada.
| | | | | |
Collapse
|
41
|
Carlyle JR, Martin A, Mehra A, Attisano L, Tsui FW, Zúñiga-Pflücker JC. Mouse NKR-P1B, a Novel NK1.1 Antigen with Inhibitory Function. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.162.10.5917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
The mouse NK1.1 Ag originally defined as NK cell receptor (NKR)-P1C (CD161) mediates NK cell activation. Here, we show that another member of the mouse CD161 family, NKR-P1B, represents a novel NK1.1 Ag. In contrast to NKR-P1C, which functions as an activating receptor, NKR-P1B inhibits NK cell activation. Association of NKR-P1B with Src homology 2-containing protein tyrosine phosphatase-1 provides a molecular mechanism for this inhibition. The existence of these two NK1.1 Ags with opposite functions suggests a potential role for NKR-P1 molecules, such as those of the Ly-49 gene family, in regulating NK cell function.
Collapse
Affiliation(s)
| | | | - Arun Mehra
- †Anatomy and Cell Biology, University of Toronto, Toronto, Ontario, Canada
| | - Liliana Attisano
- †Anatomy and Cell Biology, University of Toronto, Toronto, Ontario, Canada
| | | | | |
Collapse
|
42
|
Carlyle JR, Zúñiga-Pflücker JC. Regulation of NK1.1 Expression During Lineage Commitment of Progenitor Thymocytes. THE JOURNAL OF IMMUNOLOGY 1998. [DOI: 10.4049/jimmunol.161.12.6544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Abstract
We recently identified a stage in fetal ontogeny (NK1.1+/CD117+) that defines committed progenitors for T and NK lymphocytes. These cells are found in the fetal thymus as early as day 13 of gestation, but are absent in the fetal liver. Nonetheless, multipotent precursors derived from both the fetal thymus and fetal liver are capable of rapidly differentiating to the NK1.1+ stage upon transfer into fetal thymic organ culture (FTOC). This suggests that expression of NK1.1 marks a thymus-induced lineage commitment event. We now report that a subset of the most immature fetal thymocytes (NK1.1−/CD117+) is capable of up-regulating NK1.1 expression spontaneously upon short-term in vitro culture. Interestingly, fetal liver-derived CD117+ precursors remain NK1.1− upon similar culture. Spontaneous up-regulation of NK1.1 surface expression is minimally affected by transcriptional blockade, mitogen-induced activation, or exposure of these cells to exogenous cytokines or stromal cells. These data suggest that induction of NK1.1 expression on cultured thymocytes may be predetermined by exposure to the thymic microenvironment in vivo. Importantly, multipotent CD117+ thymocytes subdivided on the basis of NK1.1 expression after short-term in vitro culture show distinct precursor potential in lymphocyte lineage reconstitution assays. This demonstrates that even the earliest precursor thymocyte population, although phenotypically homogeneous, contains a functionally heterogeneous subset of lineage-committed progenitors. These findings characterize a thymus-induced pathway in the control of lymphocyte lineage commitment to the T and NK cell fates.
Collapse
Affiliation(s)
- James R. Carlyle
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | | |
Collapse
|