1
|
Ross JB, Myers LM, Noh JJ, Collins MM, Carmody AB, Messer RJ, Dhuey E, Hasenkrug KJ, Weissman IL. Depleting myeloid-biased haematopoietic stem cells rejuvenates aged immunity. Nature 2024; 628:162-170. [PMID: 38538791 PMCID: PMC11870232 DOI: 10.1038/s41586-024-07238-x] [Citation(s) in RCA: 62] [Impact Index Per Article: 62.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 02/26/2024] [Indexed: 04/01/2024]
Abstract
Ageing of the immune system is characterized by decreased lymphopoiesis and adaptive immunity, and increased inflammation and myeloid pathologies1,2. Age-related changes in populations of self-renewing haematopoietic stem cells (HSCs) are thought to underlie these phenomena3. During youth, HSCs with balanced output of lymphoid and myeloid cells (bal-HSCs) predominate over HSCs with myeloid-biased output (my-HSCs), thereby promoting the lymphopoiesis required for initiating adaptive immune responses, while limiting the production of myeloid cells, which can be pro-inflammatory4. Ageing is associated with increased proportions of my-HSCs, resulting in decreased lymphopoiesis and increased myelopoiesis3,5,6. Transfer of bal-HSCs results in abundant lymphoid and myeloid cells, a stable phenotype that is retained after secondary transfer; my-HSCs also retain their patterns of production after secondary transfer5. The origin and potential interconversion of these two subsets is still unclear. If they are separate subsets postnatally, it might be possible to reverse the ageing phenotype by eliminating my-HSCs in aged mice. Here we demonstrate that antibody-mediated depletion of my-HSCs in aged mice restores characteristic features of a more youthful immune system, including increasing common lymphocyte progenitors, naive T cells and B cells, while decreasing age-related markers of immune decline. Depletion of my-HSCs in aged mice improves primary and secondary adaptive immune responses to viral infection. These findings may have relevance to the understanding and intervention of diseases exacerbated or caused by dominance of the haematopoietic system by my-HSCs.
Collapse
Affiliation(s)
- Jason B Ross
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Ludwig Center for Cancer Stem Cell Research and Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Lara M Myers
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Joseph J Noh
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Ludwig Center for Cancer Stem Cell Research and Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Madison M Collins
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
- Department of Biological and Physical Sciences, Montana State University Billings, Billings, MT, USA
| | - Aaron B Carmody
- Research Technologies Branch, Rocky Mountain Laboratories, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Ronald J Messer
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Erica Dhuey
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Ludwig Center for Cancer Stem Cell Research and Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Kim J Hasenkrug
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA.
| | - Irving L Weissman
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA.
- Ludwig Center for Cancer Stem Cell Research and Medicine, Stanford University School of Medicine, Stanford, CA, USA.
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
2
|
Zhang M, Lin X, Yang Z, Li X, Zhou Z, Love PE, Huang J, Zhao B. Metabolic regulation of T cell development. Front Immunol 2022; 13:946119. [PMID: 35958585 PMCID: PMC9357944 DOI: 10.3389/fimmu.2022.946119] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 06/24/2022] [Indexed: 11/24/2022] Open
Abstract
T cell development in the thymus is tightly controlled by complex regulatory mechanisms at multiple checkpoints. Currently, many studies have focused on the transcriptional and posttranslational control of the intrathymic journey of T-cell precursors. However, over the last few years, compelling evidence has highlighted cell metabolism as a critical regulator in this process. Different thymocyte subsets are directed by distinct metabolic pathways and signaling networks to match the specific functional requirements of the stage. Here, we epitomize these metabolic alterations during the development of a T cell and review several recent works that provide insights into equilibrating metabolic quiescence and activation programs. Ultimately, understanding the interplay between cellular metabolism and T cell developmental programs may offer an opportunity to selectively regulate T cell subset functions and to provide potential novel therapeutic approaches to modulate autoimmunity.
Collapse
Affiliation(s)
- Mengdi Zhang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xiaoxi Lin
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Zhou Yang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xia Li
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Zhiguang Zhou
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Paul E. Love
- Section on Hematopoiesis and Lymphocyte Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Jiaqi Huang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
- *Correspondence: Bin Zhao, ; ; Jiaqi Huang, ;;
| | - Bin Zhao
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
- *Correspondence: Bin Zhao, ; ; Jiaqi Huang, ;;
| |
Collapse
|
3
|
Mizuno S, Iino T, Ozawa H, Arinobu Y, Chong Y, Akashi K. Notch1 expression is regulated at the post-transcriptional level by the 3' untranslated region in hematopoietic stem cell development. Int J Hematol 2017; 107:311-319. [PMID: 29098541 DOI: 10.1007/s12185-017-2358-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 10/10/2017] [Accepted: 10/11/2017] [Indexed: 01/21/2023]
Abstract
In hematopoiesis, the expression of critical genes is regulated in a stage-specific manner to maintain normal hematopoiesis. Notch1 is an essential gene involved in the commitment and development of the T-cell lineage. However, the regulation of Notch1 in hematopoiesis is controversial, particularly at the level of hematopoietic stem cell (HSC). Here, we found that the expression of Notch1 is controlled at the post-transcriptional level in HSCs. HSCs express a considerable level of Notch1 mRNA, but its protein level is very low, suggesting a post-transcriptional suppression for Notch1. Using a retroviral sensor vector expressing a fusion mRNA of GFP and 3' untranslated region (3'UTR) of a target gene, we demonstrated that the Notch1-3'UTR had a post-translational suppressive effect only at the HSC but not in the downstream progenitor stages. The sequence motif AUnA was required for this post-transcriptional regulation by the Notch1-3'UTR. Interestingly, this Notch1-3'UTR-mediated suppressive effect was relieved when HSCs were placed in the thymus, but not in the bone marrow. Thus, the expression of Notch1 in HSCs is regulated by microenvironment at the post-transcriptional level, which may control T lymphoid lineage commitment from HSCs.
Collapse
Affiliation(s)
- Shinichi Mizuno
- Center for Advanced Medical Innovation, Kyushu University, Fukuoka, Japan.,Department of Cancer Immunology and AIDs, Dana-Farber Cancer Institute, Boston, USA
| | - Tadafumi Iino
- Center for Advanced Medical Innovation, Kyushu University, Fukuoka, Japan.,Department of Cancer Immunology and AIDs, Dana-Farber Cancer Institute, Boston, USA
| | - Hidetoshi Ozawa
- Division of Hematology and Oncology, Department of Medicine, Kurume University School of Medicine, Fukuoka, Japan.,Department of Cancer Immunology and AIDs, Dana-Farber Cancer Institute, Boston, USA
| | - Yojiro Arinobu
- Center for Cellular and Molecular Medicine, Kyushu University Hospital, Fukuoka, Japan.,Department of Cancer Immunology and AIDs, Dana-Farber Cancer Institute, Boston, USA
| | - Yong Chong
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.,Department of Cancer Immunology and AIDs, Dana-Farber Cancer Institute, Boston, USA
| | - Koichi Akashi
- Center for Advanced Medical Innovation, Kyushu University, Fukuoka, Japan. .,Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan. .,Center for Cellular and Molecular Medicine, Kyushu University Hospital, Fukuoka, Japan. .,Department of Cancer Immunology and AIDs, Dana-Farber Cancer Institute, Boston, USA.
| |
Collapse
|
4
|
Abstract
I started research in high school, experimenting on immunological tolerance to transplantation antigens. This led to studies of the thymus as the site of maturation of T cells, which led to the discovery, isolation, and clinical transplantation of purified hematopoietic stem cells (HSCs). The induction of immune tolerance with HSCs has led to isolation of other tissue-specific stem cells for regenerative medicine. Our studies of circulating competing germline stem cells in colonial protochordates led us to document competing HSCs. In human acute myelogenous leukemia we showed that all preleukemic mutations occur in HSCs, and determined their order; the final mutations occur in a multipotent progenitor derived from the preleukemic HSC clone. With these, we discovered that CD47 is an upregulated gene in all human cancers and is a "don't eat me" signal; blocking it with antibodies leads to cancer cell phagocytosis. CD47 is the first known gene common to all cancers and is a target for cancer immunotherapy.
Collapse
Affiliation(s)
- Irving Weissman
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305, and Ludwig Center for Cancer Stem Cell Research and Medicine at Stanford, Stanford, CA 94305
| |
Collapse
|
5
|
Fisher IB, Ostrowski M, Muthusamy N. Role for Ets-2(Thr-72) transcription factor in stage-specific thymocyte development and survival. J Biol Chem 2011; 287:5199-210. [PMID: 22128184 DOI: 10.1074/jbc.m111.242602] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Interference of Ras signaling deregulates thymocyte development in mouse models. However, the role of Ets-2, a transcription factor that is phosphorylated on a critical threonine residue (Thr-72) by the Ras/MAPK pathway in thymocyte development, has not been defined. Transgenic mice overexpressing a phosphomutant Ets-2 (T72A) in the thymus displayed reduced thymus size associated with a 60-80% reduction in thymocyte populations. The transgenic mice exhibited a 20-fold increase in a c-Kit(+) CD4(+) CD8(+) CD3(-) population and a 5-fold increase in a unique CD5(low) population associated with a partial developmental block at the DN2-DN3 stage of thymocytes. Transgenic thymocytes exhibited increased apoptosis, and overexpression of Bcl-2 rescued the hypocellularity and associated thymocyte developmental block in double transgenic mice. The observed defects in these mice are not dependent on Ets-1 expression. These studies implicate for the first time a stage-specific Ets-1-independent regulatory role for Ets-2 in early thymocyte development and survival.
Collapse
Affiliation(s)
- Ian B Fisher
- Molecular Cellular Developmental Biology Program, Division of Hematology, Ohio State University, Columbus, Ohio 43210, USA
| | | | | |
Collapse
|
6
|
Affiliation(s)
- Irving L Weissman
- Stanford Institute of Stem Cell Biology and Regenerative Medicine, Stanford, CA, USA.
| |
Collapse
|
7
|
Combined Effects of Interleukin-7 and Stem Cell Factor Administration on Lymphopoiesis after Murine Bone Marrow Transplantation. Biol Blood Marrow Transplant 2011; 17:48-60. [DOI: 10.1016/j.bbmt.2010.07.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2010] [Accepted: 07/29/2010] [Indexed: 11/18/2022]
|
8
|
Dervović D, Zúñiga-Pflücker JC. Positive selection of T cells, an in vitro view. Semin Immunol 2010; 22:276-86. [DOI: 10.1016/j.smim.2010.04.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2010] [Accepted: 04/23/2010] [Indexed: 12/16/2022]
|
9
|
Serwold T, Hochedlinger K, Inlay MA, Jaenisch R, Weissman IL. Early TCR expression and aberrant T cell development in mice with endogenous prerearranged T cell receptor genes. THE JOURNAL OF IMMUNOLOGY 2007; 179:928-38. [PMID: 17617584 DOI: 10.4049/jimmunol.179.2.928] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The factors that regulate the rate of production of T cells by the thymus remain incompletely defined. To test whether generation of functional T cell receptors limits the rate of thymic T cell export, we made use of a line of mice, LN3alphabeta, that have endogenously prerearranged TCR genes. The prerearranged TCR genes were expressed abnormally early in hemopoietic development, indicating that RAG-mediated recombination, rather than transcription factor expression, is the key determinant of the initiation of robust TCR transcription. Thymic T cell export rates were similar between wild-type (wt) and LN3alphabeta mice, indicating that T cell maturation rates in these mice are determined by factors other than TCR gene rearrangement. In competitive bone marrow chimeras, however, LN3alphabeta thymocytes were out-competed by wt cells and failed to develop beyond the double-negative 4 stage. Furthermore, wt progenitors transplanted intrathymically into LN3alphabeta mice proliferated excessively, suggesting that increased proliferative signals in the LN3alphabeta thymus compensate for faulty T cell development driven by early TCR expression.
Collapse
MESH Headings
- Animals
- Cell Differentiation/immunology
- Flow Cytometry
- Gene Rearrangement, T-Lymphocyte/genetics
- Gene Rearrangement, T-Lymphocyte/immunology
- Genes, RAG-1
- Mice
- Mice, Mutant Strains
- Nuclear Transfer Techniques
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- T-Lymphocytes/cytology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Thymus Gland/cytology
- Thymus Gland/growth & development
- Thymus Gland/immunology
Collapse
Affiliation(s)
- Thomas Serwold
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | | | | | | | | |
Collapse
|
10
|
Abstract
B cell genesis declines with age, but at what stage and why remains unclear. Previous studies attribute the decline in B cell production in aged mice to both environmental and cell-intrinsic defects that impact mid-to-late stream B cell precursors. However, mounting evidence suggests that the aging process may also negatively affect the earliest phases of B cell development. We review past studies on the B cells and aging question, discuss recent data suggesting that age-associated defects in B cell development reflect deficiencies in hematopoietic stem cell-proximal progenitor pools, and provide an integrative model that will hopefully facilitate further studies into this complex problem.
Collapse
Affiliation(s)
- Juli P Miller
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, 269 John Morgan Building, 36th and Hamilton Walk, Philadelphia, PA 19104, USA
| | | |
Collapse
|
11
|
El Kassar N, Lucas PJ, Klug DB, Zamisch M, Merchant M, Bare CV, Choudhury B, Sharrow SO, Richie E, Mackall CL, Gress RE. A dose effect of IL-7 on thymocyte development. Blood 2004; 104:1419-27. [PMID: 15155461 DOI: 10.1182/blood-2004-01-0201] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
To study interleukin-7 (IL-7) in early thymocyte development, we generated mice transgenic (Tg) for the IL-7 gene under control of the lck proximal promoter. Founder line TgA, with the lowest level of IL-7 overexpression, showed enhanced alphabeta T-cell development. In contrast, in the highest overexpressing founder line, TgB, alphabeta T-cell development was disturbed with a block at the earliest intrathymic precursor stage. This was due to decreased progenitor proliferation as assessed by Ki-67 staining and in vivo bromodeoxyuridine (BrdU) incorporation. Bcl-2 was up-regulated in T-cell-committed progenitors in all Tg lines, and accounted for greater numbers of double positive (DP), CD4 single positive (SP), and CD8SP thymocytes in TgA mice where, in contrast to TgB mice, thymocyte progenitor proliferation was normal. Mixed marrow chimeras using TgB(+) and congenic mice as donors, and experiments using anti-IL-7 monoclonal antibody (MAb) in vivo, confirmed the role of IL-7 protein in the observed TgB phenotype. In conclusion, at low Tg overexpression, IL-7 enhanced alphabeta T-cell development by increasing thymocyte progenitor survival, while at high overexpression IL-7 reduces their proliferation, inducing a dramatic block in DP production. These results show for the first time in vivo a dose effect of IL-7 on alphabeta T-cell development and have implications for IL-7 in the clinical setting.
Collapse
MESH Headings
- Animals
- B-Lymphocytes/cytology
- B-Lymphocytes/physiology
- Cell Division/immunology
- Gene Expression Regulation, Developmental/immunology
- Interleukin-7/genetics
- Killer Cells, Natural/cytology
- Killer Cells, Natural/physiology
- Leukocyte Common Antigens/metabolism
- Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Promoter Regions, Genetic
- Proto-Oncogene Proteins c-bcl-2/metabolism
- RNA, Messenger/metabolism
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Stem Cells/cytology
- Stem Cells/physiology
- T-Lymphocytes/cytology
- T-Lymphocytes/physiology
- Thymus Gland/cytology
- Thymus Gland/embryology
- Thymus Gland/physiology
- Up-Regulation/immunology
Collapse
Affiliation(s)
- Nahed El Kassar
- Experimental Immunology Branch, National Institutes of Health, 10 Center Drive, Bldg 10 Rm 4B36, Bethesda, MD 20892-1360, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Xiao SY, Li Y, Chen WF. Kinetics of thymocyte developmental process in fetal and neonatal mice. Cell Res 2003; 13:265-73. [PMID: 12974616 DOI: 10.1038/sj.cr.7290171] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Kinetics of thymocyte development in vivo during embryogenesis was pursued. The early development of thymocytes in the fetal and neonatal BALB/c mice was discontinuous, with four waves of cell proliferation occurring at fetal day (Fd) 14 to 17, Fd 18 to day (D) 1 after birth, D 2 to D 5 and D6 thereafter. The first three proliferation waves coincided with the generation of CD4hiCD8hi (DP), TCR+CD4hiCD8-/lo (CD4 SP), and TCR+CD4-/loCD8int/hi (CD8 SP) thymocytes, respectively. The transition from DN to DP cells was further investigated and it was found out that there were two differential pathways via immature single positive (ISP) cells in the BALB/c mice, each functioning at different fetal ages. One is via TCR-CD4-CD8+ cells, occurring between Fd 15 and Fd 17 and the other is via TCR-CD4+CD8- cells, occurring from Fd 17 until birth. In contrast, the TCR-CD4-CD8+ pathway dominated overwhelmingly in the C57BL/6 mice. These findings shed new light on the hypothesis that the differential pathway preference varies with mouse strains. With respect to the shift in the intensity of CD4 and CD8 expression on thymocytes from fetal to adult mice, the TCR+CD4hiCD8-/lo, and TCR+CD4-/loCD8int/hi subsets might be equivalent to the medullary type TCR+CD4/CD8 SP cells.
Collapse
Affiliation(s)
- Shi Yun Xiao
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Beijing 100083, China
| | | | | |
Collapse
|
13
|
Domen J, Weissman IL. Hematopoietic stem cells and other hematopoietic cells show broad resistance to chemotherapeutic agents in vivo when overexpressing bcl-2. Exp Hematol 2003; 31:631-9. [PMID: 12842708 DOI: 10.1016/s0301-472x(03)00084-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Objective. Chemotherapeutic agents function by inducing apoptosis and their effectiveness depends on the balance of pro- and anti-apoptotic proteins in cells. Due to the complicated interactions of the many proteins involved, it has been difficult to determine in tumors whether overexpression of single genes is prognostic for increased resistance. Therefore, we studied the influence of bcl-2 overexpression on resistance to chemotherapeutics in a transgenic mouse system. This allowed us to study a wide variety of cells, including important but rare populations such as hematopoietic stem cells (HSC).Methods. H2K-bcl-2 transgenic and wild-type (WT) mice were treated with several agents(5-fluoruracil, cyclophosphamide, and busulfan) to determine the contribution of increased amounts of bcl-2 to the response to these chemotherapeutics in vivo. Populations were enumerated using flow cytometry. HSC were studied by FACS purification and long-term reconstitution assays in vivo and resistance was confirmed by short-term proliferation assays with different amounts of chemotherapeutics in vitro.Results. bcl-2 overexpression alone protects many cell types, though protection levels differ between populations and agents. However, even sensitive populations return to pretreatment levels faster in transgenic mice. bcl-2 overexpression also prevents the dramatic changes in HSC following 5-FU treatment (downregulation of c-kit, upregulation of Lin, less efficient long-term reconstitution). In vitro studies directly demonstrate increased resistance of bcl-2 overexpressing HSC to chemotherapeutic agents.Conclusions. Increased expression of bcl-2 in HSC and their progeny endows these cells with broad resistance to chemotherapeutic agents. The ability to (differentially) regulate sensitivity to apoptosis of bystander and tumor cells is clinically important.
Collapse
Affiliation(s)
- Jos Domen
- Department of Medicine and Immunology, Duke University Medical Center, Durham, NC 27710, USA.
| | | |
Collapse
|
14
|
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: 3.5] [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
|
15
|
Prockop SE, Palencia S, Ryan CM, Gordon K, Gray D, Petrie HT. Stromal cells provide the matrix for migration of early lymphoid progenitors through the thymic cortex. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 169:4354-61. [PMID: 12370368 DOI: 10.4049/jimmunol.169.8.4354] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
During steady state lymphopoiesis in the postnatal thymus, migration of precursors outward from the deep cortex toward the capsule is required for normal differentiation. Such migration requires, at a minimum, expression of adhesive receptors on the migrating lymphoid cells, as well as a stable matrix of their ligands persisting throughout the region of migration. In this study, we address the nature of this adhesive matrix. Although some precursor stages bound efficiently to extracellular matrix ligands, a specific requirement for the cell surface ligand VCAM-1 was also found. In situ analysis revealed that early precursors are found in intimate contact with a matrix formed by stromal cells in the cortex, a proportion of which expresses VCAM-1. In vivo administration of an anti-VCAM-1 Ab resulted in decreased thymic size and altered distribution of early precursors within the cortex. These results indicate that precursors migrating outward through the cortex may use a cellular, rather than extracellular, matrix for adhesion, and suggest that the VCAM-1(+) subset of cortical stroma may play a crucial role in supporting the migration of early precursors in the steady state thymus.
Collapse
Affiliation(s)
- Susan E Prockop
- Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10021, USA
| | | | | | | | | | | |
Collapse
|
16
|
Abstract
For me the search for hematopoietic stem cells (HSC) actually started with the discovery by Till, McCulloch, and colleagues (1-3) that bone marrow contained single cells that could give rise to myeloerythroid colonies in the spleen, and sometimes these colonies contained cells that made more spleen colonies as well as radioprotected and reconstituted lethally irradiated mice (3). But in retrospect, it should have started with the remarkable observation of Ray Owen in 1945 that bovine fraternal twins sharing a single placenta and blood circulation retained production of blood cells genetically defined to be from both throughout their life (4). It could be argued that this was the experiment that began both modern experimental hematology as well as modern cellular immunology. The Till, McCulloch, Wu, Becker, and Simonovitch experiments were elegant demonstrations that single, genetically marked cells existed (random DNA breaks and translocations induced by sublethal irradiation of the donor bone marrow) that could both self-renew and differentiate (2, 5). But these experiments did not put the pure cells in the hands of scientists, and so most of their functions for the next 25 years were implied rather than directly analyzed. Just as genetics is the complement to biochemistry (when one considers genes and gene products), cell marking is the complement to cell purification in the fields of developmental and cellular biology. The first attempts at such cellular purification came from the 'school' of Till & McCulloch (6, 7), and independently the school of Van Bekkum in the Netherlands (8). But what was lacking in those experiments and at that time were both a comprehensive approach that would take into account the clonal activity of stem cells in both self-renewal and differentiation to all blood cell outcomes, and the tools with which one could separate what turned out to be an extremely rare population in the bone marrow. And, it wasn't known until much later that most day 8-10 spleen colonies were the progeny of progenitors, not stem cells (9). Two inventions facilitated the technology of purification of HSC: the advent of monoclonal antibody technology by Kohler & Milstein (10), and the development of the multiparameter fluorescence activated cell sorter by the Herzenberg group (11). My laboratory had established assays for the clonal precursors of T cells and B cells, and we had been using the Till-McCulloch spleen colony clonal assays since the mid-1960s. In the late 1970s and early 1980s we began in earnest the search for mouse early hematopoietic progenitors, including HSCs (12-16). The purification of HSCs proved to be much like the purification of an enzyme, or a cell surface receptor, or a gene. Successive enrichments finally led to the isolation of a population, which could no longer be subdivided and which contained precursors that read out in all clonal assays as well as in radioprotection of lethally irradiated hosts (17). Our first experiments transplanting single HSC in 1991 and 1992, led to the definitive demonstration that these were indeed HSCs (18-20). But these experiments and the ideas that led to them were developed in the context of immunology and experimental hematology as they were emerging in the 1950s and 60 s. This volume of Immunological Reviews is a rich testimony to the kinds of ideas and experiments that, at least in retrospect, turned out to be critical. Many roads were taken, but only one ended up at stem cells.
Collapse
Affiliation(s)
- Irving L Weissman
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305-5329, USA
| |
Collapse
|
17
|
Christensen JL, Weissman IL. Flk-2 is a marker in hematopoietic stem cell differentiation: a simple method to isolate long-term stem cells. Proc Natl Acad Sci U S A 2001; 98:14541-6. [PMID: 11724967 PMCID: PMC64718 DOI: 10.1073/pnas.261562798] [Citation(s) in RCA: 578] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Clonogenic multipotent mouse hematopoietic stem cells (HSCs) and progenitor cells are contained within the c-kit(+) (K) lineage(-/lo) (L) Sca-1(+) (S) population of hematopoietic cells; long-term (LT) and short-term (ST) HSCs are Thy-1.1(lo). c-kit is a member of the receptor tyrosine kinase family, a class of receptors that are important in the proliferation and differentiation of hematopoietic cells. To establish whether the Flk-2/Flt3 receptor tyrosine kinase was expressed on the most primitive LT-HSCs, we sorted highly purified multipotent stem and progenitor cells on the basis of Flk-2 surface expression and used them in competitive reconstitution assays. Low numbers of Flk-2(-) HSCs gave rise to long-term multilineage reconstitution in the majority of recipients, whereas the transfer of Flk-2(+) multipotent cells resulted in mostly short-term multilineage reconstitution. The KLS subset of adult mouse bone marrow was analyzed for Flk-2 and Thy-1.1 expression. Three phenotypically and functionally distinct populations were isolated: Thy(lo) Flk-2(-) (LT-HSCs), Thy(lo) Flk-2(+) (ST-HSCs), and Thy(-) Flk-2(+) multipotent progenitors. The loss of Thy-1.1 and gain of Flk-2 expression marks the loss of self-renewal in HSC maturation. The addition of Flk-2 antibody to the lineage mix allows direct isolation of LT-HSC from adult bone marrow as c-kit(+) lin(-) Sca-1(+) Flk-2(-) from many strains of mice. Fetal liver HSCs are contained within Flk-2(-) and Flk-2(+) KTLS cells.
Collapse
Affiliation(s)
- J L Christensen
- Departments of Pathology and Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305-5324, USA.
| | | |
Collapse
|
18
|
Vasseur F, Le Campion A, Pénit C. Scheduled kinetics of cell proliferation and phenotypic changes during immature thymocyte generation. Eur J Immunol 2001; 31:3038-47. [PMID: 11592080 DOI: 10.1002/1521-4141(2001010)31:10<3038::aid-immu3038>3.0.co;2-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Precursor CD4-CD8- (DN) thymocytes rearrange their TCR-beta genes, and only those which succeed in beta-selection subsequently expand and differentiate into immature CD4+CD8+ (DP) thymocytes. The cell subsets corresponding to the successive steps of this transition can be defined in terms of CD44 and CD25 expression. We partially synchronized the differentiation process by eliminating cycling cells with the anti-mitotic agent demecolcine. Using in vivo pulse labeling with bromodeoxyuridine, we determined the order of entry into DNA synthesis of the different DN and transitory (CD4-/lo CD8+) cell subsets. Two independent proliferation phases were identified. The first cells to enter the cell cycle were CD44-CD25lo, and CD4/CD8/TCR-/BrdU four-color staining showed that they all expressed a low density of the TCR-beta chain, an element of the pre-TCR (the TCR-alpha locus is still in germ-line configuration at this stage). Cycling of CD44+CD25+ cells was detected later, and no starting point was observed at the CD44-CD25hi stage. CD8 expression was immediately detectable in cycling cells, but they took 24 h to reach the DP stage. The study of TCR-Calpha-deficient mice showed that beta gene rearrangement occurred once proliferation had ceased at the DP stage, and that it had no influence on the DN-DP transition. These data show that precursor thymocytes undergo two independent waves of expansion, and that the second wave is restricted to cells capable of pre-TCR expression.
Collapse
Affiliation(s)
- F Vasseur
- Institut National de la Santé et de la Recherche Médicale (INSERM) U 345, Institut Necker, Paris, France
| | | | | |
Collapse
|
19
|
Brown JM, Weissman IL, Shizuru JA. Immunity to infections following hematopoietic cell transplantation. Curr Opin Immunol 2001; 13:451-7. [PMID: 11498301 DOI: 10.1016/s0952-7915(00)00240-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Hematopoietic cell transplantation has progressed from the use of unpurified bone marrow cells or mobilized peripheral blood cells to the use of purified stem cells and progenitor cells. These kinds of transplants can be designed to provide not only hematopoietic rescue but also augmented innate and acquired immunity.
Collapse
Affiliation(s)
- J M Brown
- H1353, Bone Marrow Transplantation Program, Department of Medicine, 300 Pasteur Drive, Stanford, CA 94305, USA.
| | | | | |
Collapse
|
20
|
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
|
21
|
Akashi K, Kondo M, Cheshier S, Shizuru J, Gandy K, Domen J, Mebius R, Traver D, Weissman IL. Lymphoid development from stem cells and the common lymphocyte progenitors. COLD SPRING HARBOR SYMPOSIA ON QUANTITATIVE BIOLOGY 2001; 64:1-12. [PMID: 11232274 DOI: 10.1101/sqb.1999.64.1] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Affiliation(s)
- K Akashi
- Departments of Pathology and Developmental Biology, Stanford University, Stanford, California 94305, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Goh SH, Park JH, Lee YJ, Lee HG, Yoo HS, Lee IC, Park JH, Kim YS, Lee CC. Gene expression profile and identification of differentially expressed transcripts during human intrathymic T-cell development by cDNA sequencing analysis. Genomics 2000; 70:1-18. [PMID: 11087656 DOI: 10.1006/geno.2000.6342] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The development of immature thymocytes to mature T-lymphocytes is a central process for establishing a functional immune system. The gene regulatory events involved in this process are of outstanding interest in understanding the generation of the T-cell repertoire as well as the differentiation of lineage-specific cells, such as CD4(+) helper T-cells or CD8(+) cytotoxic T-lymphocytes. While some essential genes involved in lineage decision and thymocyte differentiation have been already identified, the exact regulatory mechanisms and differential gene expressions are still unknown. The present study was performed to analyze the gene expression profile during T-cell development, in particular, during the differentiation of immature thymocytes into CD4(+) mature T-cells by analyses of expressed sequence tags (ESTs), and to elucidate novel human genes involved in this process. Based on distinct developmental stages, three PCR-based cDNA libraries from immature CD3(-),4(-),8(-) triple-negative, CD4(+),8(+) double-positive, and mature CD4(+),8(-) single-positive thymocytes were constructed. A total of 1477 randomly selected clones were analyzed by automated single-pass sequencing, and the assembly of ESTs resulted in 1027 different species of contig sequences. Among them, 392 contig sequences were matched to known genes, and several novel transcripts were discovered. The matched clones were classified into seven categories according to their functional aspects, and the gene expression profiles of the three thymocyte subsets were compared. The information obtained in current study will serve as a valuable resource for elucidating the molecular mechanism of intrathymic T-cell development.
Collapse
Affiliation(s)
- S H Goh
- Genome Research Center, Korea Research Institute of Bioscience and Biotechnology, Taejon, 305-333, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Miyamoto T, Weissman IL, Akashi K. AML1/ETO-expressing nonleukemic stem cells in acute myelogenous leukemia with 8;21 chromosomal translocation. Proc Natl Acad Sci U S A 2000; 97:7521-6. [PMID: 10861016 PMCID: PMC16578 DOI: 10.1073/pnas.97.13.7521] [Citation(s) in RCA: 339] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Leukemia-specific AML1/ETO transcripts are detectable in most patients with t(8;21) acute myelogenous leukemia (AML) in long-term remission. To understand the inconsistency between the clinical cure and the presence of "residual disease" at a molecular level, we separated and identified the cells expressing AML1/ETO by phenotype and function. Here we demonstrate that AML1/ETO transcripts are present in a fraction of stem cells, monocytes, and B cells in remission marrow, and in a fraction of B cells in leukemic marrow, but not in T cells. AML1/ETO transcripts also were demonstrated in a fraction of colony-forming cells of erythroid, granulocyte-macrophage, and/or megakaryocyte lineages in both leukemic and remission marrow. These data strongly suggest that the acquisition of the t(8;21) occurs at the level of stem cells capable of differentiating into B cells as well as all myeloid lineages, and that a fraction of the AML1/ETO-expressing stem cells undergo additional oncogenic event(s) that ultimately leads to transformation into AML.
Collapse
MESH Headings
- Adult
- Antigens, CD34
- Chromosomes, Human, Pair 21
- Chromosomes, Human, Pair 8
- Core Binding Factor Alpha 2 Subunit
- DNA-Binding Proteins/biosynthesis
- DNA-Binding Proteins/genetics
- Female
- Hematopoietic Stem Cells/metabolism
- Humans
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Male
- Middle Aged
- Neoplasm, Residual/genetics
- Neoplasm, Residual/pathology
- Proto-Oncogene Proteins
- Transcription Factors/biosynthesis
- Transcription Factors/genetics
- Translocation, Genetic
Collapse
Affiliation(s)
- T Miyamoto
- Departments of Pathology and Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | |
Collapse
|
24
|
Akashi K, Richie LI, Miyamoto T, Carr WH, Weissman IL. B lymphopoiesis in the thymus. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 164:5221-6. [PMID: 10799882 DOI: 10.4049/jimmunol.164.10.5221] [Citation(s) in RCA: 103] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The thymus has been regarded as the major site of T cell differentiation. We find that in addition to alphabeta and gammadelta T cells, a significant number (approximately 3 x 104 per day) of B220+IgM+ mature B cells are exported from the thymus of C57BL/6 mice. Of these emigrating B cells, we estimate that at least approximately 2 x 104 per day are cells which developed intrathymically, whereas a maximum of approximately 0.8 x 104 per day are cells which circulated through the thymus from the periphery. The thymus possesses a significant number of pro-B and pre-B cells that express CD19, VpreB, lambda5, and pax-5. These B cell progenitors were found in the thymic cortex, whereas increasingly mature B cells were found in the corticomedullar and medullary regions. Other lymphoid cells, including NK cells and lymphoid dendritic cells, are not exported from the thymus at detectable levels. Thus, the thymus contributes to the formation of peripheral pools of B cells as well as of alphabeta and gammadelta T cells.
Collapse
Affiliation(s)
- K Akashi
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.
| | | | | | | | | |
Collapse
|
25
|
Mine T, Harada K, Matsumoto T, Yamana H, Shirouzu K, Itoh K, Yamada A. CDw108 expression during T-cell development. TISSUE ANTIGENS 2000; 55:429-36. [PMID: 10885563 DOI: 10.1034/j.1399-0039.2000.550505.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
We recently reported a gene encoding the human CDw108, a glycosylphosphatidylinositol (GPI)-anchored membrane glycoprotein that is preferentially expressed on activated T lymphocytes and erythrocytes. The present study investigated the expression of CDw108 on various tissues and cells, particularly on T cells during development. The murine CDw108 cDNA was cloned initially, and it was highly homologous to the human CDw108 (88.0% or 89.3% similarity at the nucleotide or amino acid level, respectively) or identical to the murine semaphorin K1/Sema7A. The CDw108 mRNA was demonstrated in a few tissues including thymus and brain with the highest expression coming on day 7 in whole embryo followed by relatively consistent expression during development. Cell-surface expression of the CDw108 during T-cell development was further examined by flow cytometry in the human umbilical cord blood and thymus. It was preferentially expressed on a CD34+ stem cell population of umbilical cord blood, and CD3dull CD34+/- CD117 (c-kit)+ CD4bright CDbright cells in the thymus that are involved in the stage of positive selection. These results suggest the contribution of CDw108 in T-cell development, especially in the stage of positive selection in the thymus.
Collapse
MESH Headings
- Age Factors
- Animals
- Animals, Newborn
- Antigens, CD/genetics
- Antigens, CD/immunology
- Blotting, Northern
- Brain Chemistry/immunology
- Cloning, Molecular
- DNA, Complementary
- Female
- Fetal Blood/cytology
- Fetal Blood/immunology
- Flow Cytometry
- GPI-Linked Proteins
- Gene Expression Regulation, Developmental/immunology
- Glycoproteins/genetics
- Glycoproteins/immunology
- Humans
- Lipoproteins/genetics
- Lipoproteins/immunology
- Male
- Mice
- Mice, Inbred ICR
- Molecular Sequence Data
- Placenta/chemistry
- Placenta/immunology
- Pregnancy
- RNA, Messenger/analysis
- Semaphorins
- Sequence Homology, Amino Acid
- Spleen/chemistry
- Spleen/immunology
- T-Lymphocytes/chemistry
- T-Lymphocytes/cytology
- T-Lymphocytes/immunology
- Testis/chemistry
- Testis/immunology
- Thymus Gland/chemistry
- Thymus Gland/cytology
- Thymus Gland/immunology
Collapse
Affiliation(s)
- T Mine
- Kurume University Research Center for Innovative Cancer Therapy, Department of Surgery, Kurume University School of Medicine, Japan
| | | | | | | | | | | | | |
Collapse
|
26
|
Dejbakhsh-Jones S, Strober S. Identification of an early T cell progenitor for a pathway of T cell maturation in the bone marrow. Proc Natl Acad Sci U S A 1999; 96:14493-8. [PMID: 10588733 PMCID: PMC24464 DOI: 10.1073/pnas.96.25.14493] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
We have identified a rare ( approximately 0.05-0.1%) population of cells (Thy-1(hi)CD16(+)CD44(hi)CD2(-)TCRalphabeta(-)B220(-)M ac-1(-)NK1. 1(-)) in the adult mouse bone marrow that generates CD4(+) and CD8(+) TCRalphabeta(+) T cells after tissue culture for 48 hr in the presence of Ly5 congenic marrow cells. The essential stages in the maturation of the progenitors were determined; the stages included an early transition from CD2(-)CD16(+)CD44(hi)TCRalphabeta(-) to CD2(+)CD16(int/-)CD44(int/-)TCRalphabeta(-) cells, and a later transition to CD4(+)CD8(+)TCRalphabeta(+) double-positive T cells that rapidly generate the CD4(+) and CD8(+) single-positive T cells. The maturation of the progenitors is almost completely arrested at the CD2(+)TCRalphabeta(-) stage by the presence of mature T cells at the initiation of cultures. This alternate pathway is supported by the marrow microenvironment; it recapitulates critical intermediary steps in intrathymic T cell maturation.
Collapse
Affiliation(s)
- S Dejbakhsh-Jones
- Division of Immunology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305-5111, USA
| | | |
Collapse
|
27
|
Tong J, Kishi H, Matsuda T, Muraguchi A. A bone marrow-derived stroma cell line, ST2, can support the differentiation of fetal thymocytes from the CD4+ CD8+ double negative to the CD4+ CD8+ double positive differentiation stage in vitro. Immunology 1999; 97:672-8. [PMID: 10457222 PMCID: PMC2326888 DOI: 10.1046/j.1365-2567.1999.00822.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/1998] [Revised: 03/01/1999] [Accepted: 03/18/1999] [Indexed: 11/20/2022] Open
Abstract
T-cell precursors differentiate into mature T cells predominantly in the thymus. However, it has also been reported that T-cell precursors mature in extrathymic organs such as the liver, bone marrow, or intestines. In order to investigate the nature of the extrathymic microenvironment that supports T-cell maturation, we examined the effect of a bone marrow-derived stroma cell line, ST2, on T-cell precursors by using a reaggregate thymic organ culture (RTOC) system. We found that ST2 cells supported the differentiation of fetal thymocytes at day 14.5 of gestation from a CD4- CD8- double negative (DN) to a CD4+ CD8+ double positive (DP) differentiation stage in a manner similar to that observed in thymus. Anti-interleukin-7 receptor (IL-7R) and anti-c-kit antibodies blocked the growth of thymocytes in RTOC with ST2 cells, but did not inhibit the generation of DP thymocytes. These data indicate that a bone marrow-derived stroma cell, ST2, which supports B-cell differentiation, is also able to support T-cell development and may constitute one of the microenvironmental components for extrathymic T-cell development.
Collapse
Affiliation(s)
- J Tong
- Department of Immunology, Faculty of Medicine, Toyama Medical and Pharmaceutical University, Sugitani, Toyama, Japan
| | | | | | | |
Collapse
|
28
|
Baldwin KK, Trenchak BP, Altman JD, Davis3 MM. Negative Selection of T Cells Occurs Throughout Thymic Development. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.163.2.689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Abstract
Thymic positive and negative selections govern the development of a self-MHC-reactive, yet self-tolerant, T cell repertoire. Whether these processes occur independently or sequentially remains controversial. To investigate these issues, we have employed tetrameric peptide-MHC complexes to fluorescently label and monitor polyclonal populations of thymocytes that are specific for moth cytochrome c (MCC)/I-Ek. In TCR β mice tetramer-positive thymocytes are detectable even in the most immature TCR-expressing cells. In the presence of MCC peptide, thymocytes that bind strongly to MCC/I-Ek tetramers are deleted earlier in development and more extensively than cells that bind weakly. This negative selection of the MCC/I-Ek-specific cells occurs continuously throughout development and before any evidence of positive selection. Thus, positive and negative selections are independent processes that need not occur sequentially.
Collapse
Affiliation(s)
| | - Brian P. Trenchak
- †Department of Microbiology and Immunology, and
- ‡Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305; and
| | - John D. Altman
- §Department of Microbiology and Immunology, Emory University, Atlanta, GA 30322
| | - Mark M. Davis3
- †Department of Microbiology and Immunology, and
- ‡Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305; and
| |
Collapse
|
29
|
Nomdedéu JF, Mateu R, Altès A, Llorente A, Rio C, Estivill C, López O, Ubeda J, Rubiol E. Enhanced myeloid specificity of CD117 compared with CD13 and CD33. Leuk Res 1999; 23:341-7. [PMID: 10229319 DOI: 10.1016/s0145-2126(98)00185-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The c-kit proto-oncogene encodes a 145 kd tyrosine kinase transmembrane receptor, which plays a key role in haemopoiesis. The c-kit has been classified as CD117 and is especially useful in the differential diagnosis of acute myelogenous leukemia (AML) and acute lymphoblastic leukemia (ALL). We analysed 104 consecutive cases (55 AML, 23 B-cell lineage ALL, three T-cell ALL, 11 blast crisis of chronic myeloproliferative disorders and 12 cases of myelodysplastic syndromes with more than 10% of blasts) referred to our Hospital for immunophenotypic diagnosis and compared the expression pattern of CD13, CD33 and CD117 using the same fluorochrome (phycoerythrin-PE). The recommendations of the EGIL group were followed in order to establish lineage involvement of the blastic population. The threshold used to assign positivity for CD117 was 10%. Bcr/abl, TEL/AML-1 and MLL rearrangements were assessed by molecular methods. CD117 expression was detected in 91% of AML and MDS. All the negative cases corresponded to acute monocytic leukemias. The calculated specificity for myeloid involvement was 0.86 for CD117, 0.36 for CD13 and 0.44 for CD33 (P < 0.005). CD117 was also positive in four cases of ALL. None of these cases showed bcr/abl or MLL rearrangements. In the light of these findings, CD117 expression should yield a higher score, at least one point, in the system currently applied for the diagnosis of biphenotypic acute leukemias (BAL) as its myeloid specificity is greater than that of CD13 and CD33. Moreover, its absence in AML could identify two subgroups of M5b cases. The coexpression of CD117 with cytoplasmic CD79a is often associated with CD7 reactivity, suggesting a stem cell disorder. CD117 should be included on a routine basis for the immunophenotypic diagnosis of acute leukemias.
Collapse
MESH Headings
- Acute Disease
- Adolescent
- Adult
- Aged
- Aged, 80 and over
- Antigens, CD/analysis
- Antigens, CD/biosynthesis
- Antigens, CD/genetics
- Antigens, Differentiation, Myelomonocytic/analysis
- Antigens, Differentiation, Myelomonocytic/biosynthesis
- Antigens, Differentiation, Myelomonocytic/genetics
- Biomarkers
- CD13 Antigens/analysis
- CD13 Antigens/biosynthesis
- CD13 Antigens/genetics
- Cell Lineage
- Child
- Child, Preschool
- Diagnosis, Differential
- Humans
- Immunophenotyping
- Infant
- Infant, Newborn
- Leukemia, Myeloid/immunology
- Leukemia, Myeloid/pathology
- Middle Aged
- Myelodysplastic Syndromes/immunology
- Myelodysplastic Syndromes/pathology
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology
- Proto-Oncogene Mas
- Proto-Oncogene Proteins c-kit/analysis
- Proto-Oncogene Proteins c-kit/biosynthesis
- Proto-Oncogene Proteins c-kit/genetics
- Sialic Acid Binding Ig-like Lectin 3
Collapse
Affiliation(s)
- J F Nomdedéu
- Secció de Marcadors, Department d'Hematologia, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Bhandoola A, Cibotti R, Punt JA, Granger L, Adams AJ, Sharrow SO, Singer A. Positive selection as a developmental progression initiated by alpha beta TCR signals that fix TCR specificity prior to lineage commitment. Immunity 1999; 10:301-11. [PMID: 10204486 DOI: 10.1016/s1074-7613(00)80030-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
During positive selection, immature thymocytes commit to either the CD4+ or CD8+ T cell lineage ("commitment") and convert from short-lived thymocytes into long-lived T cells ("rescue"). By formal precursor-progeny analysis, we now identify what is likely to be the initial positive selection step signaled by alpha beta TCR, which we have termed "induction". During induction, RAG mRNA expression is downregulated, but lineage commitment does not occur. Rather, lineage commitment (which depends upon the MHC class specificity of the alpha beta TCR) only occurs after downregulation of RAG expression and the consequent fixation of alpha beta TCR specificity. We propose that positive selection can be viewed as a sequence of increasingly selective developmental steps (induction-->commitment-->rescue) that are signaled by alpha beta TCR engagements of intrathymic ligands.
Collapse
MESH Headings
- Animals
- CD3 Complex/physiology
- CD5 Antigens/physiology
- Cell Differentiation/immunology
- Cell Lineage/immunology
- Clonal Deletion/immunology
- DNA-Binding Proteins/antagonists & inhibitors
- DNA-Binding Proteins/biosynthesis
- Down-Regulation/immunology
- Epitopes, T-Lymphocyte/metabolism
- Hematopoietic Stem Cells/immunology
- Hematopoietic Stem Cells/metabolism
- Histocompatibility Antigens Class I/immunology
- Homeodomain Proteins/antagonists & inhibitors
- Homeodomain Proteins/biosynthesis
- Mice
- Mice, Congenic
- Mice, Inbred C57BL
- Mice, Inbred CBA
- Receptors, Antigen, T-Cell, alpha-beta/analysis
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Signal Transduction/immunology
- T-Lymphocyte Subsets/cytology
- T-Lymphocyte Subsets/immunology
- T-Lymphocytes/chemistry
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
Collapse
Affiliation(s)
- A Bhandoola
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Kees UR, Ford J. Synergistic action of stem-cell factor and interleukin-7 in a human immature T-cell line. Immunology 1999; 96:202-6. [PMID: 10233696 PMCID: PMC2326741 DOI: 10.1046/j.1365-2567.1999.00674.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The thymus provides the microenvironment that is optimal for T-cell differentiation. The most immature cells in the human thymus express the stem-cell marker CD34 and they respond to cytokines, including stem-cell factor (SCF) and interleukin-7 (IL-7). For the normal progression of T-cell development these two cytokines appear to be vital. We have established and characterized a human pre-T-cell line, PER-487, which mirrors this requirement. This study shows that the simultaneous presence of IL-7 and SCF produces a proliferative response far exceeding additive effects. Furthermore, providing these signals in succession did not achieve the effect observed when they were provided simultaneously. This finding suggests that the effect was not mediated via secretion of molecules or modulation of surface expression. The convergence of the signal transduction pathways of the two cytokines is not known, thus cell line PER-487 provides a unique model for studying the synergistic interaction of IL-7 and SCF.
Collapse
Affiliation(s)
- U R Kees
- Division of Children's Leukaemia and Cancer Research, TVW Telethon Institute for Child Health Research, West Perth, Western Australia, Australia
| | | |
Collapse
|
32
|
Abstract
A system to innocuously visualize T cell lineage commitment is described. Using a "knock-in" approach, we have generated mice expressing a beta-galactosidase reporter in place of CD4; expression of beta-galactosidase in these animals appears to be an accurate and early indicator of CD4 gene transcription. We have exploited this knock-in line to trace CD4/CD8 lineage commitment in the thymus, avoiding important pitfalls of past experimental approaches. Our results argue in favor of a selective model of thymocyte commitment, demonstrating a fundamentally symmetrical process: engagement of either class of major histocompatibility complex (MHC) molecule by a differentiating CD4(+)CD8(+) cell can give rise to T cell antigen receptor (TCR)hi thymocytes of either lineage. Key findings include (a) direct demonstration of a substantial number of CD4-committed, receptor/coreceptor-mismatched cells in MHC class II- deficient mice, a critical prediction of the selective model; (b) highly efficient rescue of such "mismatched" intermediates by forced expression of CD8 in a TCR transgenic line, and an explanation of why previous experiments of this nature were less successful-a major past criticism of the selective model; (c) direct demonstration of an analogous, though smaller, population of CD8-committed mismatched intermediates in class I-deficient animals. Finally, we found no evidence of a CD4 default pathway.
Collapse
Affiliation(s)
- S Chan
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (CNRS/INSERM/ULP), Strasbourg, 67404 Illkirch Cedex, France
| | | | | | | | | |
Collapse
|
33
|
Abstract
The outcome of positive selection of T lymphocytes is that there is a close match between the lineage adopted by a particular cell (CD4+ or CD8+) and the specificity of the T-cell receptor for the class of Major Histocompatibility Complex molecule recognized. How this match is obtained has been a matter of debate. We review the evidence, from recent and older experiments, that indicates that the process follows a selective logic, rather than an instructive one.
Collapse
Affiliation(s)
- S Chan
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (CNRS/INSERM/ULP) Strasbourg, France.
| | | | | | | |
Collapse
|
34
|
Abstract
All lymphocytes are derived from hematopoietic stem cells (HSC). The interleukin-7 receptor (IL-7R) transduces non-redundant signals for both T and B-cell development from HSC. The upregulation of the IL-7R occurs at the stage of the clonogenic common lymphoid progenitor, a recently identified population that can give rise to all lymphoid lineages (T, B and natural killer cells) at a single cell level. The IL-7R plays a critical role in the rearrangement of immunoglobulin heavy chain genes required for B-cell development. IL-7R expression is critically regulated in developing thymocytes; thymocytes that fail the positive selection process downregulate the IL-7R, but those undergoing positive selection upregulate or maintain IL-7R expression. Recent data indicate that IL-7 signaling enhances the survival of developing thymocytes and mature T cells, presumably by its upregulating Bcl-2. Detailed analysis of the signaling cascades activated by the IL-7R may help to reveal the differential roles of IL-7 signaling in T and B-cell development.
Collapse
Affiliation(s)
- K Akashi
- Department of Pathology, Standford University School of Medicine 94305, USA.
| | | | | |
Collapse
|
35
|
Sant'Angelo DB, Lucas B, Waterbury PG, Cohen B, Brabb T, Goverman J, Germain RN, Janeway CA. A molecular map of T cell development. Immunity 1998; 9:179-86. [PMID: 9729038 DOI: 10.1016/s1074-7613(00)80600-7] [Citation(s) in RCA: 77] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Using a sensitive molecular marker for positive selection, the appearance of a particular functional TCR alpha chain sequence in cells from mice bearing a transgenic beta chain, we address several aspects of intrathymic T cell development. First, by examining specific TCR prior to and after maturation, we demonstrate how a restricted TCR repertoire is positively selected from a highly diverse immature TCR repertoire. Second, since this molecular marker is enriched in cells progressing toward the CD4 lineage and depleted in cells progressing toward the CD8 lineage, a map of the developmental pathway of alphabeta thymocytes can be inferred. Third, the first cells that show clear signs of positive intrathymic selection are identified.
Collapse
Affiliation(s)
- D B Sant'Angelo
- Section of Immunology, Yale University School of Medicine, Howard Hughes Medical Institute, New Haven, Connecticut 06520-8011, USA.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
García-Ojeda ME, Dejbakhsh-Jones S, Weissman IL, Strober S. An alternate pathway for T cell development supported by the bone marrow microenvironment: recapitulation of thymic maturation. J Exp Med 1998; 187:1813-23. [PMID: 9607922 PMCID: PMC2212319 DOI: 10.1084/jem.187.11.1813] [Citation(s) in RCA: 49] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/1997] [Revised: 03/26/1998] [Indexed: 01/19/2023] Open
Abstract
In the principal pathway of alpha/beta T cell maturation, T cell precursors from the bone marrow migrate to the thymus and proceed through several well-characterized developmental stages into mature CD4+ and CD8+ T cells. This study demonstrates an alternative pathway in which the bone marrow microenvironment also supports the differentiation of T cell precursors into CD4+ and CD8+ T cells. The marrow pathway recapitulates developmental stages of thymic maturation including a CD4+CD8+ intermediary cell and positive and negative selection, and is strongly inhibited by the presence of mature T cells. The contribution of the marrow pathway in vivo requires further study in mice with normal and deficient thymic or immune function.
Collapse
Affiliation(s)
- M E García-Ojeda
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, California 94305, USA
| | | | | | | |
Collapse
|
37
|
Systemic Overexpression of BCL-2 in the Hematopoietic System Protects Transgenic Mice From the Consequences of Lethal Irradiation. Blood 1998. [DOI: 10.1182/blood.v91.7.2272.2272_2272_2282] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A new transgenic mouse has been generated in which the proto-oncogene BCL-2 is ubiquitously overexpressed. H2K-BCL-2 transgenic mice overexpress BCL-2 in all cells of the hematolymphoid system and have been used to assess the role of BCL-2 in protecting cells of the hematolymphoid system from the consequences of ionizing radiation. We have expanded on previous studies that have demonstrated protection for specific (lymphoid) cell populations and show that systemic overexpression of BCL-2 can protect the hematopoietic system as a whole, including hematopoietic stem cells (HSC), thus increasing the radioresistance of the animal. The increase in radioresistance in H2K-BCL-2 transgenic mice has two components: an increase in the radioresistance of individual cells and, to a lesser extent, an increase in the size of certain critically important cell populations, such as HSC. Bone marrow transplantation experiments show that the increased radioresistance of the transgenic animals is provided by cells of the hematopoietic system. Protection against the consequences of irradiation is not limited to the increased expression levels of BCL-2 in transgenic mice; levels of endogenous BCL-2 are higher in lymphocyte populations that survive irradiation in wild-type mice. We show that ubiquitous overexpression of BCL-2 in the hematopoietic system can be used to increase the resistance of animals to lethal challenges such as irradiation.
Collapse
|
38
|
Systemic Overexpression of BCL-2 in the Hematopoietic System Protects Transgenic Mice From the Consequences of Lethal Irradiation. Blood 1998. [DOI: 10.1182/blood.v91.7.2272] [Citation(s) in RCA: 210] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
A new transgenic mouse has been generated in which the proto-oncogene BCL-2 is ubiquitously overexpressed. H2K-BCL-2 transgenic mice overexpress BCL-2 in all cells of the hematolymphoid system and have been used to assess the role of BCL-2 in protecting cells of the hematolymphoid system from the consequences of ionizing radiation. We have expanded on previous studies that have demonstrated protection for specific (lymphoid) cell populations and show that systemic overexpression of BCL-2 can protect the hematopoietic system as a whole, including hematopoietic stem cells (HSC), thus increasing the radioresistance of the animal. The increase in radioresistance in H2K-BCL-2 transgenic mice has two components: an increase in the radioresistance of individual cells and, to a lesser extent, an increase in the size of certain critically important cell populations, such as HSC. Bone marrow transplantation experiments show that the increased radioresistance of the transgenic animals is provided by cells of the hematopoietic system. Protection against the consequences of irradiation is not limited to the increased expression levels of BCL-2 in transgenic mice; levels of endogenous BCL-2 are higher in lymphocyte populations that survive irradiation in wild-type mice. We show that ubiquitous overexpression of BCL-2 in the hematopoietic system can be used to increase the resistance of animals to lethal challenges such as irradiation.
Collapse
|
39
|
Akashi K, Kondo M, Weissman IL. Two distinct pathways of positive selection for thymocytes. Proc Natl Acad Sci U S A 1998; 95:2486-91. [PMID: 9482912 PMCID: PMC19384 DOI: 10.1073/pnas.95.5.2486] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Most mouse thymocytes undergoing positive selection are found on one of two pathways; the c-Kit+ and the c-Kit- pathways. Here, we show that c-Kit and interleukin-7 receptor (IL-7R)-mediated signals support positive selection during the transition from the subpopulation that first expresses cell surface T cell receptor (TCR)-the TCRalpha/betaloCD4(int)/CD8(int) (DPint) c-Kit+ cells to TCRalpha/betamedc-Kit+ transitional intermediate cells (the c-Kit+ pathway). Cells that fail positive selection on the c-Kit+ pathway become TCRalpha/betaloc-Kit- (DPhi) blasts that appear to undergo alternative TCRalpha rearrangements. The rare DPhic-Kit- blast cells that thus are salvaged for positive selection by expressing a self-major histocompatibility complex selectable TCRalpha/beta up-regulate IL-7R, but not c-Kit, and are the principal progenitors on the c-Kit- pathway; this c-Kit-IL-7R+ pathway is mainly CD4 lineage committed. Cell division is a feature of the TCRlo-medc-Kit+ transition, but is not essential for CD4 lineage maturation from DPhic-Kit- blasts. In this view, positive selection on the c-Kit- path results from a salvage of cells that failed positive selection on the c-Kit+ path.
Collapse
MESH Headings
- Animals
- Antigens, CD/biosynthesis
- CD4-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/immunology
- Cell Division
- Flow Cytometry
- Gene Rearrangement, alpha-Chain T-Cell Antigen Receptor
- Genes, MHC Class I
- Genes, MHC Class II
- Haplotypes
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Proto-Oncogene Proteins c-kit/biosynthesis
- Proto-Oncogene Proteins c-kit/genetics
- Receptors, Antigen, T-Cell, alpha-beta/biosynthesis
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/physiology
- Receptors, Interleukin/biosynthesis
- Receptors, Interleukin-7
- Signal Transduction
- T-Lymphocytes/immunology
- Thymus Gland/immunology
- Up-Regulation/immunology
- beta 2-Microglobulin/genetics
Collapse
Affiliation(s)
- K Akashi
- Departments of Pathology and Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | | | | |
Collapse
|
40
|
Abstract
The existence of a common lymphoid progenitor that can only give rise to T cells, B cells, and natural killer (NK) cells remains controversial and constitutes an important gap in the hematopoietic lineage maps. Here, we report that the Lin(-)IL-7R(+)Thy-1(-)Sca-1loc-Kit(lo) population from adult mouse bone marrow possessed a rapid lymphoid-restricted (T, B, and NK) reconstitution capacity in vivo but completely lacked myeloid differentiation potential either in vivo or in vitro. A single Lin(-)IL-7R(+)Thy-1(-)Sca-1loc-Kit(lo) cell could generate at least both T and B cells. These data provide direct evidence for the existence of common lymphoid progenitors in sites of early hematopoiesis.
Collapse
Affiliation(s)
- M Kondo
- Department of Pathology, Stanford University School of Medicine, California 94305, USA
| | | | | |
Collapse
|
41
|
Altmeyer A, Simmons RC, Krajewski S, Reed JC, Bornkamm GW, Chen-Kiang S. Reversal of EBV immortalization precedes apoptosis in IL-6-induced human B cell terminal differentiation. Immunity 1997; 7:667-77. [PMID: 9390690 DOI: 10.1016/s1074-7613(00)80387-8] [Citation(s) in RCA: 53] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Cell death in B cell terminal differentiation rapidly follows cell cycle arrest in IL-6 differentiation of EBV-immortalized, IgG-bearing human lymphoblastoid cells in vitro. G1 arrest is now found to coincide with repression of EBNA2 and LMP1, two EBV genes essential for B cell transformation, without activation of the viral lytic cycle. IL-6-differentiated B cells die by apoptosis, as evidenced by increases in Annexin V binding activity, PARP cleavage, and chromatin disorganization. Expression of Mcl-1, a Bcl-2 family member, was specifically induced during IL-6 differentiation and down-regulated during apoptosis. Thus, IL-6 reverses EBV immortalization and activates the terminal differentiation program in IgG-bearing human B lymphoblastoid cells, including regulation of an anti-apoptotic gene to coordinate differentiation, cell cycle arrest, and cell death.
Collapse
Affiliation(s)
- A Altmeyer
- Department of Pathology, Cornell University Medical College, New York, New York 10021, USA
| | | | | | | | | | | |
Collapse
|
42
|
Overexpression of Human Stem Cell Factor Impairs Melanocyte, Mast Cell, and Thymocyte Development: A Role for Receptor Tyrosine Kinase-Mediated Mitogen Activated Protein Kinase Activation in Cell Differentiation. Blood 1997. [DOI: 10.1182/blood.v90.8.3018] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AbstractStem cell factor (SCF) is synthesized as both soluble (S) and membrane-associated (MA) proteins. Indirect insight into the function of MA and S isoforms of SCF has come from studies performed in Steel (Sl) mutant mice. However, the physiologic role(s) of these two isoforms remain unknown. In an attempt to better understand the in vivo role of c-kit/SCF interactions on various cell lineages, transgenic mice were generated that overexpress MA isoform of human SCF (hSCF). In murine cells, hSCF behaves as an antagonist to normal SCF function, due to interference with the interaction between endogenous murine SCF and its receptor, c-kit, encoded by the dominant white spotting (W) gene. Mice expressing the hSCF transgene display a variety of phenotypic abnormalities, which are accentuated when combined with W alleles. Here we show that mice homozygous for the hSCF transgene demonstrate a coat color deficiency seen in some mice homozygous for mild W alleles. Specifically, homozygous hSCF transgenic mice (hSCF220) display a pronounced forehead blaze, with additional white spots over the cervical region, as well as a very large belly spot. Doubly heterozygous animals that carry both a mutated W allele and the hSCF transgene also display an unusual pigment defect and a dramatic reduction in the number of dermal mast cells. Furthermore, overexpression of MA hSCF in the thymus results in abnormal thymocyte differentiation and proliferation, which is associated with reduced mitogen activated protein (MAP) kinase activation. Thus, MAP kinase activation by a receptor tyrosine kinase, such as c-kit, may be critical for the differentiation of thymocytes in vivo.
Collapse
|
43
|
Mebius RE, Rennert P, Weissman IL. Developing lymph nodes collect CD4+CD3- LTbeta+ cells that can differentiate to APC, NK cells, and follicular cells but not T or B cells. Immunity 1997; 7:493-504. [PMID: 9354470 DOI: 10.1016/s1074-7613(00)80371-4] [Citation(s) in RCA: 547] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
For a brief period during fetal lymph node organogenesis in mice, lymph node postcapillary high endothelial venules surprisingly express the Peyer's patch addressin MAdCAM-1. This expression allows initial seeding of this incipient structure by two unusual lymphocyte populations selectively expressing the Peyer's patch homing receptor integrin alpha4beta7: CD4+CD3- oligolineage progenitors and TCR gammadelta+ T cells. We show here that CD4+CD3- cells are lineage-restricted progenitors that express surface lymphotoxin-beta (LTbeta) and the chemokine receptor BLR1 and that can become natural killer cells, dendritic antigen-presenting cells, and follicular cells of unknown outcome, but these cells do not become T or B lymphocytes. Since the necessity of lymphotoxin in lymphoid organ development has been shown, we propose that the novel subset of CD4+CD3-LTbeta+ fetal cells is instrumental in the development of lymphoid tissue architecture.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Antigen-Presenting Cells/cytology
- B-Lymphocytes/cytology
- CD3 Complex/metabolism
- CD4-Positive T-Lymphocytes/cytology
- Cell Adhesion Molecules
- Cytotoxicity, Immunologic
- Fluorescent Antibody Technique, Indirect
- GTP-Binding Proteins/genetics
- Gene Expression
- Histocompatibility Antigens Class II/metabolism
- Immunity, Cellular
- Immunoglobulins/metabolism
- Integrins/metabolism
- Interleukin-2/pharmacology
- Killer Cells, Natural/cytology
- Leukocyte Common Antigens/analysis
- Leukopoiesis
- Lymph Nodes/cytology
- Lymph Nodes/embryology
- Lymphocyte Subsets/cytology
- Lymphotoxin-alpha/metabolism
- Lymphotoxin-beta
- Membrane Proteins/metabolism
- Mice
- Mice, Inbred AKR
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mucoproteins/metabolism
- RNA, Messenger/genetics
- Receptors, CXCR5
- Receptors, Chemokine
- Receptors, Cytokine/genetics
- Spleen/embryology
- Spleen/immunology
- T-Lymphocytes/cytology
Collapse
Affiliation(s)
- R E Mebius
- Department of Cell Biology and Immunology, Faculty of Medicine, Vrije Universiteit, Amsterdam, The Netherlands.
| | | | | |
Collapse
|
44
|
Yagi H, Nakamura M, Ishii T, Kasahara S, Itoh T. Ultrastructural analysis of mouse thymocyte subpopulations. Eur J Immunol 1997; 27:2680-7. [PMID: 9368626 DOI: 10.1002/eji.1830271030] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
To understand the lineage relationship and to define morphological characteristics of each thymocyte subset, we have performed ultrastructural analysis of highly purified thymocyte subpopulations. By flow cytometry, five subpopulations were sorted based on the expression of CD4 and CD8 and on cell size (forward scatter): large and small CD4+8+, CD4-8-, CD4+8-, and CD4-8+ thymocytes. Small CD4+8+ thymocytes were the smallest among lymphoid cells, and had a round and smooth cell outline with condensed nuclei, the cytoplasm was scanty and the cell organelles were not developed, suggesting the majority of this subset might be inactive by morphological criteria. CD4+8- thymocytes appeared to be similar to peripheral CD4+ T cells. The CD4-8- thymocyte subset contained morphologically immature cells in terms of cell size, presence of cell surface villi, and euchromatic appearance of the nucleus. CD4-8+ thymocytes heterogeneous in cell size, nuclear chromatin contents and amount of cytoplasm, could be divided into two distinct types. Type 1 CD4-8+ thymocytes were intermediate in size, and therefore similar to peripheral mature CD8+ T cells. Type 2 CD4-8+ thymocytes were large and irregular in shape (large CD4-8+) with irregular-shaped and euchromatic nuclei. Large CD4-8+ cells were, thus, considered to be at the transitional stage from CD4-8- to CD4+8+. At least two groups of large CD4+8+ cells were ultrastructurally classified by the nuclear chromatin content. Large CD4+8+ cells with heterochromatic nuclei were round with a smooth cell membrane, whereas large CD4+8+ cells with euchromatic nuclei were spherical with projections. Cytological features of heterochromatic large CD4+8+ cells are similar to those of small CD4+8+ thymocytes except for cell size. Euchromatic large CD4+8+ cells could be regarded as active blasts potentially leading to mature cells. Taken together, this is the first report that describes the ultrastructural characteristics of each thymocyte subset highly purified by flow cytometry.
Collapse
Affiliation(s)
- H Yagi
- Department of Immunology and Embryology, Tohoku University School of Medicine, Sendai, Japan.
| | | | | | | | | |
Collapse
|
45
|
Barthlott T, Kohler H, Pircher H, Eichmann K. Differentiation of CD4(high)CD8(low) coreceptor-skewed thymocytes into mature CD8 single-positive cells independent of MHC class I recognition. Eur J Immunol 1997; 27:2024-32. [PMID: 9295041 DOI: 10.1002/eji.1830270829] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Thymocytes with a CD4(hi)CD8(lo) coreceptor-skewed (CRS) phenotype have been shown to contain precursors for CD8 single-positive (SP) thymocytes, in addition to precursors for CD4 SP cells. The selection mechanisms that stimulate CD4(hi)CD8(lo) cells to revert to the CD8 lineage are not known. Mice transgenic (tg) for the major histocompatibility complex (MHC) class I-restricted P14 T cell receptor (TCR), on the H-2bm13 background, generate a large number of CD4(hi)CD8(lo) CRS thymocytes. We analyzed the developmental potential and the differentiation requirements of the CD4(hi)CD8(lo) population of these mice. Using reaggregate thymic organ cultures (RTOC), we observed that these cells efficiently and almost exclusively differentiate into CD8 SP cells. Differentiation occurred independent of whether or not the MHC haplotype of the thymic stroma corresponds to the MHC restriction of the tg TCR. Loss of CD4 was independent of thymic stroma, up-regulation of CD8 to full levels was dependent on thymic stroma but independent of MHC haplotype. After trypsin treatment and overnight incubation, these CRS cells re-expressed CD8 but failed to re-express CD4, indicating that they are in the process of terminating CD4 synthesis. CD8 SP cells derived from the CRS cells proliferate in response to peptide-pulsed antigen-presenting cells. Our data suggest that CD4(hi)CD8(lo) CRS thymocytes bearing the P14 tg TCR have completed positive selection and differentiate autonomously into functionally competent CD8 SP cells.
Collapse
Affiliation(s)
- T Barthlott
- Max-Planck-Institut für Immunbiologie, Freiburg, Germany
| | | | | | | |
Collapse
|
46
|
Rooke R, Waltzinger C, Benoist C, Mathis D. Targeted complementation of MHC class II deficiency by intrathymic delivery of recombinant adenoviruses. Immunity 1997; 7:123-34. [PMID: 9252125 DOI: 10.1016/s1074-7613(00)80515-4] [Citation(s) in RCA: 173] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
De novo differentiation of CD4+ T cells was provoked in mice lacking major histocompatibility complex (MHC) class II molecules by intrathymic injection of adenovirus vectors carrying class II genes. This permits a new approach to questions concerning the dynamics of CD4+ T cell compartments in the thymus and peripheral lymphoid organs. Here two issues are explored. First, we show that mature CD4+ CD8- cells reside in the thymus for a protracted period before emigrating to the periphery, highlighting the potential importance of, and our ignorance of, the postselection maturation period. Second, we demonstrate that the survival of CD4+ cells in peripheral lymphoid organs is markedly curtailed when class II molecules are absent and is not further reduced in the absence of both class II and class I molecules, raising the possibility that MHC-mediated selection may continue in the periphery.
Collapse
Affiliation(s)
- R Rooke
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Institut National de la Santé et la Recherche Médicale, Université Louis Pasteur, Illkirch, Strasbourg, France
| | | | | | | |
Collapse
|
47
|
Akashi K, Kondo M, von Freeden-Jeffry U, Murray R, Weissman IL. Bcl-2 rescues T lymphopoiesis in interleukin-7 receptor-deficient mice. Cell 1997; 89:1033-41. [PMID: 9215626 DOI: 10.1016/s0092-8674(00)80291-3] [Citation(s) in RCA: 481] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Mice lacking functional IL-7 or IL-7R alpha genes are severely deficient in developing thymocytes, T cells, and B cells. IL-7 and IL-7 receptor functions are believed to result in lymphoid cell proliferation and cell maturation, implying signal transduction pathways directly involved in mitogenesis and elaboration of developmentally specific new gene programs. Here, we show that enforced expression of the bcl-2 gene in T-lymphoid cells (by crossing in the Emu-bcl-2 transgene) in IL-7R alpha-deficient mice results in a significant restoration of thymic positive selection and T cell numbers and function. We propose cell survival signals to be the principal function of IL-7R engagement in thymic and T cell development.
Collapse
Affiliation(s)
- K Akashi
- Department of Pathology, Stanford University School of Medicine, California 94305, USA
| | | | | | | | | |
Collapse
|
48
|
Aguila HL, Akashi K, Domen J, Gandy KL, Lagasse E, Mebius RE, Morrison SJ, Shizuru J, Strober S, Uchida N, Wright DE, Weissman IL. From stem cells to lymphocytes: biology and transplantation. Immunol Rev 1997; 157:13-40. [PMID: 9255619 DOI: 10.1111/j.1600-065x.1997.tb00971.x] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
We review the development of the hematopoietic system, focusing on the transition from hematopoietic stem cells (HSCs) to T cells. This includes the isolation of HSCs, and recent progress in understanding their ontogeny, homing properties, and differentiation. HSC transplantation is reviewed, including the kinetics of reconstitution, engraftment across histocompatibility barriers, the facilitation of allogeneic engraftment, and the mechanisms of graft rejection. We describe progress in understanding T-cell development in the bone marrow and thymus as well as the establishment of lymph nodes. Finally, the role of bcl-2 in regulating homeostasis in the hematopoietic system is discussed.
Collapse
Affiliation(s)
- H L Aguila
- Department of Pathology, Stanford University School of Medicine, California 94305, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|