scholarly journals Two rare populations of mouse Thy-1lo bone marrow cells repopulate the thymus.

1988 ◽  
Vol 167 (5) ◽  
pp. 1671-1683 ◽  
Author(s):  
G J Spangrude ◽  
C E Muller-Sieburg ◽  
S Heimfeld ◽  
I L Weissman

Two-color FACS analysis of mouse bone marrow reveals a rare population, comprising 0.1-0.3% of the total, that expresses low levels of the Thy-1 antigen but does not express any of five surface markers that characterize differentiated hematolymphoid cells. We demonstrate here that this fraction of mouse bone marrow is enormously enriched in cells that can home to the thymus and differentiate into mature T lymphocytes, subsequently migrating to peripheral lymphoid organs. Only a subset of the FACS-isolated fraction (1/90 after intrathymic injection) is capable of responding to the thymic microenvironment with a productive commitment to the T cell lineage. A second fraction of mouse bone marrow, which expresses how levels of Thy-1 but is also positive for at least one of five hematolymphoid lineage-specific markers, also contains cells that home to the thymus and establish colonies of thymocytes. The two fractions each contribute approximately equal amounts of thymic colony-forming units (CFUt) to the bone marrow, and together can account for at least half of the CFUt in whole bone marrow.

1992 ◽  
Vol 176 (6) ◽  
pp. 1503-1509 ◽  
Author(s):  
P de Vries ◽  
K A Brasel ◽  
H J McKenna ◽  
D E Williams ◽  
J D Watson

The introduction of clonal assays and long-term culture systems has resulted in considerable progress in the understanding of the early events that control self-renewal and commitment to differentiation of pluripotent hematopoietic stem cells (PHSC). Relatively little is known about the factors that control the commitment of PHSC to the lymphoid lineages, especially the T cell lineage. In the present study, the expression of the proto-oncogene c-kit was used to isolate and study the capacity of highly purified day 14 colony-forming units-spleen (CFU-S) to reconstitute the thymus of sublethally irradiated Thy-1 congenic recipient mice. We demonstrate here that one c-kit positive (c-kitpos) stem cell upon intrathymic transfer can effectively reconstitute the thymus of a sublethally irradiated recipient. After a lag phase of 15 d, high levels of donor-derived thymocytes (Thy-1.1pos) could be detected until 65 d after transplantation in Thy-1.2pos host mice. Donor-derived cells were only detected in the lobe of the thymus in which cells were previously injected and not in the noninjected lobe. These data suggest that c-kitpos stem cells do not migrate from one lobe to another and that they do not re-seed the thymus after having migrated to the bone marrow. The level and duration of reconstitution was found to be cell dose dependent, suggesting that, over time, endogenous stem cells compete with donor stem cells for available sites in the thymus microenvironment. The data presented in this paper demonstrate that commitment of purified adult bone marrow-derived c-kitpos stem cells to the T cell differentiation pathway can occur in the thymus and does not have to happen in the bone marrow.


1990 ◽  
Vol 1 (2) ◽  
pp. 77-84 ◽  
Author(s):  
Naoko Nakano ◽  
Hitoshi Kikutani ◽  
Tadamitsu Kishimoto

Three distinct T-cell precursors: bone marrow cells that express low levels of the Thy-1 antigen but no lineage markers (Thy-1-lo/BM); CD4-, CD8-, and CD3-thymocytes that express low levels of the Thy-1 antigen (Thy-1-lo/Thym); and CD4-, CD8-, and CD3-thymocytes that express high levels of the Thy-1 antigen and the IL-2 Rαchain (Thy-1+/ IL2R+) were isolated by fluorescence-activated cell sorter (FACS). These three populations expanded with different kinetics in the thymus of irradiated recipient mice after intrathymic transfer. When a high dose of human recombinant IL-2 (r-IL-2) or human recombinant IL-6 (r-IL-6) was administered, r-IL-6 accelerated donor Thy-1+/IL2R+to differentiate, whereas r-IL-2 blocked normal differentiation and expansion of donor Thy-1-lo/Thym, but did not show any significant effect on donor Thy-1+/IL2R+. Neither r-IL-2 nor r-IL-6 worked directly on donor Thy-1-lo/BM in this transfer system.


1999 ◽  
Vol 190 (9) ◽  
pp. 1257-1262 ◽  
Author(s):  
Chiyu Wang ◽  
Molly A. Bogue ◽  
Jonathan M. Levitt ◽  
David B. Roth

In SCID (severe combined immunodeficient) mice, proper assembly of immunoglobulin and T cell receptor (TCR) genes is blocked by defective V(D)J recombination so that B and T lymphocyte differentiation is arrested at an early precursor stage. Treating the mice with gamma irradiation rescues V(D)J rearrangement at multiple TCR loci, promotes limited thymocyte differentiation, and induces thymic lymphomas. These effects are not observed in the B cell lineage. Current models postulate that irradiation affects intrathymic T cell precursors. Surprisingly, we found that transfer of irradiated SCID bone marrow cells to unirradiated host animals rescues both TCR rearrangements and thymocyte differentiation. These data indicate that irradiation affects precursor cells at an earlier stage of differentiation than was previously thought and suggest new models for the mechanism of irradiation rescue.


1989 ◽  
Vol 9 (6) ◽  
pp. 2665-2671 ◽  
Author(s):  
G F Tidmarsh ◽  
S Heimfeld ◽  
C A Whitlock ◽  
I L Weissman ◽  
C E Müller-Sieburg

A novel stage in early B-lymphocyte differentiation has been identified in normal mouse bone marrow cells. Earlier work had demonstrated that bone marrow cells characterized by low levels of Thy-1 and lack of a panel of lineage markers (Thy-1lo Lin- cells) were highly enriched for pluripotent hematopoietic stem cells. In this paper, we present evidence that another bone marrow population, which expressed low levels of Thy-1 and coexpressed B220, a B-lineage-specific form of the leukocyte common antigen, contained early and potent precursors for B lymphocytes upon in vivo transfer to irradiated hosts. These Thy-1lo B220+ cells, comprising 1 to 2% of bone marrow cells, were enriched for large cells in the mitotic cycle; the population lacked significant pluripotent hematopoietic stem cell activity and myeloid-erythroid progenitors. Most strikingly, Thy-1lo B220+ cells represented a highly enriched population of bone marrow cells that could be targets of Abelson murine leukemia virus transformation. We propose that Thy-1lo B220+ bone marrow cells represent the earliest stage of committed lymphocyte progenitors, intermediate in differentiation between Thy-1lo Lin- pluripotent stem cells and, in the B lineage, Thy-1- B220+ pre-B cells.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 35-35
Author(s):  
Anna Luiza Facchetti Vinhaes Assumpcao ◽  
Guoping Fu ◽  
Zhanping Lu ◽  
Ashley Kuehnl ◽  
Renren Wen ◽  
...  

T cell development originates from hematopoietic stem and progenitor cells in the bone marrow, which migrate to the thymus and obtain T cell identification. Transcription factors play critical roles in regulating early T cell development. While Notch signals are critically required at the early stage of T cell development, the completion of T cell lineage commitment is far from the initial response to Notch signaling. Other transcription factors such as PU.1, Ikaros, and RUNX1 are required to enable progenitor cells to committee T cell lineage before Notch signaling. YY1 is a ubiquitous transcription factor and mammalian Polycomb Group Protein (PcG) with important functions to regulate lymphocytes development, stem cell self-renewal, cell proliferation, and survival. Previous study showed that YY1 can interact with the Notch1 receptor intracellular domain and regulate Notch1 transactivation activities in vitro. Thus, YY1 may also belong to the core T cell lineage regulatory factors and is required for progenitor cell commitment to T cell development. To test how loss-of-function of YY1 impacts early T cell development, we utilized a conditional Yy1 knockout allele Yy1f/f with loxP sites flanking the Yy1 promoter region and exon 1. Yy1f/fmice were crossed to the inducible Mx1-Cre. In Yy1f/fMx1-Cre mice, YY1 deletion was achieved after treatment with the pI-pC. Yy1-/- mice had significantly reduced numbers of lymphoid-primed multipotent progenitor, (LMPP), common lymphoid progenitor (CLP), and double-negative (DN) T cells compared to Yy1+/+ mice. YY1 deficiency resulted in an early T cell developmental blockage at the DN1 stage. In addition, Notch1 mRNA and protein expressions were significantly reduced in Yy1-/- thymocytes compared to Yy1+/+ thymocytes. In Yy1-/- thymocytes, Notch target gene Hes1 was also downregulated. Thus, YY1 is required for early T cell development and Notch1 signaling. YY1 mediates stable PcG-dependent transcriptional repression via recruitment of PcG proteins that catalyze histone modifications. Our previous results demonstrated that YY1 PcG function is required for Igκ chain rearrangement in early B cell development, however, it is not required for YY1 functions in promoting HSC self-renewal and maintaining HSC quiescence. Many questions remain unanswered regarding how cell- and tissue-specificity is achieved by PcG proteins. Herein, we utilized a YY1 REPO domain mutant (YY1ΔREPO). The small 25 amino acid REPO domain is necessary and sufficient for recruiting other PcG proteins to YY1-bound chromatin sites in Drosophila. While YY1ΔREPO is competent for DNA binding, transcriptional activation, transient transcriptional repression, and interaction with transcriptional coregulators such as HDACs, it is defective in all YY1 PcG functions and unable to recruit other PcG proteins to DNA. This mutant is therefore a powerful tool for dissecting mechanisms governing YY1 PcG-dependent versus -independent functions. Bone marrow cells from Yy1f/f Mx1-Cre mice were transduced retrovirally with MigR1-FlagYY1, MigR1-FlagYY1ΔREPO or MigR1 vector and transplanted into lethally irradiated CD45.1+ mice. In addition, Mx1-Cre bone marrow cells infected with MigR1 vector were used as the wild-type control and transplanted into CD45.1+ mice. While YY1 is required for DN1 to DN2 transition, YY1 PcG function/REPO domain is not required for DN1 transition. Instead, in mice lack of YY1 PcG function/REPO domain, early T cells had increased cell apoptosis and failed to survive. Interestingly, although YY1 PcG function/REPO domain is critical for early T cell survival, it is not required for YY1 regulation of Notch1 expression. We concluded that YY1 is a critical regulator for early T cell development and Notch signaling. There is a lineage-specific requirement for the YY1 PcG function/REPO domain for early T cell development. While YY1 PcG function is required for early T cell survival, it is not required for YY1 regulation of Notch1 expression. YY1 PcG and non-PcG functions promotes T cell development by unique mechanisms of promoting cell survival and Notch1 expression respectively. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 3300-3300
Author(s):  
Sussan Dejbakhsh-Jones ◽  
Marcos E. Garcia-Ojeda ◽  
Devavanii Chatterjea ◽  
Aditi Mukhopadhyay ◽  
Irving L. Weissman ◽  
...  

Abstract We identified committed T cell progenitors (CTPs) in the mouse bone marrow that have not rearranged the TCR β gene, express a variety of genes associated with commitment to the T cell lineage including GATA-3 and TCF-1, Cβand Id2, and show a surface marker pattern (CD44+CD25-CD24+CD5-) that is similar to the earliest T cell progenitors in the thymus. More mature committed intermediate progenitors (CIPs) in the marrow have rearranged the TCR gene loci, express Vαand Vβgenes as well as CD3ε, but do not express surface TCR or CD3 receptors. CTPs, but not progenitors from the thymus, reconstituted the αβ T cells in the lymphoid tissues of athymic nu/nu mice. These reconstituted T cells vigorously secreted IFN- γ after stimulation in vitro, and protected the mice against lethal infection with murine cytomegalovirus (MCMV). Also CTPs from the parent strain can reconstitute an F1 MHC haplotype mismatched host. In conclusion, CTPs in wild-type bone marrow can generate functional T cells via an extrathymic pathway.


2016 ◽  
Vol 2016 ◽  
pp. 1-10 ◽  
Author(s):  
Erika Evangelina Coronado-Cerda ◽  
Moisés Armides Franco-Molina ◽  
Edgar Mendoza-Gamboa ◽  
Heriberto Prado-García ◽  
Lydia Guadalupe Rivera-Morales ◽  
...  

Chemotherapy treatments induce a number of side effects, such as leukopenia neutropenia, peripheral erythropenia, and thrombocytopenia, affecting the quality of life for cancer patients. 5-Fluorouracil (5-FU) is wieldy used as myeloablative model in mice. The bovine dialyzable leukocyte extract (bDLE) or IMMUNEPOTENT CRP® (ICRP) is an immunomodulatory compound that has antioxidants and anti-inflammatory effects. In order to investigate the chemoprotection effect of ICRP on bone marrow cells in 5-FU treated mice, total bone marrow (BM) cell count, bone marrow colony forming units-granulocyte/macrophage (CFU-GM), cell cycle, immunophenotypification, ROS/superoxide and Nrf2 by flow cytometry, and histological and hematological analyses were performed. Our results demonstrated that ICRP increased BM cell count and CFU-GM number, arrested BM cells in G0/G1 phase, increased the percentage of leukocyte, granulocytic, and erythroid populations, reduced ROS/superoxide formation and Nrf2 activation, and also improved hematological levels and weight gain in 5-FU treated mice. These results suggest that ICRP has a chemoprotective effect against 5-FU in BM cells that can be used in cancer patients.


1976 ◽  
Vol 144 (2) ◽  
pp. 456-466 ◽  
Author(s):  
J J Cohen ◽  
S S Fairchild

When mouse bone marrow cells are mixed with cortisol-resistant thymocytes and stimulated in vitro with concanavalin A, the mitogenic response observed is much greater than additive, that is, it is synergistic. Between 94 and 96% of responding cells could be identified as T cells (Thy-1 positive) and of these, 79-100% derived from the cortisol-resistant thymocyte population, not from the bone marrow. Purified macrophages could not replace bone marrow; and marrow depleted of mature T or B cells worked as well as normal marrow. Thus, T and B cells and macrophages were ruled out as the synergizing cell of bone marrow. Nude spleen contained 10 times as many precursors of T cells as did nude marrow and was 10 times better at synergy with cortisol-resistant thymocytes. This implication of the pre-T cell as synergizer was supported by the finding that the synergistic activity of marrow was lost on preincubation, but maintained if the preincubation medium contained thymosin or cyclic AMP. Thus, the ability to enhance the response of relatively mature T cells to Con A is a property of pre-T cells. It is anticipated that this property will allow more detailed studies of T-cell precursor development in mice, and possibly in man.


1989 ◽  
Vol 9 (6) ◽  
pp. 2665-2671
Author(s):  
G F Tidmarsh ◽  
S Heimfeld ◽  
C A Whitlock ◽  
I L Weissman ◽  
C E Müller-Sieburg

A novel stage in early B-lymphocyte differentiation has been identified in normal mouse bone marrow cells. Earlier work had demonstrated that bone marrow cells characterized by low levels of Thy-1 and lack of a panel of lineage markers (Thy-1lo Lin- cells) were highly enriched for pluripotent hematopoietic stem cells. In this paper, we present evidence that another bone marrow population, which expressed low levels of Thy-1 and coexpressed B220, a B-lineage-specific form of the leukocyte common antigen, contained early and potent precursors for B lymphocytes upon in vivo transfer to irradiated hosts. These Thy-1lo B220+ cells, comprising 1 to 2% of bone marrow cells, were enriched for large cells in the mitotic cycle; the population lacked significant pluripotent hematopoietic stem cell activity and myeloid-erythroid progenitors. Most strikingly, Thy-1lo B220+ cells represented a highly enriched population of bone marrow cells that could be targets of Abelson murine leukemia virus transformation. We propose that Thy-1lo B220+ bone marrow cells represent the earliest stage of committed lymphocyte progenitors, intermediate in differentiation between Thy-1lo Lin- pluripotent stem cells and, in the B lineage, Thy-1- B220+ pre-B cells.


Sign in / Sign up

Export Citation Format

Share Document