scholarly journals Ikzf1 regulates embryonic T lymphopoiesis via Ccr9 and Irf4 in zebrafish

2019 ◽  
Vol 294 (44) ◽  
pp. 16152-16163 ◽  
Author(s):  
Youkui Huang ◽  
Yafang Lu ◽  
Yuepeng He ◽  
Zhi Feng ◽  
Yandong Zhan ◽  
...  
Keyword(s):  
2016 ◽  
Vol 24 ◽  
pp. S92-S93
Author(s):  
Hiroshi Kohara ◽  
Jiyuan Liao ◽  
Shohei Miyamoto ◽  
Yoko Nagai ◽  
Tomotoshi Marumoto ◽  
...  

2019 ◽  
Author(s):  
Rongqun Guo ◽  
Fangxiao Hu ◽  
Qitong Weng ◽  
Cui Lv ◽  
Hongling Wu ◽  
...  

ABSTRACTAchievement of immunocompetent and therapeutic T lymphopoiesis from pluripotent stem cells is a central aim in T cell regenerative medicine. To date, preferentially regenerating T lymphopoiesis in vivo from pluripotent stem cells (PSC) remains a practical challenge. Here we documented that synergistic and transient expression of Runx1 and Hoxa9 restricted in the time window of endothelial to hematopoietic transition and hematopoietic maturation stages induced in vitro from PSC (iR9-PSC) preferentially generated engraftable hematopoietic progenitors capable of homing to thymus and developing into mature T (iT) cells in primary and secondary immunodeficient recipients. Single-cell transcriptome and functional analyses illustrated the cellular trajectory of T lineage induction from PSC, unveiling the T-lineage specification determined at as early as hemogenic endothelial cell stage and identifying the bona fide pre-thymic progenitors. The iT cells distributed normally in central and peripheral lymphoid organs and exhibited abundant TCRαβ repertoire. The regenerative T lymphopoiesis rescued the immune-surveillance ability in immunodeficient mice. Furthermore, gene-edited iR9-PSC produced tumor-specific-T cells in vivo that effectively eradicated tumor cells. This study provides insight into universal generation of functional and therapeutic T lymphopoiesis from the unlimited and editable PSC source.


Blood ◽  
1997 ◽  
Vol 90 (12) ◽  
pp. 4854-4864 ◽  
Author(s):  
Jason P. Gardner ◽  
Haihong Zhu ◽  
Peter C. Colosi ◽  
Gary J. Kurtzman ◽  
David T. Scadden

Abstract Recombinant adeno-associated viruses (rAAV) have been proposed to be gene transfer vehicles for hematopoietic stem cells with advantages over other virus-based systems due to their high titers and relative lack of dependence on cell cycle for target cell integration. We evaluated rAAV vector containing a LacZ reporter gene under the control of a cytomegalovirus (CMV) promoter in the context of primary human CD34+CD2− progenitor cells induced to undergo T-cell differentiation using an in vitro T-lymphopoiesis system. Target cells from either adult bone marrow or umbilical cord blood were efficiently transduced, and 71% to 79% CD2+ cells expressed a LacZ marker gene mRNA and produced LacZ-encoded protein after exposure to rAAV-CMV-LacZ. The impact of transgene expression on the differentiation of T cells was assessed by sequential quantitation of immunophenotypic subsets of virus-exposed cells and no alteration was noted compared with control. The durability of transgene expression was assessed and found to decay by day 35 with kinetics dependent on the multiplicity of infection. In addition, vector DNA was absent from CD4 or CD8 subselected CD3+ cells by DNA-polymerase chain reaction. These data suggest that rAAV vectors may result in robust transgene expression in primitive cells undergoing T-cell lineage commitment without toxicity or alteration in the pattern of T-cell differentiation. However, expression is transient and integration of the transgene unlikely. Recombinant AAV vectors are potentially valuable gene transfer tools for the genetic manipulation of events during T-cell ontogony but their potential in gene therapy strategies for diseases such as acquired immunodeficiency syndrome is limited.


2002 ◽  
Vol 73 (7) ◽  
pp. 1154-1158 ◽  
Author(s):  
Jan Storek ◽  
Ansamma Joseph ◽  
Monja A. Dawson ◽  
Daniel C. Douek ◽  
Barry Storer ◽  
...  

Blood ◽  
2004 ◽  
Vol 103 (9) ◽  
pp. 3362-3364 ◽  
Author(s):  
Yuya Kunisaki ◽  
Sadahiko Masuko ◽  
Mayuko Noda ◽  
Ayumi Inayoshi ◽  
Terukazu Sanui ◽  
...  

Abstract Clearance of apoptotic cells by macrophages is considered important for prevention of inflammatory responses leading to tissue damage. The phosphatidylserine receptor (PSR), which specifically binds to phosphatidylserine (PS) exposed on the surface of apoptotic cells, mediates uptake of apoptotic cells in vitro, yet the physiologic relevance of PSR remains unknown. This issue was addressed by generating PSR-deficient (PSR-/-) mice. PSR-/- mice exhibited severe anemia and died during the perinatal period. In the PSR-/- fetal livers, erythroid differentiation was blocked at an early erythroblast stage. In addition, PSR-/- embryos exhibited thymus atrophy owing to a developmental defect of T-lymphoid cells. Clearance of apoptotic cells by macrophages was impaired in both liver and thymus of PSR-/- embryos. However, this did not induce up-regulation of inflammatory cytokines. These results indicate that during embryonic development, PSR-mediated apoptotic cell uptake is required for definitive erythropoiesis and T lymphopoiesis, independently of the prevention of inflammatory responses. (Blood. 2004;103:3362-3364)


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 773-773
Author(s):  
James A. Kennedy ◽  
Frederic Barabe ◽  
John E. Dick

Abstract T−cell acute lymphoblastic leukemia (T−ALL) is associated with the aberrant expression of a limited number of genes, including the basic helix−loop−helix transcription factor TAL1 (SCL) and the LIM−only domain gene LMO2, in the T−cell lineage. These proteins are thought to mediate their leukemogenic effects by interfering with the transcriptional programs that regulate differentiation during normal thymocyte development. The recent X−linked SCID gene therapy trial has highlighted a role for LMO2 overexpression as an early event in T−lineage leukemogenesis, as retroviral integration into the LMO2 locus was detected in multiple patients that went on to develop T−ALL. However, our understanding of the effects of aberrant LMO2 expression upon human T−lymphopoiesis is currently limited. In order to address this area, lineage−depleted human umbilical cord blood cells were transduced with a lentivirus encoding LMO2 or a control virus, then seeded upon OP9−DL1 stroma. As expected, control cells underwent a normal stage−specific program of T cell development concluding with the emergence of a population of CD4+CD8+CD3hi TCRαβ+ cells. Interestingly, LMO2−expressing cells exhibited a differentiation block at the double negative (DN: CD8−CD4−CD7+) stage of T−cell development. These LMO2−expressing DN cells had a growth advantage compared to control cells (23 population doublings over 75 days for LMO2 vs. 15 p.d. over 60 days for control) but were not immortalized as they stopped expanding after 75 days of co−culture. In the context of T−cell leukemogenesis, these findings suggest that as an initial hit, LMO2 overexpression can induce a blockage in differentiation, resulting in the generation of a proliferative pre−leukemic pool of DN cells. These cells could subsequently accumulate additional mutations leading to the eventual development of an overt leukemia. Given that TAL1 has been shown to accelerate the development of leukemia in LMO2 transgenic mice, and that these two genes are simultaneously overexpressed in a significant percentage of T−ALL cases, this oncogene was an ideal candidate for a second genetic hit. Thus, a retrovirus encoding TAL1 was utilized to infect the LMO2+ DN T−cell population. The expression of TAL1 in these cells significantly increased their proliferative capacity and greatly extended their lifespan, as greater than 60 population doublings occurred over 220 days of culture on stroma. Of note, TAL−1 overexpression appeared to release the LMO2−induced differentiation block at the DN stage, resulting in the emergence of a population of CD4+CD8+CD3− lymphoblasts. Taken together, these findings describe the first experimental model that studies the early stages of human T−cell leukemogenesis by starting with the physiologically relevant population of primitive primary human hematopoietic cells and analyzing the impact of sequential genetic hits upon T−lymphopoiesis. These data indicate that the aberrant expression of LMO2 contributes to leukemogenesis as an early event by generating a pre−leukemic pool of DN cells and that TAL−1 overexpression in this population acts a cooperating event that leads to the emergence of a highly proliferative, immortalized clone. Given that an experimentally induced leukemia model requires the demonstration of in vivo disease, studies assessing the leukemic potential of human cells co−expressing TAL1 and LMO2 are underway in a novel NOD/SCID system that supports human T cell development.


2005 ◽  
Vol 139 (2) ◽  
pp. 210-215 ◽  
Author(s):  
U. Islander ◽  
M. C. Erlandsson ◽  
T. Chavoshi ◽  
C. Jochems ◽  
S. Moverare ◽  
...  

Immunity ◽  
1997 ◽  
Vol 7 (1) ◽  
pp. 155-162 ◽  
Author(s):  
Motonari Kondo ◽  
Koichi Akashi ◽  
Jos Domen ◽  
Kazuo Sugamura ◽  
Irving L Weissman

2006 ◽  
Vol 917 (1) ◽  
pp. 724-731 ◽  
Author(s):  
J. PLUM ◽  
M. SMEDT ◽  
B. VERHASSELT ◽  
T. KERRE ◽  
D. VANHECKE ◽  
...  

Blood ◽  
2011 ◽  
Vol 117 (21) ◽  
pp. 5652-5662 ◽  
Author(s):  
David Yao ◽  
Yuanshuai Huang ◽  
Xiaoran Huang ◽  
Weihuan Wang ◽  
Quanjian Yan ◽  
...  

Abstract Notch signaling is essential for lymphocyte development and is also implicated in myelopoiesis. Notch receptors are modified by O-fucosylation catalyzed by protein O-fucosyltransferase 1 (Pofut1). Fringe enzymes add N-acetylglucosamine to O-fucose and modify Notch signaling by altering the sensitivity of Notch receptors to Notch ligands. To address physiologic functions in hematopoiesis of Notch modified by O-fucose glycans, we examined mice with inducible inactivation of Pofut1 using Mx-Cre. These mice exhibited a reduction in T lymphopoiesis and in the production of marginal-zone B cells, in addition to myeloid hyperplasia. Restoration of Notch1 signaling rescued T lymphopoiesis and the marrow myeloid hyperplasia. After marrow transfer, both cell-autonomous and environmental cues were found to contribute to lymphoid developmental defects and myeloid hyperplasia in Pofut1-deleted mice. Although Pofut1 deficiency slightly decreased cell surface expression of Notch1 and Notch2, it completely abrogated the binding of Notch receptors with Delta-like Notch ligands and suppressed downstream Notch target activation, indicating that O-fucose glycans are critical for efficient Notch-ligand binding that transduce Notch signals. The combined data support a key role for the O-fucose glycans generated by Pofut1 in Notch regulation of hematopoietic homeostasis through modulation of Notch-ligand interactions.


Sign in / Sign up

Export Citation Format

Share Document