scholarly journals Impact of T cells on hematopoietic stem and progenitor cell function: Good guys or bad guys?

2017 ◽  
Vol 9 (2) ◽  
pp. 37 ◽  
Author(s):  
Sulima Geerman ◽  
Martijn A Nolte
2018 ◽  
Vol 57 ◽  
pp. 21-29 ◽  
Author(s):  
Maria E. Alonso-Ferrero ◽  
Niek P. van Til ◽  
Kerol Bartolovic ◽  
Márcia F. Mata ◽  
Gerard Wagemaker ◽  
...  

Blood ◽  
2020 ◽  
Vol 135 (23) ◽  
pp. 2049-2058 ◽  
Author(s):  
Christine R. Keenan ◽  
Nadia Iannarella ◽  
Gaetano Naselli ◽  
Naiara G. Bediaga ◽  
Timothy M. Johanson ◽  
...  

Abstract Loss of heterochromatin has been proposed as a universal mechanism of aging across different species and cell types. However, a comprehensive analysis of hematopoietic changes caused by heterochromatin loss is lacking. Moreover, there is conflict in the literature around the role of the major heterochromatic histone methyltransferase Suv39h1 in the aging process. Here, we use individual and dual deletion of Suv39h1 and Suv39h2 enzymes to examine the causal role of heterochromatin loss in hematopoietic cell development. Loss of neither Suv39h1 nor Suv39h2 individually had any effect on hematopoietic stem cell function or the development of mature lymphoid or myeloid lineages. However, deletion of both enzymes resulted in characteristic changes associated with aging such as reduced hematopoietic stem cell function, thymic involution and decreased lymphoid output with a skewing toward myeloid development, and increased memory T cells at the expense of naive T cells. These cellular changes were accompanied by molecular changes consistent with aging, including alterations in nuclear shape and increased nucleolar size. Together, our results indicate that the hematopoietic system has a remarkable tolerance for major disruptions in chromatin structure and reveal a role for Suv39h2 in depositing sufficient H3K9me3 to protect the entire hematopoietic system from changes associated with premature aging.


Redox Biology ◽  
2017 ◽  
Vol 12 ◽  
pp. 129-138 ◽  
Author(s):  
Y. Zhao ◽  
D.W. Carroll ◽  
Y. You ◽  
L. Chaiswing ◽  
R. Wen ◽  
...  

Leukemia ◽  
2012 ◽  
Vol 27 (7) ◽  
pp. 1613-1614 ◽  
Author(s):  
G Huls ◽  
J van Es ◽  
H Clevers ◽  
G de Haan ◽  
R van Os

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 2130-2130
Author(s):  
Zachary F. Zimmerman ◽  
Robert B. Levy

Abstract Allogeneic hematopoietic stem/progenitor cell (allo-HSPC) transplant is a potentially curative therapy for hematopoietic malignancies/disorders, however host resistance can lead to delayed reconstitution or graft rejection, especially following non-myeloablative conditioning. Our laboratory has been investigating the effector pathways utilized by host cell populations(NK, Tnaive, Tmemory), which mediate resistance to hematopoietic grafts. The effector molecules used by these host populations appear to be varied and include presently undefined pathways. This study examined the dependence of resistance on cytotoxic pathway function mediated by naïve versus memory T cells. Peripheral donor chimerism was assessed following transplant of MHC matched, minor histocompatibility antigen (mHag) mismatched C3H.SW(H-2b)àB6(H-2b) unsensitized, i.e. naive non-myeloablated (5.5 Gy TBI) cytotoxically normal (B6-wt) or defective(B6-pko and B6-gld) recipients. In wild-type recipients, transient peripheral blood donor chimerism was detected which peaked on day 7-10 post-transplant. Donor chimerism was generally undetectable 3–4 weeks post-BMT. Following a secondary transplant in these recipients, no chimerism was detected indicating that the resistance was T cell mediated and engendered a memory response. To determine the cellular requirements for resistance in naïve recipients, CD4−/− and CD8−/− mice were examined. Interestingly, although CD8 mediated B6 anti-C3H.SW responses are well documented, resistance in this model was abrogated in CD4−/−, but not CD8−/− recipients. This indicates that CD4 T cells may play a role in this resistance at the efferent phase, afferent phase, or both. To examine the requirement of cytotoxic pathways in this model, C3H.SW BM was transferred into B6-perforin−/− or B6-gld (FasL functionally deficient mice) recipients. The resistance observed was not appreciably different (ie rejection day 21–28) than that in wild-type mice. CD8 T memory cells primed to donor antigens prior to transplant were analyzed in B6-cdd (H-2b) recipients, which cannot mediate perforin and FasL dependent cytotoxicity. Anti-donor (H-2d) primed B6-cdd mice were transplanted with BALB/cTNFR1−/− bone marrow(BM) and injected with excess levels of blocking mAbs against 3 TNF family apoptosis inducing ligands, i.e. αTL1a, TRAIL and TWEAK as we previously reported. Donor CFU were assessed as a measure of early resistance(D+5). Sensitized B6-cdd mice receiving allo BM lacking TNFR1 and the 3 mAb cocktail exhibited rejection, indicating memory CD8 T cell mediated resistance remained intact despite the disruption of six candidate cytotoxic pathways. Interestingly, rejection in cytotoxically impaired (B6-cdd) recipients was a rapidly occurring event, complete within 48hrs post-BMT, an observation indistinguishable from the kinetics occurring in cytotoxically normal recipients. In total, these findings highlight the potential importance of alternative T cell effector mechanism(s) distinct from classical cytotoxic mechanisms active in allogeneic resistance against hematopoietic stem/progenitor cell grafts mediated by effector cells derived from TNaive and TMemory populations.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1411-1411
Author(s):  
Neil P Rodrigues ◽  
Ashleigh S Boyd ◽  
Cristina Fugazza ◽  
Gillian E May ◽  
YanPing Guo ◽  
...  

Abstract The zinc finger transcription factor GATA-2 has been implicated in the regulation of hematopoietic stem cells. Herein we explored the role of GATA-2 as a candidate regulator of the hematopoietic progenitor cell compartment. We showed that bone marrow from GATA-2 heterozygote (GATA-2+/-) mice displayed attenuated granulocyte-macrophage progenitor function in colony-forming cell (CFC) and serial replating CFC assays. This defect was mapped to the Lin−CD117+Sca-1−CD34+CD16/32high granulocyte-macrophage progenitor (GMP) compartment of GATA-2+/− marrow, which was reduced in size and functionally impaired in CFC assays and competitive transplantation. Similar functional impairments were obtained using a RNA interference approach to stably knockdown GATA-2 in wild-type GMP. While apoptosis and cell cycle distribution remained unperturbed in GATA-2+/− GMP, quiescent cells from GATA-2+/− GMP exhibited altered functionality. Gene expression analysis revealed attenuated expression of HES-1 mRNA in GATA-2 deficient GMPs. Binding of GATA-2 to the HES-1 locus was detected in the myeloid progenitor cell line 32Dcl3 and enforced expression of HES-1 expression in GATA-2+/− GMP rectified the functional defect, suggesting that GATA-2 regulates myeloid progenitor function through HES-1. These data collectively point to GATA-2 as novel, pivotal determinant of GMP cell fate.


Blood ◽  
2012 ◽  
Vol 120 (15) ◽  
pp. 2981-2989 ◽  
Author(s):  
Laura G. Schuettpelz ◽  
Priya K. Gopalan ◽  
Felipe O. Giuste ◽  
Molly P. Romine ◽  
Ronald van Os ◽  
...  

AbstractIncreased expression of Kruppel-like factor 7 (KLF7) is an independent predictor of poor outcome in pediatric acute lymphoblastic leukemia. The contribution of KLF7 to hematopoiesis has not been previously described. Herein, we characterized the effect on murine hematopoiesis of the loss of KLF7 and enforced expression of KLF7. Long-term multilineage engraftment of Klf7−/− cells was comparable with control cells, and self-renewal, as assessed by serial transplantation, was not affected. Enforced expression of KLF7 results in a marked suppression of myeloid progenitor cell growth and a loss of short- and long-term repopulating activity. Interestingly, enforced expression of KLF7, although resulting in multilineage growth suppression that extended to hematopoietic stem cells and common lymphoid progenitors, spared T cells and enhanced the survival of early thymocytes. RNA expression profiling of KLF7-overexpressing hematopoietic progenitors identified several potential target genes mediating these effects. Notably, the known KLF7 target Cdkn1a (p21Cip1/Waf1) was not induced by KLF7, and loss of CDKN1A does not rescue the repopulating defect. These results suggest that KLF7 is not required for normal hematopoietic stem and progenitor function, but increased expression, as seen in a subset of lymphoid leukemia, inhibits myeloid cell proliferation and promotes early thymocyte survival.


Blood ◽  
2008 ◽  
Vol 112 (13) ◽  
pp. 4862-4873 ◽  
Author(s):  
Neil P. Rodrigues ◽  
Ashleigh S. Boyd ◽  
Cristina Fugazza ◽  
Gillian E. May ◽  
YanPing Guo ◽  
...  

Abstract The zinc finger transcription factor GATA-2 has been implicated in the regulation of hematopoietic stem cells. Herein, we explored the role of GATA-2 as a candidate regulator of the hematopoietic progenitor cell compartment. We showed that bone marrow from GATA-2 heterozygote (GATA-2+/−) mice displayed attenuated granulocyte-macrophage progenitor function in colony-forming cell (CFC) and serial replating CFC assays. This defect was mapped to the Lin−CD117+Sca-1−CD34+CD16/32high granulocyte-macrophage progenitor (GMP) compartment of GATA-2+/− marrow, which was reduced in size and functionally impaired in CFC assays and competitive transplantation. Similar functional impairments were obtained using a RNA interference approach to stably knockdown GATA-2 in wild-type GMP. Although apoptosis and cell-cycle distribution remained unperturbed in GATA-2+/− GMP, quiescent cells from GATA-2+/− GMP exhibited altered functionality. Gene expression analysis showed attenuated expression of HES-1 mRNA in GATA-2–deficient GMP. Binding of GATA-2 to the HES-1 locus was detected in the myeloid progenitor cell line 32Dcl3, and enforced expression of HES-1 expression in GATA-2+/− GMP rectified the functional defect, suggesting that GATA-2 regulates myeloid progenitor function through HES-1. These data collectively point to GATA-2 as a novel, pivotal determinant of GMP cell fate.


Sign in / Sign up

Export Citation Format

Share Document