Abstract

Poor graft function (PGF) is a fatal complication following allogeneic haematopoietic stem cell transplantation. However, the underlying mechanism is unclear. Effective cross-talk between haematopoietic stem cells (HSCs) and bone marrow microenvironment is important for normal haematopoiesis. Normal HSCs reside in a hypoxic bone marrow microenvironment that protects them from oxidative stress that would otherwise inhibit their self-renewal and results in bone marrow failure. Whether an increased level of reactive oxygen species (ROS) causes PGF following allotransplant is unclear. Using a prospective case-pair study, we identified increased levels of ROS in CD34+ bone marrow cells in subjects with PGF. Elevated ROS levels was associated with an increased frequency of DNA strand breaks, apoptosis, exhaustion of quiescent CD34+ cells and defective colony-forming unit plating efficiency, particularly in the CD34+CD38− fraction. Up-regulated intracellular p53, p21, caspase-3 and caspase-9 levels (but not p38) were detected in CD34+ cells, particularly in the CD34+CD38− fraction. To further study the potential role of ROS levels in post-transplant haematopoiesis, CD34+ bone marrow cells from subjects with good graft function were treated with H2O2. This increased ROS levels resulting in defective CD34+ cells, an effect partially reversed by N-acetyl-L-cysteine. Moreover, CD34+ bone marrow cells from the donors to subjects with poor or good graft function exhibited comparable haematopoietic reconstitution capacities in the xeno-transplanted NOD-PrkdcscidIL2rgnull mice. Thus, even if the transplanted donors' bone marrow CD34+ cells are functionally normal pre-transplant, ROS-induced apoptosis may contribute to the exhaustion of CD34+ bone marrow cells in subjects with PGF following allotransplant.

Highlights

  • Poor graft function is an important, often fatal complication following allogeneic haematopoietic stem cell transplant [1,2,3,4]

  • We recently reported that the transplanted donor CD34-positive cells were quantitatively normal www.impactjournals.com/oncotarget in subjects with poor graft function, but the frequency of bone marrow CD34-positive cells were dramatically reduced and bone marrow endosteal, vascular microenvironment were impaired in subjects with poor graft function compared with those with good graft function posttransplant [2, 3], raising the question whether bone marrow CD34-positive cells in subjects with poor graft function are functionally impaired posttransplant, or the transplanted donors’ CD34-positive cells are already defective pretransplant

  • Subjects with poor graft function had significantly lower numbers of CD34-positive cells (Figure 1A; 0.21 ± 0.06×10E+6 vs. 1.09±0.18×10E+6; P=0.002) and CD34-positive, CD38-negative cells (Figure 1A; 0.07±0.03×10E+6 vs. 0.84±0.15×10E+6; P=0.0008) in the G0 phase compared with subjects with good graft function

Read more

Summary

Introduction

Poor graft function is an important, often fatal complication following allogeneic haematopoietic stem cell transplant [1,2,3,4]. The etiology of poor graft function is complex and includes many factors such as bone marrow toxic drugs, infections, graft-versus-host disease (GvHD) and an impaired bone marrow microenvironment [2, 3, 5,6,7,8,9]. Interactions between haematopoietic stem and progenitor cells and the bone marrow microenvironment are important in maintaining normal haematopoiesis [10,11,12,13]. What drives the bone marrow CD34-positive cells impairing functionally posttransplant and its molecular mechanisms remain to be elucidated in poor graft function

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call