scholarly journals Administration of interleukin-6 stimulates multilineage hematopoiesis and accelerates recovery from radiation-induced hematopoietic depression

Blood ◽  
1991 ◽  
Vol 77 (3) ◽  
pp. 472-480 ◽  
Author(s):  
ML Patchen ◽  
TJ MacVittie ◽  
JL Williams ◽  
GN Schwartz ◽  
LM Souza

Abstract Hematopoietic depression and subsequent susceptibility to potentially lethal opportunistic infections are well-documented phenomena following radiotherapy. Methods to therapeutically mitigate radiation-induced myelosuppression could offer great clinical value. In vivo studies in our laboratory have demonstrated that interleukin-6 (IL-6) stimulates pluripotent hematopoietic stem cell (CFU-s), granulocyte-macrophage progenitor cell (GM-CFC), and erythroid progenitor cell (CFU-e) proliferation in normal mice. Based on these results, the ability of IL- 6 to stimulate hematopoietic regeneration following radiation-induced hematopoietic injury was also evaluated. C3H/HeN female mice were exposed to 6.5 Gy 60Co radiation and subcutaneously administered either saline or IL-6 (1,000 micrograms/kg) on days 1 through 3 or 1 through 6 postexposure. On days 7, 10, 14, 17, and 22, femoral and splenic CFU-s, GM-CFC, and CFU-e contents and peripheral blood white cell, red cell, and platelet counts were determined. Compared with saline treatment, both 3-day and 6-day IL-6 treatments accelerated hematopoietic recovery; 6-day treatment produced the greater effects. For example, compared with normal control values (N), femoral and splenic CFU-s numbers in IL-6-treated mice 17 days postirradiation were 27% N and 136% N versus 2% N and 10% N in saline-treated mice. At the same time, bone marrow and splenic GM-CFC values were 58% N and 473% N versus 6% N and 196% N in saline-treated mice; bone marrow and splenic CFU-e numbers were 91% N and 250% N versus 31% N and 130% N in saline-treated mice; and peripheral blood white cell, red cell, and platelet values were 210% N, 60% N, and 24% N versus 18% N, 39% N, and 7% N in saline- treated mice. These studies demonstrate that therapeutically administered IL-6 can effectively accelerate multilineage hematopoietic recovery following radiation-induced hematopoietic injury.

Blood ◽  
1991 ◽  
Vol 77 (3) ◽  
pp. 472-480 ◽  
Author(s):  
ML Patchen ◽  
TJ MacVittie ◽  
JL Williams ◽  
GN Schwartz ◽  
LM Souza

Hematopoietic depression and subsequent susceptibility to potentially lethal opportunistic infections are well-documented phenomena following radiotherapy. Methods to therapeutically mitigate radiation-induced myelosuppression could offer great clinical value. In vivo studies in our laboratory have demonstrated that interleukin-6 (IL-6) stimulates pluripotent hematopoietic stem cell (CFU-s), granulocyte-macrophage progenitor cell (GM-CFC), and erythroid progenitor cell (CFU-e) proliferation in normal mice. Based on these results, the ability of IL- 6 to stimulate hematopoietic regeneration following radiation-induced hematopoietic injury was also evaluated. C3H/HeN female mice were exposed to 6.5 Gy 60Co radiation and subcutaneously administered either saline or IL-6 (1,000 micrograms/kg) on days 1 through 3 or 1 through 6 postexposure. On days 7, 10, 14, 17, and 22, femoral and splenic CFU-s, GM-CFC, and CFU-e contents and peripheral blood white cell, red cell, and platelet counts were determined. Compared with saline treatment, both 3-day and 6-day IL-6 treatments accelerated hematopoietic recovery; 6-day treatment produced the greater effects. For example, compared with normal control values (N), femoral and splenic CFU-s numbers in IL-6-treated mice 17 days postirradiation were 27% N and 136% N versus 2% N and 10% N in saline-treated mice. At the same time, bone marrow and splenic GM-CFC values were 58% N and 473% N versus 6% N and 196% N in saline-treated mice; bone marrow and splenic CFU-e numbers were 91% N and 250% N versus 31% N and 130% N in saline-treated mice; and peripheral blood white cell, red cell, and platelet values were 210% N, 60% N, and 24% N versus 18% N, 39% N, and 7% N in saline- treated mice. These studies demonstrate that therapeutically administered IL-6 can effectively accelerate multilineage hematopoietic recovery following radiation-induced hematopoietic injury.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 2133-2133
Author(s):  
Liton Francisco ◽  
Can-Lan Sun ◽  
Lester Laddaran ◽  
Melanie Sabado ◽  
Alysia Bosworth ◽  
...  

Abstract t-MDS/AML is the most common cause of non-relapse mortality in patients undergoing autologous hematopoietic cell transplantation (aHCT) for Hodgkin lymphoma (HL) or non-Hodgkin lymphoma (NHL). Although t-MDS/AML is known to result from damage to hematopoietic stem cells (HSC) as a result of genotoxic cancer treatment, the sequential cellular and molecular changes leading to its development are not clearly defined. To better understand the pathogenesis of t-MDS/AML, we conducted a prospective study in 179 patients undergoing aHCT for HL (n=41) or NHL (n=138) between 1999 and 2004, who participated in a prospective longitudinal study from pre-aHCT to five years post-aHCT, with a serial collection of bone marrow and peripheral blood samples. The median length of follow-up for this cohort was 3.9 years. This report focuses on alterations in peripheral blood parameters from pre-aHCT to the development of t-MDS/AML, and compares these trends with the patients in this cohort who did not develop t-MDS/AML. A total of 22 patients have developed t-MDS/AML in this longitudinally followed cohort thus far, resulting in a cumulative incidence of 11% at 5 years. Serial evaluation of peripheral blood parameters including hematocrit, mean corpuscular volume (MCV), hemoglobin (HGB), red cell distribution width (RDW), white blood cell (WBC) count, and platelet (PLT) count, were abstracted from medical records for the following time points: pre-aHCT, day 100, 6 month, 1 year, 2 year, 3 year, 4 year and 5 year after aHCT, for a total of 1129 time points. Values of peripheral blood parameters associated with post-aHCT relapse or persistence of the primary lymphoma or from 3 months prior to development of t-MDS/AML, were excluded from analysis. As shown in the Figure, comparison of the peripheral blood parameters in subjects who developed t-MDS/AML (cases; n=22) with those who did not (controls; n=157) revealed that hematocrit values were lower for cases compared to controls at all post-aHCT time points. HGB values were lower among cases compared to controls at all post-aHCT time points. The RDW values were higher for cases compared to controls at day 100, 6 months and 1 year post-aHCT. MCV values did not differ between cases and controls at any of the time points. WBC counts for the cases were lower than controls pre-aHCT and also at all time points from 6 months post-aHCT onwards. PLT counts for cases were lower than controls at all time points pre- and post-aHCT. A fixed effect growth curve model was fitted to the data from day 100 to 5 years post-aHCT after adjusting for age at aHCT, primary diagnosis, race/ethnicity, and sex, to examine the rate of change in the peripheral blood parameters over time. Results revealed a significantly sharper decline in MCV for cases (β per 100 days = −0.43) over time as compared to controls (β =−0.15; p = 0.006). Although hematocrit increased with time for both cases and controls, the slope for the cases was significantly less steep (controls: β per 100 days=0.31 vs. cases: β per 100 days=0.12; p =0.01). In summary, we consistently observed lower values for red cell parameters, WBC, and platelets in patients with t-MDS/ AML as compared to controls across multiple timepoints post-aHCT. These differences appeared soon after HCT, were persistent, and preceded the development of t-MDS/AML. Our previous studies indicate that there is increased turnover and reduced regenerative capacity of premalignant hematopoietic stem cells at early stages of development of t-MDS/AML. The early and persistent reduction in peripheral blood parameters observed here provides further evidence that bone marrow injury and ineffective hematopoiesis long predate the development of t-MDS/AML after aHCT. Poor hematocrit recovery and enhanced decline in MCV after aHCT were independently associated with increased risk of t-MDS/AML and warrant further development as readily applied biomarkers for disease and the need for close monitoring. Figure Figure


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 12-13
Author(s):  
Giulia Barbarito ◽  
Beruch Dejene ◽  
David C Shyr ◽  
Gopin Saini ◽  
Linda Oppizzi ◽  
...  

INTRODUCTION. While total CD34 counts in PBSC graft products have correlated with overall likelihood of hematopoietic recovery after allogeneic hematopoietic stem cell transplantation (HSCT), analyses of the HSPC composition and its relationship to relevant post-transplant clinical outcomes are lacking. In fact, the biological basis for different dynamics of hematopoietic/immune recovery, the risk of infection, and graft-versus-host disease (GvHD) is not fully understood. We have performed the first analysis of HSPC graft composition in 6 αβ T-cell/CD19 B-cell depleted haploidentical (αβhaplo) HSCT. Additionally, we correlated the HSPC graft composition with the distribution of the same HSPC subsets in serial post-HSCT bone marrow aspirates performed at Days 30, 60, and 90, with the peripheral blood counts [neutrophils, monocytes, platelet (PLT)] and with the immune recovery (CD3+, CD3+CD4+, CD3+CD8+, αβT, γδT, NK cells) at the same time points. The patients were divided in two groups: 3 patients had a robust and sustained hematopoietic recovery (Group 1) while 3 patients experienced mild cytopenia after Day 60 (Group 2). All patients were transplanted for acute leukemia and received a myeloablative TBI-based conditioning regimen. See Table 1 for details about patients and graft composition. METHODS. All patients were enrolled in the Stanford IRB approved BMT Protocols 179/351/361 and had peripheral blood (PB) and bone marrow (BM) evaluated at Day 30, 60 and 90 post HSCT for the primitive CD34+ Lin- HSPC subsets: HSC (CD38-CD45RA-CD90+), MPP (CD38+CD45RA+), CMP (CD38+CD45RA-CD123+), GMP (CD38+CD45RA+CD123+), MEP (CD38+CD45RA-CD123+) and CLP (CD38+CD127+). Aliquots of αβhaplo-HSCT grafts were cryopreserved for later analyses. Mononuclear cells were isolated from PBSC, PB and BM by Ficoll-Hypaque (Sigma-Aldrich) density gradient centrifugation. FACS analyses were performed on either fresh or frozen cells on Becton Dickinson (BD) Aria II flow cytometer. At least 5x105 events were acquired and analyzed using FlowJo software (BD). RESULTS. Despite consistent levels of αβT-cell depletion and CD34 enrichment, the frequency of the HSPC subsets varied between the grafts. Notably, while CMP and GMP were very consistent across the 6 grafts, the frequency of HSC, CLP, MEP and MPP showed a 2-fold range of variation (Fig1A). No significant correlation was observed between the HSC frequency in the graft and the hematopoietic/immune recovery. However, the frequency of HSC in the BM at Day 30 is statistically correlated (P=0.027) with the PLT at Day 90 (Fig1B). In these preliminary results, the different distribution of CMP, GMP, MEP and MPP did not impact on the hematopoietic/immune recovery. However, there was a significant correlation (P=0.02) between CLP and γδ T cells reconstitution at Day 90 in Group 1 patients (Fig1C). Additionally, the neutrophils, monocytes and phagocytic cells recovery at Day 90 is statistically correlated with the GMP frequency in the BM at Day 30 (P=0.017; P=0.018; P=0.0132, respectively) (Fig1D). Interestingly, the same strong correlation is observed between the CMP in the BM at Day 60 and the recovery of neutrophils and phagocytic cells (P=0.016, P=0.019) at Day 90 (Fig1E), but the CMP at day 30 are already predictive of a robust engraftment in the Group 1 patients (data not shown). CONCLUSIONS. In αβhaplo-HSCT, previously identified factors influencing hematopoietic recovery have been mainly limited to the enumeration of bulk CD34 counts and of mature effector cells, such as αβ/γδ T and NK cells. On the other hand, the presence of GvHD and thymic injury have been correlated to the kinetics of immune reconstitution. We hypothesized that the HSPC composition of the graft would impact lymphohematopoietic recovery in αβhaplo-HSCT recipients. Although preliminary, our data indicate that even with a consistent method of graft manipulation, the HSPC graft composition is heterogeneous. Variations in HSPC subsets frequency and number can contribute to significant differences in lymphohematopoietic recovery and, therefore, clinical outcome. The evaluation of a larger number of patients with longer follow up after HSCT are required. Comparative studies with unmanipulated T-cell replete and cord blood grafts are ongoing. Such analyses will be instrumental not only for prediction of clinical outcome, but also for optimization of novel graft engineering strategies. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1387-1387
Author(s):  
Daniel Lucas ◽  
Paul S. Frenette

Abstract The sympathetic nervous system (SNS) controls hematopoietic stem and progenitor cell (HSPC) release from their niches in the bone marrow (BM) by acting on specific niche cells: bone-lining osteoblasts during mobilization (Cell2006;124:407) and CXCL12-secreting BM stromal cells (Nature2008;452:442) in homeostasis. However, it remains unknown whether the SNS controls other processes in HSC physiology. Here, we show that an intact SNS is necessary for bone marrow reconstitution. Transplantation of lethally-irradiated recipient mice, in which the SNS was lesioned using 6-hydroxydopamine (6OHDA), with 105 healthy BM nucleated cells resulted in 65% survival of the sympathectomized mice compared to 100% survival in the control groups (6OHDA, not irradiated; Saline, not irradiated; Saline, irradiated and transplanted; p<0.01, Kaplan-Meyer survival and Logrank test). Peripheral blood analyses revealed hematopoietic failure and a severe reduction in trilineage cell counts during the recovery phase. These results indicated that HSPCs engrafted with diminished efficiency in sympathectomized mice. Since donor HSPCs were obtained from intact wild-type mice the observed phenotype suggested that the SNS regulated HSPC engraftment by acting on niche cells. The observed phenotype could be due to reduced homing (i.e. the ability of HSPCs to migrate and enter the bone marrow) or to reduced proliferation of HSPC after appropriate homing. To evaluate these two possibilities, we first injected HSPCs into lethally irradiated 6OHDA-treated or control recipients and allowed the cells to migrate to the BM for 3 hours (homing assays). We found that 2.7-fold fewer donor clonogenic progenitors (CFU-C) could be detected in the BM of sympathectomized mice compared to control animals (p<0.001), indicating that an intact SNS is required for HSPC homing to the bone marrow. To determine whether SNS signals are required for HSPC proliferation after homing, we evaluated the proliferative capacity of HSPCs following the administration of the cytotoxic drug 5-fluorouracyl (5FU). Mice treated with 6OHDA or control recipient mice received a single dose of 5FU (150 mg/kg) and their hematopoietic parameters and survival were monitored for 16 days. Peripheral blood analysis revealed reductions in both WBC (1.6 fold; p<0.05) and RBC (1.4 fold; p<0.01) in 6OHDA-treated compared to SNS-intact mice. Further analyses revealed a strong reduction (1.8 fold; p<0.01) in the BM cellular content suggesting that HSPC failed to proliferate normally in 6OHDA-treated mice. The number of CFU-C and stem cell-enriched Lin−Sca1+c-kit+ (LSK) cells in the recovery phase (12 days after 5FU) were reduced 1.9-fold (p<0.01). Moreover, 5FU produced a high mortality rate in the 6OHDA-lesioned group (76% survival at the end of the study; p<0.05 Kaplan- Meyer survival and Logrank test) when compared to 5FU-treated SNS-intact mice (100% survival). These results suggest that HSPC require SNS signals to proliferate in response to stress to restore a functional hematopoiesis. Taken together these data indicate that the sympathetic nervous system acts on the hematopoietic stem cell niche to control both homing and proliferation in the bone marrow, two critical steps that determine the success of a bone marrow transplantation procedure.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 664-664
Author(s):  
Tomer Itkin ◽  
Shiri Gur Cohen ◽  
Joel A. Spencer ◽  
Amir Schajnovitz ◽  
Saravana K. Ramasamy ◽  
...  

Abstract Bone marrow (BM) endothelial cells (BMECs) form a network of blood vessels (BVs) that regulate both leukocyte trafficking and hematopoietic stem and progenitor cell (HSPC) maintenance. However, it is not clear how BMECs balance between these dual regulatory roles and if these events occur at the same vascular site. We define the BM architecture of functionally distinct BVs, their spatial localization and association with specific stromal precursors, which cooperatively regulate HSPC fate. BM stem and progenitor cell maintenance in a metabolically non-active state and leukocyte trafficking occur at separate sites and are differentially regulated by specific BVs with distinct permeability properties. BM arteries were found to be mostly encircled by aSMA+ pericytes whereas the ensuing small-diameter endosteal and trabecular arterioles were predominantly surrounded by stem cell-niche supporting stromal precursor cells. Live imaging and flow analysis revealed that endosteal arteriole BVs exhibited high flow rate, low permeability to external plasma from the peripheral blood, and high levels of adhesion- and tight-junction molecules. Primitive HSPCs located in peri-arteriole regions were found in a non-activated, low reactive oxygen species (ROS) state. Exposure of BM HSPCs to peripheral blood plasma, enhanced their metabolic activity, exhibited by enhanced intracellular ROS levels, and glucose uptake. The same was also evident for circulating HSPCs in the blood. Plasma-exposed HSPCs displayed enhanced motility alongside with reduced long-term repopulation potential. Live imaging showed that all immature and mature leukocyte bi-directional trafficking occurred exclusively at the more permeable sinusoids, located downstream to the endosteal arterioles. Of note, BM sinusoids contained a higher prevalence of ROShigh cells in their microenvironment, including HSPCs. Rapid AMD3100-induced HSPC mobilization preferentially affected sinusoidal but not arterial BVs permeability and CXCL12 chemokine release. Endothelial specific in vivo interference with CXCL12-CXCR4 interactions, via conditional CXCR4 genetic deletion, hampered BM barrier integrity resulting in enhanced HSPC egress. In line with these results we found that during conditions favoring BM stem and progenitor cells expansion, endothelial integrity was enhanced along with reduced HSPC bi-directional trafficking. Conversely, conditional endothelial specific induced genetic or pharmacologic disruption of barrier integrity augmented ROS levels in HSPCs, enhancing their bi-directional trafficking and differentiation while reducing their BM pool size and maintenance in a metabolically non-active state. Of note, humanized mice engrafted with pre-B ALL cells exhibited reduced BM barrier permeability most probably due to BM endothelium modification via FGF-2 secretion by the leukemic clone. Interestingly, human pre-B ALL cells displayed hypersensitivity to plasmatic exposure. We hypothesize that malignant cells modify BM endothelium to provide themselves with a supportive and protective microenvironment composed of undifferentiated BM stromal progenitors and tightly sealed endothelial barrier. In conclusion, our study identifies anatomically distinct BM BVs with different barrier functions serving as systemic leukocyte trafficking or HSPC BM maintenance sites with clinical therapeutic relevance. Disclosures Rafii: Angiocrine Bioscience: Consultancy, Equity Ownership.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 4010-4010
Author(s):  
H. Angharad Watson ◽  
Rebecca J Holley ◽  
Kia J Langford-Smith ◽  
Fiona L Wilkinson ◽  
Toin H van Kuppevelt ◽  
...  

Abstract Abstract 4010 The primary axis of migration for transplanted hematopoietic stem and progenitor cells (HSPC) is CXCL12/CXCR4. Heparan sulphate (HS) is required for CXCL12 presentation and receptor binding, but the functional role of HS is poorly defined. The alpha-L-iduronidase knockout mouse (Idua−/−) accumulates HS and dermatan sulphate, recapitulating the neurodegenerative lysosomal storage disease Mucopolysaccharidosis I Hurler (MPSIH). MPSIH is primarily treated with HSPC transplant, but clinical experience suggests a historical engraftment defect in patients. We show significantly reduced HSPC migration in Idua−/− recipients and under limiting engraftment conditions we show a significant haematopoietic engraftment defect in Idua−/− recipients. No significant donor cell effect was observed. Bone marrow but not peripheral blood CXCL12 levels are slightly elevated in Idua−/− mice. CFU frequency in BM is unchanged between genotypes but reduced significantly in peripheral blood of Idua−/− mice. In whole bone marrow, and on mesenchymal stem cells from Idua−/− mice, HS is present in significant excess, particularly in extracellular matrix, and cell surface locations, with significant increases in all sulphation modifications, especially 2-O-sulphation. Finally we show that excess HS, and particularly HS with increased 2-O -sulphation, functionally inhibit haematopoietic progenitor cell migration in vitro. These data provide novel insight into the influence of highly sulphated HS in CXCL12 mediated haematopoietic progenitor cell migration and help to explain why HSCT engraftment has been historically low in MPSIH. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
1969 ◽  
Vol 34 (6) ◽  
pp. 739-746 ◽  
Author(s):  
THOMAS M. KILBRIDGE ◽  
PAUL HELLER

Abstract Serial determinations of red cell volumes were made with an electronic sizing device in 30 patients with hepatic cirrhosis. Variations in red cell volumes were correlated with other hematologic and clinical findings. The results of these studies suggest that volume macrocytosis in patients with alcoholic cirrhosis is either due to megaloblastosis of the bone marrow or to an accelerated influx of young red cells into the peripheral blood.


Blood ◽  
1997 ◽  
Vol 89 (11) ◽  
pp. 3919-3924 ◽  
Author(s):  
Jean C.Y. Wang ◽  
Monica Doedens ◽  
John E. Dick

Abstract We have previously reported the development of in vivo functional assays for primitive human hematopoietic cells based on their ability to repopulate the bone marrow (BM) of severe combined immunodeficient (SCID) and nonobese diabetic/SCID (NOD/SCID) mice following intravenous transplantation. Accumulated data from gene marking and cell purification experiments indicate that the engrafting cells (defined as SCID-repopulating cells or SRC) are biologically distinct from and more primitive than most cells that can be assayed in vitro. Here we demonstrate through limiting dilution analysis that the NOD/SCID xenotransplant model provides a quantitative assay for SRC. Using this assay, the frequency of SRC in cord blood (CB) was found to be 1 in 9.3 × 105 cells. This was significantly higher than the frequency of 1 SRC in 3.0 × 106 adult BM cells or 1 in 6.0 × 106 mobilized peripheral blood (PB) cells from normal donors. Mice transplanted with limiting numbers of SRC were engrafted with both lymphoid and multilineage myeloid human cells. This functional assay is currently the only available method for quantitative analysis of human hematopoietic cells with repopulating capacity. Both CB and mobilized PB are increasingly being used as alternative sources of hematopoietic stem cells in allogeneic transplantation. Thus, the findings reported here will have important clinical as well as biologic implications.


Cells ◽  
2021 ◽  
Vol 10 (3) ◽  
pp. 523
Author(s):  
Abhirup Bagchi ◽  
Aneesha Nath ◽  
Vasanth Thamodaran ◽  
Smitha Ijee ◽  
Dhavapriya Palani ◽  
...  

Reliable human erythroid progenitor cell (EPC) lines that can differentiate to the later stages of erythropoiesis are important cellular models for studying molecular mechanisms of human erythropoiesis in normal and pathological conditions. Two immortalized erythroid progenitor cells (iEPCs), HUDEP-2 and BEL-A, generated from CD34+ hematopoietic progenitors by the doxycycline (dox) inducible expression of human papillomavirus E6 and E7 (HEE) genes, are currently being used extensively to study transcriptional regulation of human erythropoiesis and identify novel therapeutic targets for red cell diseases. However, the generation of iEPCs from patients with red cell diseases is challenging as obtaining a sufficient number of CD34+ cells require bone marrow aspiration or their mobilization to peripheral blood using drugs. This study established a protocol for culturing early-stage EPCs from peripheral blood (PB) and their immortalization by expressing HEE genes. We generated two iEPCs, PBiEPC-1 and PBiEPC-2, from the peripheral blood mononuclear cells (PBMNCs) of two healthy donors. These cell lines showed stable doubling times with the properties of erythroid progenitors. PBiEPC-1 showed robust terminal differentiation with high enucleation efficiency, and it could be successfully gene manipulated by gene knockdown and knockout strategies with high efficiencies without affecting its differentiation. This protocol is suitable for generating a bank of iEPCs from patients with rare red cell genetic disorders for studying disease mechanisms and drug discovery.


Sign in / Sign up

Export Citation Format

Share Document