Does a Chronic Inflammatory Process Underlie Clonal Progression In Aplastic Anemia?- In Vitro and In Vivo Evidence That Inflammation Produces Aneuploidy for Chromosomes 7 and 8 In Replicating Cells

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 641-641
Author(s):  
Elaine M Sloand ◽  
Kelsey Loeliger ◽  
Loretta Pfannes ◽  
Andrea Poon ◽  
Rodrigo Calado ◽  
...  

Abstract Abstract 641 Inflammatory conditions such as ulcerative colitis or Barrett's esophagus predispose to chromosomal instability, microsatellite instability, gene hypermethylation, loss of tumor suppressor genes such as p53 leading to malignant transformation. Aplastic anemia (AA) also shows some features of chronic inflammation – immunosuppressive treatment (IST) responders show persisting activation of Th1 lymphocytes (Sloand et al Blood 2002) and it is well recognized that AA patients can develop clonal hematologic disorders. Bone marrow (BM) at presentation may show clonal abnormalities by fluorescence in situ hybridization (FISH) or SNP array analysis. We postulated that chromosomal abnormalities in severe AA may be a consequence of genomic damage caused by effector T-cells as well as telomere shortening resulting from accelerated cell turnover. Selective survival of aneuploid cells such as trisomy 8 and monosomy 7 in an inflammatory environment may occur subsequently due to upregulation of anti-apoptotic genes. We examined the relationship between inflammation and clonal expansion in bone marrow (BM) by culturing normal bone marrow mononuclear cells (BMMNCs) for two days with mis-matched allogeneic lymphocytes, autologous lymphocytes or interferon gamma (IFN γ) prior to short-term methylcellulose culture. After two weeks, colonies were picked, pooled, and stained with annexin; annexin-negative cells were subjected to FISH and spectral karyotyping (SKY). Telomere length was determined by qPCR. To assess the role of free-radicals in the inflammatory setting, we performed separate studies using the free radical scavengers, alpha-tocopherol, and desferrioxamine. We also cultured BMMNCs in low oxygen (5%) to decrease free radical generation. P53 gene was examined by FISH, sequenced, and protein expression measured by immunoblot. We also examined the in vivo effect of inflammation in the AA mouse model (Bloom ML et al Exp Hematol 2004). Luminex for cytokines was performed on the supernatants of all samples. Annexin negative human BMMNCs showed significant aneuploidy after short-term culture with allogeneic lymphocytes or with IFNγ (Fig 1; N=10; p<0.01). SKY showed aneuploidy in chromosomes 7 and 8. Lymphocytes preincubated with 200ng/mL of cyclosporine prior to co-cultivation with BMMNCs did not induce aneuploidy, nor did autologous lymphocytes or allogeneic lymphocytes separated from bone marrow by a semi-permeable membrane. Aneuploidy-induced by allogeneic lymphocytes was ameliorated by alpha-tocopherol and desferrioxamine in a dose-dependent manner (Fig 2) and eliminated by MHC blocking antibody. BMMNCs cultured in 5% O2 had significantly less aneuploidy after culture with allogeneic lymphocytes than BMMNCs cultured in 21% O2 (mean 4% and 15% aneuploidy respectively N=3;). BMMNCs, the samples cultured with CD8 cells but not CD4 cells developed aneuploidy (N=3; mean 10% and 3% respectively) and had a 36% decrease in telomere length compared to control. P53 protein expression was decreased in BMMNCs cultured with mis-matched lymphocytes, but was upregulated ten-fold by alpha-tocopherol and low O2. Aneuploidy was also detected in samples exposed to H2O2 (N=4) suggesting a role for free radicals. No abnormalities in p53 gene sequence could be detected in any of the cultured BMMNC. Haploinsufficiency for p53 was noted in 70% of the aneuploid cells but none of the diploid cells (N=3). CXCL5 and IL10, IFNγ and TFNα concentrations were greatest in the supernants of BMMNCs cultured with lymphocytes. We assessed whether lymphocyte-infusion induced aneuploidy in the murine model of AA. Mice receiving lymphocyte infusions but not controls showed clonal populations of aneuploid cells by SKY. Chromosomes 11 (containing p53 gene), 19 and 7 clones were most frequently affected. These data suggest that inflammatory changes can facilitate clonal evolution in AA by causing DNA damage, telomere erosion and selection of cells resistant to apoptosis. The relationship of inflammatory changes to clonal progression in AA deserves further study. Disclosures: No relevant conflicts of interest to declare.

1987 ◽  
Vol 5 (3) ◽  
pp. 231-241 ◽  
Author(s):  
Vincent S. Gallicchio ◽  
Thomas D. Watts ◽  
George P. Casale ◽  
Philip M. Bartholomew

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 2867-2867
Author(s):  
M. Fischer ◽  
M. Schmidt ◽  
S. Klingenberg ◽  
C. Eaves ◽  
C. von Kalle4 ◽  
...  

Abstract The multidrug resistance transporter, ABCG2, is expressed in primitive hematopoietic stem cells from a variety of sources. These cells are detected in dual wave-length fluorescent FACS profiles as a “side population” (SP cells) on the basis of their ability to efflux the fluorescent dye, Hoechst 33342. We have previously shown that 2 types of human short term repopulating cells (STRC) can be enumerated by limiting dilution analysis of their efficient ability to regenerate exclusively myeloid cells after 3 weeks (STRC-Ms), or both myeloid and lymphoid cells after 6–12 weeks (STRC-MLs) in NOD/SCID-b2microglobulin-/- (b2m-/-) mice. Previous findings also implicated these STRCs as determinants of the rapidity of early hematologic recovery in patients transplanted with cultured mobilized peripheral blood (mPB) cells. Here we asked whether any human STRCs have an SP phenotype and hence whether the isolation of SP cells would retain the rapid repopulating activity of a clinical transplant. CD3- SP and non-SP cells were isolated by FACS from low-density (LD) mPB cells after Hoechst staining and transplanted at limiting dilutions into 117 sublethally irradiated b2m-/- mice. The numbers and types of human hematopoietic cells present in the bone marrow of these mice were subsequently monitored by FACS analysis of bone marrow cells aspirated serially, 3, 8 and 12 wks post-transplant. A verapamil-sensitive SP population was reproducibly detected in all 5 patients’ samples studied (0.039 ± 0.012% of the CD3- LD cells). The in vivo assays failed to detect either STRC-Ms or STRC-MLs in the SP fraction and all these activities were obtained from the non-SP cells. If even a single recipient of the largest dose of SP cells transplanted had been positive, this would have detected 10% of the STRCs present. Thus, &gt;90% of all STRC-M and STRC-ML in mPB are non-SP cells. However, 4 of 40 mice transplanted with SP mPB cells produced some B-lymphoid cells only starting 12 wks post-transplant. However, this result is difficult to interpret since subjecting the STRC-Ms to the Hoechst 33342 staining and FACS isolation procedure alone eliminated their ability to generate megakaryocytic progeny in vivo, although this did not occur when these cells were just stained for CD34 and then isolated by FACS. In addition, the differentiation behaviour of STRC-MLs was not affected by the Hoechst staining and subsequent FACS isolation procedure. In summary, we demonstrate that purification of SP cells depletes human mPB transplants of STRCs, thereby raising serious concerns about the safety of any clinical use of SP cell-enriched transplants as stem cell support after myeloablation. Our results also suggest that the staining and enrichment procedure for isolating SP human cells may differentially affect the lineage potential of some types of STRCs, including those whose activity may be indispensable for rapid and multi-lineage hematologic recovery.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 2674-2674
Author(s):  
Seiji Fukuda ◽  
Hal E. Broxmeyer ◽  
Louis M. Pelus

Abstract The Flt3 receptor tyrosine kinase (Flt3) is expressed on primitive normal and transformed hematopoietic cells and Flt3 ligand (FL) facilitates hematopoietic stem cell mobilization in vivo. The CXC chemokine SDF-1α(CXCL12) attracts primitive hematopoietic cells to the bone marrow microenvironment while disruption of interaction between SDF-1α and its receptor CXCR4 within bone marrow may facilitate their mobilization to the peripheral circulation. We have previously shown that Flt3 ligand has chemokinetic activity and synergistically increases migration of CD34+ cells and Ba/F3-Flt3 cells to SDF-1α in short-term migration assays; this was associated with synergistic phosphorylation of MAPKp42/p44, CREB and Akt. Consistent with these findings, over-expression of constitutively active ITD (internal tandem duplication) Flt3 found in patients with AML dramatically increased migration to SDF-1α in Ba/F3 cells. Since FL can induce mobilization of hematopoietic stem cells, we examined if FL could antagonize SDF-1α/CXCR4 function and evaluated the effect of FL on in vivo homing of normal hematopoietic progenitor cells. FL synergistically increased migration of human RS4;11 acute leukemia cells, which co-express wild-type Flt3 and CXCR4, to SDF-1α in short term migration assay. Exogenous FL had no effect on SDF-1α induced migration of MV4-11 cells that express ITD-Flt3 and CXCR4 however migration to SDF-1α was partially blocked by treatment with the tyrosine kinase inhibitor AG1296, which inhibits Flt3 kinase activity. These results suggest that FL/Flt3 signaling positively regulates SDF-1α mediated chemotaxis of human acute leukemia cells in short-term assays in vitro, similar to that seen with normal CD34+ cells. In contrast to the enhancing effect of FL on SDF-1α, prolonged incubation of RS4;11 and THP-1 acute myeloid leukemia cells, which also express Flt3 and CXCR4, with FL for 48hr, significantly inhibited migration to SDF-1α, coincident with reduction of cell surface CXCR4. Similarly, prolonged exposure of CD34+ or Ba/F3-Flt3 cells to FL down-regulates CXCR4 expression, inhibits SDF-1α-mediated phosphorylation of MAPKp42/p44, CREB and Akt and impairs migration to SDF-1α. Despite reduction of surface CXCR4, CXCR4 mRNA and intracellular CXCR4 in Ba/F3-Flt3 cells were equivalent in cells incubated with or without FL, determined by RT-PCR and flow cytometry after cell permeabilization, suggesting that the reduction of cell surface CXCR4 expression is due to accelerated internalization of CXCR4. Furthermore, incubation of Ba/F3-Flt3 cells with FL for 48hr or over-expression of ITD-Flt3 in Ba/F3 cells significantly reduced adhesion to VCAM1. Consistent with the negative effect of FL on in vitro migration and adhesion to VCAM1, pretreatment of mouse bone marrow cells with 100ng/ml of FL decreased in vivo homing of CFU-GM to recipient marrow by 36±7% (P&lt;0.01), indicating that FL can negatively regulate in vivo homing of hematopoietic progenitor cells. These findings indicate that short term effect of FL can provide stimulatory signals whereas prolonged exposure has negative effects on SDF-1α/CXCR4-mediated signaling and migration and suggest that the FL/Flt3 axis regulates hematopoietic cell trafficking in vivo. Manipulation of SDF-1α/CXCR4 and FL/Flt3 interaction could be clinically useful for hematopoietic cell transplantation and for treatment of hematopoietic malignancies in which both Flt3 and CXCR4 are expressed.


2007 ◽  
Vol 292 (6) ◽  
pp. H2582-H2588 ◽  
Author(s):  
Masayuki Kubo ◽  
Tao-Sheng Li ◽  
Ryo Suzuki ◽  
Mako Ohshima ◽  
Shu-Lan Qin ◽  
...  

Therapeutic angiogenesis can be induced by the implantation of bone marrow cells (BMCs). Hydrogen peroxide (H2O2) has been shown to increase VEGF expression and to be involved in angiogenesis. We tested the hypothesis that pretreatment with H2O2 enhances the efficacy of BMCs for neovascularization. H2O2 pretreatment was done by incubating mouse BMCs in 5 μM H2O2 for 30 min, followed by washing twice with PBS. The H2O2-pretreated and untreated BMCs were then studied in vitro and in vivo. RT-PCR analysis showed that expression of VEGF and Flk-1 mRNA was significantly higher in H2O2-pretreated BMCs than in untreated BMCs after 12 and 24 h of culture ( P < 0.01). Pretreatment with H2O2 also effectively enhanced the VEGF production and endothelial differentiation from BMCs after 1 and 7 days of culture ( P < 0.05). To estimate the angiogenic potency in vivo, H2O2-pretreated or untreated BMCs were intramuscularly implanted into the ischemic hindlimbs of mice. After 14 days of treatment, many of the H2O2-pretreated BMCs were viable, showed endothelial differentiation, and were incorporated in microvessels. Conversely, the survival and incorporation of the untreated BMCs were relatively poor. Microvessel density and blood flow in the ischemic hindlimbs were significantly greater in the mice implanted with H2O2-pretreated BMCs than in those implanted with untreated BMCs ( P < 0.05). These results show that the short-term pretreatment of BMCs with low-dose H2O2 is a novel, simple, and feasible method of enhancing their angiogenic potency.


Blood ◽  
2004 ◽  
Vol 103 (8) ◽  
pp. 2965-2972 ◽  
Author(s):  
Heath L. Bradley ◽  
Christine Couldrey ◽  
Kevin D. Bunting

Abstract Signal transducer and activator of transcription-5 (STAT5) plays an important role in repopulating activity of hematopoietic stem cells (HSCs). However, the relationship of STAT5 activation with early acting cytokine receptors is not well established. We have directly compared bone marrow (BM) from mice mutant for STAT5a and STAT5b (STAT5ab-/-) with that from mice lacking c-Mpl (c-Mpl-/-), the thrombopoietin receptor. Both STAT5 and c-Mpl deficiency only mildly affected committed myeloid progenitors assayed in vitro, but STAT5ab-/- BM showed lower Gr-1+ (4.4-fold), B220+ (23-fold), CD4+ (20-fold), and Ter119+ (17-fold) peripheral blood repopulating activity than c-Mpl-/- BM against wild-type competitor in long-term repopulating assays in vivo. Direct head-to-head competitions of STAT5ab-/- BM and c-Mpl-/- BM showed up to a 25-fold reduction in STAT5ab-/- contribution. Differences affecting reconstitution of primitive c-Kit+Lin-Sca-1+ multipotent progenitor (MPP)/HSC (1.8-fold) and c-Kit+Lin-Sca-1- oligopotent progenitor BM fractions (3.3-fold) were more modest. In serial transplantation experiments, STAT5ab-/- and c-Mpl-/- BM both failed to provide consistent engraftment in tertiary hosts and could not radioprotect lethally irradiated quaternary recipients. These results indicate substantial overlap in c-Mpl-STAT5 signaling defects at the MPP/HSC level but indicate that STAT5 is activated independent of c-Mpl to promote multilineage hematopoietic differentiation. (Blood. 2004;103:2965-2972)


Blood ◽  
1997 ◽  
Vol 90 (11) ◽  
pp. 4354-4362 ◽  
Author(s):  
Nobuko Uchida ◽  
Annabelle M. Friera ◽  
Dongping He ◽  
Michael J. Reitsma ◽  
Ann S. Tsukamoto ◽  
...  

Abstract The DNA synthesis inhibitor hydroxyurea (HU) was administered to determine whether it induces changes in the cell-cycle status of primitive hematopoietic stem cells (HSCs)/progenitors. Administration of HU to mice leads to bone marrow accumulation of c-kit+Thy-1.1loLin−/loSca-1+ (KTLS) cells in S/G2/M phases of the cell cycle. HU is a relatively nontoxic, reversible cell-cycle agent that can lead to approximately a threefold expansion of KTLS cells in vivo and approximately an eightfold increase in the number of KTLS cells in S/G2/M. HSCs in HU-treated mice have undiminished multilineage long-term and short-term clonal reconstitution activity.


1976 ◽  
Vol 231 (1) ◽  
pp. 52-55 ◽  
Author(s):  
R Batt ◽  
JM McKenzie

With the use of microelectrodes, membrane potential (MP) was measured in mouse thyroid glands in vitro. A basal resting MP of about -39 mV was confirmed. The initial effect of feeding a low-iodine diet (6-12 days) was hyperpolarization, up to -47 m V; chronic low-iodine diet led to depolarization. Low concentrations of thyrotropin (less than 3 mU/ml superfusate) caused hyperpolarization and high ones (greater than 10 mU/ml) led to depolarization. Cyclic AMP (10(-3) M), dibutyryl cyclic AMP (1.2 X 10(-4) M or 1.2 X 10(-3) M) and theophylline (10(-2) or 10(-3) M) caused similar hyperpolarization: D- and DL-propranolol (5 X 10(-5) -5 X 10(-4) M) produced depolarization and inhibited hyperpolarization by thyrotropin. Conclusions are that hyperpolarization is a consequence of short-term increased secretion of thyrotropin in vivo or of low (near physiological) concentrations in vitro; these effects are probably mediated by cyclic AMP. The relationship to and mechanism of depolarization resulting from chronic enhanced endogenous secretion or high in vitro concentrations of thyrotropin are unknown.


Blood ◽  
2009 ◽  
Vol 114 (19) ◽  
pp. 4054-4063 ◽  
Author(s):  
Benjamin J. Frisch ◽  
Rebecca L. Porter ◽  
Benjamin J. Gigliotti ◽  
Adam J. Olm-Shipman ◽  
Jonathan M. Weber ◽  
...  

Abstract Microenvironmental signals can determine hematopoietic stem cell (HSC) fate choices both directly and through stimulation of niche cells. In the bone marrow, prostaglandin E2 (PGE2) is known to affect both osteoblasts and osteoclasts, whereas in vitro it expands HSCs and affects differentiation of hematopoietic progenitors. We hypothesized that in vivo PGE2 treatment could expand HSCs through effects on both HSCs and their microenvironment. PGE2-treated mice had significantly decreased number of bone trabeculae, suggesting disruption of their microarchitecture. In addition, in vivo PGE2 increased lineage− Sca-1+ c-kit+ bone marrow cells without inhibiting their differentiation. However, detailed immunophenotyping demonstrated a PGE2-dependent increase in short-term HSCs/multipotent progenitors (ST-HSCs/MPPs) only. Bone marrow cells transplanted from PGE2 versus vehicle-treated donors had superior lymphomyeloid reconstitution, which ceased by 16 weeks, also suggesting that ST-HSCs were preferentially expanded. This was confirmed by serial transplantation studies. Thus in vivo PGE2 treatment, probably through a combination of direct and microenvironmental actions, preferentially expands ST-HSCs in the absence of marrow injury, with no negative impact on hematopoietic progenitors or long-term HSCs. These novel effects of PGE2 could be exploited clinically to increase donor ST-HSCs, which are highly proliferative and could accelerate hematopoietic recovery after stem cell transplantation.


Author(s):  
Л.П. Сычева ◽  
Л.М. Рождественский ◽  
Н.И. Лисина ◽  
Т.Г. Шлякова ◽  
В.В. Зорин

Изучена антимутагенная активность противолучевых препаратов разного механизма действия: беталейкина, флагеллина, индралина и рибоксина в краткосрочных и хроническом экспериментах на мышах in vivo. Все препараты проявили антимутагенные свойства в клетках костного мозга, а также гепатопротекторное действие. Наибольший эффект при используемых условиях эксперимента отмечен для беталейкина и нового противолучевого препарата флагеллина. The antimutagenic activity of anti-radiation drugs betaleukin, flagellin, indraline and riboxin with different mechanisms of action was studied in short-term and chronic experiments in mice in vivo. All drugs showed antimutagenic properties in bone marrow cells, as well as hepatoprotective effect. The greatest effect under the used experimental conditions was noted for betaleukin and the new anti-radiation drug flagellin.


Sign in / Sign up

Export Citation Format

Share Document