The micronucleus test in rat erythrocytes from bone marrow, spleen and peripheral blood: the response to low doses of ionizing radiation, cyclophosphamide and vincristine determined by flow cytometry

Author(s):  
Lilianne Abramsson-Zetterberg ◽  
Jan Grawé ◽  
Gösta Zetterberg
Author(s):  
H. Norppa ◽  
M. Hayashi ◽  
T. Sofuni ◽  
Y. Kodama ◽  
M. Ishidate

2008 ◽  
Vol 132 (5) ◽  
pp. 813-819
Author(s):  
Xiaohong Han ◽  
Jeffrey L. Jorgensen ◽  
Archana Brahmandam ◽  
Ellen Schlette ◽  
Yang O. Huh ◽  
...  

Abstract Context.—The immunophenotypic profile of basophils is not yet fully established, and the immunophenotypic changes in chronic myelogenous leukemia are not fully characterized. Objective.—To establish a comprehensive immunophenotypic spectrum of normal basophils and to assess the range of immunophenotypic aberrations of basophils in chronic myelogenous leukemia. Design.—Using 4-color flow cytometry, we compared the immunophenotypic profile of basophils in peripheral blood or bone marrow samples from 20 patients with no evidence of neoplasia to basophils from 15 patients with chronic myelogenous leukemia. Results.—Basophils in control cases were all positive for CD9, CD13, CD22, CD25 (dim), CD33, CD36, CD38 (bright), CD45 (dimmer than lymphocytes and brighter than myeloblasts), and CD123 (bright), and were negative for CD19, CD34, CD64, CD117, and HLA-DR. Basophils in all chronic myelogenous leukemia patients possessed 1 to 5 immunophenotypic aberrancies. The most common aberrancies were underexpression of CD38, followed by aberrant expression of CD64 and underexpression of CD123. CD34 and CD117 were present in cases with basophilic precursors. Myeloblasts showed a distinct immunophenotypic profile, as they typically expressed CD34 and CD117, showed dimmer expression (compared with basophils) of CD38, CD45, and CD123, and lacked expression of CD22. Conclusions.—Flow cytometric immunophenotyping can identify immunophenotypic aberrations of basophils in chronic myelogenous leukemia, and discriminate basophils from myeloblasts.


2013 ◽  
Vol 7 (1) ◽  
pp. 99-111 ◽  
Author(s):  
Hai-Su Yang ◽  
Min Yang ◽  
Xiaoyu Li ◽  
Sorina Tugulea ◽  
Henry Dong

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2312-2312
Author(s):  
Dean A. Lee ◽  
William C. Choi

Abstract BACKGROUND: Inoculation of human normal or leukemic myeloblasts into sublethaly irradiated NOD/SCID mice often results in persistent low-level engraftment (< 5%), but significant proliferation (≥ 5-fold expansion) rarely occurs. Most malignant samples that engraft and proliferate are of FAB M4 subtype and exhibit rapid extramedullary growth at the site of injection without significant marrow or spleen involvement. We hypothesized that low engraftment and proliferation of less mature FAB subtypes results from an increased requirement of these cells for a marrow environment of cytokine and contact-dependent growth and survival factors not adequately provided across species by the mouse bone marrow stroma. Here we show that the subcutaneous injection of minimally-differentiated human mesenchymal stem cells (MSC) in a Matrigel matrix creates an artificial human marrow environment resulting in improved survival and proliferation of human myeloblasts. METHODS: Human leukemic myeloblasts were obtained from the marrow or peripheral blood of 14 newly diagnosed pediatric patients under an IRB-approved collection and banking protocol. MSC were obtained from sterile filters following processing of human marrow from healthy donors or from the NIH-funded MSC bank at Tulane University. 6-to-12 week old NOD-SCID mice were injected IV with 5x106 AML blasts via the retro-orbital sinus (N=38), subcutaneously in 0.5mL Matrigel (N=18), or subcutaneously with 5x105 MSC in 0.5mL of Matrigel (N=14). Mice were euthanized when evidence of tumor burden was present. Peripheral blood, bone marrow, spleen, and subcutaneous nodules were obtained for flow immunophenotyping, FISH, and histopathology. Percent engraftment was determined by flow cytometry for human CD33-APC and mouse H2Kd-PE. RESULTS: Median time from injection to necropsy was 12.5 weeks. 18% died of spontaneous murine thymomas. No animals died of progressive human AML if myeloblasts were injected IV or subcutaneously with Matrigel, and all had < 5% involvement of bone marrow, spleen, and blood. Six animals injected with AML and MSC (43%) developed visible tumors at a median of 8.5 weeks. These tumors were easily reduced to single cell suspensions of > 98% CD33+ by flow cytometry, with mean estimated recovery of 1.3x108 human myeloblasts per mouse tumor (mean 36-fold expansion, range 4 to 52-fold). For cases in which the AML and MSC were derived from subjects of disparate gender, the origin of the cells (leukemic donor vs. MSC donor) was validated by FISH for human X/Y chromosomes. Histopathology of the resulting mass revealed the central development of a stromal chondroid matrix similar to trabecular bone. Marrow, spleen, and blood for all these animals contained < 5% human myeloblasts. CONCLUSIONS: Here we describe an effective method for expanding immature human leukemic myeloblasts in the NOD/SCID mouse. These findings suggest that less mature myeloblasts require human MSC for survival and proliferation and appear to lack significant homing to or expansion in mouse marrow even in the presence of a significant ectopic tumor burden. This is a useful technique for expanding human AML cells for research, may be a model for more broad-based patient-oriented testing of chemotherapeutic and biologic therapies for AML, and represents a novel animal model for studying the stromal interactions and growth requirements of malignant and non-malignant myeloid precursors.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 1284-1284
Author(s):  
Christiane Pott ◽  
Sebastian Boettcher ◽  
Stefan Gesk ◽  
Reiner Siebert ◽  
Wolfram Klapper ◽  
...  

Abstract Two prospectively randomized intergroup trials of the European MCL Network investigating the impact of different combined immuno-chemotherapy protocols for patients with stage II–IV mantle cell lymphoma (MCL) >< 65 yrs followed by autologous stem cell transplantation (PBSCT) for patients <65 yrs and a rituximab or interferone maintenance treatment for patients >65 yrs are currently performed. Patients and methods: 180 German patients with diagnostic peripheral blood (PB) involvement detectable by consensus IGH PCR were analysed for t(11;14) translocation by FISH and 4-colour Flow Cytometry (FC) at diagnosis as well as molecular response (MR) by quantitative IGH-RQ-PCR after induction. The results were compared to clinical parameters at diagnosis and PFS. Results: Patients had a median age of 61 years, an elevated LDH in 37%, B-symptoms in 41% and extranodal involvement in 37%. According to the MIPI risk factor score 26% had an adverse, 34% an intermediate and 40% a good prognosis. PB involvement by MCL was demonstrated in 67% (74/111) by t(11;14) FISH and in 97% (152/156) by FC. Detection of MCL cells by FC in PB did not correlate with clinical parameters as stage, LDH, extranodal involvement or bone marrow infiltration. However, median % of MCL cells measured by FC differed significantly with clinical stage (p<0.0001), LDH (p=0.0096) bone marrow infiltration (<0.0001) and MIPI prognostic index (0.0002). MR after induction treatment was achieved in 60% (60/100) of patients and correlated with BM infiltration at diagnosis (p=0.0117). There was a trend for improved PFS in patients achieving MR (PFS 90% vs. 75% at 15 months after induction) however, the observation time is still too short for definitive evaluation. Conclusion: FC is highly sensitive for detection of PB involvement in MCL and improves staging accuracy. Achievement of MR might be an early indicator for treatment outcome after immuno-chemotherapy in MCL with the potential of defining prognostic subgroups.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 4130-4130
Author(s):  
Daniel L. Kraft ◽  
Vartan Ghazarossian ◽  
Mike Crocker ◽  
Sergio Najar ◽  
Antonio A. Carrasco-Yalðn

Abstract INTRODUCTION: Bone marrow (BM) contains a rich supply of adult stem and progenitor cells, including hematopoieitic and mesenchymal stem cells which are used in Bone Marrow Transplantation (BMT) and an increasing array of regenerative therapies. Traditional marrow harvest methods utilize percutaneous large bore needle aspiration, result in marrow highly diluted by peripheral blood, and are crude, tedious, labor intensive and expensive, usually requiring general anesthesia, and &gt;100 serial small volume aspirates to obtain adequate cell numbers for BMT. BM is showing increasing long-term advantages over mobilized PBSC for many alloegeneic BMTs, in terms of less cGVHD and in some cases improved survival. Improved BM harvest methods are needed. A novel device, the “MarrowMiner” (MM), was developed for the minimally invasive harvest of BM to enable the rapid, convenient, outpatient harvest of large quantities of BM under local anesthesia for use in allogeneic and autologous BMT and cell therapies utilizing autologous marrow derived cells. The MarrowMiner utilizes a single marrow entry site into the anterior or posterior iliac, through which the flexible, powered, guidable FlexShaft catheter can access the majority of the marrow space and aspirate rich marrow. Extensive testing in human cadavers and porcine models demonstrated a 10X increase in stem cells activity/ml (by CFU) compared to that of traditional needle harvests. The MM recently received both FDA and CE Mark regulatory approved, and ‘First In Human’ trials were successfully completed under local anesthesia, demonstrating safety, efficacy and higher stem cell yields compared to traditional methods. METHODS: In an ongoing prospective study, 10 patients undergoing autologous marrow derived therapy for use in regenerative medicine, had marrow harvested from their anterior or posterior ileac by the MM under local anesthesia on one hip, with direct comparison to standard needle serial marrow aspirates on the patients opposite hip (up to 350 ml per side). Cell viability, counts, CD34+, T cell, and MSC populations were assessed by flow cytometry. RESULTS: The MM successfully harvested marrow from a single entry sites and 2–3 paths under local anesthesia, without complications. Compared to standard harvest in the same patients, MM harvests had significantly number of Total Nucleated Cells ml compared to marrow harvested from the same patient by standard needle ( mean 1.98 fold greater TNC (range 0.87–3.36, p&lt;.05). Viability was equivalent at (&gt;99). In addition to higher TNC/ml, significantly higher levels (mean 3.56 fold) of Aldeflour/ALDH+ cells/ml, CD34+, and phenotypic MSC (CD45−,34−,90+,105+) and endothelial progenitor cells were obtained, as measured by flow cytometry. Mean CD3+ T-cell counts per ml were lower with MM harvests. CONCLUSIONS: The novel FDA approved MarrowMiner system demonstrated safety and efficacy in clinical use, harvesting more stem cells per unit volume in a single entry compared to standard harvest methods. These results suggests the MM may enable improved clinical stem cell harvests in a more rapid and minimally invasive manner in the outpatient setting, while harvesting a richer marrow product with less peripheral blood contamination. Such a system, facilitating convenient, on demand stem cell collection may have significant application for BMT and other marrow based cellular therapies.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 2803-2803
Author(s):  
Xiaohui Zhang ◽  
Lynn Moscinski ◽  
John M. Bennett ◽  
Reza Setoodeh ◽  
Deniz Peker ◽  
...  

Abstract Abstract 2803 Myelodysplastic syndrome (MDS) and T-cell large granular (T-LGL) leukemia are both bone marrow failure disorders. It has been reported in a small number of cases that clonal T-LGL proliferation or leukemia can coincidentally occur with MDS. Also, clonal CD8+/CD57+ effector T cells expansion was detected in as many as 50% of MDS bone marrows [Epling-Burnette, 2007]. How clonal LGL cells that reside in the bone marrow interfere with hematopoiesis remains unclear, particularly in the setting of MDS. We analyzed the clinicopathological features of concomitant MDS and T-LGL, and evaluated bone marrow status for lineage or pan-hypoplasia in these patients. Design: Clinical and pathologic data from patients with a diagnosis of MDS and flow cytometry performed on the peripheral blood between 1/2005 and 12/2009 were reviewed. The concurrent bone marrow biopsies from each patient at the time of flow cytometric analysis were reviewed by two hematopathologists. Bone marrow cellularity, lineage hypoplasia (M:E >5: 1 or <1:2) were documented. Peripheral lymphocyte count and CD3+/CD57+ and CD8+/CD57+ populations by flow cytometry were calculated and T cell gene receptor (TCR) rearrangements were correlated. Results: We performed LGL flow cytometry panel on 76 MDS patients (high grade MDS, n=23; low grade, n=54), as well as TCR gene rearrangements, and identified clonal T-LGL cells in peripheral blood of 37 patients (48.7%), including 15 high grade MDS (40.5%, RAEB-I and RAEB-II), and 22 low grade MDS (59.4%), including RCMD(13), RA(1), RS(1), RCMD-RA(1), RCMD-RS (2), 5q- MDS(1), and MDS unclassifiable(3). The immunophenotype of the T-LGL cells was typically CD3+/CD57+/CD7 dim+/CD5 dim+/CD8+ with variable CD11b,CD11c, CD16, CD56 and HLA-DR. A frequent variant in these MDS patients was CD11b-,CD11c -, CD16+/−, CD56+/−, HLA-DR- and CD62L+.The TCRβ or/and TCRγ gene rearrangements were positive in 35 of the 38 cases (92.1%). The peripheral blood lymphocyte counts were 300–3820 cells/μL (1199±799 cells/μL); the CD3+/CD8+/CD57+ T-LGL cell counts were 30–624 cells/μL (229±154 cells/μL). In comparison, the remaining 39 patients with non-clonal T-LGL included 11 high grade MDS cases, and 28 low grade MDS cases. The peripheral blood lymphocyte counts were 308–2210 cells/μL (1030±461 cells/μL). CD3+/CD57+ cells were 1–425 cells/μL (105±98 cells/μL). There was no identifiable phenotypic features suggestive of clonal T-LGL cells such as dim CD5 and/or dim CD7 with aforementioned aberrant expressions on T-cells, although 7 of the 39 cases had TCRβ or/and TCRγ gene rearrangements. Thirty healthy donors were included for controls with absolute lymphocyte counts of 2136±661 cells/μL and baseline CD3+/CD57+ cells of 162±109 cells/μL. All showed no clonal LGL phenotype and negative TCR gene rearrangements. Since the presence of T-LGL cells may impair bone marrow hematopoiesis, we examined if there are bone marrow status differences between these two groups. All the bone marrows were obtained at diagnosis or not on chemotherapy. The bone marrow cellularity of the MDS patients with clonal T-LGL ranged from <3% to almost 100%, averaging 56%, with 8 cases with dramatic hypocellularity (<3%-20%), while the bone marrow cellularity of the MDS patients without clonal T-LGL ranged from 20% to 90%, averaging 62%, with only 2 cases with mild hypocellularity (20% in 73- and 65-year-old). In addition, among MDS patients with clonal T-LGL cells, 14 of 37 (37.8%; 5 high grade, and 9 low grade) bone marrows had certain lineage hypoplasia, including 3 cases of trilineal hypoplasia, 9 cases of erythroid hypoplasia, and 2 cases of myeloid hypoplasia. In contrast, among 39 MDS patients without T-LGL, there were only 1 bone marrow with trilineal hypoplasia and 3 others with erythroid hypoplasia (10.2%). The difference between the two groups was statistically significant (p=0.004, chi square test). In conclusion, our studies indicate that clonal T-LGL cells expansion is a fairly common finding in high grade as well as low grade MDS. The clonal T-LGL cells have more than one variant immunophenotypes and are typically positive for TCR gene rearrangements. Additionally, we observed that the clonal LGL cells present in MDS bone marrows could be associated with lineage hypoplasia, which, in this respect, might impact clinical treatment. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document