scholarly journals Growth of Human Mast Cells from Bone Marrow and Peripheral Blood-Derived CD34+ Pluripotent Hematopoietic Cells

Author(s):  
Geethani Bandara ◽  
Dean D. Metcalfe ◽  
Arnold S. Kirshenbaum
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.


1966 ◽  
Vol 14 (2) ◽  
pp. 167-170 ◽  
Author(s):  
ROBERT S. BRIGGS ◽  
PASQUALE E. PERILLIE ◽  
STUART C. FINCH

By means of an indirect histochemical technique, the intracellular lysozyme of the formed elements of the peripheral blood and bone marrow was estimated. Evidence is presented that monocytes, as well as mature neutrophils and their precursors extending back to the progranulocyte, contain significant amounts of this enzyme. A rare mature eosinophil demonstrated a trace of lysozyme activity. There was no evidence of lysozyme activity in basophils, erythrocytes, megakaryocytes, platelets, plasma cells, tissue mast cells or bone marrow reticuloendothelial cells.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 2441-2441 ◽  
Author(s):  
Steven J. Lemery ◽  
Jamie A. Robyn ◽  
J. Philip McCoy ◽  
Joseph Kubofcik ◽  
YaeJean Kim ◽  
...  

Abstract Hypereosinophilic syndrome is a rare disorder characterized by hypereosinophilia and eosinophil-mediated tissue injury. An imatinib sensitive myeloproliferative variant (MHES) has been described which has a male predominance, and is associated with elevated serum tryptase levels, tissue fibrosis, increased atypical mast cells, and the presence of the fusion oncogene FIP1L1-PDGFRα which has tyrosine kinase activity. The FIP1L1-PDGFRα mutation has been detected in peripheral blood mononuclear cells, however, the hypercellular bone marrow and elevated serum tryptase levels suggest that multiple lineages might be involved in the clonal process. We analyzed peripheral blood from eight patients with the FIP1L1-PDGFRα mutation. Individual patient samples were sorted by flow cytometry to collect greater than 95% pure populations of CD3, CD14, and CD19 cells. Density gradient centrifugation followed by negative selection for CD16, CD3, CD14, and CD19 using an immunomagnetic bead column was used to purify eosinophils to > 99% purity. Bone marrow from one patient was obtained, and mast cells were cultured from CD34 positive cells. Three techniques were used to assay for the presence of the FIPL1-PDGFRα fusion gene: nested RT-PCR, TaqMan quantitative PCR, and FISH. Eosinophils were positive for the fusion gene in all patient samples that were analyzed. Monocytes were also positive in all but one instance. Surprisingly some patients showed positivity in lymphoid lineages as well. The bone marrow derived pure mast cell culture was positive for the mutation, consistent with the elevation of serum tryptase and atypical appearance of mast cells in MHES. In conclusion, although MHES seems to have a multilineage predilection, specific lineages involved may vary between patients. This may reflect differences in the progenitor stage at which the mutation occurs. Whether the pattern of lineage involvement has any relation to the phenotypic expression of the disease remains to be elucidated.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 3064-3064
Author(s):  
Fengchun Yang

Abstract Mutations in the NF1 tumor suppressor gene cause neurofibromatosis type 1 (NF1), a GTPase activating protein for Ras called neurofibromin. NF1 is a genetic disorder that affects approximately 250,000 individuals in the US, Europe, and Japan alone. Neurofibromas, the hallmark of NF1, are complex tumors characterized by tumorigenic Schwann cells, neoangiogenesis, fibrosis, and degranulating mast cells. Studies in experimental models have emphasized the role of inflammatory cells in altering the microenvironment and facilitating malignant outgrowth. Similarly, Parada (Science, 2002) found that nullizygosity of Nf1 in Schwann cells of conditional knockout mice (Krox20;Nf1flox/flox) was necessary but not sufficient for neurofibroma formation and haploinsufficiency of Nf1 in lineages within the tumor microenvironment was required for neurofibroma progression. We previously provided the first genetic, cellular, and biochemical evidence that haploinsufficiency of Nf1 alters Ras activity and cell fates in mast cells (JEM, 2000, 2001) and identified a mechanism underlying the recruitment of mast cells to tumorigenic Schwann cells (JCI 2003). However, it remains unclear whether Nf1 +/− bone marrow derived hematopoietic cells can directly contribute to neurofibroma formation in vivo. To address this question, Nf1+/− or wildtype (WT) EGFP+ bone marrow (BM) was adoptively transferred into lethally irradiated Krox20;Nf1flox/flox mice and cohorts were followed prospectively for tumor formation using positron emission tomography and computerized axial tomography. Mice transplanted with Nf1+/− but not WT BM developed progressive enlargement of the trigeminal nerve, dorsal root ganglia, peripheral nerves, and motor paralysis similar to Krox20;Nf1flox/− mice that are haploinsufficient at Nf1 in all lineages of the tumor microenvironment. Postmortem analysis revealed that Krox20;Nf1flox/flox mice transplanted with Nf1+/− BM had cellular neurofibromas containing Schwann cells, fibroblasts, blood vessels and mast cells, which closely resembled the cellular architecture of human neurofibromas. Mice transplanted with WT BM did not develop neurofibromas. These studies establish that recruitment of Nf1 +/− BM derived cells to the neurofibroma microenvironment is directly linked to neurofibroma formation and progression. Given our observations, therapies which prevent both the recruitment and the tumor promoting functions of Nf1 +/− hematopoietic cells in neurofibroma formation are currently being tested in vivo as pre-clinical trials.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 2789-2789
Author(s):  
Ioanna Bazdiara ◽  
Despina Pantelidou ◽  
Athanasios Anastasiadis ◽  
Vassilios Papadopoulos ◽  
Dimitrios Margaritis ◽  
...  

Abstract Evolving data demonstrate the pathogenetic significance of chromosomal ends telomeres and telomerase activity in the molecular pathogenesis of many hematological disorders. Furthermore, the presence of eroded telomeres and enhanced telomerase activity in hematopoietic cells has been associated with poor prognosis both in myeloid and lymphoid malignancies. The aim of the present study was to evaluate telomere length and telomerase activity in patients with Ph1-negative Chronic Myeloproliferative Disorders (Ph−-CMPD) either at diagnosis or during the course of the disease and to assess their possible clinical utility. Sixty-six bone marrow and 60 peripheral blood samples were obtained from 80 Ph−-CMPD patients (aged 58.57±16.42 years) and 18 healthy age-matched controls (aged 53.94±15.16 years). Thirty-six patients diagnosed suffering from Polycythemia Vera, 36 from Essential Thombocythemia, 4 from Idiopathic Myelofibrosis and 4 from Unclassified CMPD. Twenty-six samples were studied at diagnosis, whereas 54 during the course of the disease. Telomere length analysis of individual chromosome ends was performed on bone marrow metaphases using Telomere/Centromere Quantitative-Fluorescence In Situ Hybridization (T/C Q-FISH) (Dako A/S, Denmark). Telomerase activity was determined in bone marrow purified CD34(+) and CD20(+) cells as well as in peripheral blood CD3(+) T-lymphocytes and granulocytes with the PCR-based Telomeric Repeat Amplification Protocol (TRAP) assay (Roche, Germany). Gene expression of telomerase-associated proteins (hTERT, hTER, TEP1, TRF-1 and TRF-2) was assayed by Real-Time Multiplex PCR (Maximbio, USA). Ph−-CMPD patients showed significantly more eroded telomeres (P=0.010) and increased telomerase activity in CD34(+) cells (P=0.005) compared to healthy age-matched individuals. However, there was no statistical difference in telomere length (P=0.451) and enzyme activity (P=0.538) among different groups of Ph−-CMPD. Telomerase activity was not detected in the remaining hematopoietic cells both in patients and healthy controls, which was closely correlated with downregulation in hTERT mRNA expression. hTER, TEP1, TRF-1 and TRF-2 showed no apparent differential expression of mRNA in all hematopoietic cell fractions. Chromosomal aberrations (+8, +9, del13q14, del20q12) were found by FISH in 37% Ph−-CMPD patients with reduced telomere lengths (P=0.001) and enhanced telomerase activity (P=0.014), especially during the course of the disease (P=0.028). The patients with shortened telomeres displayed a higher incidence of having thrombotic or hemorrhagic events during follow-up (P=0.011), treatment failure (P=0.024) and disease progression to myelofibrosis, myelodysplastic syndromes, secondary leukemia or death (P=0.137). Nevertheless, telomerase expression was not correlated with the above complications. The event free survival (survival without complications, e.g. myelofibrosis, myelodysplastic syndromes, secondary leukemia and death) was significantly shorter in patients with reduced telomere lengths (Log Rank P=0.033), who demonstrated a 7,71-fold higher probability of having complications within five years from the initial diagnosis (95% CI=2,04–31,49 P<0.001). In conclusion, accelerated telomere shortening may not be prevented or restored by telomerase activity in most of the Ph−-CMPD myeloid cells. Loss of telomere stability seems to predispose to further genetic events such as chromosomal rearrangement and consequently to trigger off a multistage neoplastic transformation of these diseases. Moreover, the negative correlation between telomere length and survival probability of Ph−-CMPD patients is indicative that telomere dynamics may serve as a useful prognostic tool for these patients.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2267-2267
Author(s):  
Eva Sahakian ◽  
John Powers ◽  
Jie Chen ◽  
Allison Distler ◽  
Jennifer Rock-Klotz ◽  
...  

Abstract Histone Deacetylase 11 (HDAC11) is the newest member of the HDAC family of enzymes, which we have previously reported to function as a negative regulator of IL-10 expression in macrophages and dendritic cells. Thus far, its role in other hematopoietic cells has not been completely elucidated. We hereby report for the first time a lineage-restricted over-expression of HDAC11 in neutrophils, committed neutrophil precursors and myeloid leukemias exhibiting neutrophilic differentiation demonstrating a novel physiological role of HDAC11 as a negative regulator of neutrophil cytokine production. Leukocyte subpopulations from murine bone marrow and spleen were flow-sorted and analyzed by qRT-PCR for HDAC11 mRNA, revealing a higher level of mRNA expression on neutrophils and promyelocytes, as compared to monocytes and lymphoid subsets. Similarly, sorted human peripheral blood leukocytes from normal donors, showed higher levels of HDAC11 mRNA in neutrophils, as compared to monocytes. To further investigate the transcriptional activity of HDAC11 in myeloid and lymphoid cells, we utilized a HDAC11 promoter-driven eGFP reporter mice, where eGFP expression indicates HDAC11 transcription (Heintz, N Nat. Rev. Neuroscience 2001). Using multiparametric flow cytometry with lineage-specific markers on this mouse model, we confirmed a marked over-expression of HDAC11 on neutrophils, compared to other subpopulations including monocytes, B-cell, T-cells, NK cells and plasma cells. Furthermore, analysis of bone marrow hematopoietic cells revealed a swift over-expression of HDAC11 at the promyelocyte stage of neutrophil differentiation, with low to undetectable expression in upstream uncommitted common myeloid progenitors and lineage-unrelated monocytic precursors. To study whether this lineage-specific overexpression applies to malignant processes, we studied human cell lines and found overt overexpression of HDAC11 in the acute promyelocytic leukemia cell line NB4, as compared to the myeloblastic cell line Kasumi and two monocyte/macrophage cell lines U937 and THP1. Moreover, in-vitro maturation of the differentiation-inducible myeloid cell line HL60 demonstrated a marked increase in HDAC11 mRNA, paralleling the acquisition of nuclear segmentation characteristic of neutrophil maturation. In order to investigate the physiologic role of HDAC11 overexpression on neutrophils, we utilized a model of germline-HDAC11KO mice. Surprisingly, highly purified neutrophils lacking HDAC11 showed an overt overproduction of TNF-alpha and IL-6 upon stimulation with LPS, as compared to their wild type counterparts. We hereby report a previously un-described lineage-specific over-expression of HDAC11 in neutrophils and its precursors, which actively functions as a physiological repressor of cytokine production and possibly involved in their regulation. Given the predominance of neutrophils which account for 70% of leukocytes in the peripheral blood, and their pivotal role in the first line of defense, results highlight a novel mechanism for HDAC11, as a key regulator and modulator of neutrophil cytokine production with potential implications for autoimmunity, inflammation, and infection. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
1998 ◽  
Vol 92 (5) ◽  
pp. 1728-1734 ◽  
Author(s):  
Susanne Schnittger ◽  
Bernhard Wörmann ◽  
Wolfgang Hiddemann ◽  
Frank Griesinger

Partial tandem duplication within the MLL gene has recently been described as a novel genetic alteration in acute myeloid leukemia (AML). It has been associated with trisomy of chromosome 11, but was also identified in AML patients with normal karyotypes. The current study was performed to investigate whether MLL duplications are restricted to AML, and hence whether they may also occur in normal hematopoietic cells. MLL-duplication transcripts were analyzed by nested reverse-transcriptase polymerase chain reaction (RT-PCR) in peripheral blood in two groups of 45 and 20 patients, respectively, as well as in two bone marrow samples from healthy volunteers. Duplications were detected in two independent nested RT-PCR experiments in the peripheral blood samples of 38 of 45 (84%) and 20 of 20 (100%) of the two groups and in both bone marrow samples. On this basis, MLL duplications seem to occur frequently in a subset of cells in normal hematopoiesis. The type of partially duplicated MLL transcripts varied substantially. Three transcripts were identical to those known from AML. In addition, four new transcripts were characterized. Three of these four were in frame and potentially translatable. MLL duplications were also detected by seminested genomic PCR with intron 9– and intron 1–specific primers in 20 of 20 peripheral blood samples studied, indicating that the duplications are genomically fixed at the DNA level and are not an RT-PCR artifact. In summary, MLL duplications are regularly generated by homologous ALU recombination in a small number of hematopoietic cells of most or even all healthy donors. These data suggest that MLL duplications are not implicated in the malignant transformation in AML, or alternatively, that only a few cells will acquire additional oncogenic mutations necessary to establish the malignant phenotype of AML. © 1998 by The American Society of Hematology.


Sign in / Sign up

Export Citation Format

Share Document