GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
Filter
  • American Society of Hematology  (5)
  • 1
    In: Blood, American Society of Hematology, Vol. 120, No. 21 ( 2012-11-16), p. 1195-1195
    Abstract: Abstract 1195 Background: In the murine primitive hematopoietic stem cell (HSC) compartment, MPL/thrombopoietin (THPO) signaling plays an important role for the maintenance of adult quiescent HSCs. However, the role of MPL/THPO signaling in the human primitive HSC compartment has not yet been elucidated. We have previously identified very primitive human cord blood (CB)-derived CD34-negative (CD34−) severe combined immunodeficiency (SCID)-repopulating cells (SRCs) using the intra-bone marrow injection (IBMI) method (Blood 2003:101;2924). Recently, we developed a high-resolution purification method for the primitive CD34− SRCs using 18 lineage (Lin)-specific antibodies (Exp Hematol 2011:39:203). In this study, we investigated the functional significance of the MPL expression in the human primitive CD34− SRCs (HSCs). Materials and Methods: First, we sorted 18Lin−CD34+/−MPL+/−cells by FACS. Thereafter, these four fractions of cells were analyzed for their hematopoietic colony-forming capacities (CFCs), the maintenance/production of CD34+ cells in cocultures with human bone marrow-derived mesenchymal stromal cells (DP MSCs) (Blood 2010:116:1575). Finally, these four fractions of cells were transplanted by IBMI into NOD/Shi-scid/IL-2R ƒÁcnull (NOG) mice to investigate their long-term (LT) repopulating capacities. We performed primary, secondary, and tertiary transplantations for up to 18 months. Results: The CFCs of highly purified 18Lin−CD34−cells were quite unique, since they yielded mainly erythroid-bursts (BFU-E) and erythro/megakaryocytes-containing mixed colonies (CFU-EM). Interestingly, they showed very weak myeloid CFCs. Moreover, 200 18Lin−CD34−MPL+cells yielded about 150 colonies, consisting of 20% BFU-E, 20% colony-forming unit-megakaryocytes, and 60% CFU-EM in the presence of 10% platelet-poor plasma, THPO, IL-3, and erythropoietin. The phenotypic and functional characteristics of the 18Lin−CD34+/−MPL+/−cells were further investigated by cocultures with DP MSCs. After 7 days of coculturing the 18Lin−CD34+MPL+/−cells, the total number of cells was observed to expand by about 500 times, 40 % of which consisted of CD34+ cells. On the other hand, the 18Lin−CD34−MPL+/−cells expanded only by about 100 times, 10 to 20% of which consisted of CD34+ cells. Interestingly, the lineage differentiation potentials of the observed 18Lin−CD34+/−MPL+/−cells were significantly different. The percentages of myeloid cells generated from the 18Lin−CD34+MPL+/−cells were significantly greater than those of the 18Lin−CD34−MPL+/−cells. On the contrary, 18Lin−CD34−MPL+cells generated significantly higher percentages of CD41+ cells compared to the other fractions. These observations suggested that the differentiation potentials of 18Lin−CD34+/−MPL+/− cells were different and that these four cell populations contained distinct classes of hematopoietic progenitors as well as HSCs. We next investigated the SRC activities of the 18Lin−CD34+/−MPL+/−cells using NOG mice. In the primary recipient mice, all 18 mice (9 for each cell type) received CD34+MPL+/−SRCs were highly repopulated with human CD45+ cells. On the other hand, 11/13 mice receiving CD34−MPL+SRCs and 12/12 mice receiving CD34−MPL−SRCs showed human cell repopulation. Interestingly, these CD34+/−MPL+/−SRCs showed different repopulation kinetics. The CD34+MPL+ SRCs showed a rapid and sustained repopulating pattern (Fig.1-A). In contrast, the repopulation rates in the mice receiving CD34+MPL− and CD34−MPL+/− SRCs gradually increased, and thereafter reached a high level of repopulation at 18–24 weeks after the transplantation (Fig.1-A, B). All of the primary recipient mice that received CD34+MPL+/− SRCs showed a secondary repopulating capacity. However, only the mice that received CD34+MPL− SRCs showed a tertiary repopulating capacity. Very interestingly, the primary recipient mice that received CD34−MPL− SRCs showed a distinct secondary repopulating capacity. Tertiary transplantation experiments in these mice are now underway in our laboratory. Conclusion: These results clearly indicated that both the CD34+/− SRCs not expressing MPL sustained a LT ( 〉 1 year) human cell repopulation in NOG mice. Therefore, these findings suggest that the functional significance of the MPL expression in the human primitive HSCs is different from that in murine primitive HSCs. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2012
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 4707-4707
    Abstract: Abstract 4707 Reduced-intensity stem cell transplantation (RIST) has come to be generally accepted as a method of allogeneic stem cell transplantation (SCT) for patients considered ineligible for myeloablative preparative regimens because of advanced age or comorbidities. We have recently reported unmanipulated nonmyeloablative HLA-haploidentical SCT using a conditioning treatment consisting of fludarabine, busulfan and anti-T-lymphocyte globulin and graft-versus-host disease (GVHD) prophylaxis consisting of tacrolimus and methylprednisolone (1 mg/kg) (Biol Blood Marrow Transplant 2006; 12:1073). In that study, the incidence of severe GVHD was only 10%. One of the mechanisms for such a low incidence of GVHD may have been caused by a reduced intensity of conditioning. Less intensive regimens should be associated with lower toxicity, a lower release of inflammatory cytokines, and potentially less GVHD; however, the mechanisms remain to be determined. Thus, using a murine MHC-haploidentical BMT model, BDF1(H-2b/d)®B6C3F1 (H-2b/k), that we established, we examined the influence of an intensity of conditioning treatment on GVHD. Recipient mice received T-cell-depleted bone marrow (5×106) and spleen cells (2×107) after total body irradiation (TBI) 13 Gy (myeloablative group) or 5 Gy (RIST group). Both groups of mice rapidly achieved donor engraftment. Recipients in the RIST group showed significantly fewer GVHD signs than those in the myeloablatve group. Histopathological examination of the myeloablative group on day 14 revealed various pathological changes in intestine (in particular large intestine) compatible to GVHD. In contrast, intestine samples from the RIST group showed few pathological changes with much less infiltration of donor T cells. Consequently, all recipients in the myeloablative group had died of GVHD by day 60, while all recipients survived for more than 3 months. These results clearly showed that the intensity of conditioning treatment influenced on the severity of GVHD and survival of recipients. Next, we investigated the mechanisms by which reduced intensity of conditioning ameliorated GVHD. Transplantation was performed using spleen cells that were labeled with the fluorescent cytoplasmic dye, carboxyfluorescein diacetate succinimidyl ester (CFSE), and cells in secondary lymphoid organs were analyzed by flow cytometry. The number of donor T cells in mesenteric lymph nodes on day 7 of the RIST group was significantly lower than that of the myeloablative group. In addition, a significantly increased number of host CD4+ T cells were recruited to secondary lymphoid organs on day 4 in the RIST group compared with the myeloablative group. An increased number of donor or host regulatory (Foxp3+CD4+) T cells were also observed in the RIST group. The levels of IFN gamma or IL-4 in lymphoid organs of the RIST group were higher than those of the myeloablative group. These results strongly suggest that host immune cells that survived conditioning treatment or cytokine milieu in secondary lymphoid organs contributed to the suppression of donor T cells during the initiation of GVHD. In addition, the expression of Th1 chemokine receptor, CXCR3, on donor T cells in secondary lymphoid organs and the expression levels of CXCL9, CXCL10, and CXCL11, ligands for CXCR3, in the large intestines were relatively lower in the RIST group, suggesting that the migration ability of donor T cells into GVHD target organs was negatively influenced by the intensity of conditioning. In conclusion, we showed that reduced intensity of conditioning improved the severity of GVHD, and that recipient immune cells, including regulatory T cells, together with reduced expression of inflammatory cytokines or chemokines, contributed to the improvement of GVHD in RIST. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2010
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 120, No. 21 ( 2012-11-16), p. 2345-2345
    Abstract: Abstract 2345 Background: Ratajczak and his colleagues identified a unique population of very small embryonic-like (VSEL) stem cells in adult mouse bone marrow (BM) (Leukemia 2006:20;857). These VSELs are; 1) very small (∼4 μm); 2) express pluripotent stem cell markers, such as Oct4, Nanog, SSEA-1, and Rex-1; 3); responsive to a SDF-1 gradient; 4) possess large nuclei that contain euchromatin. It is very interesting to note that VSELs possess the potential to differentiate into 3 germ layers in vitro and in vivo, thereby contributing to tissue/organ regeneration. These VSELs were isolated as lineage-negative (Lin−), Sca-1-positive (Sca-1+), CD45-negative (CD45−) cells by FACS. However, the incidence of VSELs in BM-derived mononuclear cells is ∼0.01%. Therefore, it is difficult to isolate VSELs very effectively. This study describes our recently developed highly efficient method for isolating VSELs using enzymatic treatment of murine bone. Materials and Methods: Murine BM nucleated cells (BMNC) were isolated from BM flushed from the pairs of femurs and tibiae of 8 week-old C57BL/6 mice. Erythrocytes were removed using a hypotonic solution. Then the remaining bone tissues were thoroughly washed using PBS- with 2% FCS. These bone tissue specimens were crushed in a mortar and then incubated in cell dissociation buffer containing a-medium with 5% FCS supplemented with 1.5 mg/ml type I collagenase and 2 mg/ml dispase at 37°C for 1 hour. Next, the BMNCs and bone-derived nucleated cells (BDNCs) were stained with various monoclonal antibodies, including anti-lineages, anti-CD45, anti-Sca-1, anti-CXCR4, anti-CD133, and anti-PDGFRα, and then were used for subsequent FACS analyses. Results: The R1 gate was set on the FSC channel using 4 and 10 μm synthetic beads, based on the predicted very small size of VSELs. The VSELs were isolated from BMNCs and BDNCs by multicolor FACS, as a population of Lin−Sca-1+CD45− cells (Fig. 1A). The incidences of VSELs in the BMNCs and BDNCs were 0.001% and 0.1%, respectively. Therefore, the enzymatic treatment of bone tissues yielded about 100 times the efficiency for the isolation of VSELs (Fig. 1B). The bone-derived (BD) VSELs were small ( 〈 5 μm) and possessed a relatively large nucleus surrounded by a narrow rim of cytoplasm. They expressed CD133, but not PDGFRα. However, they weakly expressed CXCR4. The gene expression profiles were analyzed using real time quantitative PCR (RQ-PCR) to evaluate the expression of ES cell markers (Oct4, Nanog, Rex1, Dppa3), HSC (KSL) markers (c-kit, Tal1, GATA2), and MSC markers (Nestin, Ang1, CXCL12, VE-Cadherin). Unexpectedly, BD VSELs expressed high levels of Nestin and Cadherin. However, they expressed weak levels of Oct4 and Nanog. The gene expression profile of the BD VSELs was clearly distinct from the well-defined populations of ES cells, KSL cells, and MSCs. Interestingly, the number of these BD VSELs significantly increased after the induction of liver injury by carbon tetrachloride administration. They were then most likely mobilized into the peripheral blood (PB). G-CSF did mobilize KSL cells into PB, as previously reported. However, G-CSF did not mobilize the BD VSELs. The effects of sRANKL on the mobilization of BD VSELs were examined in vivo. Interestingly, the number of BD VSELs significantly increased 2–3 days after the administration of sRANKL. However, the number of VSELs in PB did not increase. These results suggest that BD VSELs actively proliferated after liver injury and bone resorption. Conclusion: The present data suggest that the majority of the Lin−Sca-1+CD45− cells reside in the bone tissue. BD VSELs resemble BM-derived VSELs. However, a RQ-PCR analysis revealed that the gene expression profile of BD VSELs was different from those of the previously reported BM-derived VSELs. Further studies will therefore be required to elucidate their stem cell characteristics and the potential relationship between BD VSELs and BM-derived VSELs. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2012
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 4117-4117
    Abstract: Abstract 4117 Related haploidentical donors, as cord blood, can be alternative donor sources in stem cell transplantation (SCT). Severe GVHD, however, has interfered the progress of haploidentical SCT (haploSCT). To deal with this strong GVHD, T cell depletion has usually been used in US and European countries. In order to pursue the controllable GVL effect by T cells, we have performed unmanipulated haploSCT using myeloablative or reduced intensity preconditioning regimen accompanied with intensified GVHD prophylaxis, including steroids. In this meeting, we will summarize our experience of haploSCT for more than ten years. From August 1998 to September 2010, we have performed 351 cases of haploSCT (all cases were HLA 2–3 antigen mismatched in GVH direction). Patients' characteristics are sex: male 186, female 168, age: 16–65 years old (median 39), disease: AML/MDS 149, ALL 81, ML 67, others 54. Eighty-three percent of cases underwent SCT in non-complete remission (non-CR) status. Patients under 45 years old underwent myeloablative preconditioning regimen consisting of FLU/CA/CY/TBI 8Gy (haplo-full, n=100), and patients over 45 years old or with comorbidities or repetitive SCT (including second to fifth SCT) underwent reduced intensity preconditioning regimen consisting of FLU/(CA)/BU/ATG or FLU/(CA)/MEL/ATG (haplo-mini, n=251). High dose Ara-C (CA) was optional to reduce tumor burden. As ATG, ATG (Fresenius) 8mg/kg, or thymoglubulin (genzyme) 2–4mg/kg were integrated into conditioning treatments mainly for reduced-intensity transplantation. GVHD prophylaxis consisted of taclolimus (TAC), methylprednisolone (mPSL) 2mg/kg/day, short term MTX, and mycophenolate mofetil (MMF) 15mg/kg/day in haplo-full, and TAC, and mPSL 1mg/kg/day in haplo-mini, respectively. For elderly patients over 50 years old in haplo-mini, MMF was added. Hematopoietic engraftment in haploSCT was as rapid as that in HLA-identical SCT, except 10 cases of graft rejection. The median time to reach a neutrophil account of 〉 0.5 × 109/l was 10 days for haplo-mini and 13 days for haplo-full. Platelet recovery was achieved in 66 % and 60% of patients undergoing haplo-mini and haplo-full, respectively. The median time to reach a nontransfused platelet count of 3 20 × 109/l was 22 days for haplo-mini and 33 days for haplo-full. Sixty percent of haplo-mini patients and 54 % of haplo-full patients did not develop acute GVHD. Acute GVHD (grade II-IV) was observed in 20% for haplo-mini and 36 % for haplo-full. Overall survival at five years was 30% for haplo-full and 40% for haplo-mini, respectively. If limited to CR cases, overall survival reached over 60% in haplo-mini. There is no difference in survival rate among patients' diseases. In multivariate analysis on survival using variables, including disease status before transplantation, haplo-full vs haplo-mini, mismatches in GVH direction, mismatches in HVG direction, patients' age, and the number of transplantation times, the disease status (CR) was found to be only a significantly favorable factor (P= 0.0026). Unmanipulated haploSCT is feasible and effective for refractory diseases. ATG dose used in haplo-mini is critical, and rather low compared with that of European cases reported so far. Although it should be too early to refer long term outcome, unmanipulated haploSCT could be considered as an option to control refractory diseases. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2011
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 122, No. 21 ( 2013-11-15), p. 1177-1177
    Abstract: We have previously identified very primitive human cord blood (CB)-derived CD34-negative (CD34-) severe combined immunodeficiency (SCID)- repopulating cells (SRCs) using the intra-bone marrow injection (IBMI) method (Blood 2003:101;2924). A series of our studies suggests that the identified CD34- SRCs are a distinct class of primitive hematopoietic stem cell (HSC) and that they are at the apex of human HSC hierarchy. Recently, we developed a high-resolution purification method for primitive CD34- SRCs using 18 lineage (Lin)-specific antibodies, which can enrich CD34- SRC at 1/1,000 level (Exp Hematol 2011: 39:203). In the present study, we tried to identify the positive marker of CD34- SRCs in order to further purify and characterize the CD34- SRCs (HSCs). Materials and Methods First, we extensively analyzed candidate positive markers, including known HSC markers and various adhesion molecules by FACS using highly purified CB-derived 18Lin-CD34+/- cells. Finally, we identified CD133 as a positive marker of human CB-derived CD34- SRCs. Then, CB-derived 18Lin- CD34+/-CD133+/- cells were sorted by FACS, and hematopoietic stem/progenitor cell (HSPC) capacities of these four fractions of cells were extensively investigated. HSPC capacities were evaluated using (1) colony-forming cell (CFC) assays, (2) measurement of maintenance/production of CD34+ cell capacities in co-cultures with human bone marrow-derived mesenchymal stromal cells (BM-MSCs) (Blood 2010:24:162), (3) SRC activities using NOG mice, (4) limiting dilution analyses (LDA) to determine the SRC frequency in the 18Lin-CD34-CD133+ fractions, and (5) comparison of gene expression profiles between 18Lin-CD34+/-CD133+/- cells by real-time RT-PCR. Results Seventy-five percent of 18Lin-CD34+ and 13.5% of 18Lin-CD34- cells highly expressed CD133. In the CFC assays, the plating efficiencies of 18Lin-CD34+CD133+, CD34+CD133-, CD34-CD133+ and CD34-CD133- cells were 57%, 65%, 39% and 19%, respectively. Interestingly, most of 18Lin-CD34-CD133+/- cells formed erythroid-bursts (71% and 73%) and erythro/megakaryocytes-containing mixed colonies (25% and 27%). On the contrary, they formed few granulocyte/macrophage colonies (4.2% and 0%). Then, we co-cultured these four fractions of cells with human BM-MSCs. One thousand of 18Lin-CD34+/-CD133+/- cells were seeded into each well and cells were co-cultured for 7 days in the presence of SCF+TPO+FL+IL-3+IL-6 +G-CSF. Both the 18Lin-CD34-CD133+/- cells produced CD34+ cells. However, the percentage and absolute number of CD34+ cells produced from 18Lin-CD34-CD133+ cells (31.7 % and 3.2 x 104 cells) were greater than those of 18Lin-CD34-CD133- cells (13.2 % and 0.4 x 104 cells). In addition, both the 18Lin-CD34- CD133+/- cells generated higher percentages (13.5 % and 11.5%) of CD41+ cells compared to those of the 18Lin- CD34+CD133+/- (1.8% and 4.2%) cells. Collectively, 18Lin-CD34+/-CD133+/- cells showed different in vitro lineage differentiation potentials. Then, these four fractions of cells were transplanted into NOG mice by IBMI. We performed primary and secondary transplantations for up to 36 weeks. In the results, all of the mice received 18Lin-CD34+CD133+ cells (n = 5) or 18Lin-CD34-CD133+ cells (n = 9) showed primary and secondary human CD45+ cell repopulations. However, neither 18Lin-CD34+CD133- cells nor 18Lin-CD34-CD133- cells showed human cell repopulations (n = 6 in each group). These results clearly demonstrated that the CD133 expression clearly segregated SRC activities in the 18Lin-CD34+/- cells. Moreover, LDA demonstrated that the frequency of SRCs in the 18Lin-CD34-CD133+ fraction was 1/142. Interestingly, HSC self-renewal maintenance genes, such as Notch1, HoxB4, HoxA9, and Bmi-1, were highly expressed in both 18Lin-CD34+/-CD133+ cells. Conclusion These results clearly demonstrated that CD133 is a positive marker of human CB-derived CD34- SRCs (HSCs). Furthermore, CD133 segregated SRC activities of 18Lin-CD34- as well as 18Lin-CD34+ cells in its positive fractions. More importantly, these findings suggest that number of CD133+ cells in cord blood units is a more appropriate marker to detect/predict HSC potentials in cord blood stem cell transplantation in comparison to currently used CD34+ cell numbers. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2013
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...