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  (2)
  • Medicine  (2)
Material
Publisher
  • American Society of Hematology  (2)
Language
Years
Subjects(RVK)
  • Medicine  (2)
RVK
  • 1
    In: Blood, American Society of Hematology, Vol. 114, No. 22 ( 2009-11-20), p. 2766-2766
    Abstract: Abstract 2766 Poster Board II-742 (Introduction) B7-H1 (CD274) is a costimulatory/coinhibitory molecule that plays a crucial role in T cell regulation in various immune responses. B7-H1 expression is detected not only on professional antigen-presenting cells but also on some tumor cells. It has been reported that B7-H1 molecules on tumor cells inhibit the proliferation of tumor-specific cytotoxic T-lymphocytes and are associated with poor prognosis in patients with some tumors such as renal and breast cancers. In this study, we investigated whether functional B7-H1 molecules are expressed on blasts from myelodysplastic syndromes (MDS) and, if so, are associated with pathophysiology in specific immune evasion in MDS. (Materials and methods) (1) First, we analyzed the expression of B7-H1 mRNA and protein in MDS cell lines (OIH-1, SKM-1, and F36P) using reverse transcription-PCR and flow cytometry (FCM), respectively. Although F36P cells alone expressed B7-H1 molecules at mRNA and protein levels, interferon (IFN)-γ and more efficiently a combination of IFN-γ and tumor necrosis factor (TNF)-α enhanced or induced B7-H1 expression in all of these cell lines. Notably, the combination of IFN-γ and TNF-α induced nuclear factor (NF) γB activation of MDS cell lines, and the blocking of NFκB activation by pyrrolidine dithiocarbamate (PDTC) or NFκB inhibitor peptide turned off the B7-H1 induction. (2) To investigate whether B7-H1+ MDS blasts have a proliferative advantage, we compared the cell cycle and colony formation between B7-H1+ and B7-H1– cell fractions in F36P cells. B7-H1+ cells had fewer G0/G1-phase cells and more G2/M-phase cells compared with B7-H1– cells. Furthermore, purified B7-H1+ cells formed more colonies than B7-H1– cells in the colony-forming assay using the methylcellulose. (3) We investigated the immunomodulatory effects of B7-H1+ blasts on T cells. When normal CD3+ T cells were cultured with or without irradiated B7-H1+ F36P cells, the B7-H1+ F36P cells had markedly increased percentages of T cells showing apoptosis. The blocking antibody for B7-H1 or PD-1 inhibited the T-cell apoptosis induced by B7-H1+ cells, decreased the percentage of caspase-3+ cells in T cells, and increased T-cell proliferation. (4) We then examined B7-H1 expression on blasts from 41 MDS patients, 20 patients with acute myeloid leukemia transformed from MDS (AML-MDS), and 10 hematologically normal individuals using FCM. B7-H1+ cases, in whom more than 5% of blasts expressed B7-H1 molecules, were observed in 6 of 14 (43%) patients with refractory anemia with excess blasts (RAEB), 6 of 12 (50%) with RAEB in transformation (RAEB-t), and 4 of 20 (20%) AML-MDS patients, but in none of 15 RA patients and normal individuals. Similar to the data obtained using MDS cell lines, B7-H1 expression on blasts purified from MDS patients was induced by stimulation with IFN-γ or TNF-α, and more strikingly by their combination. PDTC inhibited the induction of B7-H1 expression. The percentages of G0/G1-phase cells were lower and those of G2/M-phase cells higher in B7-H1+ blasts than in B7-H1– blasts from patients. Furthermore, in allogeneic coculture, in which B7-H1+ blasts from an MDS patient and CD3+ T cells from healthy volunteers were cultured, T-cell proliferation was significantly augmented by blocking B7-H1 molecules. (5) Finally, we investigated the expression of PD-1, a counterreceptor of B7-H1, on circulating T cells in 34 MDS patients. PD-1 expression on CD3+, CD4+, and CD8+ T cells in MDS patients was significantly higher than that in healthy volunteers. (CONCLUSIONS) Taking the results together, in MDS: 1) B7-H1 expression on blasts occurs more often in the advanced disease stages; 2) B7-H1 expression on blasts may be induced by cytokines derived from the bone marrow environment via NFκB activation; 3) cell cycle and proliferation were more active in B7-H1+ blasts than in B7-H1– blasts; and 4) B7-H1 molecules on blasts suppress T-cell immunity. These findings indicate that B7-H1 molecules on blasts may be involved in the pathophysiology of MDS. 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: 2009
    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. 112, No. 11 ( 2008-11-16), p. 2722-2722
    Abstract: (INTRODUCTION) The B7 family molecules, which play an important role in the immune response by costimulating or coinhibiting T cells via antigen–T-cell receptor interactions, are expressed not only on professional antigen-presenting cells but also on some tumor cells. We previously reported that the expression of B7.2 and B7-H2 on leukemic cells was associated with poor prognosis in acute myeloid leukemia. In this study, we investigated whether functional B7.2 and B7-H2 molecules are expressed on myeloma cells and, if so, whether these molecules are associated with pathophysiology in multiple myeloma (MM). (METHODS AND RESULTS) Using flow cytometry (FCM), we examined the expression of B7.2 and B7-H2 molecules on fresh plasma cells in bone marrow samples from 10 normal individuals, 21 monoclonal gammopathy of undetermined significance (MGUS) patients, and 40 MM patients. The percentages of B7.2+ cells in plasma cells were much higher in MM patients than in MGUS patients or in normal individuals (MM vs. MGUS, P = 0.0128; MM vs. normals, P = 0.0053). When MM patients were divided into two groups, those in whom more than 40% of myeloma cells expressed B7.2 (n = 20, called B7.2high+ MM patients in this study) showed significantly lower levels of hemoglobin and platelets and higher levels of M-protein compared with other MM patients (B7.2−/low MM patients in this study, n = 20). Meanwhile, B7-H2 expression on plasma cells was clearly documented only in 4 patients with intractable disease: 1 plasma cell leukemia, 1 progressive disease, and 2 chemotherapy-resistant MM. Notably, the expression of B7.2 and B7-H2 on myeloma cells was augmented at the refractory stage compared with expression at the initial diagnosis in 2 patients examined. All of the above findings support the concept that these molecules are associated with disease progression in MM. Based on the above data, we speculated that B7.2 or B7-H2 expression on myeloma cells was associated with their proliferative potential. When KMS-27 cells (a human myeloma cell line [HMCL]), part of which expressed these B7 family molecules, were analyzed in FCM, B7.2+ and B7-H2+ cells had significantly fewer G0/G1-phase cells and more G2/M-phase cells compared with B7.2− and B7-H2− cells, respectively. Consistent with these results, when four cell populations (B7.2+, B7.2−, B7-H2+, and B7-H2− cells) were isolated and examined, B7.2+ and B7-H2+ KMS-27 cells proliferated more rapidly in liquid cultures and formed more colonies in semisolid cultures compared with B7.2− and B7-H2− KMS- 27 cells, respectively. The same growth advantage of myeloma cells expressing B7.2 and B7-H2 molecules was also documented in all other HMCLs examined. When B7.2 and B7-H2 expression was induced on HMCL RPMI8226 cells by cultivation with tumor necrosis factor-a, the cell cycle was clearly stimulated. Furthermore, when 293T cells were transfected with either B7.2 or B7-H2 gene or Mock, B7.2 or B7-H2 gene induction induced cell cycle activation. Finally, we examined whether B7.2 and B7-H2 molecules on myeloma cells affect T cell immunology. In the mixed lymphocyte-myeloma reaction using KMS-27 cells and CD4+ T cells, an antagonistic monoclonal antibody (mAb) against B7.2 decreased the production of interleukin (IL)-10 as well as that of interferon (IFN)-g and IL-2. Meanwhile, an antagonistic mAb against inducible costimulator (ICOS), which blocks the B7-H2-ICOS pathways, decreased the production of IL-10 and IFN-g but not that of IL-2. The finding that both B7.2 and B7-H2 molecules on myeloma cells enhanced IL-10 production is particularly interesting because IL-10 not only reduces the antitumor immune response in general but also is a growth factor for myeloma cells. (CONCLUSIONS) Our results show that functional B7.2 and B7-H2 molecules are expressed on myeloma cells and that these molecules may be associated with a proliferative advantage of MM cells and therefore contribute to the pathophysiology of MM.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2008
    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...