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  (11)
  • 1
    In: Blood, American Society of Hematology, Vol. 120, No. 21 ( 2012-11-16), p. 3280-3280
    Abstract: Abstract 3280 CD4+CD25+Foxp3+ regulatory T cells (Treg) play an important role in the maintenance of self-tolerance and immune homeostasis and Treg deficiency contributes to the development of autoimmune diseases. CD4Treg, conventional CD4 T cells (Tcon) and CD8 T cells are derived from lymphocyte progenitor cells that differentiate into distinct functional subsets in the thymus before export to the peripheral circulation. As T cells differentiate and expand in the periphery, each T cell subset is differentially regulated and subjected to distinct homeostatic signals. For example, interleukin-2 (IL-2) is a critical regulator of Treg development, expansion and survival and lack of IL-2 results in selective Treg deficiency. In regulating Treg homeostasis, IL-2 has multiple and distinct effects on Treg differentiation, proliferation and susceptibility to apoptosis. To determine the mechanism whereby IL-2 affects susceptibility of Treg to apoptosis, we used a new flow cytometry-based assay (BH3 profiling) to measure the mitochondrial membrane depolarization in response to a panel of pro-apoptotic BH3 peptides (BIM, BID, BAD, NOXA, PUMA, BMF, HRK). This assay allowed us to compare “priming” which we define as susceptibility to BH3 peptide-induced mitochondrial membrane depolarization in different T cell subsets, including CD4 Treg, CD4 Tcon and CD8 T cells. We also examined cell surface expression of CD95 death receptor (Fas) and cytoplasmic expression of Bcl-2 and Ki67 as additional measures of susceptibility to apoptosis and proliferation in each subset. In resting blood obtained from healthy donors (n=10), CD4 Treg were more “primed” than either CD4 Tcon or CD8 T cells when exposed to several BH3 peptides (PUMA, BMF and the combination of BAD+NOXA). CD4 Treg were also found to have decreased expression of Bcl-2 and increased expression of CD95 and Ki67 compared to CD4 Tcon or CD8 T cells. Thus, Treg in healthy individuals have higher proliferative activity and are more susceptible to apoptosis than other major T cell subsets through both mitochondrial and death receptor pathways. To establish the functional effects of TCR stimulation and IL-2, CD4 Treg, CD4 Tcon and CD8 T cells were purified by cell sorting and cultured for 5–6 days with or without TCR stimulation (1μg/ml anti-CD3 + 1μg/ml anti-CD28) and IL-2 (100 IU/ml). Results were compared to cells cultured in media alone. Results are summarized in the table below. CD4 Tcon and CD8 T cells responded in a similar fashion to either TCR stimulation alone or TCR plus IL-2. This response included increased BH3 priming, reduced expression of Bcl-2, increased expression of CD95 and increased proliferation (Ki-67). IL-2 alone had no effect on CD4 Tcon or CD8 T cells. In contrast, TCR stimulation alone had no effect on CD4 Treg but IL-2 alone reduced BH3 priming and increased expression of Bcl-2. Combined TCR stimulation plus IL-2 in Treg increased BH3 priming, reduced expression of Bcl-2, increased expression of CD95 and increased proliferation. Thus, TCR stimulation reversed the anti-apoptotic effects of IL-2 alone and markedly increased susceptibility of Treg to apoptosis. When compared with CD4 Tcon and CD8 T cells, these studies demonstrate distinct effects of TCR stimulation and IL-2 on both mitochondrial and death receptor pathways of apoptosis in CD4 Treg and define mechanisms whereby TCR stimulation and IL-2 interact to regulate Treg homeostasis. Table 1. Effects of in vitro TCR stimulation and IL-2 on apoptotic pathways of T cell subsets TCR Stimulation IL-2 TCR + IL2 BH3 priming Bcl-2 CD95 Ki67 BH3 priming Bcl-2 CD95 Ki67 BH3 priming Bcl-2 CD95 Ki67 CD4 Treg – – – – ↓ ↑ – – ↑ ↓ ↑ ↑ CD4 Tcon ↑ ↓ ↑ ↑ – – – – ↑ ↓ ↑ ↑ CD8 ↑ ↓ ↑ ↑ – – – – ↑ ↓ ↑ ↑ 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. 122, No. 21 ( 2013-11-15), p. 3113-3113
    Abstract: We have previously demonstrated that Th17 cells, which produce IL-17A, are significantly elevated in peripheral blood and bone marrow (BM) of patients with Multiple Myeloma (MM) and IL-17A promotes MM cell growth and survival, both in vitro and in vivo via IL-17A receptor. We have recently evaluated and observed that anti-IL-17A monoclonal antibody (mAb) significantly inhibited MM cell growth in vitro, while IL-17A induced proliferation of MM cells compared to control. We have also observed significant down-regulation of IL-6 production by anti-IL-17A mAb in MM-BMSC co-culture. Importantly, the administration of anti-IL-17A mAb weekly for 4 weeks in the SCIDhu model of human myeloma, where MM cells grow within the human microenvironment in mice led to a significant inhibition of tumor growth compared to the control mice. This remarkable activity of anti-IL17 mAb raised the question of whether the myeloma cells themselves are a possible source of IL-17. In this study, we used transcriptome sequencing (RNA-Seq) data to evaluate the expression of IL-17A in primary CD138+ myeloma cells (N=17) compared to normal plasma cells (NPC) (N=5). Whereas none of the NPCs expressed IL-17A, it was significantly over-expressed in majority of MM cells. In addition, these data also showed that the expression of other IL-17 family members (IL-7B, C, D, E & F) and Th17-associated pro-inflammatory cytokines (IL-21, IL-22 & IL-23) were not significantly elevated in primary myeloma cells compared to normal donor plasma cells. We further validated these observations by IL-17 immunoblot showing IL17 expression in all MM cell lines and 10 out of 14 primary patient MM cells; confirmed IL-17 expression in MM cells by quantitative RT-PCR, and flow cytometry and by immuno-histochemistry and confocal microscopy. We observed that IL-17 knock down by IL-17-specific siRNA inhibited MM cell growth as well as their ability to induce IL-6 production in co-cultures with BMSC. Finally, expression profile data from 172 uniformly treated patients showed that patients with lower IL-17A expression had superior overall survival compared to those with higher expression. These data confirms that MM cells express IL-17 and targeting it with a mAb will abrogate the autocrine loop making it an attractive therapeutic target. 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 ...
  • 3
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 2496-2496
    Abstract: Reconstitution of T cell function after allogeneic HSCT is dependent on the balanced recovery of CD4+Foxp3+ regulatory T cells (Treg) and CD4+Foxp3- conventional T cells (Tcon). While Tcon are required for effector T cell function, Treg play an essential role in the maintenance of immune tolerance after allogeneic HSCT and prevention of graft versus host disease (GVHD). To examine the reconstitution of Treg and Tcon after HSCT and identify mechanisms that contribute to homeostatic imbalance of these T cell subsets, we undertook a prospective analysis of 188 adult patients (median age 54y) with hematologic malignances who underwent T-cell replete allogeneic HSCT. Patients received either myeloablative (MAC; n=80) or reduced-intensity conditioning (RIC; n=108). GVHD prophylaxis differed in these two cohorts as RIC patients received tacrolimus/methotrexate/sirolimus-based regimens and MAC patients received tacrolimus/methotrexate-based regimens. Serial blood samples (total n=739) obtained at 1, 2, 3, 6, 9 and 12 months after transplant were characterized by flow cytometry with a panel of intracellular and surface markers designed to quantify phenotypically and functionally distinct subsets of Treg, Tcon and CD8 T cells in each sample. Likely reflecting the prophylactic administration of sirolimus for the first 6 months post-HSCT after RIC conditioning, recovery of absolute CD3+, CD4+ and CD8+ T cell counts was significantly greater after MAC-HSCT throughout the first year after HSCT. Total CD4+ Tcon recovery was significantly decreased in RIC patients at all time points but Treg recovery was significantly lower only in the first 2 months after HSCT. Central memory cells (CM; 45RA-62L+) comprise the majority of Treg and Tcon throughout the first year after HSCT. The percentage of Treg-CM is greater than Tcon-CM and the fraction of CM cells within Treg and Tcon is increased in the RIC cohort (Figure 1A). Effector memory cells (EM; 45RA-62L-) also represent a large fraction of Treg and Tcon. Reconstitution of Treg-EM and Tcon-EM is similar but recovery of this subset appears to be strongly affected by sirolimus (Figure 1B). In RIC patients who receive sirolimus, the percentage of EM cells within Treg and Tcon subsets is significantly lower in the first 6 months after HSCT. Naïve Treg and Tcon (CD45RA+CD62L+) represent a relatively small fraction of recovering CD4 T cells during this period. Reconstitution of naïve CD4 T cells identified as recent thymic emigrants (RTE; CD45RA+CD31+) is markedly different in Treg and Tcon (Figure 1C). Within Tcon, the RTE fraction rapidly recovers to levels observed in healthy donors. In contrast, the fraction of RTE-Treg remains very low, with no evidence of improvement for at least 1 year. For both Treg-RTE and Tcon-RTE, recovery is significantly greater after RIC suggesting that either the reduced-intensity of conditioning or sirolimus is thymus protective. In vivo proliferation of each T cell subset was monitored by expression of Ki67. As previously observed in healthy donors, proliferation of Treg was significantly greater than Tcon at all time points. This primarily reflects homeostatic proliferation of Treg memory cells since very few naïve Treg are present. Both Treg and Tcon proliferation was higher in the MAC cohort in the first 1-3 months after HSCT. In vivo susceptibility to apoptosis was monitored by expression of pro-survival Bcl-2 and pro-apoptotic CD95 (FAS) expression. All Treg subsets expressed lower levels of Bcl-2 and higher levels of CD95 compared to Tcon. RIC/sirolimus was associated with higher levels of Bcl-2 and lower levels of CD95 predominately in the first 3 months after transplant. This effect was evident in all Treg and Tcon subsets. These results demonstrate distinctly different patterns of reconstitution of Treg and Tcon after allogeneic HSCT. Reconstitution of Tcon is characterized by rapid recovery of thymic generation, moderate homeostatic proliferation of memory subsets and relative resistance to apoptosis. Reconstitution of Treg is characterized by prolonged impaired thymic generation, high levels of homeostatic proliferation of memory subsets and increased susceptibility to both intrinsic and extrinsic pathways of apoptosis. RIC followed by administration of sirolimus for GVHD prophylaxis appears to selectively delay recovery of Treg and Tcon EM cells while sparing naïve, RTE and CM subsets. Figure 1 Figure 1. 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: 2014
    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. 122, No. 21 ( 2013-11-15), p. 3475-3475
    Abstract: CD4+CD25+Foxp3+ regulatory T cells (Treg) play an important role in the maintenance of self-tolerance and immune homeostasis. Interleukin-2 (IL-2) is critical for Treg development, expansion, activity and survival. Lack of either IL-2 or IL-2Rα (CD25) results in Treg deficiency and Treg impairment is associated with loss of tolerance, autoimmunity, and chronic GVHD. We previously demonstrated that daily administration of low-dose IL-2 resulted in selective expansion of Treg in vivo and clinical improvement of chronic GVHD (Koreth et al. NEJM 2011). We also reported that low-dose IL-2 enhanced Treg resistance to FAS-mediated apoptosis through the extrinsic pathway (Matsuoka et al. Sci Transl Med 2013). However susceptibility of Treg to apoptosis through the intrinsic pathway has not been examined. To examine the mechanisms whereby IL-2 affects susceptibility of Treg to apoptosis through the intrinsic pathway, we used a functional flow cytometry-based assay (BH3 profiling) to measure mitochondrial membrane depolarization in response to a panel of pro-apoptotic BH3 peptides (BIM, BID, BAD, NOXA, PUMA, BMF, HRK). Mitochondria that are more sensitive to the BH3 peptides are more 'primed' for apoptosis, and more sensitive to pro-apoptotic signaling. This assay allowed us to compare “priming” which we define as susceptibility to BH3 peptide-induced mitochondrial membrane depolarization in Treg and Tcon. We also examined cytoplasmic expression of Bcl-2, BclxL, Mcl1, Bim, XIAP, Ki67 and Staurosporine (STS) induced apoptosis as additional measures of susceptibility to apoptosis and proliferation in each subset. In resting blood obtained from healthy donors (n=25), Treg were more “primed” than Tcon when exposed to several BH3 peptides (Bim, Bad, PUMA and BMF). This BH3 peptide pattern suggests that the anti-apoptotic protein, Bcl2 plays a primary role in the priming of these cells. Analysis of pro- and anti-apoptotic proteins revealed that Treg expressed higher levels Bim and BclxL and lower levels of Bcl2 and XIAP than Tcon, but there was no significant difference in Mcl1. Treg expressed higher levels of Ki67 and were more susceptible to both STS- and FAS-induced apoptosis than Tcon. Thus, Treg in healthy individuals have higher proliferative activity and are more susceptible to apoptosis than Tcon through both mitochondrial and death receptor pathways. To examine the functional effects of IL-2 on T cell homeostasis, Treg and Tcon were purified by cell sorting and cultured with different concentrations of IL-2 (10, 100, 1000 IU/ml) with and without TCR stimulation (1ug/ml anti-CD3 + 1ug/ml anti-CD28). BH3 priming was measured after 3 days and results were compared to cells cultured in media alone. Low-concentration IL-2 (10 U/ml) lowered BH3 priming and increased Bcl2 expression in Treg. Similar effects were observed in Tcon, but higher concentrations of IL-2 ( 〉 100 U/ml) were required to increase Bcl2 expression and decrease BH3 priming in Tcon. TCR stimulation alone induced higher BH3 priming in Tcon but had little effect in Treg. The combination of TCR stimulation plus IL-2 increased BH3 priming in both Tcon and Treg. TCR stimulation increased proliferation only in Tcon, but the TCR + IL-2 combination increased proliferation in both Tcon and Treg. Thus, IL-2 alone reduced BH3 priming in unstimulated cells but the TCR + IL-2 combination induced higher proliferation and higher BH3 priming. STS-induced apoptosis assays revealed that low-dose IL-2 reduced susceptibility to apoptosis only in Treg. IL-2 did not change the expression of BclxL, Mcl1, and Bim but XIAP expression was increased in both Treg and Tcon. To confirm the effects of low-dose IL-2 on Treg priming were mediated by Bcl2, we examined the effect of ABT-199, a selective Bcl2 inhibitor on BH3 priming in Treg and Tcon. As shown in Figure 1, ABT-199 enhanced priming and spontaneous apoptosis of both Treg and Tcon. IL-2 had no effect on ABT-199-induced priming or apoptosis in Tcon. In contrast, IL-2 reversed the effects of ABT-199 on Treg priming and apoptosis providing further evidence that the inhibition of intrinsic pathway apoptosis mediated by IL-2 in Treg is dependent on Bcl2. Disclosures: Letai: AbbVie: Consultancy; Dana-Farber Cancer Institute: Patents & Royalties.
    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 ...
  • 5
    In: Blood, American Society of Hematology, Vol. 109, No. 11 ( 2007-06-01), p. 4964-4972
    Abstract: Waldenström macroglobulinemia (WM) is an incurable lymphoplasmacytic lymphoma with limited options of therapy. Protein kinase Cβ (PKCβ) regulates cell survival and growth in many B-cell malignancies. In this study, we demonstrate up-regulation of PKCβ protein in WM using protein array techniques and immunohistochemistry. Enzastaurin, a PKCβ inhibitor, blocked PKCβ activity and induced a significant decrease of proliferation at 48 hours in WM cell lines (IC50, 2.5-10 μM). Similar effects were demonstrated in primary CD19+ WM cells, without cytotoxicity on peripheral blood mononuclear cells. In addition, enzastaurin overcame tumor cell growth induced by coculture of WM cells with bone marrow stromal cells. Enzastaurin induced dose-dependent apoptosis at 48 hours mediated via induction of caspase-3, caspase-8, caspase-9, and PARP cleavage. Enzastaurin inhibited Akt phosphorylation and Akt kinase activity, as well as downstream p-MARCKS and ribosomal p-S6. Furthermore, enzastaurin demonstrated additive cytotoxicity in combination with bortezomib, and synergistic cytotoxicity in combination with fludarabine. Finally, in an in vivo xenograft model of human WM, significant inhibition of tumor growth was observed in the enzastaurin-treated mice (P = .028). Our studies therefore show that enzastaurin has significant antitumor activity in WM both in vitro and in vivo, providing the framework for clinical trials to improve patient outcome in WM.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2007
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 108, No. 11 ( 2006-11-16), p. 439-439
    Abstract: Hematopoietic development and long-term homeostasis are tightly regulated by a dynamic balance of stem cell self-renewal and differentiation. To understand the role of the FoxO family of transcription factors in these processes, we studied the impact of somatic deletion of all FoxO genes in the adult hematopoietic system of mice engineered with the interferon-inducible Mx-Cre transgene and various conditional FoxO alleles: FoxO1L/L; FoxO3 L/L; and/or FoxO4 L/L. Cre-mediated excision of FoxO1, FoxO3 and FoxO4 (FoxO1/O3/O4L/L), resulted in myeloid lineage expansion, lymphoid developmental abnormalities and a marked decrease of the lineage-negative, Sca-1+, c-Kit+ (LSK) compartment, containing the hematopoietic stem cell (HSC) population, but normal numbers of myeloid progenitors. Furthermore, FoxO1/O3/O4L/L null bone marrow cells were defective in competitive and noncompetitive repopulation assays. This FoxO deficient defect correlated with enhanced cell cycling and increased apoptosis of HSC cells. Notably, there was an HSC-restricted elevation in the level of reactive oxygen species (ROS) that was not observed in committed progenitors, and was associated with decreased catalase and MnSOD expression. Furthermore, in vivo treatment with the anti-oxidative agent N-acetyl-cysteine (NAC) resulted in complete reversion of the FoxO deficient HSC phenotype. Taken together, these results demonstrate that, in the HSC compartment, FoxO proteins are mediators of quiescence and enhanced survival and play essential regulatory roles in the response to physiologic oxidative stress, a function that may contribute to the long-term regenerative potential of the hematopoietic stem cell compartment.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2006
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 1257-1257
    Abstract: Abstract 1257 CD4+CD25+Foxp3+ regulatory T cells (Treg) are known to play an important role in the maintenance of peripheral tolerance and control of cGVHD. Recent studies have shown that Treg undergo extensive homeostatic proliferation after allogeneic HSCT. However, highly proliferating Treg are also susceptible to apoptosis and the inability to maintain adequate Treg survival may contribute to the development of cGVHD. Thus, mechanisms of Treg survival and persistence are of great importance. Telomeres are repetitive DNA sequences, located at the ends of each chromosome. Telomeres gradually shorten with each cell division and eventually lead to cell senescence and death. In rapidly dividing cells, telomere length is maintained by telomerase, a reverse transcriptase that prevents premature senescence. Previous studies have shown that abnormal telomerase activity is found in T cells from patients with autoimmune diseases such as rheumatoid arthritis and systemic lupus erythematosus, suggesting that telomerase may play a role in the maintenance of immune tolerance. In the present study, we examined whether telomerase activity and telomere length were associated with Treg homeostasis after allogeneic HSCT, particularly in the context of cGVHD. We examined Treg and conventional CD4 T cells (Tcon) in 35 patients with hematologic malignancies (median age 54 yr) who survived without relapse for more than 2 years after HSCT. Patient samples were obtained at 43 months (median, range: 25–99 months) after HSCT. The severity of cGVHD was classified according to NIH criteria. At the time of analysis, 2 patients had no cGVHD, 19 had mild cGVHD, 9 had moderate cGVHD, and 5 had severe cGVHD. CD4+CD25-CD127+ Tcon and CD4+CD25+CD127- Treg were isolated by flow cytometric cell sorting. Relative telomere length was measured by real time PCR. Telomerase activity was measured by PCR-ELISA. Telomere length was significantly shorter in Treg compared to Tcon in all patients (0.05 vs 0.28; p 〈 0.01), but there was no correlation between telomere length and severity of cGVHD (see Table below) in both Treg and Tcon. Telomerase activity was increased in Treg compared to Tcon (29.73 vs 8.95; p 〈 0.01). In contrast to telomere length, telomerase activity of Treg is significantly associated with severity of cGVHD (38.89 in patients with no or mild cGVHD vs. 8.19 in patients with moderate or severe GVHD, p=0.0002). These data indicate that induction of telomerase activity in Treg after allogeneic HSCT does not prevent overall telomere shortening. However, activation of telomerase appears to increase the replicative life span of Treg and provides a mechanism for maintaining survival of these cells in vivo. In contrast, failure to activate telomerase in some patients may lead to shortened survival of Treg, inability to maintain peripheral tolerance and development of severe cGVHD. 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 ...
  • 8
    In: Blood, American Society of Hematology, Vol. 120, No. 21 ( 2012-11-16), p. 652-652
    Abstract: Abstract 652 Long-term survival of patients with acute myeloid leukemia (AML) is poor, and new forms of therapy are needed. Many genetic lesions have been identified and studied, and most patients have chromosome translocations or other mutations that promote self-renewal of leukemic stem cells, block differentiation, enhance growth, and block apoptosis. Only a few of these mutations result in druggable targets (e.g., PML-RARa, Kit, PDGFR, FLT3 for instance). In addition to genetic lesions, epigenetic abnormalities have been shown to be very common in AML, and provide opportunities for novel treatments. Using genome-wide approaches to identify alternative splicing, we have recently shown that AML cells have a high level of aberrantly regulated genome-wide alternative splicing (AS) as a frequent epigenetic event. By comparing samples from 62 AML patients with 10 normal donors (NDs) we identified 428 genes differentially spliced in AML. A list of differentially spliced genes includes 50 oncogenes and 52 tumor suppressor genes, as well as genes encoding proteins involved in cell proliferation and differentiation, and apoptosis. We evaluated splicing event frequency in AML compared to NDs and we observed that on average 527 (range 137–1657) genes were identified as differentially spliced in any given patient, out of 62 analyzed. Also, we found that any given differentially spliced gene, of the 3,108 detected, were spliced on average in 26 (range 1–54) AML patients. Thus, splicing aberrations are highly recurrent in AML patients. To identify the causes of aberrant splicing in AML, we evaluated transcript levels of the 24 major splicing factors (SFs) that are involved in the first and second splicing transesterification reactions. These splicing factors are important proteins involved in spliceosomalassembly. Expression levels of these SFs were evaluated in 20 AML patients exhibiting high levels of AS. Quantitative RT-PCR analysis showed significant (up to 30 fold) upregulation of U2AF2 (P 〈 2.00E-07), PTBP (P=3.00E-04) and SFRS12 (P=0.002) SF transcript levels in AML patient samples compared to CD34+ cells from NDs. In preliminary studies, we also detected elevated expression of U2AF2 and PTBP proteins in several patient samples. These results suggest the intriguing possibility that aberrant splicing in AML may be the result of alterations of these SFs. To test this hypothesis we generated stably transfected HEK293 cell lines overexpressing U2AF2 or PTBP. We have developed a synthetic semi-quantitative splicing assay to evaluate the effects of overexpression of these SFs. We have obtained a minigene cassette of the p53 inducible PIG3 gene based on previous splicing studies. The minigene cassette was cloned between RFP (red fluorescent prtoein) and GFP (green fluorescent prtoein) in such a way that translation of the normally spliced transcript results in expression of RFP and GFP, while aberrant splicing results in the expression of RFP only. Production of a similar minigene cassette that includes exons/introns of a gene that is subjected to aberrant splicing in AML (NOTCH2, FLT3 and CD13) is in progress. In studies, completed so far, with the PIG3 minigene cassette construct transiently transfected into the HEK293 cells lines, overexpression of PTBP increased aberrant splicing of the PIG3 minigene. Similar studies testing the effects of elevated levels of U2AF2 and PTBP on NOTCH2 and other genes misspliced in AML (such as FLT3 and CD13) will be presented. Our results indicate that aberrant splicing could be an important event in AML, and development of an in vitro, synthetic splicing assay will enable us to better understand the underlying causes of this process in AML. 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 ...
  • 9
    In: Blood, American Society of Hematology, Vol. 120, No. 21 ( 2012-11-16), p. 225-225
    Abstract: Abstract 225 CD4+CD25+Foxp3+ regulatory T cells (Treg) are known to play a central role in the maintenance of self-tolerance and immune homeostasis. After allogeneic stem cell transplantation, impaired recovery of Treg is associated with the development of cGVHD. Interleukin-2 (IL-2) is a critical regulator of Treg development, expansion and survival and lack of IL-2 results in Treg deficiency. In patients with cGVHD, we previously demonstrated that Treg proliferate at high levels but this subset is also highly susceptible to apoptosis leading to inadequate Treg numbers (Matsuoka et al. JCI 2010). We also reported that low-dose IL-2 administration resulted in selective expansion of Treg in vivo and clinical improvement of cGVHD (Koreth et al. NEJM 2011). To identify mechanisms responsible for increased Treg susceptibility to apoptosis in cGVHD we used a new flow cytometry-based assay to measure mitochondrial membrane depolarization in response to a panel of pro-apoptotic BH3 peptides (BIM, BID, BAD, NOXA, PUMA, BMF, HRK). This assessment allowed us to compare BH3 peptide-induced mitochondrial membrane depolarization (“priming”) in different T cell subsets, including CD4 Treg, conventional CD4 T cells (CD4 Tcon), and CD8 T cells. Expression of Bcl-2, CD95 and Ki67 were also studied in each T cell subset. We studied peripheral blood samples from 36 patients with hematologic malignancies (median age 59 yr) who are 〉 2 years post HSCT (27 patients with cGVHD and 9 patients without cGVHD) and 15 patients who received daily subcutaneous IL-2 for 8 weeks for treatment of steroid-refractory cGvHD. Severity of cGVHD was classified according to NIH criteria. In patients without cGVHD, BH3 priming was similar in all 3 T cell subsets (CD4 Treg, CD4 Tcon and CD8). In patients with cGVHD, CD4 Treg were more primed than CD4 Tcon when challenged with BIM, BAD, PUMA, BMF and the combination of BAD + NOXA peptides (p 〈 0.01 – 0.0001). Treg were more primed than CD8 T cells when challenged with PUMA peptide (p 〈 0.0001), but priming in Treg and CD8 T cells was similar for other BH3 peptides in patients with cGVHD. We also compared BH3 priming of each T cell subset in patients with different grades of cGVHD. When challenged with BH3 peptides, Treg, Tcon and CD8 T cells were less primed in patients with severe cGVHD. In patients with cGVHD, Treg expressed higher levels of Ki-67, higher levels of CD95 and lower levels of Bcl-2 than Tcon. Expression of CD95 did not vary with severity of GVHD in any T cell subset, but expression of Bcl-2 was significantly increased in all subsets in patients with severe cGVHD. Increased BH3 priming and high expression of CD95 indicate that Treg are more susceptible to apoptosis than Tcon in cGVHD. However, both Treg and Tcon become less primed and Bcl-2 levels increase in severe cGVHD suggesting that these cells are less susceptible to mitochondrial pathway apoptosis. Since the total number of Treg and Tcon are significantly reduced in patients with cGVHD, these findings suggest that the remaining circulating cells are relatively resistant to mitochondrial pathway apoptosis. CD95 expression in Treg remains high indicating no change in death receptor pathway apoptosis. Daily treatment with low-dose IL-2 for 8 weeks selectively expands Treg in vivo in patients with severe cGVHD. As the number of Treg increase, BH3 profiling shows that these cells gradually become more primed and therefore more susceptible to mitochondrial pathway apoptosis. Taken together, these studies help define the complex and distinct pathways that regulate survival in different T cell subsets and changes in these pathways that occur in patients with chronic GVHD. These pathways play important roles in the maintenance of T cell homeostasis and targeting these complex pathways can provide new opportunities to promote immune tolerance after allogeneic HSCT. 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 ...
  • 10
    In: Blood, American Society of Hematology, Vol. 112, No. 13 ( 2008-12-15), p. 5161-5170
    Abstract: An attractive target for therapeutic intervention is constitutively activated, mutant FLT3, which is expressed in a subpopulation of patients with acute myelocyic leukemia (AML) and is generally a poor prognostic indicator in patients under the age of 65 years. PKC412 is one of several mutant FLT3 inhibitors that is undergoing clinical testing, and which is currently in late-stage clinical trials. However, the discovery of drug-resistant leukemic blast cells in PKC412-treated patients with AML has prompted the search for novel, structurally diverse FLT3 inhibitors that could be alternatively used to override drug resistance. Here, we report the potent and selective antiproliferative effects of the novel mutant FLT3 inhibitor NVP-AST487 on primary patient cells and cell lines expressing FLT3-ITD or FLT3 kinase domain point mutants. NVP-AST487, which selectively targets mutant FLT3 protein kinase activity, is also shown to override PKC412 resistance in vitro, and has significant antileukemic activity in an in vivo model of FLT3-ITD+ leukemia. Finally, the combination of NVP-AST487 with standard chemotherapeutic agents leads to enhanced inhibition of proliferation of mutant FLT3-expressing cells. Thus, we present a novel class of FLT3 inhibitors that displays high selectivity and potency toward FLT3 as a molecular target, and which could potentially be used to override drug resistance in AML.
    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...