Academic literature on the topic 'Nalm6 cells'

Create a spot-on reference in APA, MLA, Chicago, Harvard, and other styles

Select a source type:

Consult the lists of relevant articles, books, theses, conference reports, and other scholarly sources on the topic 'Nalm6 cells.'

Next to every source in the list of references, there is an 'Add to bibliography' button. Press on it, and we will generate automatically the bibliographic reference to the chosen work in the citation style you need: APA, MLA, Harvard, Chicago, Vancouver, etc.

You can also download the full text of the academic publication as pdf and read online its abstract whenever available in the metadata.

Journal articles on the topic "Nalm6 cells"

1

Ramírez Saldaña, Maricela, Miguel Aguilar Santelises, Martha Moreno Lafont, Leopoldo Santos Argumedo, Vladimir Paredes Cervantes, and Rubén López Santiago. "Human B cells are targets for Brucella abortus infection. (P3130)." Journal of Immunology 190, no. 1_Supplement (May 1, 2013): 186.12. http://dx.doi.org/10.4049/jimmunol.190.supp.186.12.

Full text
Abstract:
Abstract It has been demonstrated that Brucella abortus is capable to multiply into professional antigen presenting cells such as macrophages and dendritic cells. However, there are a few reports about Brucella infection into B cells. In this work, we evaluated the ability of B. abortus 2308 for infect the human B cell line, NALM6 R. B cells cultured in RPMI medium supplied with L-glutamine and 10% of fetal calf serum were infected with several MOIs of Brucella abortus 2308 transformed by electroporation with the pBBR1MCS-2-gfp-mut3 plasmid to evaluate the entry of green fluorescent Brucella. The percentage of cells containing bacteria was evaluated by flow cytometry at three time points. To evaluate whether Brucella was on the surface or into the B cells, kinetics of infection was performed and analyzed by transmission electron microscopy. Association of Brucella abortus 2308-gfp-mut3 with NALM6 R cells increased in a dose-time dependent manner to reach a maximum of 50% of NALM6 R infected cells. Electronic microscopy revealed that NALM 6R B cells produce filopodia over B. abortus. Once inside, the bacteria resided into small vesicles. Apparently bacteria underwent some bactericidal mechanism, because the electron microscopy showed bacteria degraded into vesicles. It was not observed evidence of replication of bacteria inside B cells. Brucella abortus 2308 is capable to interact with NALM6 R membrane, to induce filopodia formation and to entry into B cells inside vesicles.
APA, Harvard, Vancouver, ISO, and other styles
2

Tang, Yongmin, Lixia Li, Di Wang, Hongqiang Shen, Baiqin Qian, Chunfang Luo, and Haizhong Zhang. "Study on the Preparation of the Immunotoxin 2E8-NCTD and Its Targeting Killing Effect In Vitro." Blood 110, no. 11 (November 16, 2007): 4186. http://dx.doi.org/10.1182/blood.v110.11.4186.4186.

Full text
Abstract:
Abstract Objective: Monoclonal antibody (mAb) conjugated with certain toxin to generate immunotoxin bears an important and promising new therapy for patients with hematopoietic malignancies. However, most toxic moieties conjugated to antibody proteins reported were toxic proteins which presented immunogenicity to patients capable of producing anti-toxin antibody. Norcantharidin (NCTD) is a small molecule of toxin derived from a Chinese medicine Mylabris phalerata Pallas. The chemical name of this agent is 7-Oxabicyclo(2,2,1) heptane-2,3-dicarboxylic anhydride with a molecular weight of 168.15 CAS. It does not have the immunogenicity to human body so that it bears a promising potential for development of new targeting drug. In this study, a new clone of self-made anti-CD19 mAb named ZCH-4-2E8 conjugated with NCTD was used to investigate its targeting efficacy against CD19+ lymphoid malignant Nalm-6 cells in vitro in order to provide the experimental fundamentals for the further development of this new targeting agent. Methods: 2E8 monoclonal antibody was prepared from mouse ascites and purified by gel chromatography. The purity of the antibody protein was checked by SDS-PAGE assay. Immunotoxin 2E8-NCTD was successfully generated through conjugating CD19 mAb protein and Norcantharidin by the activated ester method. The binding activity of the immunoconjugate (2E8-NCTD) to CD19 antigens on cell surface and the expression levels of CD19 antigens on Nalm-6 and K562 cells were examined by flow cytometery. Comparisons of the inhibitory effects among PBS, purified 2E8 antibody, Norcantharidin and immunotoxin 2E8-NCTD groups on cell growth of either Nalm6 cells or K562 cells were made. Results: The purity of the purified 2E8 antibody was more than 99% demonstrated by SDS-PAGE assay. 2E8 antibody in the supernatant reacted with 99.34% of Nalm6 cells, while only 0.98% of K562 cells were reacted with this antibody. The new generated immunotoxin (2E8-NCTD) had a positive rate of 99.90% on Nalm6 cells with little reduction of binding activity. From the in vitro study, both 2E8-NCTD and Norcantharidin were shown to have significant inhibitory effects on the growth of CD19+Nalm-6 cells after 96 h of culture with inhibitory rates of 71.25% and 97.62%, respectively (P < 0.001), while the purified 2E8 antibody (inhibitory rate of 30.29, P > 0.05) didn’t show any significant influences on the growth of Nalm6 cells as compared to that of negative control. No significant inhibitory effects were identified among immunotoxin 2E8-NCTD and 2E8 antibody as compared to that of control group on CD19-K562 cells with inhibitory rates of 15.41% and 4.17%, respectively, P > 0.05, indicating that a significant targeting effect of the 2E8-NCTD against Nalm6 cells was noticed. Conclusions: The immunotoxin 2E8-NCTD was successfully synthesized by activated ester method with an excellent targeting killing efficacy on CD19+ Nalm-6 leukemia cells in vitro, which lays the experimental fundamentals for the further development of this new targeting agent.
APA, Harvard, Vancouver, ISO, and other styles
3

Iwasa, Masaki, Yasuo Miura, Aya Fujishiro, Sumie Fujii, Noriko Sugino, Satoshi Yoshioka, Akihiro Tamura, et al. "Bortezomib Attenuates Adhesion of B Cell Precursor Acute Lymphoblastic Lleukemia Cells to Bone Marrow Mesenchymal Stromal/Stem Cells Via Regulating SPARC Expression." Blood 126, no. 23 (December 3, 2015): 786. http://dx.doi.org/10.1182/blood.v126.23.786.786.

Full text
Abstract:
The prognosis of adults with B cell precursor acute lymphoblastic leukemia (BCP-ALL) is poor. Many cases in complete remission experience relapse of leukemia despite intensive chemotherapy or hematopoietic stem cell transplantation. This clinical observation suggests that minimal residual disease (MRD) still exists after these intensive therapies. Cell adhesion-mediated drug resistance (CAM-DR) is one of the mechanisms to support MRD in the bone marrow microenvironment (Clin Cancer Res 14:9, 2008). Mesenchymal stromal/stem cells (MSCs) are a cellular component of bone marrow (BM) and maintain physiological precursor B lymphopoiesis (Nature Rev Immunol 6:107, 2006). We investigated our hypothesis that survival of BCP-ALL cells is supported by their direct adhesion to BM-MSCs in BM microenvironment. First, we confirmed that BCP-ALL cells exhibited their drug resistant phenotype through adhesion to human BM-MSCs using human BCP-ALL cell line, Nalm6. We isolated human BM-MSCs from normal bone marrow samples (AllCells, Emeryville, CA). When Nalm6 cells were co-cultured with human BM-MSCs, they were prone to adhere to BM-MSCs. Nalm6 adhered to BM-MSCs (Nalm6/ad) were more resistant to the treatment with various anti-cancer drugs including doxorubicin than Nalm6 in suspension (Nalm6/su). Immunoblot analysis showed that expression level of anti-apoptotic protein Bcl-2 and phosphorylation level of pro-survival kinase Akt are higher in Nalm6/ad compared with those in Nalm6/su. In cell cycle analysis using Ki67/PI fluorescence-activated cell sorter (FACS) assay, the percentage of populations in G0 phase and S/G2/M phase was higher in Nalm6/ad compared with that in Nalm6/su, which indicated the increase of MRD and the high proliferation of leukemic cells in adhesive population, respectively. Therefore, we considered that detachment of Nalm6 from BM-MSCs is important to restore chemosensitivity of BCP-ALL cells and to eliminate these cells. Accordingly, we screened drugs that are capable of disrupting the adhesion of Nalm6 to BM-MSCs, and found that several proteasome inhibitors had a such activity. BM-MSCs were treated with bortezomib (10nM for 24h or 100nM for 1h), carfilzomib (3 nM for 24 h or 100 nM for 1 h) or oprozomib (10 nM for 24 h or 300 nM for 4 h), and then washed with PBS and co-cultured with Nalm6 cells. We confirmed that the number of BM-MSCs is not affected with these drugs irrespective of their concentration and incubation time by MTT assay. Co-cultured cells were divided into suspension cells and adherent cells. Each population was stained with monoclonal antibodies against CD19 and CD90 to detect Nalm6 and BM-MSCs, respectively, and analyzed by FACS. When BM-MSCs were pre-treated with bortezomib, carfilzomib or oprozomib, the number of Nalm6/ad was significantly decreased, and inversely, the number of Nalm6/su was increased, as compared to that of co-cultures with untreated BM-MSCs. Intriguingly, BM-MSCs treated with bortezomib showed aberrant expression of secreted protein acidic and rich in cysteine (SPARC). Immunoblot analysis showed that SPARC expression of BM-MSCs was transiently increased after bortezomib treatment. In co-cultures with BM-MSCs transfected with siRNA targeting SPARC, the number of Nalm6/ad was increased. In addition, the number of Nalm6 adhered to BM-MSCs pre-treated with recombinant human SPARC was lower than that adhered to control non-treated BM-MSCs. Therefore, SPARC showed anti-adhesive property and was one of the molecules associated with adhesion of BM-MSCs to Nalm6. Finally, we tested whether bortezomib shows therapeutic effects in vivo. Bortezomib administration supported survival of BCP-ALL xenograft model mice utilizing Nalm6, which was concomitant with the decrease in leukemia cells in the femur and in the size of spleen, as compared to those in non-treated control mice. In summary, Nalm6 cells that adhered to BM-MSCs contributed to construction of chemoresistant population similar to MRD. Here, we found that this population could be detached by treatment of BM-MSCs with bortezomib through transient increase in their SPARC expression. Our findings suggest that bortezomib or other proteasome inhibitors might be promising drugs to treat BCP-ALL through attenuating the adhesion of leukemic cells to BM-MSCs, and shed new insight into a therapeutic strategy in BCP-ALL by targeting BM-MSCs. Disclosures No relevant conflicts of interest to declare.
APA, Harvard, Vancouver, ISO, and other styles
4

Hermanova, Ivana, Karel Valis, Karel Fiser, Hana Nuskova, Jan Trka, and Julia Starkova. "L-Asparaginase Strongly Affects Bioenergetics in Leukemic Cells." Blood 120, no. 21 (November 16, 2012): 779. http://dx.doi.org/10.1182/blood.v120.21.779.779.

Full text
Abstract:
Abstract Abstract 779 L-asparaginase (L-asp) is an important component of childhood acute lymphoblastic leukemia (ALL) therapy. Its cytotoxic effect is based on the depletion of extracellular asparagine and glutamine. Leukemic cells are sensitive to this depletion due to the lower activity of asparagine synthetase compared to healthy cells. However, the mechanism of resistance development remains unclear. The aim of this study was to obtain further insights into the mechanisms underlying the cytotoxic effect of L-asp. We used two models: resistant preB ALL leukemic cells derived from the REH (TEL/AML1[+]; very sensitive) and NALM6 (TEL/PDGFRB[+]; medium sensitive) cell lines by long-term incubation with L-asp. As a second model we used REH and NALM6 incubated with L-asp for 24 hrs, mimicking acute phase of treatment. We performed GEP of REH, res-REH, NALM6 and res-NALM6. There was an overlap of 30 genes changed in both cases. Applying the pathway analysis we found that L-asp influences the regulation of lipid metabolism and apoptosis regulated by mitochondrial proteins. Next we merged our data with GEP data published by Holleman et al. (NEJM, 2004) of ALL patients′ samples sensitive/resistant to L-Asp. Two pathways, one regulating protein translation, the other metabolism, scored very highly. Next to glucose, glutamine is the other major cellular energy source, it is also important for activation of PI3K/Akt/mTOR pathway - the key regulator of translation. Since L-asp also depletes glutamine, we focused on the effect of L-asp treatment on bioenergetics and translation in leukemic cells. To confirm the effect of L-asp on PI3K/Akt/mTOR pathway we measured the amount of Akt protein and P70S6K/p-P70S6K (downstream target of mTOR) in REH and NALM6 treated for 24 hours with L-asp. Akt and p-P70S6K expression fell in both cell lines. Next we were interested if PI3K/mTOR inhibition affects the sensitivity of resistant cell lines. We measured the cytostatic effect of co-treatment: L-asp with mTORC1 inhibitor (rapamycin); with dual inhibitor of mTORC1 and PI3K (LY294002); and with specific inhibitor of PI3K (PX866) on REH, res-REH, NALM6 and res-NALM6. The MTS assay proved the L-asp/rapamycin combination the most effective. It strongly diminished viability of all cell lines, even in those resistant to L-asp. c-Myc as an activator of glutamine catabolism showed a significant decrease in REH and NALM6 after incubation with L-asp. In addition, c-Myc was also decreased in res-REH compared with REH. Importantly, c-Myc activates glycolysis which is a main energy generator in cancer cells rather than oxidative phosphorylation (OXPHOS). We showed diminishement of Glucose transporter type 1 protein expression and 2.5-times reduced level of lactate, the product of cancer cell glycolysis, in media after L-asp exposure. Next we measured cell respiration that mirrors oxidative fosforylation (OXPHOS). REH and NALM6 showed increased capacity of respiratory chain after L-asp exposure, which was observed as an increase of endogenous respiration in uncoupled state. REH boosted the capacity of the respiratory chain from 163 to 236 pmol O2/s/mg (p<0.01). NALM6 have increased the capacity of the respiratory chain from 78 to 155 pmol O2/s/mg (p<0.01). Interestingly, REH has a significantly higher capacity of respiratory chain compared with NALM6 and a lower ability to regulate it. Moreover, res-REH had elevated the level of the endogenous oxygen consumption whereas res-NALM6 levels did not change. Our results indicate that leukemic cells, which normally depend on glycolysis for energy generation rather than OXPHOS, are able to swap these two mechanisms of energy generation under amino acid deprivation stress. The data show that L-asp inhibits mTORC1 and strongly affects bioenergetics pathways of leukemic cells. It decreased c-Myc-regulated glycolysis and increased OXPHOS. We believe that these metabolic changes are mainly caused by the effort of the cells to mobilize another mitochondrial pathway as Krebs cycle and β-oxidation, with the aim to supply depleted amino acids. We conclude from these data that resistance of the cells is caused by better biochemical adaptability to the nutrient deprived environment. Support: GAUK 92710, IGA NT1249, UNCE204012 Disclosures: No relevant conflicts of interest to declare.
APA, Harvard, Vancouver, ISO, and other styles
5

Gu, Yan, Chunhua Song, Sinisa Dovat, Qinglong Guo, Qinyu Ge, and Zheng Ge. "Oncogenesis of CRLF2 Overexpression and Effect of JAK2 Inhibitor in CRLF2 Overexpressed B-Cell Acute Lymphoblastic Leukemia." Blood 134, Supplement_1 (November 13, 2019): 2757. http://dx.doi.org/10.1182/blood-2019-125259.

Full text
Abstract:
Backgroud: Cytokine receptor-like factor 2 (CRLF2) plays an important role in the development of normal B lymphocytes, which can mediate the proliferation of early B cells. However, the diect oncogenic effect of CRLF2 overexpression in acute lymhpoblastic leukemia (ALL) is far yet to be clarified. Here, we explored the effect of CRLF2 overexpression on cell proliferation and the effect of the novel JAK2 inhibitor on B-ALL cells with CRLF2 overexpression. Methods: The 83 patients with newly-diagnosed ALL (56 B-cell and 27 T-cell ALL; range from 14 to 77 years old) between June 2008 and June 2016 were studied at Zhongda Hospital Southeast University. The 21 normal bone marrow subjects were enrolled as controls. The qPCR method is developed for detection CRLF2 expression and the CRLF2 overexpression was determined with a cutoff value more than the highest sample of normal bone marrow control. Median differences between the cohorts were evaluated using a Mann-Whitney U-test. Frequency differences were analyzed using uni- and multivariate Cox model. Event-free survival (EFS) and overall survival (OS) were estimated by the Kaplan-Meier method and compared by log-rank test. CRLF2 F232C gain-of-function mutant which we previously reported or CRLF2 were expressed in Nalm6 and 697 B-ALL cells with lentiviral transduction. WST-1 cell proliferation assay and in vitro clonogenic assay were performed upon JAK2 inhibitor (BBT594) treatment. Nalm6-CRLF2-luc, Nalm6-F232C-luc, and Nalm6-vector-luc cells were injected via tail vein into the NSG mice. The leukemia engraftment was monitored once a week by living imaging. Results: The expression of CRLF2 in patietns with ALL was significantly higher than the normal control (P<0.0001). Patients with CRLF2 overexpression had a significantly higher WBC count (53*10^9/L vs. 29.5*10^9/L, P=0.041). Survival analysis showed that the patients with CRLF2 overexpression had a worse EFS and OS, the differences were statistically significant (11 months vs. 26 months, P=0.043 and 15 months vs. 32 months, P=0.015). Also, the CRLF2 expression is determined with flow cytometry after staining with FITC-CRLF2 antibody in 28 samples. The correlation analysis was performed on the CRLF2 expression detected by qPCR and flow cytometry, respectively. A significant positive correlation of the two methods was observed(r=0.957, P<0.0001). These data not only indicate that CRLF2 overexpression is a marker of poor outcome, but also reveal the qPCR might be a simple and quick method for screening CRLF2 overexpression in the clinic compared to flow cytometry which is commonly used. We further found that expression of CRLF2 or CRLF2 F232C mutant into Nalm6 and 697 B-ALL cells dramatically increase the CRLF2 mRNA level, which is 69 times than vector-only control. Moreover, CRLF2 or CRLF2 F232C significantly promotes the cell proliferation of Nalm6 and 697 cells compared to vector only (P<0.001). In addition, JAK2 inhibitor (BBT594) treatment showed the significant dose-dependent cell proliferation arrest and clonogenic inhibition in CRLF2 or CRLF2 F232C overexpressed Nalm6 and 697 cells compared to vector-only control. Furthermore, in vivo we observed the 5-fold higher signal intensity of leukemia engraftment in the mice injected with Nalm6-CRLF2-luc or Nalm6-F232C-luc compared to that of Nalm6-vector-luc control 1-3 weeks after the injection(P<0.001). The Nalm6-CRLF2-luc and Nalm6-F232C-luc infiltrations were observed in bone marrow, central nervous system, liver and spleen of the mice. Conclusion: We showed that CRLF2 overexpression could enhance the proliferation and infiltration of human B-ALL cells, and for the first time indicated that JAK2 inhibitor could suppress the cell proliferation and clonogenesis of the CRLF2 overexpressed B-ALL cells. Our data provide direct evidence of the oncogenic role of CRLF2 overexpression and the new therapeutic potential for targeting CRLF2 overexpressed B-ALL with JAK2 inhibitor. Disclosures No relevant conflicts of interest to declare.
APA, Harvard, Vancouver, ISO, and other styles
6

Barredo, Julio C., Sanja Altman-Hamandzic, and Guy J. Leclerc. "Differences in Folylpoly-γ-Glutamate Synthetase (FPGS) Expression in Childhood ALL Result from Cell Lineage of Origin and Presence of Non-Random Translocations." Blood 104, no. 11 (November 16, 2004): 2087. http://dx.doi.org/10.1182/blood.v104.11.2087.2087.

Full text
Abstract:
Abstract Methotrexate (MTX) is a universal component of chidlhood ALL therapies and its conversion to long chain polyglutamates (PG) by folylpoly-γ-glutamate synthetase (FPGS) is essential for its antileukemic acitivity. Expression of FPGS appears to be controlled by tissue/lineage specific and proliferation-dependent mechanisms. Levels of FPGS mRNA, protein, and enzyme activity are 2-3 fold higher in B-precursor (Bp) ALL cells when compared to T-lineage ALL, and these differences correlate with intracellular accumulation of long chain MTX-PG and lymphoblast sensitivity to MTX. To characterize these lineage differences in FPGS expression between B and T lymphoblasts we examined its mRNA expression in Nalm6 (Bp-ALL) and CCRF-CEM (T-ALL) cells during cellular growth and cell cycle checkpoints. During early exponential growth (24 hrs), FPGS expression was 6-fold higher in Nalm6 when compared to CCRF-CEM but decreased significantly after 72 hrs while it was unchanged in CCRF-CEM cells. During G1/G0 phase we found that FPGS expression was 15-fold higher in Nalm6 when compared to CCRF-CEM cells. Taken together, these data suggest that during proliferation and cell cycle progression FPGS gene expression is regulated differently in Bp-ALL and T-ALL cells. To determine whether this lineage-specific regulation occurs at the transcriptional level we performed nuclear run-on assays. We found that FPGS mRNA transcription initiation rate was 1.7-fold higher in Nalm6 when compared to CCRF-CEM cells, indicating that differences in promoter regulation lead to the observed lineage differences in FPGS expression in Bp- vs. T-ALL. We then used a methylation specific PCR assay to investigate the methylation status of the FPGS proximal promoter region from both Nalm6 and CCRF-CEM cells. Our data indicate that the FPGS proximal promoter region is unmethylated in both cell lines and therefore can not explain the observed lineage-specific differences. 5′-RACE experiments demonstrated that Nalm6 and CCRF-CEM have the same FPGS transcriptional start sites, but a reporter gene assay indicated that the minimal promoter region that directs FPGS transcription in CCRF-CEM cells is insufficient to drive FPGS mRNA transcription in NALM6 cells. In order to identify potential regulatory regions directing FPGS transcription in Bp-lymphoblasts, we used DNaseI hypersensitivity assays. We identified a hypersensitive region located 8.5 kbp upstream to exon 1 in Nalm6 cells suggesting that tissue-specific regulatory elements responsible for lineage-specific FPGS expression in Bp-ALL cells may be localized within this region. Finally, we detected reduced levels of FPGS mRNA expression in RCH-ACV (Bp-ALL, t(1:19)/E2A-PBX1) and REH (Bp-ALL, t(12:21)/TEL-AML1) cells expressing chromosomal translocated fusions when compared to control (Nalm6). To characterize the molecular basis of the E2A-PBX1 and TEL-AML1 interactions with FPGS mRNA expression in Bp lymphoblasts we used antisense and RNAi technology to downregulate these two genetic fusions. Our data lead us to hypothesize that in addition to lineage-specific regulatory differences in FPGS expression, molecular mechanisms associated with non-random translocations may alter FPGS mRNA expression and influence MTX sensitivity in Bp-ALL lymphoblasts.
APA, Harvard, Vancouver, ISO, and other styles
7

Borges, Christopher M., Kevin Wasko, Jared M. Nasser, Kelly Donahue, Amanda Pfautz, Lincy P. Antony, Glenn Leary, Steven Sexton, Richard A. Morgan, and Karrie K. Wong. "Preclinical Development of Edit-201, a Multigene Edited Healthy Donor NK Cell with Enhanced Anti-Tumor Function and Superior Serial Killing Activity in an Immunosuppressive Environment." Blood 136, Supplement 1 (November 5, 2020): 33. http://dx.doi.org/10.1182/blood-2020-139988.

Full text
Abstract:
Natural killer (NK) cells distinguish tumor from healthy tissue via multiple mechanisms, including recognition of stress ligands and loss of MHC class I expression. For example, KIR mismatching enables allogenic NK cells to respond to MHC positive tumors in a similar manner to MHC negative tumors, making allogeneic NK cell therapy a promising approach for broad oncology indications. Accordingly, allogenic human HD-NK cells, including gene-modified cells, have demonstrated an impressive safety and efficacy profile when administered to patients with advanced hematologic malignancies. However, effector function of allogeneic NK cells can be diminished by the lack of functional persistence, as well as tumor-intrinsic immunosuppressive mechanisms, such as production of TGF-β. To this end, we developed a next-generation allogeneic NK cell therapy using CRISPR-Cas12a gene editing to enhance NK cell function through knockout of the genes CISH and TGFBR2. Both single and simultaneous targeting (DKO) of TGFBR2 and CISH in NK cells using CRISPR-Cas12a produced in/dels at both targets in greater than 80% of NK cells, with greater than 90% of edited NK cells viable at 72 hours post-editing. Importantly, we find that DKO NK cells do not phosphorylate the SMAD2/3 protein downstream of the TGF-b receptor complex and demonstrate increased phosphorylation of pSTAT3 and pSTAT5 upon IL-15 stimulation, consistent with protein level knockout of TGFBR2 and CISH. To determine whether DKO NK cells exhibited superior function relative to control NK cells, we first measured the ability of DKO NK cells to kill Nalm6 cells (adult B cell ALL) relative to unedited control NK cells. We find that in the presence of exogenous TGF-b, DKO NK cells demonstrate improved cytotoxicity against Nalm6 tumor targets by delaying tumor re-growth in comparison to control NK cells. To better characterize the ability of DKO NK cells to kill tumor cells, we developed an in vitro serial killing assay. In this long-duration assay, up to 30 days, control and DKO NK cells (grown in the presence of IL-15) were challenged every 48 hours with a new bolus of Nalm6 tumor targets. Both DKO and unedited NK cells control Nalm6 target cell growth for greater than 18 days (9 additions of new Nalm6 target cells), demonstrating a surprising ability for the same NK cells to serially kill new Nalm6 target cells for a prolonged period of time in vitro. We find that DKO NK cells produce higher levels of IFN-γ and TNF-α relative to control NK cells over the duration of the entire serial killing assay, suggesting that DKO NK cells can continue to produce these inflammatory cytokines even after serial killing. As many tumors, including hematologic malignancies, have high concentrations of TGF-β in their microenvironments, we next tested the ability of DKO NK cells to control the growth of Nalm6 cells in our serial killing assay in the presence of TGF-b. 10ng/mL TGF-β was added at the start of the assay as well as at each addition of new Nalm6 target cells. We observed that control NK cells fail to restrict Nalm6 target cell growth beyond 4 days (after 1 addition of new Nalm6 target cells) whereas DKO NK cells control Nalm6 target cell growth for greater than 18 days (after 9 additions of new Nalm6 target cells). Similar to the serial killing assay without TGF-b, we find that DKO NK cells produce higher concentrations of IFN-γ and TNF-α relative to control NK cells over the duration of the entire serial killing assay. Broadening our repertoire of target cells beyond B cell malignancies is now in progress, including the AML-like cell lines HL-60 and THP-1, the multiple myeloma cell line RPMI 8226, and various solid tumor targets. In summary, using CRISPR-Cas12a we demonstrated highly efficient gene editing of primary human NK cells at two unique targets designed to augment NK cell anti-tumor activity across a variety of malignancies. Most significantly, we demonstrate sustained anti-tumor serial-killing activity in the presence of the potent immunosuppressive cytokine TGF-β. Together, the increased overall effector function of CISH/TGFBR2 DKO primary human NK cells and their ability to serial kill, support their development as a potent allogeneic cell-based medicine for cancer. This potential medicine, termed EDIT-201, is being advanced to clinical study. Disclosures Borges: Editas Medicine: Current Employment, Current equity holder in publicly-traded company. Wasko:Editas Medicine: Current Employment, Current equity holder in publicly-traded company. Nasser:Editas Medicine: Current Employment, Current equity holder in publicly-traded company. Donahue:Editas Medicine: Current Employment, Current equity holder in publicly-traded company. Pfautz:Editas Medicine: Current Employment, Current equity holder in publicly-traded company. Antony:Editas Medicine: Current Employment, Current equity holder in publicly-traded company. Leary:Editas Medicine: Current Employment, Current equity holder in publicly-traded company. Sexton:Editas Medicine: Current Employment, Current equity holder in publicly-traded company. Morgan:Editas Medicine: Current Employment, Current equity holder in publicly-traded company. Wong:Editas Medicine: Current Employment, Current equity holder in publicly-traded company.
APA, Harvard, Vancouver, ISO, and other styles
8

Wang, Lingyan, Yue Zhang, Eden Anderson, Rajesh Kumar, Adam J. Lamble, and Rimas J. Orentas. "CD22 CAR-T Induces Both CD19 and CD22 Surface Down-Modulation: Defining a Mechanism of Generalized Immune Evasion and the Effects of Epigenetic Modifiers." Blood 136, Supplement 1 (November 5, 2020): 22–23. http://dx.doi.org/10.1182/blood-2020-134930.

Full text
Abstract:
The success of chimeric antigen receptor modified T lymphocyte (CAR-T cell) therapy for pediatric and adult leukemia and lymphoma has been impressive, but far from universal. Even for the most responsive disease, pediatric pre-B-ALL, one-year remission rates are 50% or less. This brings fundamental questions as to how leukemia resists CAR-T activity front and center. CD19- and CD22-specific CAR-T escape mechanisms appear to differ. For CD19, antigen loss and RNA-splicing variants are common. For CD22, decreasing the number of target molecules on the surface of the leukemia appears to be a primary means of escape (Fry, et al., 2018). Murine models have demonstrated that bryostatin increases CD22 density and improves CAR-T activity (Ramakrishna, et al., 2019). We explored the activity of bryostatin and four FDA-approved epigenetic modifiers (panobinostat, vorinostat, 5-azacytidine, ATRA) for the ability to impact CD19 and CD22 expression in Raji, NALM6, and REH cell lines. Dose ranges tested were not toxic. For CD22: a) bryostatin, increased expression for all lines (surface molecules expressed per cell), b) 5-azacytidine, increased expression only in NALM6, and c) ATRA, increased expression only in Raji. For CD19: a) only bryostatin, and only in NALM6, increased expression. When normal B cells from peripheral blood were tested (n=3) bryostatin did not increase the number of CD19 or CD22 molecules on the cell surface. When cytolysis assays with CD22 CAR-T were carried out, bryostatin potentiated NALM6 killing, but paradoxically inhibited killing of Raji cells. To see if this effect was due to differential target molecule expression, we quantified the number of target (CD22) and non-target (CD19) molecules on the surface of each leukemia line during CTL assays. At high effector to target cell ratios, (E:T 1:1 or greater), culturing of NALM6 or Raji with CD22-CAR-T resulted in a marked reduction in both CD19 and CD22 surface expression at 24 hours, indicating a general mechanism of immune evasion. Even though CD19 and CD22 were elevated in bryostatin-treated NALM6, and CD19 was elevated in bryostatin-treated Raji, CD22 CAR-T activity still decreased target antigen expression 40% to greater than 100%. To see if this effect was due to permanent clonal selection, CAR-T were removed and purified leukemia cells re-cultured on their own. For Raji leukemia cells surviving CD22 CAR-T, the number of CD19 molecules per cell recovered within 24 hours. Full recovery was not seen for CD22, unless Raji cells had also been treated with bryostatin during the CTL assay. Our data highlights that unless bryostatin is included, CAR-T exposure induces a durable down-modulation of CD22 levels on Raji cells. We are currently defining epigenetic control of CD19 and CD22 expression upon exposure to CD22 CAR-T, bryostatin and other modifiers. These findings still do not explain why Raji cell killing is not potentiated by bryostatin, while NALM-6 is. The exposure of leukemia lines to epigenetic modifiers may also induce the expression of other molecules on the cell surface that either potentiate or inhibit CAR-T mediated killing. Preliminary data support that hypothesis that bryostatin may not only modify CD19 and CD22 expression, it may also induce both negative checkpoint molecule and positive T cell activating ligand expression on the leukemia cell surface. Thus, CAR-T induce both general and target-antigen specific changes in leukemia, depending in part on the leukemia subtype analyzed. Bryostatin increased CD22 expression in leukemia cell lines but not in normal B cells, indicating that control of CD22 levels on the cell surface is altered by leukemic transformation. A 24-hour exposure to CD22 CAR-T can induce dramatic transient changes in CD19 and CD22 expression levels, and durable changes in CD22 expression in Raji cells that can be surmounted by the inclusion of bryostatin. The difference in Raji and NALM6 sensitization to killing may be due to the origin of these cells as Burkitt lymphoma (Raji) is more differentiated with respect to B cell lineage than pre-B ALL (NALM6). We can conclude that analysis of CAR-T target ligand number is not sufficient to predict the efficacy of CAR-T therapy. We have demonstrated that mechanisms beyond alteration in target antigen number on the cell surface, such as the induction of positive and negative T cell ligands, must also be considered in the context of the leukemia types targeted by CAR-T therapy. Figure Disclosures Orentas: Lentigen Technology, a Miltenyi Biotec Company: Research Funding.
APA, Harvard, Vancouver, ISO, and other styles
9

Barredo, Julio C., Tingting Kinser, and Guy J. Leclerc. "TEL/AML1 and E2A/PBX1 Tranlsocations Lead to Altered Folylpoly-γ-Glutamate Synthetase (FPGS) Expression in ALL." Blood 106, no. 11 (November 16, 2005): 539. http://dx.doi.org/10.1182/blood.v106.11.539.539.

Full text
Abstract:
Abstract Expression of the human folylpoly-γ-glutamate synthetase (FPGS) gene is controlled by tissue/lineage specific and proliferation-dependent mechanisms. Levels of FPGS mRNA, protein, and enzyme activity are 2–3 fold higher in B-precursor (Bp) ALL cell lines and primary cells when compared to T-lineage ALL. These differences correlate with intracellular accumulation of long chain methotrexate (MTX) polyglutamates (PG), and more important with clinical sensitivity to MTX. However, significant heterogeneity of FPGS expression exists within hematopoietic cells of the same lineage, suggesting additional factors influence FPGS expression in Bp- and T-ALL. cDNA microarray data was analyzed for expression of folate-related genes (DHFR, FPGS, RFC, MTHFR, and γ-GGH) in six prognostic leukemia subtypes (T-ALL, hyperdiploid (&gt; 50 chromosomes), BCR-ABL, E2A-PBX1, TEL-AML1, and MLL) (Yeoh et al, Cancer Cell, 2002). It was found that only the FPGS gene exhibited significant heterogeneity of expression compared the others analyzed. Therefore, we investigated the effect of distinct non-random genomic translocations on FPGS gene expression to determine whether gene fusions may be responsible for the heterogeneity of FPGS expression in ALL cells. Using quantitative fluorescence real-time RT-PCR, we first detected reduced levels of FPGS mRNA expression in the RCH-ACV (Bp-ALL, t(1:19)/E2A-PBX1) and REH (Bp-ALL, t(12:21)/TEL-AML1) human leukemia cell lines expressing non-random chromosomal gene fusions when compared to control (NALM6). To determine if expression of fusion proteins encoded by the E2A-PBX1 and TEL-AML1 translocations lead to decrease FPGS promoter activity and mRNA expression, we used a FPGS-luciferase reporter gene assay. The CHO FPGS null mutant cell line AUXB1, was triple transfected with constructs expressing luciferase (pFPGS-luc), β-galactosidase (pCMVβ), and E2A-PBX1 (pCMVKJ7) or TEL-AML1 (pTEL-AML1) and assayed for luciferase and β-galactosidase activity. Expression of E2A-PBX1 and TEL-AML1, determined by quantitative RT-PCR and Western blot, resulted in 45% and 30% decrease in the level of normalized FPGS-luciferase activity, respectively. These data indicate that FPGS gene transcription is decreased by both translocations, either by direct binding to regulatory elements or indirectly via downstream pathways influenced by E2A-PBX1 and TEL-AML1. Therefore, to confirm these findings and to identify genes or pathways in Bp-ALL regulated by E2A-PBX1 and TEL-AML1 gene fusions, we constructed NALM6 stable cell lines expressing E2A-PBX1 or TEL-AML1 using vectors pCI-neo/E2A-PBX1 and pCI-neo/TEL-AML1. In both NALM6 stable cell lines the level of FPGS mRNA expression was decreased by more than 50% compared to wild type (NALM6). Total RNA was then extracted from NALM6, NALM6/E2A-PBX1, and NALM6/TEL-AML1 cells and analyzed by cDNA microarray. Gene expression profile analysis from existing databases and our NALM6/E2A-PBX1 and NALM6/TEL-AML1 stable cell lines identified genes differentially expressed compared to NALM6 control. These included transcription factors (CSDA, AKAP12, ARL2, PBX1, TEL), cell cycle regulators (CDK2P1), signal transduction (PIK3C3), proliferation and cell migration (PTPRK), zinc finger binding protein (PHF2), among others. This study is the first to demonstrate the causal relationship between the presence of E2A-PBX1 and TEL-AML1 gene fusions and altered FPGS expression and may explain the differential sensitivity to MTX exhibited by these childhood ALL phenotypes.
APA, Harvard, Vancouver, ISO, and other styles
10

Tukaramrao, Diwakar Bastihalli, Arati Sharma, Dhimant Desai, and Sinisa Dovat. "Abstract 6052: Metabolic consequences of casein kinase 2α inhibition in lymphoid leukemia." Cancer Research 83, no. 7_Supplement (April 4, 2023): 6052. http://dx.doi.org/10.1158/1538-7445.am2023-6052.

Full text
Abstract:
Abstract Overexpression of casein kinase 2α (CK2α) is a common feature in lymphoid leukemias. Constitutively active CK2α can disable transcriptional activity of lymphoid transcription factors like IKAROS that act as metabolic gatekeeper and limit the energy supply needed for oncogenic transformation of B cells. Our studies have shown that pharmacological inhibition of CK2α can restore the transcriptional activity of IKAROS and ablate leukemia. However, the role of CK2α in glucose metabolism has not been fully studied in B and T acute lymphoblastic leukemia (ALL). Therefore, in the present study we explored the metabolic alterations induced by the pharmacological inhibition of CK2α in B and T ALL. We studied the effect of CK2α inhibition using a specific inhibitor - CX4945 on cell proliferation, glucose utilization, lactate production, and intracellular ATP levels using established methods in human B-ALL (NALM6) cell line. Effect of CX4945 on glycolysis was studied using the Seahorse cell analyzer in human B (NALM6 and 697) and T (MOLT4 and 697) ALL cell lines. Metabolomics study was undertaken to analyze differential metabolite profiling in NALM6 cells treated with CX4945 compared to vehicle using LC-MS/MS based methods. Results showed that CX4945 induced apoptotic cell death in ALL cell lines with IC50 concentrations ranging between 4-10 µM. CX4945 treatment significantly affected the glucose consumption in NALM6 cells. Similarly, substantial decrease in intracellular ATP and lactate levels compared to vehicle was recorded. CK2α inhibition significantly decreased the glycolytic activity in B- and T-ALL cell lines. It was observed that glycolytic reserves were significantly decreased in 697 (3-fold), MOLT4 (4-fold), CEM (3-fold) cells incubated with CX4945 in comparison to vehicle. The principal component analysis from metabolomics study showed a clear separation between CX4945 and vehicle-treated NAML6 cells. Sixty-four statistically significant, differentially expressed metabolites were recorded in the study. Analyte classes included TCA cycle intermediates, nucleic acids and their precursors as well as glycolysis intermediates that were significantly affected by CK2α inhibition. In conclusion, our study shows that selective inhibition of CK2α by CX4945 caused energy deficiency and cell death in ALL cell lines. CK2 inhibition targeted the key energy dependent pathway by rendering ALL cells inefficient in utilizing glucose and operating glycolysis for generation of cellular energy. These results offer a new mechanistic understanding of CK2α inhibition mediated ablation of ALL. Citation Format: Diwakar Bastihalli Tukaramrao, Arati Sharma, Dhimant Desai, Sinisa Dovat. Metabolic consequences of casein kinase 2α inhibition in lymphoid leukemia [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 6052.
APA, Harvard, Vancouver, ISO, and other styles

Dissertations / Theses on the topic "Nalm6 cells"

1

Henry, Danielle. "The role of the human INO80 complex in telomere maintenance." Thesis, 2020. http://hdl.handle.net/1866/24728.

Full text
Abstract:
Les extrémités des chromosomes contiennent des répétitions de séquences d’ADN appelées télomères qui empêchent l’activation inopportune de la réponse aux dommages de l'ADN afin de préserver l'intégrité génomique. Les télomères raccourcissent à chaque cycle de réplication d’ADN et la télomérase a pour fonction de contrebalancer cette érosion en allongeant les télomères. Les cellules somatiques n’expriment pas la télomérase, donc leur durée de vie est normalement limitée par ce raccourcissement progressif des télomères qui conduit à l'activation de la voie p53 entraînant un arrêt de la croissance cellulaire. En revanche, les cellules cancéreuses acquièrent l'immortalité cellulaire principalement en réactivant la télomérase ou en utilisant des méthodes alternatives d'allongement des télomères basées sur la recombinaison d’ADN. Auparavant, dans notre laboratoire, un criblage CRISPR à l'échelle du génome a été réalisé dans la lignée cellulaire pré-B NALM-6 traitée avec la molécule BIBR1532, un inhibiteur de la télomérase. Ces résultats suggéraient que cinq sous-unités du complexe de remodelage de la chromatine INO80, lorsque supprimées indépendamment, réduisaient la prolifération des cellules ayant un raccourcissement des télomères induit par le BIBR1532. Mon objectif était d'étudier cette interaction génétique afin de comprendre les processus biologiques impliqués dans cette létalité synthétique. Après l'élimination des gènes codant à la fois pour la sous-unité enzymatique de la télomérase humaine (hTERT) ainsi que les sous-unités spécifiques du complexe INO80 humain, nous avons constaté que les cellules double-négatives avaient une capacité proliférative réduite, ce qui démontre que l’interaction génétique mesurée par criblage CRISPR est bel et bien spécifique. Étant donné le rôle du facteur de transcription p53 dans la réponse cellulaire au raccourcissement télomérique, nous avons exploré l’importance de cette voie de signalisation pour l’interaction entre le complexe INO80 humain et la télomérase. Après l’activation de p53 avec un traitement avec la molécule nutlin-3a, les niveaux d'expression de plusieurs cibles de p53 tels que MDM2 et CDKN1A ont augmenté dans les cellules ayant une délétion du gène NFRKB, codant pour une sous-unité du complexe INO80 humain. Les cellules ayant une délétion du gène UCHL5, codant pour le partenaire d’interaction de NFRKB, ont également montré une augmentation de l’expression de MDM2 lorsque traitées avec nutlin-3a. Enfin, la perte de télomérase (hTERT) modifie les niveaux d'expression des composants de la 2 voie p53 CDKN1A, BAX et MDM2. En conclusion, la suppression des gènes codant pour des sous-unités du complexe INO80 telles que NFRKB ou UCHL5 est nuisible aux cellules ayant une délétion de la télomérase. Le complexe INO80 humain peut être impliqué dans l'inhibition de la voie p53, en réponse à l'activation de p53 soit par des télomères courts ou avec un traitement avec nutlin-3a. Des recherches plus approfondies sur cette interaction génétique pourraient mener au développement de nouvelles thérapies combinatoires afin d’inhiber la croissance des cellules cancéreuses.
The ends of chromosomes contain telomeric repeats that prevent the DNA damage response from being activated in order to preserve genomic integrity. Telomerase functions to alleviate incomplete DNA replication at telomeres, and to repair those telomeres damaged by various means including oxidative damage. The lifespan of telomerase negative somatic cells is normally restricted by gradual telomere shortening which can lead to the activation of the p53 pathway resulting in cellular growth arrest. Cancer cells often elongate their telomeres in order to acquire cellular immortality predominantly by reactivating telomerase or by using recombination-based, alternative telomere lengthening methods. Previously in our lab, a genome-wide CRISPR screen was conducted in the pre-B cell line NALM-6 treated with a small molecule inhibitor of telomerase, BIBR1532. These previous results suggested that five subunits of the INO80 chromatin-remodeling complex, when independently deleted, reduced cellular proliferation in cells with BIBR1532 induced telomere shortening. My goal was to investigate this genetic interaction in order to understand the biological processes implicated in this synthetic lethal relationship. After the knockout of the genes encoding both the enzymatic subunit of human telomerase (hTERT) and specific subunits of the human INO80 complex, I found that the proliferative capacity of NALM-6 cells was reduced. This result indicates the genetic interaction identified by CRISPR screening is in fact specific. In addition, after p53 stimulation with nutlin-3a treatment, expression levels of the p53 pathway component MDM2 were altered after the knockout of the genes encoding specific subunits of the human INO80 complex, NFRKB and UCHL5, individually. CDKN1A expression was also altered after nutlin-3a treatment and NFRKB knockout. Finally, the loss of telomerase (hTERT) alters the expression levels of the p53 pathway components CDKN1A, BAX and MDM2. In conclusion, the deletion of the genes encoding specific subunits of the INO80 complex, including NFRKB and UCHL5, is harmful to cells after hTERT knockout. The human INO80 complex may be involved in inhibiting the p53 pathway, in response to p53 activation by short telomeres or nutlin-3a treatment. Further investigation into this synthetic lethal relationship may shed light on new combinatorial therapeutics in cancer.
APA, Harvard, Vancouver, ISO, and other styles

Book chapters on the topic "Nalm6 cells"

1

Adachi, Noritaka, Aya Kurosawa, and Hideki Koyama. "Highly Proficient Gene Targeting by Homologous Recombination in the Human Pre-B Cell Line Nalm-6." In Chromosomal Mutagenesis, 17–29. Totowa, NJ: Humana Press, 2008. http://dx.doi.org/10.1007/978-1-59745-232-8_2.

Full text
APA, Harvard, Vancouver, ISO, and other styles
2

Drexler, Hans G. "NALM-1." In The Leukemia-Lymphoma Cell Line FactsBook, 115–16. Elsevier, 2001. http://dx.doi.org/10.1016/b978-012221970-2/50041-3.

Full text
APA, Harvard, Vancouver, ISO, and other styles
3

Drexler, Hans G. "NALM-6." In The Leukemia-Lymphoma Cell Line FactsBook, 117–18. Elsevier, 2001. http://dx.doi.org/10.1016/b978-012221970-2/50042-5.

Full text
APA, Harvard, Vancouver, ISO, and other styles
4

Drexler, Hans G. "NALM-16." In The Leukemia-Lymphoma Cell Line FactsBook, 119. Elsevier, 2001. http://dx.doi.org/10.1016/b978-012221970-2/50043-7.

Full text
APA, Harvard, Vancouver, ISO, and other styles
5

Drexler, Hans G. "NALM-19." In The Leukemia-Lymphoma Cell Line FactsBook, 120. Elsevier, 2001. http://dx.doi.org/10.1016/b978-012221970-2/50044-9.

Full text
APA, Harvard, Vancouver, ISO, and other styles
6

Drexler, Hans G. "NALM-20." In The Leukemia-Lymphoma Cell Line FactsBook, 121–22. Elsevier, 2001. http://dx.doi.org/10.1016/b978-012221970-2/50045-0.

Full text
APA, Harvard, Vancouver, ISO, and other styles
7

Drexler, Hans G. "NALM-24." In The Leukemia-Lymphoma Cell Line FactsBook, 123–24. Elsevier, 2001. http://dx.doi.org/10.1016/b978-012221970-2/50046-2.

Full text
APA, Harvard, Vancouver, ISO, and other styles
8

Drexler, Hans G. "NALM-26." In The Leukemia-Lymphoma Cell Line FactsBook, 125. Elsevier, 2001. http://dx.doi.org/10.1016/b978-012221970-2/50047-4.

Full text
APA, Harvard, Vancouver, ISO, and other styles
9

Drexler, Hans G. "NALM-27." In The Leukemia-Lymphoma Cell Line FactsBook, 126–27. Elsevier, 2001. http://dx.doi.org/10.1016/b978-012221970-2/50048-6.

Full text
APA, Harvard, Vancouver, ISO, and other styles
10

Drexler, Hans G. "NALM-29." In The Leukemia-Lymphoma Cell Line FactsBook, 128–29. Elsevier, 2001. http://dx.doi.org/10.1016/b978-012221970-2/50049-8.

Full text
APA, Harvard, Vancouver, ISO, and other styles

Conference papers on the topic "Nalm6 cells"

1

Lu, Jun Q., Huafeng Ding, Carissa L. Reynolds, Yuanming Feng, Li V. Yang, Fred E. Bertrand, Tom J. McConnell, and Xin-Hua Hu. "Angle-resolved Light Scattering Study of NALM-6 and HL-60 Cells for White Blood Cell Differentiation." In Biomedical Optics. Washington, D.C.: OSA, 2010. http://dx.doi.org/10.1364/biomed.2010.btud85.

Full text
APA, Harvard, Vancouver, ISO, and other styles
We offer discounts on all premium plans for authors whose works are included in thematic literature selections. Contact us to get a unique promo code!

To the bibliography