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
  • Gutierrez, Alejandro  (127)
Material
Language
  • 1
    In: Electroanalysis, Wiley, Vol. 26, No. 8 ( 2014-08), p. 1694-1701
    Abstract: This work reports the analytical applications of a graphene paste electrode (GrPE) for the quantification of dopamine, ethanol and phenolic compounds. Dopamine was detected by differential pulse voltammetry‐adsorptive stripping with medium exchange at submicromolar levels even in the presence of high excess of ascorbic acid and serotonin. The electrocatalytic activity of graphene towards the oxidation of NADH and the reduction of quinones allowed the sensitive amperometric determination of ethanol and phenols using GrPE modified with alcohol dehydrogenase/NAD + or polyphenol oxidase, respectively, with successful applications in real samples like alcoholic beverages and tea.
    Type of Medium: Online Resource
    ISSN: 1040-0397 , 1521-4109
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2014
    detail.hit.zdb_id: 1483564-2
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 7_Supplement ( 2023-04-04), p. 3500-3500
    Abstract: Amplification of chromosomal material derived from 12q is pathognomonic in a subset of soft tissue sarcomas (STS), including well-differentiated and dedifferentiated liposarcoma. However, it remains unclear which of the amplified genes are oncogenic drivers, obstacles to rational therapeutic development. We defined a minimal 12q amplicon in human liposarcoma, and tested the oncogenic activity of each recurrently amplified gene to accelerate the onset of AKT-induced STS in a genetically engineered zebrafish model of the disease. This revealed several genes whose overexpression accelerated the onset of AKT-induced STS, including METTL1, best-known for its ability to catalyze methylation of tRNAs. Surprisingly, catalytically defective mutants of METTL1 exhibited oncogenic activity that was at least as potent as the wild-type gene in zebrafish and human STS, indicating enzyme-independent oncogenic functions. We found that METTL1 promotes ribosome assembly and oncogenic translation independent of its enzymatic activity. In human liposarcoma cells and mouse embryonic fibroblasts, METTL1 overexpression was sufficient to stimulate polysome formation, and this effect was phenocopied by catalytically dead mutants of METTL1. METTL1 overexpression also robustly stimulated the rate of protein synthesis in cells (P & lt; 0.0001), and in a cell-free biochemical assay for translation of a reporter mRNA (P & lt; 0.001). The ability of METTL1 to stimulate ribosomal function triggered a therapeutically targetable dependency, because small molecule inhibitors of ribosome biogenesis had potent therapeutic activity against METTL1-amplified (P & lt; 0.0001), but not METTL1 wild-type, sarcoma cells. Thus, METTL1 is a soft tissue sarcoma oncogene that stimulates oncogenic translation independent of its enzymatic activity, an oncogenic function that can be targeted therapeutically. Citation Format: Raja Hussain Ali, Brianna Silverman, Esteban A. Orellana, Alyssa Kennedy, Caitlin Bowers, Ashley Gutierrez, David Papke, Christopher D. Fletcher, Antonio P. Atayde, Akiko Shimamura, Richard I. Gregory, Alejandro Gutierrez. A noncatalytic role for mettl1 in oncogenic translation in soft tissue sarcoma [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 3500.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 3155-3155
    Abstract: Abstract 3155 The often aggressive and unpredictable behavior of T-cell lymphoblastic malignancies continues to pose both major clinical challenges in children and adults. To discover oncogenic pathways downstream of critical genetic abnormalities that are characteristically deregulated in T-cell acute lymphoblastic leukemia (T-ALL), and to identify novel molecular targets for anti-leukemic agents with T-cell specificity, we carried out a genome-wide functional screen in T-ALL cell lines using a retroviral library of inducible short-hairpin RNAs (shRNAs). Among the genes that are required for the growth of T-ALL cells, we found that loss of TYK2, a JAK family tyrosine kinase, was specifically lethal in each of three T-ALL cell lines that we tested in this screen. By contrast, TYK2 knock-down did not affect the growth of cell lines from diffuse large B-cell lymphoma or multiple myeloma, indicating that TYK2 is specifically required for the growth of T-ALL cells. We confirmed by knock-down with multiple independent shRNAs that the loss of TYK2 induces apoptosis in T-ALL, whereas knock-down of other JAK proteins (JAK1, JAK2 or JAK3) had no effect. We found that the TYK2 protein is constitutively phosphorylated in many T-ALL cell lines, and that these cells are sensitive to small molecule JAK/TYK2 inhibitors, including JAK inhibitor-I, AG-490 and CEP-701. To identify upstream receptors involved in TYK2 activation, we knocked down IFNAR, IFNGR, IL10R and IL12RB, and found that loss of IFNAR1 and IFNAR2 specifically inhibit the growth of T-ALL cells, as strongly as TYK2 knock-down. In addition, targeted knock-down analysis of downstream STAT proteins revealed that loss of STAT1 also inhibits the growth of T-ALL cells, indicating that this transcription factor is involved in the TYK2 pathway and required for cell survival. In fact, STAT1 protein was constitutively phosphorylated in many T-ALL cell lines and this phosphorylation was inhibited by both TYK2 knock-down and treatment with JAK/TYK2 inhibitors. Although interferon-mediated pathways are cytostatic in many cell types, our results indicate a requirement for the IFNAR-TYK2-STAT1 pathway in promoting the growth and survival of T-ALL cells. Dependence on this pathway confers unique sensitivity of T-ALL cells to TYK2 inhibition by small molecule inhibitors, thus providing a novel therapeutic target for clinical testing in patients with this disease. Disclosures: Druker: Molecular MD: Equity Ownership.
    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 ...
  • 4
    In: Blood, American Society of Hematology, Vol. 115, No. 9 ( 2010-03-04), p. 1735-1745
    Abstract: To identify dysregulated pathways in distinct phases of NOTCH1-mediated T-cell leukemogenesis, as well as small-molecule inhibitors that could synergize with or substitute for γ-secretase inhibitors (GSIs) in T-cell acute lymphoblastic leukemia (T-ALL) therapy, we compared gene expression profiles in a Notch1-induced mouse model of T-ALL with those in human T-ALL. The overall patterns of NOTCH1-mediated gene expression in human and mouse T-ALLs were remarkably similar, as defined early in transformation in the mouse by the regulation of MYC and its target genes and activation of nuclear factor-κB and PI3K/AKT pathways. Later events in murine Notch1-mediated leukemogenesis included down-regulation of genes encoding tumor suppressors and negative cell cycle regulators. Gene set enrichment analysis and connectivity map algorithm predicted that small-molecule inhibitors, including heat-shock protein 90, histone deacetylase, PI3K/AKT, and proteasome inhibitors, could reverse the gene expression changes induced by NOTCH1. When tested in vitro, histone deacetylase, PI3K and proteasome inhibitors synergized with GSI in suppressing T-ALL cell growth in GSI-sensitive cells. Interestingly, alvespimycin, a potent inhibitor of the heat-shock protein 90 molecular chaperone, markedly inhibited the growth of both GSI-sensitive and -resistant T-ALL cells, suggesting that its loss disrupts signal transduction pathways crucial for the growth and survival of T-ALL cells.
    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 ...
  • 5
    In: Blood, American Society of Hematology, Vol. 132, No. Supplement 1 ( 2018-11-29), p. 891-891
    Abstract: Asparaginase, a bacterial enzyme that depletes the nonessential amino acid asparagine, is an integral component of acute leukemia therapy. However, asparaginase resistance is a common clinical problem whose biologic basis is poorly understood. We hypothesized, based on the concept of synthetic lethality, that gain-of-fitness alterations in the drug-resistant cells had conferred a survival advantage that could be exploited therapeutically. To identify molecular pathways that promote fitness of leukemic cells upon treatment with asparaginase, we performed a genome-wide CRISPR/Cas9 loss-of-function screen in the asparaginase-resistant T-ALL cell line CCRF-CEM. Cas9-expressing CCRF-CEM cells were transduced with a genome-wide guide RNA library (Shalem et al. Science343, 84-87, 2014), treated with either vehicle or asparaginase (10 U/L), and guide RNA representation was assessed. Our internal positive control, asparagine synthetase, was the gene most significantly depleted in asparaginase-treated cells (RRA significance score = 1.56 x 10-7), followed closely by two regulators of Wnt signaling, NKD2 and LGR6 (RRA score = 6 x 10-6and 2.19 x 10-5, respectively). To test how these genes regulate Wnt signaling in T-ALL, we transduced CCRF-CEM cells with shRNAs targeting NKD2 or LGR6, or with an shLuciferase control. Knockdown of NKD2 or LGR6 increased levels of active β-catenin, as well as the activity of a TopFLASH reporter of canonical Wnt/β-catenin transcriptional activity (P 〈 0.0001), indicating that NKD2 and LGR6 are negative regulators of Wnt signaling in these cells. We then validated the screen results using shRNA knockdown of NKD2 or LGR6, which profoundly sensitized these cells to asparaginase (P 〈 0.0001) and potentiated asparaginase-induced apoptosis (P 〈 0.0001). Inhibition of glycogen synthase kinase 3 (GSK3) is a key event in Wnt-induced signal transduction. Thus, we tested whether CHIR99021, an ATP-competitive inhibitor of both GSK3 isoforms (GSK3α and GSK3β), could phenocopy the effect of Wnt pathway activation. Pharmacologic inhibition of GSK3 induced significant sensitization to asparaginase across a panel of cell lines representing distinct subtypes of treatment-resistant acute leukemia, including T-ALL, AML and hypodiploid B-ALL (Fig. 1a, b). Importantly, GSK3 inhibition did not sensitize normal hematopoietic progenitors to asparaginase, suggesting a leukemia-specific effect. Wnt-induced sensitization to asparaginase was independent of β-catenin and mTOR activation, because genetic and pharmacologic manipulation of these Wnt targets had no effect on asparaginase response. Instead, it was mediated by Wnt-dependent stabilization of proteins (Wnt/STOP), which inhibits GSK3-dependent protein ubiquitination and proteasomal degradation (Acebron et al. Mol Cell54, 663-674, 2014, Taelman et al. Cell143, 1136-1148, 2010). Indeed,Wnt-induced sensitization to asparaginase was completely blocked by the transduction of leukemia cells with FBXW7 (P 〈 0.0001), whose overexpression can reverse Wnt/STOP (Acebron et al. Mol Cell54, 663-674; 2014), or by expression of a hyperactive proteasomal subunit ΔN-PSMA4 (P 〈 0.0001), which globally increases protein degradation (Choi et al. Nat Commun7, 10963, 2016). Although GSK3α and GSK3βare redundant for many of their biologic functions, genetic or pharmacologic inhibition of GSK3α fully phenocopied Wnt-induced sensitization to asparaginase (P 〈 0.0001), whereas selective inhibition of GSK3β had no effect. We then leveraged the recently developed GSK3α-selective small molecule inhibitor BRD0705 (Wagner et al. Sci Transl Med10, 2018) to test the in vivo therapeutic potential of our findings. Immunodeficient NRG mice were injected with leukemic cells from a primary asparaginase-resistant T-ALL patient derived xenograft, and treatment was begun after confirmation of leukemic engraftment (n=16 mice per group). In vivo, this PDX proved completely resistant to asparaginase or BRD0705 monotherapy, whereas the combination was highly efficacious (median survival of 17 days in vehicle, vs. median not reached at 60 days in combo-treated mice; P 〈 0.0001; Fig. 2a, b). The combination was also well-tolerated, with no appreciable weight changes or increases in serum bilirubin levels. Our findings provide a molecular rationale for activating Wnt/STOP signaling to improve the therapeutic index of asparaginase. 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: 2018
    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. 1408-1408
    Abstract: Using comparative genome hybridization (array CGH), we have discovered a small area of increased copy number on the long arm of chromosome 6 in 8 out of 20 (40%) T-ALL cell lines. The region of increased copy number is very small, containing only one gene, MYB, the cellular homolog of the avian oncogene v-myb. By performing fiber-FISH on these cell lines, we have shown that the increased copy number results from a discrete tandem duplication of the MYB gene on one allele. Although myb is a frequent target of retroviral insertional activation in screens for oncogenes whose overexpression accelerates the onset of murine T-ALL, its overexpression and increased copy number has not previously been implicated in human T-ALL. Using gene expression profiling and Western blotting, we have demonstrated that the duplication in human T-ALL results in increased levels of MYB expression. Furthermore, using quantitative PCR we have confirmed that this tandem duplication occurs in primary human T-ALL samples. In our studies to date, MYB tandem duplication and overexpression appears to occur as part of the major multistep molecular pathway in T-ALL that affects a majority of cases, in which the leukemic cells also have TAL1/SCL and LMO1/2 overexpression, and NOTCH1 gene activating mutations, together with homozygous deletion of the INK4A/ARF locus. We are currently determining the mechanism through which MYB overexpression contributes to the pathogenesis of T-ALL by siRNA knockdown in T-ALL cell lines. Our finding of MYB tandem gene duplication differs from classic forms of oncogene amplification involving double minutes or homogenously staining regions. It is possible that MYB copy number is increased through a novel somatic mechanism of allele-specific, tandem duplication of a small genomic region during the process of malignant transformation. Another possibility is suggested by recent studies documenting that inherited large-scale copy number variation (CNV) accounts for much of the phenotypic diversity within human populations. Further studies will be needed to determine whether MYB tandem duplication is present in the germline DNA of T-ALL patients, which if identified, would provide the first example of an inherited CNV functioning as a mechanism of cancer susceptibility.
    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. 120, No. 21 ( 2012-11-16), p. 3558-3558
    Abstract: Abstract 3558 Despite the use of intensive and toxic treatment regimens, T-cell acute lymphoblastic leukemia (T-ALL) remains fatal in 25% of children and 60% of adults, highlighting the need for novel therapeutic strategies. We performed two complementary small molecule screens to identify novel T-ALL therapeutic agents: i) an in vivo zebrafish screen for small molecules toxic to MYC-overexpressing thymocytes, and ii) an in vitro screen for drugs that synergize with NOTCH inhibitors in human T-ALL cell lines. Hits common to both screens included perphenazine, an FDA-approved phenothiazine antipsychotic. Perphenazine potently induced mitochondrial apoptosis and had antileukemic activity in zebrafish T-ALL, in human T-ALL cell lines, and in primary human T-ALL lymphoblasts. However, the molecular target mediating the antileukemic activity of perphenazine appeared to be unrelated to any of its known cellular targets. To identify the relevant biologic target of perphenazine, we developed a novel method for drug target discovery, termed activity correlation proteomics, which combines native protein extraction of isotope-labeled proteomes, fluorous affinity chromatography, and quantitative mass spectrometry proteomics to relate phenotypic activities of drugs with their binding targets. Using activity correlation proteomics, we identified protein phosphatase 2A (PP2A) as the protein whose binding affinity for different phenothiazines most closely correlated with their antileukemic activity. Perphenazine was shown to activate PP2A in T-ALL cells, as assessed by rapid drug-induced dephosphorylation of multiple PP2A substrates including MYC, AKT, p70S6K, ERK, and BAD, and its effects were phenocopied by FTY720, a PP2A activator. Moreover, shRNA knockdown of PP2A scaffolding or catalytic subunits attenuated the activity of perphenazine, indicating that PP2A is required for its antileukemic activity. Our findings thus establish a novel strategy for pharmacologic activation of the PP2A tumor suppressor, and highlight its therapeutic potential in T-ALL and other cancers driven by hyperphosphorylated PP2A substrates. Disclosures: Off Label Use: Discovery of antileukemic activity of perphenazine mediated via activation of PP2A.
    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 ...
  • 8
    In: Blood, American Society of Hematology, Vol. 118, No. 15 ( 2011-10-13), p. 4169-4173
    Abstract: The BCL11B transcription factor is required for normal T-cell development, and has recently been implicated in the pathogenesis of T-cell acute lymphoblastic leukemia (T-ALL) induced by TLX overexpression or Atm deficiency. To comprehensively assess the contribution of BCL11B inactivation to human T-ALL, we performed DNA copy number and sequencing analyses of T-ALL diagnostic specimens, revealing monoallelic BCL11B deletions or missense mutations in 9% (n = 10 of 117) of cases. Structural homology modeling revealed that several of the BCL11B mutations disrupted the structure of zinc finger domains required for this transcription factor to bind DNA. BCL11B haploinsufficiency occurred across each of the major molecular subtypes of T-ALL, including early T-cell precursor, HOXA-positive, LEF1-inactivated, and TAL1-positive subtypes, which have differentiation arrest at diverse stages of thymocyte development. Our findings provide compelling evidence that BCL11B is a haploinsufficient tumor suppressor that collaborates with all major T-ALL oncogenic lesions in human thymocyte transformation.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2011
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 115, No. 14 ( 2010-04-08), p. 2845-2851
    Abstract: To further unravel the molecular pathogenesis of T-cell acute lymphoblastic leukemia (T-ALL), we performed high-resolution array comparative genomic hybridization on diagnostic specimens from 47 children with T-ALL and identified monoallelic or biallelic LEF1 microdeletions in 11% (5 of 47) of these primary samples. An additional 7% (3 of 44) of the cases harbored nonsynonymous sequence alterations of LEF1, 2 of which produced premature stop codons. Gene expression microarrays showed increased expression of MYC and MYC targets in cases with LEF1 inactivation, as well as differentiation arrest at an early cortical stage of thymocyte development characterized by expression of CD1B, CD1E, and CD8, with absent CD34 expression. LEF1 inactivation was associated with a younger age at the time of T-ALL diagnosis, as well as activating NOTCH1 mutations, biallelic INK4a/ARF deletions, and PTEN loss-of-function mutations or activating mutations of PI3K or AKT genes. These cases generally lacked overexpression of the TAL1, HOX11, HOX11L2, or the HOXA cluster genes, which have been used to define separate molecular pathways leading to T-ALL. Our findings suggest that LEF1 inactivation is an important step in the molecular pathogenesis of T-ALL in a subset of young children.
    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 ...
  • 10
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 1001-1001
    Abstract: Abstract 1001 Background: Tyrosine kinases are widely implicated in the genesis of hematologic malignancies and tyrosine kinase inhibitors have achieved remarkable clinical outcomes. However, most cancer patients are ineligible for this form of therapy because we lack knowledge of the oncogenes driving the growth of their malignant cells. In particular, first-line therapy for T-cell acute lymphoblastic leukemia (T-ALL) fails in 25% of children and more than 50% of adults despite improvements in clinical outcomes due to intensification of therapy, and these relapsed patients have a very poor prognosis. Most current T-ALL therapy regimens lack tyrosine kinase inhibitors suggesting that addition of effective kinase inhibitors to T-ALL therapy may improve clinical outcomes. We report that tyrosine kinase 2 (TYK2), signaling through signal transducer and activator of transcription 1 (STAT1), is vital for the maintenance of T-ALL growth and viability, suggesting that incorporation of TYK2 inhibitors into T-ALL therapy regimens may improve patient outcomes. Methods: T-ALL diagnostic specimens were collected with informed consent and neoplastic cells were isolated on a Ficoll gradient. Cells were treated with an siRNA library that individually silences each member of the tyrosine kinase gene family and cultured for 4 days at which time an MTS assay was used to assess cell viability. DNA was isolated from 16 T-ALL cell lines as well as 50 T-ALL patient specimens. TYK2 was sequenced and identified point mutant were cloned and introduced into Ba/F3 cells for assessment of transformative capacity. Transformed Ba/F3 cells were lysed and immunoblotted with antibodies specific for total or phospho-TYK2, STAT1, STAT3, and ERK1/2. Results: RNAi functional profiling of cells from a T-ALL patient specimen demonstrated dependence on TYK2 for cell viability. Sequence analysis of TYK2 in T-ALL cell lines and patient samples revealed a diversity of TYK2 point mutations. These mutations were located just outside of or within the FERM domain (V15A, G36D, G36R, S47N, R425H), the pseudokinase domain (V731I), and the kinase domain (E957D, and R1027H). Introduction of these point mutants into Ba/F3 cells revealed that 7/8 (all except R425H) transformed these cells to factor-independent growth. Analysis of whole cell extracts from transformed cells revealed constitutive phosphorylation of TYK2, STAT1, STAT3, and ERK1/2. Treatment of transformed cells with JAK Inhibitor I revealed extreme sensitivity of cells to this TYK2 kinase inhibitor as well as reduction of TYK2 AND STAT1 phosphorylation. Conclusions: We demonstrate for the first time that the TYK2-STAT1 pathway is constitutively active and required for viability in the setting of T-ALL, sometimes due to gain-of-function point mutations in TYK2. Small-molecule kinase inhibitors with activity against TYK2 reduce the growth of these TYK2-dependent cells. Hence, we have defined a new, oncogenic signaling pathway in T-ALL cells that can be readily modulated by kinase inhibitors. Interestingly, this pathway has previously been associated with tumor suppression, therefore, our findings indicate that activation of this pathway can have varying effects depending on cellular context. Cumulatively, these data offer new therapeutic options for T-ALL patients while simultaneously providing novel insights into the biology of T-cells and the TYK2-STAT1 signaling cascade. Disclosures: Druker: Molecular MD: Equity Ownership.
    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 ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...