Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Clin Cancer Res ; 29(16): 3189-3202, 2023 08 15.
Article in English | MEDLINE | ID: mdl-37339179

ABSTRACT

PURPOSE: Many cancers lack argininosuccinate synthetase 1 (ASS1), the rate-limiting enzyme of arginine biosynthesis. This deficiency causes arginine auxotrophy, targetable by extracellular arginine-degrading enzymes such as ADI-PEG20. Long-term tumor resistance has thus far been attributed solely to ASS1 reexpression. This study examines the role of ASS1 silencing on tumor growth and initiation and identifies a noncanonical mechanism of resistance, aiming to improve clinical responses to ADI-PEG20. EXPERIMENTAL DESIGN: Tumor initiation and growth rates were measured for a spontaneous Ass1 knockout (KO) murine sarcoma model. Tumor cell lines were generated, and resistance to arginine deprivation therapy was studied in vitro and in vivo. RESULTS: Conditional Ass1 KO affected neither tumor initiation nor growth rates in a sarcoma model, contradicting the prevalent idea that ASS1 silencing confers a proliferative advantage. Ass1 KO cells grew robustly through arginine starvation in vivo, while ADI-PEG20 remained completely lethal in vitro, evidence that pointed toward a novel mechanism of resistance mediated by the microenvironment. Coculture with Ass1-competent fibroblasts rescued growth through macropinocytosis of vesicles and/or cell fragments, followed by recycling of protein-bound arginine through autophagy/lysosomal degradation. Inhibition of either macropinocytosis or autophagy/lysosomal degradation abrogated this growth support effect in vitro and in vivo. CONCLUSIONS: Noncanonical, ASS1-independent tumor resistance to ADI-PEG20 is driven by the microenvironment. This mechanism can be targeted by either the macropinocytosis inhibitor imipramine or the autophagy inhibitor chloroquine. These safe, widely available drugs should be added to current clinical trials to overcome microenvironmental arginine support of tumors and improve patient outcomes.


Subject(s)
Sarcoma , Soft Tissue Neoplasms , Humans , Animals , Mice , Sarcoma/drug therapy , Hydrolases/pharmacology , Polyethylene Glycols/pharmacology , Polyethylene Glycols/therapeutic use , Cell Line, Tumor , Argininosuccinate Synthase/genetics , Arginine/metabolism , Soft Tissue Neoplasms/drug therapy , Tumor Microenvironment
2.
J Clin Oncol ; : JCO2018792549, 2018 10 29.
Article in English | MEDLINE | ID: mdl-30372397

ABSTRACT

Clear cell renal cell carcinoma (ccRCC) is the most common renal cell carcinoma subtype, and metastatic ccRCC is associated with 5-year survival rates of 10% to 20%. Genetically, ccRCC originates from sequential losses of multiple tumor suppressor genes. Remarkably, chromosome 3p loss occurs in more than 90% of sporadic ccRCCs. This results in concurrent one-copy loss of four tumor suppressor genes that are also mutated individually at high frequency in ccRCC (ie, VHL, 80%; PBRM1, 29% to 46%; BAP1, 6% to 19%; and SETD2, 8% to 30%). Pathogenically, 3p loss probably represents the first genetic event that occurs in sporadic ccRCC and the second genetic event in VHL-mutated hereditary ccRCC. VHL constitutes the substrate recognition module of the VCB-Cul2 E3 ligase that degrades HIF1/2α, whereas PBRM1, BAP1, and SETD2 are epigenetic modulators that regulate gene transcription. Because 3p loss and VHL inactivation are nearly universal truncal events in ccRCC, the resulting HIF1/2 signaling overdrive and accompanied tumor hypervascularization probably underlie the therapeutic benefits observed with vascular endothelial growth factor receptor inhibitors, including sorafenib, sunitinib, pazopanib, axitinib, bevacizumab, cabozantinib, and lenvatinib. Furthermore, recent marked advances in ccRCC genomics, transcriptomics, proteomics, metabolomics, molecular mechanisms, mouse models, prognostic and predictive biomarkers, and clinical trials have rendered invaluable translational insights concerning precision kidney cancer therapeutics. With an armamentarium encompassing 13 drugs that exploit seven unique therapeutic mechanisms (ie, cytokines, vascular endothelial growth factor receptor, mTORC1, cMET/AXL, fibroblast growth factor receptor, programmed cell death-1 and programmed death-ligand 1, and cytotoxic T-cell lymphocyte associated-4) to treat metastatic renal cell carcinoma, one of the imminent clinical questions concerning care of patients with metastatic ccRCC is how a personalized treatment strategy, through rationally combining and sequencing different therapeutic modalities, can be formulated to offer the best clinical outcome for individual patients. Here, we attempt to integrate recent discoveries of immediate translational impacts and discuss future translational challenges and opportunities.

3.
Cell Rep ; 18(12): 2893-2906, 2017 03 21.
Article in English | MEDLINE | ID: mdl-28329682

ABSTRACT

PBRM1 is the second most commonly mutated gene after VHL in clear cell renal cell carcinoma (ccRCC). However, the biological consequences of PBRM1 mutations for kidney tumorigenesis are unknown. Here, we find that kidney-specific deletion of Vhl and Pbrm1, but not either gene alone, results in bilateral, multifocal, transplantable clear cell kidney cancers. PBRM1 loss amplified the transcriptional outputs of HIF1 and STAT3 incurred by Vhl deficiency. Analysis of mouse and human ccRCC revealed convergence on mTOR activation, representing the third driver event after genetic inactivation of VHL and PBRM1. Our study reports a physiological preclinical ccRCC mouse model that recapitulates somatic mutations in human ccRCC and provides mechanistic and therapeutic insights into PBRM1 mutated subtypes of human ccRCC.


Subject(s)
Carcinoma, Renal Cell/metabolism , HMGB Proteins/metabolism , Kidney Neoplasms/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Animals , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , DNA-Binding Proteins , Down-Regulation/genetics , Gene Deletion , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , HMGB Proteins/deficiency , Humans , Hydronephrosis/genetics , Hydronephrosis/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Integrases/metabolism , Kidney/metabolism , Kidney/pathology , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Oxidative Phosphorylation , STAT3 Transcription Factor/metabolism , Signal Transduction , Transcription, Genetic
4.
J Clin Invest ; 126(9): 3526-40, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27482884

ABSTRACT

Genomic studies have linked mTORC1 pathway-activating mutations with exceptional response to treatment with allosteric inhibitors of mTORC1 called rapalogs. Rapalogs are approved for selected cancer types, including kidney and breast cancers. Here, we used sequencing data from 22 human kidney cancer cases to identify the activating mechanisms conferred by mTOR mutations observed in human cancers and advance precision therapeutics. mTOR mutations that clustered in focal adhesion kinase targeting domain (FAT) and kinase domains enhanced mTORC1 kinase activity, decreased nutrient reliance, and increased cell size. We identified 3 distinct mechanisms of hyperactivation, including reduced binding to DEP domain-containing MTOR-interacting protein (DEPTOR), resistance to regulatory associated protein of mTOR-mediated (RAPTOR-mediated) suppression, and altered kinase kinetics. Of the 28 mTOR double mutants, activating mutations could be divided into 6 complementation groups, resulting in synergistic Rag- and Ras homolog enriched in brain-independent (RHEB-independent) mTORC1 activation. mTOR mutants were resistant to DNA damage-inducible transcript 1-mediated (REDD1-mediated) inhibition, confirming that activating mutations can bypass the negative feedback pathway formed between HIF1 and mTORC1 in the absence of von Hippel-Lindau (VHL) tumor suppressor expression. Moreover, VHL-deficient cells that expressed activating mTOR mutants grew tumors that were sensitive to rapamycin treatment. These data may explain the high incidence of mTOR mutations observed in clear cell kidney cancer, where VHL loss and HIF activation is pathognomonic. Our study provides mechanistic and therapeutic insights concerning mTOR mutations in human diseases.


Subject(s)
Kidney Neoplasms/genetics , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/metabolism , Animals , Antibiotics, Antineoplastic/pharmacology , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/genetics , DNA Damage , Female , Genome, Human , Humans , Kidney Neoplasms/drug therapy , Kinetics , Male , Mice , Mice, SCID , Molecular Dynamics Simulation , Mutation , Mutation, Missense , Plasmids/metabolism , Protein Domains , RNA, Small Interfering/metabolism , TOR Serine-Threonine Kinases/genetics , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
5.
J Clin Invest ; 125(3): 1203-14, 2015 Mar 02.
Article in English | MEDLINE | ID: mdl-25664857

ABSTRACT

Head morphogenesis requires complex signal relays to enable precisely coordinated proliferation, migration, and patterning. Here, we demonstrate that, during mouse head formation, taspase1-mediated (TASP1-mediated) cleavage of the general transcription factor TFIIA ensures proper coordination of rapid cell proliferation and morphogenesis by maintaining limited transcription of the negative cell cycle regulators p16Ink4a and p19Arf from the Cdkn2a locus. In mice, loss of TASP1 function led to catastrophic craniofacial malformations that were associated with inadequate cell proliferation. Compound deficiency of Cdkn2a, especially p16Ink4a deficiency, markedly reduced the craniofacial anomalies of TASP1-deficent mice. Furthermore, evaluation of mice expressing noncleavable TASP1 targets revealed that TFIIA is the principal TASP1 substrate that orchestrates craniofacial morphogenesis. ChIP analyses determined that noncleaved TFIIA accumulates at the p16Ink4a and p19Arf promoters to drive transcription of these negative regulators. In summary, our study elucidates a regulatory circuit comprising proteolysis, transcription, and proliferation that is pivotal for construction of the mammalian head.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/genetics , Endopeptidases/physiology , Transcription Factor TFIIA/metabolism , Transcription, Genetic , Animals , Brain/embryology , Brain/pathology , Cell Proliferation , Cells, Cultured , Craniofacial Abnormalities/enzymology , Craniofacial Abnormalities/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Facial Bones/abnormalities , Facial Bones/embryology , Genetic Loci , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Morphogenesis , Skull/abnormalities , Skull/embryology
6.
Cell Res ; 24(11): 1354-66, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25267403

ABSTRACT

Taspase1, a highly conserved threonine protease, cleaves nuclear transcriptional regulators mixed-lineage leukemia (MLL, MLL1), MLL2, TFIIA, and ALF to orchestrate a wide variety of biological processes. In vitro studies thus far demonstrated that Taspase1 plays important roles in the proliferation of various cancer cell lines, including HER2-positive breast cancer cells. To investigate the role of Taspase1 in breast tumorigenesis in vivo, we deleted Taspase1 from mouse mammary glands by generating MMTV-neu;MMTV-cre;Tasp1(F/-) mice. We demonstrate that initiation of MMTV-neu- but not MMTV-wnt-driven breast cancer is blocked in the absence of Taspase1. Importantly, Taspase1 loss alone neither impacts normal development nor pregnancy physiology of the mammary gland. In mammary glands Taspase1 deficiency abrogates MMTV-neu-induced cyclins E and A expression, thereby preventing tumorigenesis. The mechanisms were explored in HER2-positive breast cancer cell line BT474 and HER2-transformed MCF10A cells and validated using knockdown-resistant Taspase1. As Taspase1 was shown to cleave MLL which forms complexes with E2F transcription factors to regulate Cyclins E, A, and B expression in mouse embryonic fibroblasts (MEFs), we investigated whether the cleavage of MLL by Taspase1 constitutes an essential in vivo axis for HER2/neu-induced mammary tumorigenesis. To this end, we generated MMTV-neu;MLL(nc/nc) transgenic mice that carry homozygous non-cleavable MLL alleles. Remarkably, these mice are also protected from HER2/neu-driven breast tumorigenesis. Hence, MLL is the primary Taspase1 substrate whose cleavage is required for MMTV-neu-induced tumor formation. As Taspase1 plays critical roles in breast cancer pathology, it may serve as a therapeutic target for HER2-positive human breast cancer.


Subject(s)
Cyclin E/metabolism , Endopeptidases/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , Receptor, ErbB-2/metabolism , Alleles , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Line , Cell Proliferation , Cell Transformation, Neoplastic , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Endopeptidases/chemistry , Endopeptidases/genetics , Female , Genotype , Histone-Lysine N-Methyltransferase , Humans , Lactation , Mammary Glands, Animal/enzymology , Mice , Mice, Transgenic , Mutagenesis, Site-Directed , Myeloid-Lymphoid Leukemia Protein/genetics , Pregnancy , RNA Interference , Receptor, ErbB-2/genetics
7.
Dev Cell ; 27(2): 188-200, 2013 Oct 28.
Article in English | MEDLINE | ID: mdl-24176642

ABSTRACT

The evolution of tissue-specific general transcription factors (GTFs), such as testis-specific TBP-related factor 2 (TRF2), enables the spatiotemporal expression of highly specialized genetic programs. Taspase1 is a protease that cleaves nuclear factors MLL1, MLL2, TFIIAα-ß, and ALFα-ß (TFIIAτ). Here, we demonstrate that Taspase1-mediated processing of TFIIAα-ß drives mammalian spermatogenesis. Both Taspase1(-/-) and noncleavable TFIIAα-ßnc/nc testes release immature germ cells with impaired transcription of Transition proteins (Tnp) and Protamines (Prm), exhibiting chromatin compaction defects and recapitulating those observed with TRF2(-/-) testes. Although the unprocessed TFIIA still complexes with TRF2, this complex is impaired in targeting and thus activating Tnp1 and Prm1 promoters. The current study presents a paradigm in which a protease (Taspase1) cleaves a ubiquitously expressed GTF (TFIIA) to enable tissue-specific (testis) transcription, meeting the demand for sophisticated regulation of distinct subsets of genes in higher organisms.


Subject(s)
Endopeptidases/metabolism , Spermatogenesis/physiology , Telomeric Repeat Binding Protein 2/metabolism , Transcription Factor TFIIA/metabolism , Animals , Chromosomal Proteins, Non-Histone/genetics , Endopeptidases/genetics , Enzyme Activation , Gene Expression Regulation, Developmental , Histone-Lysine N-Methyltransferase/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid-Lymphoid Leukemia Protein/metabolism , Promoter Regions, Genetic , Protamines/genetics , Spermatozoa , Telomeric Repeat Binding Protein 2/genetics
8.
PLoS Genet ; 9(1): e1003132, 2013.
Article in English | MEDLINE | ID: mdl-23326237

ABSTRACT

Mastermind-like 1 (MAML1) is a transcriptional co-activator in the Notch signaling pathway. Recently, however, several reports revealed novel and unique roles for MAML1 that are independent of the Notch signaling pathway. We found that MAML1 enhances the transcriptional activity of runt-related transcription factor 2 (Runx2), a transcription factor essential for osteoblastic differentiation and chondrocyte proliferation and maturation. MAML1 significantly enhanced the Runx2-mediated transcription of the p6OSE2-Luc reporter, in which luciferase expression was controlled by six copies of the osteoblast specific element 2 (OSE2) from the Runx2-regulated osteocalcin gene promoter. Interestingly, a deletion mutant of MAML1 lacking the N-terminal Notch-binding domain also enhanced Runx2-mediated transcription. Moreover, inhibition of Notch signaling did not affect the action of MAML1 on Runx2, suggesting that the activation of Runx2 by MAML1 may be caused in a Notch-independent manner. Overexpression of MAML1 transiently enhanced the Runx2-mediated expression of alkaline phosphatase, an early marker of osteoblast differentiation, in the murine pluripotent mesenchymal cell line C3H10T1/2. MAML1(-/-) embryos at embryonic day 16.5 (E16.5) had shorter bone lengths than wild-type embryos. The area of primary spongiosa of the femoral diaphysis was narrowed. At E14.5, extended zone of collagen type II alpha 1 (Col2a1) and Sox9 expression, markers of chondrocyte differentiation, and decreased zone of collagen type X alpha 1 (Col10a1) expression, a marker of hypertrophic chondrocyte, were observed. These observations suggest that chondrocyte maturation was impaired in MAML1(-/-) mice. MAML1 enhances the transcriptional activity of Runx2 and plays a role in bone development.


Subject(s)
Bone Development/genetics , Chondrocytes , Core Binding Factor Alpha 1 Subunit , Embryonic Development/genetics , Nuclear Proteins , Transcription Factors , Animals , Cell Differentiation , Cell Proliferation , Chondrocytes/cytology , Chondrocytes/metabolism , Chondrocytes/pathology , Collagen Type II/metabolism , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Gene Expression Regulation, Developmental , Humans , Mice , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Osteoblasts/pathology , Osteocalcin/genetics , Osteocalcin/metabolism , Receptors, Notch/genetics , Receptors, Notch/metabolism , SOX9 Transcription Factor/metabolism , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism
9.
Cancer Res ; 72(11): 2913, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-24619300
10.
Development ; 138(23): 5235-46, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22069191

ABSTRACT

Mastermind (Mam) is one of the elements of Notch signaling, a system that plays a pivotal role in metazoan development. Mam proteins form transcriptionally activating complexes with the intracellular domains of Notch, which are generated in response to the ligand-receptor interaction, and CSL DNA-binding proteins. In mammals, three structurally divergent Mam isoforms (MamL1, MamL2 and MamL3) have been identified. There have also been indications that Mam interacts functionally with various other transcription factors, including the p53 tumor suppressor, ß-catenin and NF-κB. We have demonstrated previously that disruption of MamL1 causes partial deficiency of Notch signaling in vivo. However, MamL1-deficient mice did not recapitulate total loss of Notch signaling, suggesting that other members could compensate for the loss or that Notch signaling could proceed in the absence of Mam in certain contexts. Here, we report the generation of lines of mice null for MamL3. Although MamL3-null mice showed no apparent abnormalities, mice null for both MamL1 and MamL3 died during the early organogenic period with classic pan-Notch defects. Furthermore, expression of the lunatic fringe gene, which is strictly controlled by Notch signaling in the posterior presomitic mesoderm, was undetectable in this tissue of the double-null embryos. Neither of the single-null embryos exhibited any of these phenotypes. These various roles of the three Mam proteins could be due to their differential physical characteristics and/or their spatiotemporal distributions. These results indicate that engagement of Mam is essential for Notch signaling, and that the three Mam isoforms have distinct roles in vivo.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Nuclear Proteins/metabolism , Receptors, Notch/metabolism , Signal Transduction/physiology , Trans-Activators/metabolism , Transcription Factors/metabolism , Animals , Blotting, Southern , Blotting, Western , DNA Primers/genetics , Fibroblasts , Flow Cytometry , Gene Expression Regulation, Developmental/genetics , Glycosyltransferases/metabolism , In Situ Hybridization , Luciferases , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Nuclear Proteins/genetics , Plasmids/genetics , Polymerase Chain Reaction , Real-Time Polymerase Chain Reaction , Signal Transduction/genetics , Trans-Activators/genetics , Transcription Factors/genetics
11.
Acta Histochem Cytochem ; 43(6): 163-8, 2010 Dec 29.
Article in English | MEDLINE | ID: mdl-21245983

ABSTRACT

Rho family G proteins including Rac regulate a variety of cellular functions, such as morphology, motility, and gene expression. Here we developed a fluorescence resonance energy transfer-based analysis in which we could monitor the activity of Rac1. To detect fluorescence resonance energy transfer, yellow fluorescent protein fused Rac1 and cyan fluorescent protein fused Cdc42-Rac1-interaction-binding domain of Pak1 protein were used as intermolecular probes of FRET. The fluorophores were separated with linear unmixing method. The fluorescence resonance energy transfer efficiency was measured by acceptor photobleaching assisted assay. With these methods, the Rac1 activity was visualized in a cell. The present findings indicate that this approach is sensitive enough to achieve results similar to those from ratiometric fluorescence resonance energy transfer analysis.

12.
Proc Natl Acad Sci U S A ; 104(23): 9764-9, 2007 Jun 05.
Article in English | MEDLINE | ID: mdl-17535917

ABSTRACT

Mastermind (Mam) is one of the elements of Notch signaling, an ancient system that plays a pivotal role in metazoan development. Genetic analyses in Drosophila and Caenorhabditis elegans have shown Mam to be an essential positive regulator of this signaling pathway in these species. Mam proteins bind to and stabilize the DNA-binding complex of the intracellular domains of Notch and CBF-1, Su(H), Lag-1 (CSL) DNA-binding proteins in the nucleus. Mammals have three Mam proteins, which show remarkable similarities in their functions while having an unusual structural diversity. There have also been recent indications that Mam-1 functionally interacts with other transcription factors including p53 tumor suppressor. We herein describe that Mam-1 deficiency in mice abolishes the development of splenic marginal zone B cells, a subset strictly dependent on Notch2, a CSL protein and Delta1 ligand. Mam-1 deficiency also causes a partially impaired development of early thymocytes, while not affecting the generation of definitive hematopoiesis, processes that are dependent on Notch1. We also demonstrate the transcriptional activation of a target promoter by constitutively active forms of Notch to decrease severalfold in cultured Mam-1-deficient cells. These results indicate that Mam-1 is thus required to some extent for Notch-dependent stages in lymphopoiesis, thus supporting the notion that Mam is an essential component of the canonical Notch pathway in mammals.


Subject(s)
Lymphocytes/physiology , Nuclear Proteins/deficiency , Nuclear Proteins/metabolism , Signal Transduction/genetics , Transcription Factors/deficiency , Transcription Factors/metabolism , Animals , Blotting, Western , Cell Line , Flow Cytometry , Hematopoiesis/genetics , Mice , Mice, Knockout , Nuclear Proteins/genetics , Receptor, Notch1/metabolism , Transcription Factors/genetics
13.
J Biol Chem ; 277(52): 50612-20, 2002 Dec 27.
Article in English | MEDLINE | ID: mdl-12386158

ABSTRACT

Mastermind (Mam) is one of the evolutionarily conserved elements of Notch signaling. Genetic analyses in Drosophila implicated it as an important positive regulator of the pathway. We show here identification of two new members of human Mam family (human Mastermind-2 (hMam-2) and human Mastermind-3 (hMam-3)), which retain characteristics similar to human Mastermind-1 (hMam-1) and Drosophila Mastermind. Both hMam-2 and hMam-3 stabilize and participate in the DNA-binding complex RBP-J/CBF-1 protein and the Notch intracellular domains that serve as intermediates of the signaling. Both hMam-2 and hMam-3 enhanced the activation of transcription from a target promoter by Notch signaling. However, we also show evidence that the activation of the target promoter by Notch3 and Notch4 is more efficiently potentiated by hMam-2 than by hMam-1 or -3. The multiplicity of Mam proteins in the mammalian system may help provide divergence to the strength of the Notch signals in different cell types.


Subject(s)
DNA-Binding Proteins/genetics , Membrane Proteins/physiology , Nuclear Proteins/genetics , Signal Transduction , Transcription, Genetic , Amino Acid Sequence , Cell Line , Cell Nucleus/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/isolation & purification , Gene Expression Regulation , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Nuclear Proteins/chemistry , Nuclear Proteins/isolation & purification , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Receptors, Cell Surface/physiology , Receptors, Notch , Recombinant Proteins/metabolism , Restriction Mapping , Sequence Alignment , Sequence Homology, Amino Acid , T-Lymphocytes , Trans-Activators , Transcription Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...