Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Transl Med ; 11(494)2019 05 29.
Article in English | MEDLINE | ID: mdl-31142678

ABSTRACT

Given the high frequency of activating NOTCH1 mutations in T cell acute lymphoblastic leukemia (T-ALL), inhibition of the γ-secretase complex remains an attractive target to prevent ligand-independent release of the cytoplasmic tail and oncogenic NOTCH1 signaling. However, four different γ-secretase complexes exist, and available inhibitors block all complexes equally. As a result, these cause severe "on-target" gastrointestinal tract, skin, and thymus toxicity, limiting their therapeutic application. Here, we demonstrate that genetic deletion or pharmacologic inhibition of the presenilin-1 (PSEN1) subclass of γ-secretase complexes is highly effective in decreasing leukemia while avoiding dose-limiting toxicities. Clinically, T-ALL samples were found to selectively express only PSEN1-containing γ-secretase complexes. The conditional knockout of Psen1 in developing T cells attenuated the development of a mutant NOTCH1-driven leukemia in mice in vivo but did not abrogate normal T cell development. Treatment of T-ALL cell lines with the selective PSEN1 inhibitor MRK-560 effectively decreased mutant NOTCH1 processing and led to cell cycle arrest. These observations were extended to T-ALL patient-derived xenografts in vivo, demonstrating that MRK-560 treatment decreases leukemia burden and increased overall survival without any associated gut toxicity. Therefore, PSEN1-selective compounds provide a potential therapeutic strategy for safe and effective targeting of T-ALL and possibly also for other diseases in which NOTCH signaling plays a role.


Subject(s)
Molecular Targeted Therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Receptors, Notch/antagonists & inhibitors , Animals , Cell Line, Tumor , Cell Proliferation , Disease Progression , Gastrointestinal Tract/pathology , Gene Deletion , Gene Targeting , Humans , Male , Mice , Presenilin-1/metabolism , Receptors, Notch/metabolism , Signal Transduction , T-Lymphocytes/metabolism
2.
Oncogene ; 38(27): 5457-5468, 2019 07.
Article in English | MEDLINE | ID: mdl-30967635

ABSTRACT

T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive cancer arising from T-cell progenitors. Although current treatments, including chemotherapy and glucocorticoids, have significantly improved survival, T-ALL remains a fatal disease and new treatment options are needed. Since more than 60% of T-ALL cases bear oncogenic NOTCH1 mutations, small molecule inhibitors of NOTCH1 signalling; γ-secretase inhibitors (GSI), are being actively investigated for the treatment of T-ALL. Unfortunately, GSI have shown limited clinical efficacy and dose-limiting toxicities. We hypothesized that by combining known drugs, blocking NOTCH activity through another mechanism, may synergize with GSI enabling equal efficacy at a lower concentration. Here, we show that the clinically used anti-malarial drug chloroquine (CQ), an inhibitor of lysosomal function and autophagy, decreases T-ALL cell viability and proliferation. This effect of CQ was not observed in GSI-resistant T-ALL cell lines. Mechanistically, CQ impairs the redox balance, induces ds DNA breaks and activates the DNA damage response. CQ also interferes with intracellular trafficking and processing of oncogenic NOTCH1. Interestingly, we show for the first time that the addition of CQ to γ-secretase inhibition has a synergistic therapeutic effect on T-ALL and reduces the concentration of GSI required to obtain a reduction in cell viability and a block of proliferation. Overall, our results suggest that CQ may be a promising repurposed drug in the treatment of T-ALL, as a single treatment or in combination with GSI, increasing the therapeutic ratio.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Antimalarials/pharmacology , Chloroquine/pharmacology , Enzyme Inhibitors/pharmacology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Receptor, Notch1/metabolism , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Damage , Humans , Ligands , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
3.
Cell ; 166(1): 193-208, 2016 Jun 30.
Article in English | MEDLINE | ID: mdl-27293189

ABSTRACT

γ-Secretases are a family of intramembrane-cleaving proteases involved in various signaling pathways and diseases, including Alzheimer's disease (AD). Cells co-express differing γ-secretase complexes, including two homologous presenilins (PSENs). We examined the significance of this heterogeneity and identified a unique motif in PSEN2 that directs this γ-secretase to late endosomes/lysosomes via a phosphorylation-dependent interaction with the AP-1 adaptor complex. Accordingly, PSEN2 selectively cleaves late endosomal/lysosomal localized substrates and generates the prominent pool of intracellular Aß that contains longer Aß; familial AD (FAD)-associated mutations in PSEN2 increased the levels of longer Aß further. Moreover, a subset of FAD mutants in PSEN1, normally more broadly distributed in the cell, phenocopies PSEN2 and shifts its localization to late endosomes/lysosomes. Thus, localization of γ-secretases determines substrate specificity, while FAD-causing mutations strongly enhance accumulation of aggregation-prone Aß42 in intracellular acidic compartments. The findings reveal potentially important roles for specific intracellular, localized reactions contributing to AD pathogenesis.


Subject(s)
Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/analysis , Amyloid beta-Peptides/metabolism , Peptide Fragments/metabolism , Presenilin-2/analysis , Adaptor Protein Complex 1/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amino Acid Motifs , Amyloid Precursor Protein Secretases/metabolism , Animals , Cell Line, Tumor , Endosomes/chemistry , Humans , Lysosomes/chemistry , Mice , Presenilin-1/analysis , Presenilin-1/chemistry , Presenilin-1/genetics , Presenilin-1/metabolism , Presenilin-2/chemistry , Presenilin-2/genetics , Presenilin-2/metabolism , Rats , Substrate Specificity
4.
Oncotarget ; 7(27): 41251-41264, 2016 Jul 05.
Article in English | MEDLINE | ID: mdl-27183910

ABSTRACT

Glioblastoma multiforme (GBM) is the most common malignant brain tumor in adults. The current standard of care includes surgery followed by radiotherapy (RT) and chemotherapy with temozolomide (TMZ). Treatment often fails due to the radiation resistance and intrinsic or acquired TMZ resistance of a small percentage of cells with stem cell-like behavior (CSC). The NOTCH signaling pathway is expressed and active in human glioblastoma and NOTCH inhibitors attenuate tumor growth in vivo in xenograft models. Here we show using an image guided micro-CT and precision radiotherapy platform that a combination of the clinically approved NOTCH/γ-secretase inhibitor (GSI) RO4929097 with standard of care (TMZ + RT) reduces tumor growth and prolongs survival compared to dual combinations. We show that GSI in combination with RT and TMZ attenuates proliferation, decreases 3D spheroid growth and results into a marked reduction in clonogenic survival in primary and established glioma cell lines. We found that the glioma stem cell marker CD133, SOX2 and Nestin were reduced following combination treatments and NOTCH inhibitors albeit in a different manner. These findings indicate that NOTCH inhibition combined with standard of care treatment has an anti-glioma stem cell effect which provides an improved survival benefit for GBM and encourages further translational and clinical studies.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Benzazepines/administration & dosage , Brain Neoplasms/therapy , Chemoradiotherapy/methods , Dacarbazine/analogs & derivatives , Glioblastoma/therapy , Receptors, Notch/antagonists & inhibitors , Animals , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Cell Line, Tumor , Dacarbazine/administration & dosage , Glioblastoma/mortality , Glioblastoma/pathology , Humans , Mice , Mice, Nude , Survival Analysis , Temozolomide , Xenograft Model Antitumor Assays
5.
Structure ; 23(7): 1227-35, 2015 Jul 07.
Article in English | MEDLINE | ID: mdl-26051713

ABSTRACT

Notch receptors are transmembrane proteins that undergo activating proteolysis in response to ligand stimulation. A negative regulatory region (NRR) maintains receptor quiescence by preventing protease cleavage prior to ligand binding. We report here the X-ray structure of the NRR of autoinhibited human Notch3, and compare it with the Notch1 and Notch2 NRRs. The overall architecture of the autoinhibited conformation, in which three LIN12-Notch repeat (LNR) modules wrap around a heterodimerization domain, is preserved in Notch3, but the autoinhibited conformation of the Notch3 NRR is less stable. The Notch3 NRR uses a highly conserved surface on the third LNR module to form a dimer in the crystal. Similar homotypic interfaces exist in Notch1 and Notch2. Together, these studies reveal distinguishing structural features associated with increased basal activity of Notch3, demonstrate increased ligand-independent signaling for disease-associated mutations that map to the Notch3 NRR, and identify a conserved dimerization interface present in multiple Notch receptors.


Subject(s)
Receptors, Notch/chemistry , Cell Line, Tumor , Crystallography, X-Ray , HEK293 Cells , Humans , Models, Molecular , Mutation, Missense , Protein Binding , Protein Interaction Domains and Motifs , Protein Multimerization , Proteolysis , Receptor, Notch3 , Receptors, Notch/genetics , Receptors, Notch/metabolism
6.
J Biol Chem ; 290(23): 14705-16, 2015 Jun 05.
Article in English | MEDLINE | ID: mdl-25918160

ABSTRACT

Cell surface receptors of the NOTCH family of proteins are activated by ligand induced intramembrane proteolysis. Unfolding of the extracellular negative regulatory region (NRR), enabling successive proteolysis by the enzymes Adam10 and γ-secretase, is rate-limiting in NOTCH activation. Mutations in the NOTCH1 NRR are associated with ligand-independent activation and frequently found in human T-cell malignancies. In mammals four NOTCH receptors and five Delta/Jagged ligands exist, but mutations in the NRR are only rarely reported for receptors other than NOTCH1. Using biochemical and functional assays, we compared the molecular mechanisms of ligand-independent signaling in NOTCH1 and the highly related NOTCH2 receptor. Both murine Notch1 and Notch2 require the metalloprotease protease Adam17, but not Adam10 during ligand-independent activation. Interestingly, the human NOTCH2 receptor is resistant to ligand-independent activation compared with its human homologs or murine orthologs. Taken together, our data reveal subtle but functionally important differences for the NRR among NOTCH paralogs and homologs.


Subject(s)
ADAM Proteins/metabolism , Receptor, Notch2/metabolism , ADAM17 Protein , Amino Acid Sequence , Animals , Calcium/metabolism , Cell Line , Humans , Leukemia/genetics , Leukemia/metabolism , Mice , Molecular Sequence Data , Mutation , Protein Unfolding , Receptor, Notch1/chemistry , Receptor, Notch1/metabolism , Receptor, Notch2/chemistry , Receptor, Notch2/genetics
7.
Mol Cell Biol ; 34(15): 2822-32, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24842903

ABSTRACT

In mammals, there are four NOTCH receptors and five Delta-Jagged-type ligands regulating many aspects of embryonic development and adult tissue homeostasis. NOTCH proteins are type I transmembrane receptors that interact with ligands on adjacent cells and are activated by regulated intramembrane proteolysis (RIP). The activation mechanism of NOTCH1 receptors upon ligand binding is well understood and requires cleavage by ADAM10 metalloproteases prior to intramembranous cleavage by γ-secretase. How the other human NOTCH receptor homologues are activated upon ligand binding is not known. Here, we dissect the proteolytic activation mechanism of the NOTCH2 and NOTCH3 receptors. We show that NOTCH2 and NOTCH3 signaling can be triggered by both Delta-Jagged-type ligands and requires ADAM10 and presenilin-1 or -2. Importantly, we did not find any role for the highly related ADAM17/TACE (tumor necrosis factor alpha-converting enzyme) protease in ligand-induced NOTCH2 or NOTCH3 signaling. These results demonstrate that canonical ligand-induced proteolysis of the NOTCH1, -2, and -3 receptors strictly depends on consecutive cleavage of these receptors by ADAM10 and the presenilin-containing γ-secretase complex, leading to transcriptional activation.


Subject(s)
ADAM Proteins/metabolism , Presenilin-1/metabolism , Presenilin-2/metabolism , Receptor, Notch2/metabolism , Receptors, Notch/metabolism , ADAM17 Protein , Amyloid Precursor Protein Secretases/metabolism , Animals , Cell Line , HEK293 Cells , Humans , Membrane Proteins/metabolism , Metalloproteases/metabolism , Mice , NIH 3T3 Cells , Proteolysis
8.
Radiother Oncol ; 108(3): 440-445, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23891097

ABSTRACT

BACKGROUND AND PURPOSE: Patients with advanced NSCLC have survival rates <15%. The NOTCH pathway plays an important role during lung development and physiology but is often deregulated in lung cancer, making it a potential therapeutic target. We investigated NOTCH signaling in NSCLC and hypothesized that high NOTCH activity contributes to radiation resistance. MATERIALS AND METHODS: NOTCH signaling in NSCLC patient samples was investigated using quantitative RT-PCR. H460 NSCLC cells with either high or blocked NOTCH activity were generated and their radiation sensitivity monitored using clonogenic assays. In vivo, xenograft tumors were irradiated and response assessed using growth delay. Microenvironmental parameters were analyzed by immunohistochemistry. RESULTS: Patients with high NOTCH activity in tumors showed significantly worse disease-free survival. In vitro, NOTCH activity did not affect the proliferation or intrinsic radiosensitivity of NSCLC cells. In contrast, xenografts with blocked NOTCH activity grew slower than wild type tumors. Tumors with high NOTCH activity grew significantly faster, were more hypoxic and showed a radioresistant phenotype. CONCLUSIONS: We demonstrate an important role for NOTCH in tumor growth and correlate high NOTCH activity with poor prognosis and radioresistance. Blocking NOTCH activity in NSCLC might be a promising intervention to improve outcome after radiotherapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung/radiotherapy , Lung Neoplasms/radiotherapy , Radiation Tolerance , Receptors, Notch/physiology , Animals , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/radiation effects , Humans , Lung Neoplasms/pathology , Mice , Signal Transduction/physiology
9.
Radiother Oncol ; 99(3): 392-397, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21680037

ABSTRACT

BACKGROUND AND PURPOSE: Hypoxia is a hallmark of solid cancers and associated with metastases and treatment failure. During tumor progression epithelial cells often acquire mesenchymal features, a phenomenon known as epithelial-to-mesenchymal transition (EMT). Intratumoral hypoxia has been linked to EMT induction. We hypothesized that signals from the tumor microenvironment such as growth factors and tumor oxygenation collaborate to promote EMT and thereby contribute to radioresistance. MATERIALS AND METHODS: Gene expression changes under hypoxia were analyzed using microarray and validated by qRT-PCR. Conversion of epithelial phenotype upon hypoxic exposure, TGFß addition or oncogene activation was investigated by Western blot and immunofluorescence. Cell survival following ionizing radiation was assayed using clonogenic survival. RESULTS: Upon hypoxia, TGFß addition or EGFRvIII expression, MCF7, A549 and NMuMG epithelial cells acquired a spindle shape and lost cell-cell contacts. Expression of epithelial markers such as E-cadherin decreased, whereas mesenchymal markers such as vimentin and N-cadherin increased. Combining hypoxia with TGFß or EGFRvIII expression, lead to more rapid and pronounced EMT-like phenotype. Interestingly, E-cadherin expression and the mesenchymal appearance were reversible upon reoxygenation. Mesenchymal conversion and E-cadherin loss were associated with radioresistance. CONCLUSIONS: Our findings describe a mechanism by which the tumor microenvironment may contribute to tumor radioresistance via E-cadherin loss and EMT.


Subject(s)
Cadherins/metabolism , Neoplasms/metabolism , Radiation Tolerance , Analysis of Variance , Blotting, Western , Cell Hypoxia , Cell Line, Tumor , Cell Survival/radiation effects , Cell Transformation, Neoplastic , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Expression Regulation, Neoplastic , Humans , Immunoenzyme Techniques , Mesoderm/metabolism , Mesoderm/pathology , Neoplasms/genetics , Neoplasms/pathology , Phenotype , Real-Time Polymerase Chain Reaction , Signal Transduction , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL
...