Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
PLoS One ; 19(5): e0303145, 2024.
Article in English | MEDLINE | ID: mdl-38728268

ABSTRACT

Water stress can adversely affect seed germination and plant growth. Seed osmopriming is a pre-sowing treatment in which seeds are soaked in osmotic solutions to undergo the first stage of germination prior to radicle protrusion. Seed osmopriming enhances germination performance under stressful environmental conditions, making it an effective method to improve plant resistance and yield. This study analyzed the effect of seed osmopriming with polyethylene glycol (PEG) on seed germination and physiological parameters of Coronilla varia L. Priming treatments using 10% to 30% PEG enhanced germination percentage, germination vigor, germination index, vitality index, and seedling mass and reduced the time to reach 50% germination (T50). The PEG concentration that led to better results was 10%. The content of soluble proteins (SP), proline (Pro), soluble sugars (SS), and malondialdehyde (MDA) in Coronilla varia L. seedlings increased with the severity of water stress. In addition, under water stress, electrolyte leakage rose, and peroxidase (POD) and superoxide dismutase (SOD) activities intensified, while catalase (CAT) activity increased at mild-to-moderate water stress but declined with more severe deficiency. The 10% PEG priming significantly improved germination percentage, germination vigor, germination index, vitality index, and time to 50% germination (T50) under water stress. Across the water stress gradient here tested (8 to 12% PEG), seed priming enhanced SP content, Pro content, and SOD activity in Coronilla varia L. seedlings compared to the unprimed treatments. Under 10% PEG-induced water stress, primed seedlings displayed a significantly lower MDA content and electrolyte leakage than their unprimed counterparts and exhibited significantly higher CAT and POD activities. However, under 12% PEG-induced water stress, differences in electrolyte leakage, CAT activity, and POD activity between primed and unprimed treatments were not significant. These findings suggest that PEG priming enhances the osmotic regulation and antioxidant capacity of Coronilla varia seedlings, facilitating seed germination and seedling growth and alleviating drought stress damage, albeit with reduced efficacy under severe water deficiency.


Subject(s)
Germination , Polyethylene Glycols , Seedlings , Seeds , Polyethylene Glycols/pharmacology , Germination/drug effects , Seedlings/drug effects , Seedlings/growth & development , Seeds/drug effects , Seeds/growth & development , Dehydration , Catalase/metabolism , Malondialdehyde/metabolism , Proline/metabolism , Superoxide Dismutase/metabolism , Water/metabolism
2.
bioRxiv ; 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38659938

ABSTRACT

Chimeric antigen receptor (CAR) T cell therapy targeting CD19 elicits remarkable clinical efficacy in B-cell malignancies, but many patients relapse due to failed expansion and/or progressive loss of CAR-T cells. We recently reported a strategy to potently restimulate CAR-T cells in vivo, enhancing their functionality by administration of a vaccine-like stimulus comprised of surrogate peptide ligands for a CAR linked to a lymph node-targeting amphiphilic PEG-lipid (termed CAR-T-vax). Here, we demonstrate a general strategy to generate and optimize peptide mimotopes enabling CAR-T-vax generation for any CAR. Using the clinical CD19 CAR FMC63 as a test case, we employed yeast surface display to identify peptide binders to soluble IgG versions of FMC63, which were subsequently affinity matured by directed evolution. CAR-T vaccines using these optimized mimotopes triggered marked expansion of both murine CD19 CAR-T cells in a syngeneic model and human CAR-T cells in a humanized mouse model of B cell acute lymphoblastic leukemia (B-ALL), and enhanced control of leukemia progression. This approach thus enables vaccine boosting to be applied to any clinically-relevant CAR-T cell product.

3.
Cancer Cell ; 41(12): 2100-2116.e10, 2023 12 11.
Article in English | MEDLINE | ID: mdl-38039964

ABSTRACT

Selection of the best tumor antigen is critical for the therapeutic success of chimeric antigen receptor (CAR) T cells in hematologic malignancies and solid tumors. The anaplastic lymphoma kinase (ALK) receptor is expressed by most neuroblastomas while virtually absent in most normal tissues. ALK is an oncogenic driver in neuroblastoma and ALK inhibitors show promising clinical activity. Here, we describe the development of ALK.CAR-T cells that show potent efficacy in monotherapy against neuroblastoma with high ALK expression without toxicity. For neuroblastoma with low ALK expression, combination with ALK inhibitors specifically potentiates ALK.CAR-T cells but not GD2.CAR-T cells. Mechanistically, ALK inhibitors impair tumor growth and upregulate the expression of ALK, thereby facilitating the activity of ALK.CAR-T cells against neuroblastoma. Thus, while neither ALK inhibitors nor ALK.CAR-T cells will likely be sufficient as monotherapy in neuroblastoma with low ALK density, their combination specifically enhances therapeutic efficacy.


Subject(s)
Neuroblastoma , Humans , Anaplastic Lymphoma Kinase/genetics , Anaplastic Lymphoma Kinase/metabolism , Neuroblastoma/drug therapy , Neuroblastoma/genetics , Neuroblastoma/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Antigens, Neoplasm , T-Lymphocytes , Cell Line, Tumor
4.
Cell ; 186(15): 3148-3165.e20, 2023 07 20.
Article in English | MEDLINE | ID: mdl-37413990

ABSTRACT

Chimeric antigen receptor (CAR) T cell therapy effectively treats human cancer, but the loss of the antigen recognized by the CAR poses a major obstacle. We found that in vivo vaccine boosting of CAR T cells triggers the engagement of the endogenous immune system to circumvent antigen-negative tumor escape. Vaccine-boosted CAR T promoted dendritic cell (DC) recruitment to tumors, increased tumor antigen uptake by DCs, and elicited the priming of endogenous anti-tumor T cells. This process was accompanied by shifts in CAR T metabolism toward oxidative phosphorylation (OXPHOS) and was critically dependent on CAR-T-derived IFN-γ. Antigen spreading (AS) induced by vaccine-boosted CAR T enabled a proportion of complete responses even when the initial tumor was 50% CAR antigen negative, and heterogeneous tumor control was further enhanced by the genetic amplification of CAR T IFN-γ expression. Thus, CAR-T-cell-derived IFN-γ plays a critical role in promoting AS, and vaccine boosting provides a clinically translatable strategy to drive such responses against solid tumors.


Subject(s)
Cancer Vaccines , Neoplasms , Receptors, Chimeric Antigen , Humans , Neoplasms/therapy , T-Lymphocytes , Immunotherapy, Adoptive , Receptors, Antigen, T-Cell/metabolism
5.
Nat Biomed Eng ; 7(9): 1081-1096, 2023 09.
Article in English | MEDLINE | ID: mdl-37095318

ABSTRACT

In solid tumours, the abundance of macrophages is typically associated with a poor prognosis. However, macrophage clusters in tumour-cell nests have been associated with survival in some tumour types. Here, by using tumour organoids comprising macrophages and cancer cells opsonized via a monoclonal antibody, we show that highly ordered clusters of macrophages cooperatively phagocytose cancer cells to suppress tumour growth. In mice with poorly immunogenic tumours, the systemic delivery of macrophages with signal-regulatory protein alpha (SIRPα) genetically knocked out or else with blockade of the CD47-SIRPα macrophage checkpoint was combined with the monoclonal antibody and subsequently triggered the production of endogenous tumour-opsonizing immunoglobulin G, substantially increased the survival of the animals and helped confer durable protection from tumour re-challenge and metastasis. Maximizing phagocytic potency by increasing macrophage numbers, by tumour-cell opsonization and by disrupting the phagocytic checkpoint CD47-SIRPα may lead to durable anti-tumour responses in solid cancers.


Subject(s)
CD47 Antigen , Neoplasms , Mice , Animals , CD47 Antigen/metabolism , Receptors, Immunologic/metabolism , Phagocytosis , Macrophages , Antibodies, Monoclonal/metabolism
6.
Nat Mater ; 21(6): 710-720, 2022 06.
Article in English | MEDLINE | ID: mdl-35606429

ABSTRACT

Activation of the innate immune STimulator of INterferon Genes (STING) pathway potentiates antitumour immunity, but systemic delivery of STING agonists to tumours is challenging. We conjugated STING-activating cyclic dinucleotides (CDNs) to PEGylated lipids (CDN-PEG-lipids; PEG, polyethylene glycol) via a cleavable linker and incorporated them into lipid nanodiscs (LNDs), which are discoid nanoparticles formed by self-assembly. Compared to state-of-the-art liposomes, intravenously administered LNDs carrying CDN-PEG-lipid (LND-CDNs) exhibited more efficient penetration of tumours, exposing the majority of tumour cells to STING agonist. A single dose of LND-CDNs induced rejection of established tumours, coincident with immune memory against tumour rechallenge. Although CDNs were not directly tumoricidal, LND-CDN uptake by cancer cells correlated with robust T-cell activation by promoting CDN and tumour antigen co-localization in dendritic cells. LNDs thus appear promising as a vehicle for robust delivery of compounds throughout solid tumours, which can be exploited for enhanced immunotherapy.


Subject(s)
Nanoparticles , Neoplasms , Humans , Immunotherapy , Lipids , Membrane Proteins/metabolism , Membrane Proteins/pharmacology , Nanoparticles/therapeutic use , Neoplasms/drug therapy
7.
Cancer Immunol Res ; 9(11): 1245-1251, 2021 11.
Article in English | MEDLINE | ID: mdl-34544686

ABSTRACT

Recent success in the use of immunotherapy for a broad range of cancers has propelled the field of cancer immunology to the forefront of cancer research. As more and more young investigators join the community of cancer immunologists, the Arthur L. Irving Family Foundation Cancer Immunology Symposium provided a platform to bring this expanding and vibrant community together and support the development of the future leaders in the field. This commentary outlines the lessons that emerged from the inaugural symposium highlighting the areas of scientific and career development that are essential for professional growth in the field of cancer immunology and beyond. Leading scientists and clinicians in the field provided their experience on the topics of scientific trajectory, career trajectory, publishing, fundraising, leadership, mentoring, and collaboration. Herein, we provide a conceptual and practical framework for career development to the broader scientific community.


Subject(s)
Allergy and Immunology/education , Biomedical Research/methods , Neoplasms/epidemiology , Physicians/organization & administration , Humans , Leadership
8.
Blood ; 134(22): 1960-1972, 2019 11 28.
Article in English | MEDLINE | ID: mdl-31515251

ABSTRACT

Leukemia stem cells are a rare population with a primitive progenitor phenotype that can initiate, sustain, and recapitulate leukemia through a poorly understood mechanism of self-renewal. Here, we report that Krüppel-like factor 4 (KLF4) promotes disease progression in a murine model of chronic myeloid leukemia (CML)-like myeloproliferative neoplasia by repressing an inhibitory mechanism of preservation in leukemia stem/progenitor cells with leukemia-initiating capacity. Deletion of the Klf4 gene severely abrogated the maintenance of BCR-ABL1(p210)-induced CML by impairing survival and self-renewal in BCR-ABL1+ CD150+ lineage-negative Sca-1+ c-Kit+ leukemic cells. Mechanistically, KLF4 repressed the Dyrk2 gene in leukemic stem/progenitor cells; thus, loss of KLF4 resulted in elevated levels of dual-specificity tyrosine-(Y)-phosphorylation-regulated kinase 2 (DYRK2), which were associated with inhibition of survival and self-renewal via depletion of c-Myc protein and p53 activation. In addition to transcriptional regulation, stabilization of DYRK2 protein by inhibiting ubiquitin E3 ligase SIAH2 with vitamin K3 promoted apoptosis and abrogated self-renewal in murine and human CML stem/progenitor cells. Altogether, our results suggest that DYRK2 is a molecular checkpoint controlling p53- and c-Myc-mediated regulation of survival and self-renewal in CML cells with leukemic-initiating capacity that can be targeted with small molecules.


Subject(s)
Kruppel-Like Transcription Factors/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Neoplastic Stem Cells/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Signal Transduction , Animals , Cell Survival/drug effects , Cell Survival/genetics , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Gene Deletion , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mice , Mice, Knockout , Neoplastic Stem Cells/pathology , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Vitamin K 3/pharmacology , Dyrk Kinases
9.
Science ; 365(6449): 162-168, 2019 07 12.
Article in English | MEDLINE | ID: mdl-31296767

ABSTRACT

Chimeric antigen receptor-T cell (CAR-T) therapy has been effective in the treatment of hematologic malignancies, but it has shown limited efficacy against solid tumors. Here we demonstrate an approach to enhancing CAR-T function in solid tumors by directly vaccine-boosting donor cells through their chimeric receptor in vivo. We designed amphiphile CAR-T ligands (amph-ligands) that, upon injection, trafficked to lymph nodes and decorated the surfaces of antigen-presenting cells, thereby priming CAR-Ts in the native lymph node microenvironment. Amph-ligand boosting triggered massive CAR-T expansion, increased donor cell polyfunctionality, and enhanced antitumor efficacy in multiple immunocompetent mouse tumor models. We demonstrate two approaches to generalizing this strategy to any chimeric antigen receptor, enabling this simple non-human leukocyte antigen-restricted approach to enhanced CAR-T functionality to be applied to existing CAR-T designs.


Subject(s)
Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Immunotherapy, Adoptive , Neoplasms/therapy , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , Animals , Disease Models, Animal , HEK293 Cells , Humans , Immunization, Secondary , K562 Cells , Mice
10.
Sci Transl Med ; 11(498)2019 06 26.
Article in English | MEDLINE | ID: mdl-31243150

ABSTRACT

The clinical application of cytokine therapies for cancer treatment remains limited due to severe adverse reactions and insufficient therapeutic effects. Although cytokine localization by intratumoral administration could address both issues, the rapid escape of soluble cytokines from the tumor invariably subverts this effort. We find that intratumoral administration of a cytokine fused to the collagen-binding protein lumican prolongs local retention and markedly reduces systemic exposure. Combining local administration of lumican-cytokine fusions with systemic immunotherapies (tumor-targeting antibody, checkpoint blockade, cancer vaccine, or T cell therapy) improves efficacy without exacerbating toxicity in syngeneic tumor models and the BrafV600E /Ptenfl/fl genetically engineered melanoma model. Curative abscopal effects on noncytokine-injected tumors were also observed as a result of a protective and systemic CD8+ T cell response primed by local therapy. Cytokine collagen-anchoring constitutes a facile, tumor-agnostic strategy to safely potentiate otherwise marginally effective systemic immunotherapies.


Subject(s)
Cytokines/administration & dosage , Immunotherapy , Neoplasms/immunology , Neoplasms/therapy , Animals , Antibodies, Neoplasm/immunology , Cell Line, Tumor , Collagen , Disease Models, Animal , Interleukin-12/therapeutic use , Interleukin-2/therapeutic use , Lumican/metabolism , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice, Inbred C57BL , Neoadjuvant Therapy , PTEN Phosphohydrolase/metabolism , Programmed Cell Death 1 Receptor/metabolism , Proto-Oncogene Proteins B-raf/metabolism , Serum Albumin/metabolism , T-Lymphocytes/immunology , Weight Loss
11.
Proc Natl Acad Sci U S A ; 116(21): 10482-10487, 2019 05 21.
Article in English | MEDLINE | ID: mdl-31068472

ABSTRACT

A major obstacle to curing chronic myeloid leukemia (CML) is the intrinsic resistance of CML stem cells (CMLSCs) to the drug imatinib mesylate (IM). Prosurvival genes that are preferentially expressed in CMLSCs compared with normal hematopoietic stem cells (HSCs) represent potential therapeutic targets for selectively eradicating CMLSCs. However, the discovery of such preferentially expressed genes has been hampered by the inability to completely separate CMLSCs from HSCs, which display a very similar set of surface markers. To overcome this challenge, and to minimize confounding effects of individual differences in gene expression profiles, we performed single-cell RNA-seq on CMLSCs and HSCs that were isolated from the same patient and distinguished based on the presence or absence of BCR-ABL. Among genes preferentially expressed in CMLSCs is PIM2, which encodes a prosurvival serine-threonine kinase that phosphorylates and inhibits the proapoptotic protein BAD. We show that IM resistance of CMLSCs is due, at least in part, to maintenance of BAD phosphorylation by PIM2. We find that in CMLSCs, PIM2 expression is promoted by both a BCR-ABL-dependent (IM-sensitive) STAT5-mediated pathway and a BCR-ABL-independent (IM-resistant) STAT4-mediated pathway. Combined treatment with IM and a PIM inhibitor synergistically increases apoptosis of CMLSCs, suppresses colony formation, and significantly prolongs survival in a mouse CML model, with a negligible effect on HSCs. Our results reveal a therapeutically targetable mechanism of IM resistance in CMLSCs. The experimental approach that we describe can be generally applied to other malignancies that harbor oncogenic fusion proteins or other characteristic genetic markers.


Subject(s)
Biphenyl Compounds/therapeutic use , Drug Resistance, Neoplasm , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Neoplastic Stem Cells/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Thiazolidines/therapeutic use , Animals , Drug Screening Assays, Antitumor , Fusion Proteins, bcr-abl/metabolism , Humans , Imatinib Mesylate , Leukemia, Experimental/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Mice , Molecular Targeted Therapy , Phosphorylation , Protein Kinase Inhibitors , STAT Transcription Factors/metabolism , bcl-Associated Death Protein/metabolism
12.
Proc Natl Acad Sci U S A ; 114(44): 11751-11756, 2017 10 31.
Article in English | MEDLINE | ID: mdl-29078326

ABSTRACT

Developing tools to accurately predict the clinical prevalence of drug-resistant mutations is a key step toward generating more effective therapeutics. Here we describe a high-throughput CRISPR-Cas9-based saturated mutagenesis approach to generate comprehensive libraries of point mutations at a defined genomic location and systematically study their effect on cell growth. As proof of concept, we mutagenized a selected region within the leukemic oncogene BCR-ABL1 Using bulk competitions with a deep-sequencing readout, we analyzed hundreds of mutations under multiple drug conditions and found that the effects of mutations on growth in the presence or absence of drug were critical for predicting clinically relevant resistant mutations, many of which were cancer adaptive in the absence of drug pressure. Using this approach, we identified all clinically isolated BCR-ABL1 mutations and achieved a prediction score that correlated highly with their clinical prevalence. The strategy described here can be broadly applied to a variety of oncogenes to predict patient mutations and evaluate resistance susceptibility in the development of new therapeutics.


Subject(s)
CRISPR-Cas Systems/genetics , Drug Resistance, Neoplasm/genetics , Mutagenesis/genetics , Animals , Antineoplastic Agents/pharmacology , CRISPR-Cas Systems/drug effects , Cell Line, Tumor , Clustered Regularly Interspaced Short Palindromic Repeats/drug effects , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Drug Resistance, Neoplasm/drug effects , Fusion Proteins, bcr-abl/genetics , Leukemia/drug therapy , Leukemia/genetics , Mice , Mutagenesis/drug effects , Oncogenes/genetics , Point Mutation/drug effects , Point Mutation/genetics
13.
Methods Mol Biol ; 1465: 159-73, 2016.
Article in English | MEDLINE | ID: mdl-27581147

ABSTRACT

Genome-wide RNA interference (RNAi) screening in mammalian cells has proven to be a powerful tool for identifying new genes and molecular pathways relevant to many cellular processes and diseases. For example, screening for genes that, when inactivated, lead to resistance to cancer therapeutic drugs can reveal new mechanisms for how resistance develops and identify potential targetable strategies to overcome drug resistance. Here, we describe a detailed procedure for performing a high-throughput RNAi screen using a genome-wide human short hairpin RNA (shRNA) library for identifying tyrosine kinase inhibitor (TKI)-resistance genes in a human CML cell line model.


Subject(s)
Drug Resistance, Neoplasm , High-Throughput Screening Assays/methods , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , RNA, Small Interfering/genetics , Fusion Proteins, bcr-abl/genetics , Gene Library , Humans , Imatinib Mesylate/pharmacology , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Protein Kinase Inhibitors/pharmacology , Sequence Analysis, DNA
14.
Sci Transl Med ; 6(252): 252ra121, 2014 Sep 03.
Article in English | MEDLINE | ID: mdl-25186176

ABSTRACT

Resistance to the BCR-ABL inhibitor imatinib mesylate (IM) poses a major problem for the treatment of chronic myeloid leukemia (CML). IM resistance often results from a secondary mutation in BCR-ABL that interferes with drug binding. However, in many instances, there is no mutation in BCR-ABL, and the basis of such BCR-ABL-independent IM resistance remains to be elucidated. To gain insight into BCR-ABL-independent IM resistance mechanisms, we performed a large-scale RNA interference screen and identified IM-sensitizing genes (IMSGs) whose knockdown renders BCR-ABL(+) cells IM-resistant. In these IMSG knockdown cells, RAF/mitogen-activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) signaling is sustained after IM treatment because of up-regulation of PRKCH, which encodes the protein kinase C (PKC) family member PKCη, an activator of CRAF. PRKCH is also up-regulated in samples from CML patients with BCR-ABL-independent IM resistance. Combined treatment with IM and trametinib, a U.S. Food and Drug Administration-approved MEK inhibitor, synergistically kills BCR-ABL(+) IMSG knockdown cells and prolongs survival in mouse models of BCR-ABL-independent IM-resistant CML. Finally, we showed that CML stem cells contain high levels of PRKCH, and this contributes to their intrinsic IM resistance. Combined treatment with IM and trametinib synergistically kills CML stem cells with negligible effect on normal hematopoietic stem cells. Collectively, our results identify a therapeutically targetable mechanism of BCR-ABL-independent IM resistance in CML and CML stem cells.


Subject(s)
Benzamides/therapeutic use , Drug Resistance, Neoplasm/drug effects , Fusion Proteins, bcr-abl/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Molecular Targeted Therapy , Piperazines/therapeutic use , Pyrimidines/therapeutic use , Animals , Benzamides/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Disease Progression , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Knockdown Techniques , Genes, Neoplasm , Humans , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Mice , Mitogen-Activated Protein Kinase Kinases/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Phosphorylation/drug effects , Piperazines/pharmacology , Protein Kinase C/metabolism , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , RNA, Small Interfering/metabolism , Survival Analysis , Up-Regulation/drug effects , raf Kinases/metabolism
15.
Pigment Cell Melanoma Res ; 27(1): 124-33, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24112705

ABSTRACT

Resistance to the BRAF inhibitor vemurafenib poses a significant problem for the treatment of BRAFV600E-positive melanomas. It is therefore critical to prospectively identify all vemurafenib resistance mechanisms prior to their emergence in the clinic. The vemurafenib resistance mechanisms described to date do not result from secondary mutations within BRAFV600E. To search for possible mutations within BRAFV600E that can confer drug resistance, we developed a systematic experimental approach involving targeted saturation mutagenesis, selection of drug-resistant variants, and deep sequencing. We identified a single nucleotide substitution (T1514A, encoding L505H) that greatly increased drug resistance in cultured cells and mouse xenografts. The kinase activity of BRAFV600E/L505H was higher than that of BRAFV600E, resulting in cross-resistance to a MEK inhibitor. However, BRAFV600E/L505H was less resistant to several other BRAF inhibitors whose binding sites were further from L505 than that of PLX4720. Our results identify a novel vemurafenib-resistant mutant and provide insights into the treatment for melanomas bearing this mutation.


Subject(s)
Drug Resistance, Neoplasm/genetics , Indoles/pharmacokinetics , Melanoma/genetics , Melanoma/metabolism , Mutation, Missense , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Sulfonamides/pharmacokinetics , Amino Acid Substitution , Animals , Cell Line, Tumor , Heterografts , Humans , Melanoma/drug therapy , Melanoma/pathology , Mice , Neoplasm Transplantation , Protein Structure, Tertiary , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Vemurafenib
16.
Proc Natl Acad Sci U S A ; 110(6): 2312-7, 2013 Feb 05.
Article in English | MEDLINE | ID: mdl-23345428

ABSTRACT

Hematopoietic stem cells (HSCs) are the source of all blood lineages, and HSCs must balance quiescence, self-renewal, and differentiation to meet lifelong needs for blood cell development. Transformation of HSCs by the breakpoint cluster region-ABL tyrosine kinase (BCR-ABL) oncogene causes chronic myelogenous leukemia (CML). The E-twenty six (ets) transcription factor GA binding protein (GABP) is a tetrameric transcription factor complex that contains GABPα and GABPß proteins. Deletion in bone marrow of Gabpa, the gene that encodes the DNA-binding component, caused cell cycle arrest in HSCs and profound loss of hematopoietic progenitor cells. Loss of Gabpα prevented development of CML, although mice continued to generate BCR-ABL-expressing Gabpα-null cells for months that were serially transplantable and contributed to all lineages in secondary recipients. A bioinformatic screen identified the serine-threonine kinase protein kinase D2 (PRKD2) as a potential effector of GABP in HSCs. Prkd2 expression was markedly reduced in Gabpα-null HSCs and progenitor cells. Reduced expression of PRKD2 or pharmacologic inhibition decreased cell cycling, and PRKD2 rescued growth of Gabpα-null BCR-ABL-expressing cells. Thus, GABP is required for HSC cell cycle entry and CML development through its control of PRKD2. This offers a potential therapeutic target in leukemia.


Subject(s)
GA-Binding Protein Transcription Factor/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/etiology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Protein Kinases/metabolism , Animals , Antineoplastic Agents/pharmacology , Benzamides , Cell Cycle , GA-Binding Protein Transcription Factor/deficiency , GA-Binding Protein Transcription Factor/genetics , Gene Expression , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Mice , Mice, Transgenic , Piperazines/pharmacology , Protein Kinase D2 , Protein Kinase Inhibitors/pharmacology , Protein Kinases/genetics , Pyrimidines/pharmacology
17.
Gene ; 503(1): 147-51, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22575730

ABSTRACT

Lycopene Beta-cyclase (LCY-B) is thought to play a critical role in Beta-carotene synthesis in fruit. A full-length cDNA clone encoding Lycopene Beta-cyclase was isolated from muskmelon (Cucumis melo L.) by RT-PCR and RACE. The clone, designated CmLcyb1, contains 1871 nucleotides, with an open reading frame of 1512 nucleotides. The deduced 504-amino-acid sequence showed high identities with other plant Lycopene Beta-cyclases. Real time quantitative RT-PCR analysis indicated that CmLcyb1 was expressed in all tissues and organs of muskmelon inbred M01-3 with white mesocarp and, 'Homoka', an orange mesocarp cultivar. The expression levels of CmLcyb1 in roots, stems, leaves and flowers in the two genotypes differed little. The expression level was highest in mature fruit of 'Homoka' and was much higher than that in mature fruit of M01-3. Moreover, the mRNA level of CmLcyb1 was very low in fruits before fruit-size fixation and increased dramatically in the size-fixed fruits of these two genotypes. The mRNA levels of CmLcyb1 during fruit development of 'Homoka' were all higher than those of M01-3. Interestingly, Beta-carotene content showed almost the same change trend as mRNA levels during fruit development in these two genotypes, suggesting that Beta-carotene accumulation may be linked to the CmLcyb1 transcript level in muskmelon fruit.


Subject(s)
Cucumis melo/genetics , DNA, Complementary/genetics , Intramolecular Lyases/genetics , Sequence Analysis, DNA/methods , Amino Acid Sequence , Base Sequence , Cucumis melo/enzymology , Intramolecular Lyases/biosynthesis , Molecular Sequence Data , Sequence Homology, Amino Acid
18.
Mol Biol Rep ; 39(4): 4283-90, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21786156

ABSTRACT

Nitrate reductase is a key enzyme in the overall process of nitrate assimilation by plants. A full-length cDNA clone encoding nitrate reductase (NR; EC 1.6.6.1) was isolated from cucumber (Cucumis sativus L.) by RT-PCR and RACE techniques. The NR of cucumber (CsNR), a full-length cDNA sequence of 3032 bp contains an open reading frame of 2748 bp encoding 915 amino acids. The deduced 915 amino acid sequence showed high identities with NR from other plants. Quantitative real-time PCR analysis indicated that CsNR expression was different in root, stem, leaf, flower and mature fruit tissues. CsNR transcript level and nitrate reductase activity (NRA) was down-regulated and the change in NO(3) (-) concentration showed a negative trend with NRA in leaves when subjected to the 182 mM NO(3) (-) treatment. However, the CsNR transcript level was up-regulated in roots by 182 mM NO(3) (-) treatment. Furthermore, NRA in roots lagged behind CsNR expression and there was no obvious lag of NRA in leaves. This study found that in roots, there was no obvious relationship between NRA and NO(3) (-) content. These results indicated that NRA was not only controlled by the level of CsNR mRNA and there was an obvious negative relationship between NO(3) (-) content and NRA in leaves but not in roots.


Subject(s)
Cucumis sativus/enzymology , Cucumis sativus/genetics , Genes, Plant/genetics , Nitrate Reductase/genetics , Nitrates/pharmacology , Stress, Physiological/genetics , Cloning, Molecular , Cucumis sativus/drug effects , DNA, Complementary/genetics , Gene Expression Profiling , Gene Expression Regulation, Plant/drug effects , Molecular Sequence Data , Nitrate Reductase/metabolism , Organ Specificity/drug effects , Organ Specificity/genetics , Plant Leaves/drug effects , Plant Leaves/enzymology , Plant Leaves/genetics , Plant Roots/drug effects , Plant Roots/enzymology , Plant Roots/genetics , Real-Time Polymerase Chain Reaction , Stress, Physiological/drug effects , Time Factors
19.
Blood ; 118(10): 2840-8, 2011 Sep 08.
Article in English | MEDLINE | ID: mdl-21715304

ABSTRACT

The oncoprotein BCR-ABL transforms myeloid progenitor cells and is responsible for the development of chronic myeloid leukemia (CML). In transformed cells, BCR-ABL suppresses apoptosis as well as autophagy, a catabolic process in which cellular components are degraded by the lysosomal machinery. The mechanism by which BCR-ABL suppresses autophagy is not known. Here we report that in both mouse and human BCR-ABL-transformed cells, activating transcription factor 5 (ATF5), a prosurvival factor, suppresses autophagy but does not affect apoptosis. We find that BCR-ABL, through PI3K/AKT/FOXO4 signaling, transcriptionally up-regulates ATF5 expression and that ATF5, in turn, stimulates transcription of mammalian target of rapamycin (mTOR; also called mechanistic target of rapamycin), a well-established master negative-regulator of autophagy. Previous studies have shown that the BCR-ABL inhibitor imatinib mesylate induces both apoptosis and autophagy, and that the resultant autophagy modulates the efficiency by which imatinib kills BCR-ABL-transformed cells. We demonstrate that imatinib-induced autophagy is because of inhibition of the BCR-ABL/PI3K/AKT/FOXO4/ATF5/mTOR pathway that we have identified in this study.


Subject(s)
Activating Transcription Factors/metabolism , Autophagy , Fusion Proteins, bcr-abl/metabolism , Gene Expression Regulation, Neoplastic , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , TOR Serine-Threonine Kinases/genetics , Activating Transcription Factors/antagonists & inhibitors , Activating Transcription Factors/genetics , Animals , Antineoplastic Agents/pharmacology , Benzamides , Blotting, Western , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Chromatin Immunoprecipitation , Humans , Imatinib Mesylate , Immunosuppressive Agents/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Luciferases/metabolism , Mice , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Piperazines/pharmacology , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Pyrimidines/pharmacology , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/metabolism , Transcription, Genetic
20.
Biochem Biophys Res Commun ; 393(3): 365-70, 2010 Mar 12.
Article in English | MEDLINE | ID: mdl-20138160

ABSTRACT

To unravel the roles of sucrose phosphate synthase (SPS) in muskmelon (Cucumis melo L.), we reduced its activity in transgenic muskmelon plants by an antisense approach. For this purpose, an 830 bp cDNA fragment of muskmelon sucrose phosphate synthase was expressed in antisense orientation behind the 35S promoter of the cauliflower mosaic virus. The phenotype of the antisense plants clearly differed from that of control plants. The transgenic plant leaves were markedly smaller, and the plant height and stem diameter were obviously shorter and thinner. Transmission electron microscope observation revealed that the membrane degradation of chloroplast happened in transgenic leaves and the numbers of grana and grana lamella in the chloroplast were significantly less, suggesting that the slow growth and weaker phenotype of transgenic plants may be due to the damage of the chloroplast ultrastructure, which in turn results in the decrease of the net photosynthetic rate. The sucrose concentration and levels of sucrose phosphate synthase decreased in transgenic mature fruit, and the fruit size was smaller than the control fruit. Together, our results suggest that sucrose phosphate synthase may play an important role in regulating the muskmelon plant growth and fruit development.


Subject(s)
Cucumis melo/growth & development , Fruit/growth & development , Glucosyltransferases/physiology , Plants, Genetically Modified/growth & development , Cucumis melo/enzymology , Cucumis melo/genetics , DNA, Antisense/genetics , Fruit/enzymology , Fruit/genetics , Glucosyltransferases/antagonists & inhibitors , Glucosyltransferases/genetics , Plants, Genetically Modified/enzymology , Plants, Genetically Modified/genetics , Sucrose/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...