18 results on '"Glorymar Ibáñez"'
Search Results
2. Translational Strategies for Repotrectinib in Neuroblastoma
- Author
-
Nestor Rosales, Andoyo Ndengu, Filemon S. Dela Cruz, Daoqi You, Stephen S. Roberts, Matthew Long, Armaan Siddiquee, Paul Calder, Tara O'Donohue, Andrew L. Kung, Audrey Mauguen, Glorymar Ibáñez, and Diego F. Coutinho
- Subjects
Cancer Research ,Chemotherapy ,Macrocyclic Compounds ,business.industry ,medicine.medical_treatment ,medicine.disease ,Article ,stat ,Mice ,Neuroblastoma ,Oncology ,Cell culture ,In vivo ,Trk receptor ,Cancer research ,Animals ,Humans ,Pyrazoles ,Medicine ,Cytotoxic T cell ,business ,Proto-oncogene tyrosine-protein kinase Src - Abstract
Limited clinical data are available regarding the utility of multikinase inhibition in neuroblastoma. Repotrectinib (TPX-0005) is a multikinase inhibitor that targets ALK, TRK, JAK2/STAT, and Src/FAK, which have all been implicated in the pathogenesis of neuroblastoma. We evaluated the preclinical activity of repotrectinib monotherapy and in combination with chemotherapy as a potential therapeutic approach for relapsed/refractory neuroblastoma. In vitro sensitivity to repotrectinib, ensartinib, and cytotoxic chemotherapy was evaluated in neuroblastoma cell lines. In vivo antitumor effect of repotrectinib monotherapy, and in combination with chemotherapy, was evaluated using a genotypically diverse cohort of patient-derived xenograft (PDX) models of neuroblastoma. Repotrectinib had comparable cytotoxic activity across cell lines irrespective of ALK mutational status. Combination with chemotherapy demonstrated increased antiproliferative activity across several cell lines. Repotrectinib monotherapy had notable antitumor activity and prolonged event-free survival compared with vehicle and ensartinib in PDX models (P < 0.05). Repotrectinib plus chemotherapy was superior to chemotherapy alone in ALK-mutant and ALK wild-type PDX models. These results demonstrate that repotrectinib has antitumor activity in genotypically diverse neuroblastoma models, and that combination of a multikinase inhibitor with chemotherapy may be a promising treatment paradigm for translation to the clinic.
- Published
- 2021
- Full Text
- View/download PDF
3. Validation of a non-oncogene encoded vulnerability to exportin 1 inhibition in pediatric renal tumors
- Author
-
Diego F. Coutinho, Prabhjot S. Mundi, Lianna J. Marks, Chelsey Burke, Michael V. Ortiz, Daniel Diolaiti, Lauren Bird, Kelly L. Vallance, Glorymar Ibáñez, Daoqi You, Matthew Long, Nestor Rosales, Adina Grunn, Andoyo Ndengu, Armaan Siddiquee, Ervin S. Gaviria, Allison R. Rainey, Ladan Fazlollahi, Hajime Hosoi, Andrea Califano, Andrew L. Kung, and Filemon S. Dela Cruz
- Subjects
Child, Preschool ,Cell Line, Tumor ,Humans ,General Medicine ,Child ,Xenograft Model Antitumor Assays ,Kidney Neoplasms - Abstract
Malignant rhabdoid tumors (MRTs) and Wilms' tumors (WTs) are rare and aggressive renal tumors of infants and young children comprising ∼5% of all pediatric cancers. MRTs are among the most genomically stable cancers, and although WTs are genomically heterogeneous, both generally lack therapeutically targetable genetic mutations.Comparative protein activity analysis of MRTs (n = 68) and WTs (n = 132) across TCGA and TARGET cohorts, using metaVIPER, revealed elevated exportin 1 (XPO1) inferred activity. In vitro studies were performed on a panel of MRT and WT cell lines to evaluate effects on proliferation and cell-cycle progression following treatment with the selective XPO1 inhibitor selinexor. In vivo anti-tumor activity was assessed in patient-derived xenograft (PDX) models of MRTs and WTs.metaVIPER analysis identified markedly aberrant activation of XPO1 in MRTs and WTs compared with other tumor types. All MRT and most WT cell lines demonstrated baseline, aberrant XPO1 activity with in vitro sensitivity to selinexor via cell-cycle arrest and induction of apoptosis. In vivo, XPO1 inhibitors significantly abrogated tumor growth in PDX models, inducing effective disease control with sustained treatment. Corroborating human relevance, we present a case report of a child with multiply relapsed WTs with prolonged disease control on selinexor.We report on a novel systems-biology-based comparative framework to identify non-genetically encoded vulnerabilities in genomically quiescent pediatric cancers. These results have provided preclinical rationale for investigation of XPO1 inhibitors in an upcoming investigator-initiated clinical trial of selinexor in children with MRTs and WTs and offer opportunities for exploration of inferred XPO1 activity as a potential predictive biomarker for response.This work was funded by CureSearch for Children's Cancer, Alan B. Slifka Foundation, NIH (U01 CA217858, S10 OD012351, and S10 OD021764), Michael's Miracle Cure, Hyundai Hope on Wheels, Cannonball Kids Cancer, Conquer Cancer the ASCO Foundation, Cycle for Survival, Paulie Strong Foundation, and the Grayson Fund.
- Published
- 2022
- Full Text
- View/download PDF
4. Exploiting a PAX3-FOXO1-induced synthetic lethal ATR dependency for rhabdomyosarcoma therapy
- Author
-
Yi Bei, Christian Furth, Koshi Imami, Jana Rolff, Angelika Eggert, Johannes H. Schulte, Jennifer von Stebut, Monika Scheer, Konstantin Helmsauer, Matthias Selbach, Victor Bardinet, Michael V. Ortiz, Heathcliff Dorado Garcia, Andrej Lissat, Dennis Gürgen, Georg Seifert, Patrick Hundsoerfer, Anton G. Henssen, Kerstin Haase, Joachim G. Schulz, Antje Margret Wengner, Fabian Pusch, Marieluise Kirchner, Philipp Mertins, Glorymar Ibáñez, Birgit Lala, Natalie Timme, Ian C. MacArthur, Rocío Chamorro González, and Celine Y. Chen
- Subjects
medicine.anatomical_structure ,Oncogene ,Chemistry ,In vivo ,Poly ADP ribose polymerase ,Cell ,Cancer research ,medicine ,Alveolar rhabdomyosarcoma ,Progenitor cell ,Rhabdomyosarcoma ,medicine.disease ,In vitro - Abstract
Pathognomonic PAX3-FOXO1 fusion oncogene expression is associated with poor outcome in rhabdomyosarcoma. Combining genome-wide CRISPR screening with cell- based functional genetic approaches, we here provide evidence that PAX3-FOXO1 induces replication stress, resulting in a synthetic lethal dependency to ATR-mediated DNA damage-response signaling in rhabdomyosarcoma. Expression of PAX3-FOXO1 in muscle progenitor cells was not only sufficient to induce hypersensitivity to ATR inhibition, but PAX3-FOXO1-expressing rhabdomyosarcoma cells also exhibited increased sensitivity to structurally diverse inhibitors of ATR, a dependency that could be validated genetically. Mechanistically, ATR inhibition led to replication stress exacerbation, decreased BRCA1 phosphorylation and reduced homologous recombination-mediated DNA repair pathway activity. Consequently, ATR inhibitor treatment increased sensitivity of rhabdomyosarcoma cells to PARP inhibitionin vitro, and combined ATR and PARP inhibition induced regression of primary patient-derived alveolar rhabdomyosarcoma xenograftsin vivo. Moreover, a genome-wide CRISPR activation screen (CRISPRa) identifiedFOSgene family members as inducers of resistance against ATR inhibitors. Mechanistically,FOSgene family members reduced replication stress in rhabdomyosarcoma cells. Lastly, compassionate use of ATR inhibitors in two pediatric patients suffering from relapsed PAX3-FOXO1-expressing alveolar rhabdomyosarcoma showed signs of tolerability, paving the way to clinically exploit this novel synthetic lethal dependency in rhabdomyosarcoma.
- Published
- 2020
- Full Text
- View/download PDF
5. TPX-0005 (Repotrectinib), a next-generation ALK/ROS1/NTRK1–3 inhibitor, has potent antiproliferative and anti-tumor activity as monotherapy and in combination with chemotherapy in neuroblastoma cell lines and pediatric patient derived xenograft models
- Author
-
Audrey Mauguen, A. Ndengu, Andrew L. Kung, Nestor Rosales, Stephen S. Roberts, F. Dela Cruz, Armaan Siddiquee, Paul Calder, Glorymar Ibáñez, and Tara O'Donohue
- Subjects
Antitumor activity ,Neuroblastoma cell ,Cancer Research ,Chemotherapy ,Pediatric patient ,Oncology ,business.industry ,medicine.medical_treatment ,Cancer research ,ROS1 ,Medicine ,business - Published
- 2020
- Full Text
- View/download PDF
6. A chemical probe of CARM1 alters epigenetic plasticity against breast cancer cell invasion
- Author
-
Yuling Chen, Hong Zeng, Xiao-Chuan Cai, Magda Szewczyk, Ludmila Dombrovsky, Linas Mazutis, Ming Jiang, Aiping Dong, Taraneh Hajian, Glorymar Ibáñez, Hong Wu, Peter Brown, Fengling Li, Cheryl H. Arrowsmith, Lei Shi, Masoud Vedadi, Victor Vivcharuk, Jenny Xiang, Haiteng Deng, Nawei Zhang, Carlo C. dela Seña, Tuo Zhang, Li-Xuan Qin, Jinrong Min, Zhenyu Zhang, Dalia Barsyte, Wei Xu, Junyi Wang, Jonghan P Lee, Ke Wang, Weihong Zheng, Shi Chen, Eui-jun Kim, Minkui Luo, and Xiang Niu
- Subjects
0301 basic medicine ,genetic structures ,CARM1 ,QH301-705.5 ,Science ,PRMT ,Cell ,Chemical biology ,Cancer metastasis ,mechanism ,Chemical probe ,Plasticity ,behavioral disciplines and activities ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,0302 clinical medicine ,Biochemistry and Chemical Biology ,Transcription (biology) ,medicine ,Epigenetics ,Biology (General) ,030304 developmental biology ,0303 health sciences ,General Immunology and Microbiology ,Chemistry ,General Neuroscience ,General Medicine ,Methylation ,single cell ,3. Good health ,Cell biology ,inhibitor ,030104 developmental biology ,Membrane ,medicine.anatomical_structure ,nervous system ,030220 oncology & carcinogenesis ,Medicine ,Breast cancer cells ,methylation ,human activities ,epigenetic ,psychological phenomena and processes ,Research Article ,Human - Abstract
CARM1 is a cancer-relevant protein arginine methyltransferase that regulates many aspects of transcription. Its pharmacological inhibition is a promising anti-cancer strategy. Here SKI-73 (6a in this work) is presented as a CARM1 chemical probe with pro-drug properties. SKI-73 (6a) can rapidly penetrate cell membranes and then be processed into active inhibitors, which are retained intracellularly with 10-fold enrichment for several days. These compounds were characterized for their potency, selectivity, modes of action, and on-target engagement. SKI-73 (6a) recapitulates the effect of CARM1 knockout against breast cancer cell invasion. Single-cell RNA-seq analysis revealed that the SKI-73(6a)-associated reduction of invasiveness acts by altering epigenetic plasticity and suppressing the invasion-prone subpopulation. Interestingly, SKI-73 (6a) and CARM1 knockout alter the epigenetic plasticity with remarkable difference, suggesting distinct modes of action for small-molecule and genetic perturbations. We therefore discovered a CARM1-addiction mechanism of cancer metastasis and developed a chemical probe to target this process., eLife digest Drugs that are small molecules have the potential to block the individual proteins that drive the spread of cancer, but their design is a challenge. This is because they need to get inside the cell and find their target without binding to other proteins on the way. However, small molecule drugs often have an electric charge, which makes it hard for them to cross the cell membrane. Additionally, most proteins are not completely unique, making it harder for the drugs to find the correct target. CARM1 is a protein that plays a role in the spread of breast cancer cells, and scientists are currently looking for a small molecule that will inhibit its action. The group of enzymes that CARM1 belongs to act by taking a small chemical group, called a methyl group, from a molecule called SAM, and transferring it to proteins that switch genes on and off. In the case of CARM1, this changes cell behavior by turning on genes involved in cell movement. Genetically modifying cells so they will not produce any CARM1 stops the spread of breast cancer cells, but developing a drug with the same effects has proved difficult. Existing drugs that can inhibit CARM1 in a test tube struggle to get inside cells and to distinguish between CARM1 and its related enzymes. Now, Cai et al. have modified and tested a CARM1 inhibitor to address these problems, and find out how these small molecules work. At its core, the inhibitor has a structure very similar to a SAM molecule, so it can fit into the SAM binding pocket of CARM1 and its related enzymes. To stop the inhibitor from binding to other proteins, Cai et al. made small changes to its structure until it only interacted with CARM1.Then, to get the inhibitor inside breast cancer cells, Cai et al. cloaked its charged area with a chemical shield, allowing it to cross the cell membrane. Inside the cell, the chemical shield broke away, allowing the inhibitor to attach to CARM1. Analysis of cells showed that this inhibition only affected the cancer cells most likely to spread. Blocking CARM1 switched off genes involved in cell movement and stopped cancer cells from travelling through 3D gels. This work is a step towards making a drug that can block CARM1 in cancer cells, but there is still further work to be done. The next stages will be to test whether the new inhibitor works in other types of cancer cells, in living animals, and in human patient samples.
- Published
- 2019
- Full Text
- View/download PDF
7. Author response: A chemical probe of CARM1 alters epigenetic plasticity against breast cancer cell invasion
- Author
-
Xiao-Chuan Cai, Tuo Zhang, Eui-jun Kim, Ming Jiang, Ke Wang, Junyi Wang, Shi Chen, Nawei Zhang, Hong Wu, Fengling Li, Carlo C dela Seña, Hong Zeng, Victor Vivcharuk, Xiang Niu, Weihong Zheng, Jonghan P Lee, Yuling Chen, Dalia Barsyte, Magda Szewczyk, Taraneh Hajian, Glorymar Ibáñez, Aiping Dong, Ludmila Dombrovski, Zhenyu Zhang, Haiteng Deng, Jinrong Min, Cheryl H Arrowsmith, Linas Mazutis, Lei Shi, Masoud Vedadi, Peter J Brown, Jenny Xiang, Li-Xuan Qin, Wei Xu, and Minkui Luo
- Published
- 2019
- Full Text
- View/download PDF
8. Evaluation of Compound Optical Interference in High-Content Screening
- Author
-
Hakim Djaballah, Glorymar Ibáñez, Christophe Antczak, Bhavneet Bhinder, Constantin Radu, Paul Calder, and David Shum
- Subjects
0301 basic medicine ,Green Fluorescent Proteins ,Biosensing Techniques ,Interference (wave propagation) ,01 natural sciences ,Biochemistry ,Fluorescence ,Analytical Chemistry ,03 medical and health sciences ,Cell Line, Tumor ,Humans ,Microscopy ,Chemistry ,0104 chemical sciences ,Chemical screening ,High-Throughput Screening Assays ,010404 medicinal & biomolecular chemistry ,MicroRNAs ,030104 developmental biology ,Pharmaceutical Preparations ,Cheminformatics ,High-content screening ,Molecular Medicine ,Biological Assay ,MiRNA biogenesis ,Biosensor ,Biogenesis ,Biotechnology ,HeLa Cells - Abstract
Compound optical interference remains an inherent problem in chemical screening and has been well documented for biochemical assays and less so for automated microscopy-based assays. It has also been the assumption that the latter should not suffer from such interference because of the washing steps involved in the process, thus eliminating the residual nonspecific compound effects. Instead, these compounds may have no relevance to the actual target, and as such, compound optical interference contributes to a number of false-positives, resulting in a high attrition rate during subsequent follow-up studies. In this report, we analyze the outcome of a high-content screen using enhanced green fluorescent protein as a reporter in a gain-of-function cell-based assay in search of modulators of the micro RNA (miRNA) biogenesis pathway. Using a previously validated image-based biosensor, we screened a diverse library collection of ~315,000 compounds covering natural and synthetic derivatives in which 1130 positives were identified to enhance green fluorescence expression. Lateral confirmation and dose-response studies revealed that all of these compounds were the result of optical interference and not specific inhibition of miRNA biogenesis. Here, we highlight the chemical classes that are susceptible to compound optical interference and discuss their implications in automated microscopy-based assays.
- Published
- 2017
9. Sinefungin Derivatives as Inhibitors and Structure Probes of Protein Lysine Methyltransferase SETD2
- Author
-
Fengling Li, Aiping Dong, Masoud Vedadi, Wenyu Yu, Weihong Zheng, Alena Siarheyeva, Jinrong Min, Gil Blum, Abdellah Allali-Hassani, Glorymar Ibáñez, Peter Brown, Maria F. Amaya, Hong Zeng, Matthieu Schapira, Minkui Luo, Taraneh Hajian, and Hong Wu
- Subjects
Models, Molecular ,Adenosine ,Methyltransferase ,Chemistry ,Lysine ,Histone-Lysine N-Methyltransferase ,General Chemistry ,Computational biology ,Biochemistry ,Small molecule ,Article ,Catalysis ,Structure-Activity Relationship ,Sinefungin ,Colloid and Surface Chemistry ,SETD2 ,Molecular Probes ,Humans ,Epigenetics ,Enzyme Inhibitors ,Protein Lysine Methyltransferase - Abstract
Epigenetic regulation is involved in numerous physiological and pathogenic processes. Among the key regulators that orchestrate epigenetic signaling are over 50 human protein lysine methyltransferases (PKMTs). Interrogation of the functions of individual PKMTs can be facilitated by target-specific PKMT inhibitors. Given the emerging need for such small molecules, we envisioned an approach to identify target-specific methyltransferase inhibitors by screening privileged small-molecule scaffolds against diverse methyltransferases. In this work, we demonstrated the feasibility of such an approach by identifying the inhibitors of SETD2. N-propyl sinefungin (Pr-SNF) was shown to interact preferentially with SETD2 by matching the distinct transition-state features of SETD2's catalytically active conformer. With Pr-SNF as a structure probe, we further revealed the dual roles of SETD2's post-SET loop in regulating substrate access through a distinct topological reconfiguration. Privileged sinefungin scaffolds are expected to have broad use as structure and chemical probes of methyltransferases.
- Published
- 2012
- Full Text
- View/download PDF
10. A High Throughput Scintillation Proximity Imaging Assay for Protein Methyltransferases
- Author
-
David Shum, Constantin Radu, Christophe Antczak, Gil Blum, Bhavneet Bhinder, Hakim Djaballah, Glorymar Ibáñez, and Minkui Luo
- Subjects
chemistry.chemical_classification ,Streptavidin ,Drug discovery ,Organic Chemistry ,Lysine ,Peptide ,Histone-Lysine N-Methyltransferase ,General Medicine ,Methylation ,Biology ,Small molecule ,Article ,High-Throughput Screening Assays ,Computer Science Applications ,chemistry.chemical_compound ,chemistry ,Biochemistry ,Biotinylation ,Drug Discovery ,Humans ,Scintillation Counting ,Protein Methyltransferases ,Enzyme Inhibitors - Abstract
Protein methyltransferases (PMTs) orchestrate epigenetic modifications through post-translational methylation of various protein substrates including histones. Since dysregulation of this process is widely implicated in many cancers, it is of pertinent interest to screen inhibitors of PMTs, as they offer novel target-based opportunities to discover small molecules with potential chemotherapeutic use. We have thus developed an enzymatic screening strategy, which can be adapted to scintillation proximity imaging assay (SPIA) format, to identify these inhibitors. We took advantage of S-adenosyl-L-[3H-methyl]-methionine availability and monitored the enzymatically catalyzed [3H]-methyl addition on lysine residues of biotinylated peptide substrates. The radiolabeled peptides were subsequently captured by streptavidin coated SPA imaging PS beads. We applied this strategy to four PMTs: SET7/9, SET8, SETD2, and EuHMTase1, and optimized assay conditions to achieve Z' values ranging from 0.48 to 0.91. The robust performance of this SPIA for the four PMTs was validated in a pilot screen of approximately 7,000 compounds. We identified 80 cumulative hits across the four targets. NF279, a suramin analogue, was found to specifically inhibit SET7/9 and SETD2 with IC50 values of 1.9 and 1.1 μ M, respectively. Another identified compound, Merbromin, a topical antiseptic, was classified as a pan-active inhibitor of the four PMTs. These findings demonstrate that our proposed SPIA strategy is generic for multiple PMTs and can be successfully implemented to identify novel and specific inhibitors of PMTs. The specific PMT inhibitors may constitute a new class of anti-proliferative agents for potential therapeutic use.
- Published
- 2012
- Full Text
- View/download PDF
11. Drug Discovery and Repurposing for Retinoblastoma
- Author
-
Christophe Antczak, Hakim Djaballah, Bhavneet Bhinder, Kathryn G. Champ, Jeni P. Mahida, and Glorymar Ibáñez
- Subjects
Drug ,Retinoblastoma ,Computer science ,Drug discovery ,media_common.quotation_subject ,Value proposition ,Translational research ,medicine.disease ,Risk analysis (engineering) ,Return on investment ,Drug delivery ,medicine ,Repurposing ,media_common - Abstract
Retinoblastoma, a rare cancer of the eye, presents an urgent and unmet clinical need for an improved and effective treatment. Discovery of novel drugs and therapy would require an emphasis and refocus on adopting approaches that are fast, efficacious, and innovative, so as to bridge the growing gap between research labs and clinic. To this end, three main approaches with a potential of high return on investment and better value proposition have been described. First, use of high-throughput phenotypic screens to test a focused set of FDA-approved drugs presents an excellent opportunity to repurpose existing drugs and foster translational research, with minimal time commitment. This is also complemented by the advent of alternate routes of localized drug delivery, which opens doors to revisit shelved drugs previously ignored due to their high systemic toxicity. In addition, testing natural products offer the capability to broaden the limited structural diversity within the commercially available chemical collections and can fuel identification of new chemical entities. Second, non-toxic nutraceuticals can be evaluated for their applicability as companion effectors to enhance ongoing chemotherapy, improving the health of young patients. Third, use of three-dimensional models to test chemotherapeutic agents for retinoblastoma is proposed; such models are true prototypes of an in vivo tumor, and perhaps more realistic indicators of a drug’s efficacy and higher potential of clinical success. Taken together, these three approaches can be viewed as the rapid routes towards drug discovery with hopes to establish a quick and potent therapy for retinoblastoma.
- Published
- 2015
- Full Text
- View/download PDF
12. Small-molecule inhibitors of SETD8 with cellular activity
- Author
-
Constantin Radu, Judd C. Rice, David Shum, Xiangjun Rao, Minkui Luo, Glorymar Ibáñez, Hakim Djaballah, and Gil Blum
- Subjects
0303 health sciences ,Methyltransferase ,DNA damage ,HEK 293 cells ,DNA replication ,General Medicine ,Histone-Lysine N-Methyltransferase ,Biology ,Cell cycle ,Biochemistry ,Small molecule ,3. Good health ,High-Throughput Screening Assays ,Histone H4 ,03 medical and health sciences ,0302 clinical medicine ,HEK293 Cells ,Transcription (biology) ,030220 oncology & carcinogenesis ,Molecular Medicine ,Humans ,Letters ,030304 developmental biology - Abstract
SETD8/SET8/Pr-SET7/KMT5A is the sole protein lysine methyltransferase (PKMT) known to monomethylate lysine 20 of histone H4 in vivo. SETD8's methyltransferase activity has been implicated in many essential cellular processes including DNA replication, DNA damage response, transcription modulation, and cell cycle regulation. Developing SETD8 inhibitors with cellular activity is a key step toward elucidating the diverse roles of SETD8 via convenient pharmacological perturbation. From the hits of a prior high throughput screen (HTS), SPS8I1-3 (NSC663284, BVT948, and ryuvidine) were validated as potent SETD8 inhibitors. These compounds contain different structural motifs and inhibit SETD8 via distinct modes. More importantly, these compounds show cellular activity by suppressing the H4K20me1 mark of SETD8 and recapitulate characteristic S/G2/M-phase cell cycle defects as observed for RNAi-mediated SETD8 knockdown. The commonality of SPS8I1-3 against SETD8, together with their distinct structures and mechanisms for SETD8 inhibition, argues for the collective application of these compounds as SETD8 inhibitors.
- Published
- 2014
13. Discovery of a dicer-independent, cell-type dependent alternate targeting sequence generator: implications in gene silencing & pooled RNAi screens
- Author
-
Mu Li, Hakim Djaballah, Bhavneet Bhinder, Susan Magdaleno, Glorymar Ibáñez, Alexander V. Vlassov, and David Shum
- Subjects
Ribonuclease III ,Small interfering RNA ,Science ,Trans-acting siRNA ,Computational biology ,DEAD-box RNA Helicases ,Small hairpin RNA ,RNA interference ,Genetics ,Humans ,RNA, Small Interfering ,Drosha ,Multidisciplinary ,Biology and life sciences ,biology ,Sequence Analysis, RNA ,Computational Biology ,Gene targeting ,Genomics ,Functional Genomics ,RNA silencing ,Genetic interference ,Gene Targeting ,biology.protein ,Medicine ,Gene expression ,Gene Function ,HeLa Cells ,Research Article ,Dicer - Abstract
There is an acceptance that plasmid-based delivery of interfering RNA always generates the intended targeting sequences in cells, making it as specific as its synthetic counterpart. However, recent studies have reported on cellular inefficiencies of the former, especially in light of emerging gene discordance at inter-screen level and across formats. Focusing primarily on the TRC plasmid-based shRNA hairpins, we reasoned that alleged specificities were perhaps compromised due to altered processing; resulting in a multitude of random interfering sequences. For this purpose, we opted to study the processing of hairpin TRCN#40273 targeting CTTN; which showed activity in a miRNA-21 gain-of-function shRNA screen, but inactive when used as an siRNA duplex. Using a previously described walk-through method, we identified 36 theoretical cleavage variants resulting in 78 potential siRNA duplexes targeting 53 genes. We synthesized and tested all of them. Surprisingly, six duplexes targeting ASH1L, DROSHA, GNG7, PRKCH, THEM4, and WDR92 scored as active. QRT-PCR analysis on hairpin transduced reporter cells confirmed knockdown of all six genes, besides CTTN; revealing a surprising 7 gene-signature perturbation by this one single hairpin. We expanded our qRT-PCR studies to 26 additional cell lines and observed unique knockdown profiles associated with each cell line tested; even for those lacking functional DICER1 gene suggesting no obvious dependence on dicer for shRNA hairpin processing; contrary to published models. Taken together, we report on a novel dicer independent, cell-type dependent mechanism for non-specific RNAi gene silencing we coin Alternate Targeting Sequence Generator (ATSG). In summary, ATSG adds another dimension to the already complex interpretation of RNAi screening data, and provides for the first time strong evidence in support of arrayed screening, and questions the scientific merits of performing pooled RNAi screens, where deconvolution of up to genome-scale pools is indispensable for target identification.
- Published
- 2014
14. Formulating a fluorogenic assay to evaluate S-adenosyl-L-methionine analogues as protein methyltransferase cofactors
- Author
-
Glorymar Ibáñez, Weihong Zheng, Gil Blum, Rui Wang, Caitlin Sengelaub, Kabirul Islam, and Minkui Luo
- Subjects
Protein-Arginine N-Methyltransferases ,S-Adenosylmethionine ,Methyltransferase ,CARM1 ,High-throughput screening ,Lysine ,Coenzymes ,Methylation ,Biology ,Small molecule ,Article ,High-Throughput Screening Assays ,Biochemistry ,Luminescent Measurements ,Humans ,Protein Methyltransferases ,Molecular Biology ,Biotechnology - Abstract
Protein methyltransferases (PMTs) catalyze arginine and lysine methylation of diverse histone and nonhistone targets. These posttranslational modifications play essential roles in regulating multiple cellular events in an epigenetic manner. In the recent process of defining PMT targets, S-adenosyl-L-methionine (SAM) analogues have emerged as powerful small molecule probes to label and profile PMT targets. To examine efficiently the reactivity of PMTs and their variants on SAM analogues, we transformed a fluorogenic PMT assay into a ready high throughput screening (HTS) format. The reformulated fluorogenic assay is featured by its uncoupled but more robust character with the first step of accumulation of the commonly-shared reaction byproduct S-adenosyl-L-homocysteine (SAH), followed by SAH-hydrolase-mediated fluorogenic quantification. The HTS readiness and robustness of the assay were demonstrated by its excellent Z′ values of 0.83–0.95 for the so-far-examined 8 human PMTs with SAM as a cofactor (PRMT1, PRMT3, CARM1, SUV39H2, SET7/9, SET8, G9a and GLP1). The fluorogenic assay was further implemented to screen the PMTs against five SAM analogues (allyl-SAM, propargyl-SAM, (E)-pent-2-en-4-ynyl-SAM (EnYn-SAM), (E)-hex-2-en-5-ynyl-SAM (Hey-SAM) and 4-propargyloxy-but-2-enyl-SAM (Pob-SAM)). Among the examined 8 × 5 pairs of PMTs and SAM analogues, native SUV39H2, G9a and GLP1 showed promiscuous activity on allyl-SAM. In contrast, the bulky SAM analogues, such as EnYn-SAM, Hey-SAM and Pob-SAM, are inert toward the panel of human PMTs. These findings therefore provide the useful structure–activity guidance to further evolve PMTs and SAM analogues for substrate labeling. The current assay format is ready to screen methyltransferase variants on structurally-diverse SAM analogues.
- Published
- 2011
15. An enzyme-coupled ultrasensitive luminescence assay for protein methyltransferases
- Author
-
Minkui Luo, Yaritzy M. Astudillo, Jamie L. McBean, and Glorymar Ibáñez
- Subjects
Adenosine monophosphate ,S-Adenosylmethionine ,Methyltransferase ,Biophysics ,Biochemistry ,Sensitivity and Specificity ,chemistry.chemical_compound ,Histocompatibility Antigens ,medicine ,Protein methylation ,Humans ,Luciferase ,Protein Methyltransferases ,Molecular Biology ,Enzyme Assays ,chemistry.chemical_classification ,Cell Biology ,Methylation ,Histone-Lysine N-Methyltransferase ,Luciferin ,Adenosine ,Molecular biology ,S-Adenosylhomocysteine ,Enzyme ,chemistry ,Luminescent Measurements ,medicine.drug - Abstract
Epigenetic regulation through protein posttranslational modifications is essential in development and disease. Among the key chemical modifications is protein methylation carried out by protein methyltransferases (PMTs). Quantitative and sensitive PMT activity assays can provide valuable tools to investigate PMT functions. Here we developed an enzyme-coupled luminescence assay for S-adenosyl-l-methionine (AdoMet/SAM)-based PMTs. In this assay, S-adenosyl-l-homocystine (AdoHcy/SAH), the by-product of PMT-involved methylation, is sequentially converted to adenine, adenosine monophosphate, and then adenosine 5'-triphosphate (ATP) by 5'-methylthio-adenosine/AdoHcy nucleosidase (MTAN), adenine phosphoribosyl transferase (APRT), and pyruvate orthophosphate dikinase (PPDK), respectively. The resultant ATP can be readily quantified with a luciferin/luciferase kit. This assay is featured for its quantitative linear response to AdoHcy and the ultrasensitivity to 0.3 pmol of AdoHcy. With this assay, the kinetic parameters of SET7/9 methylation were characterized and unambiguously support an ordered mechanism with AdoMet binding as the initial step, followed by the substrate binding and the rate-limiting methylation. The luminescence assay is also expected to be generally applicable to many other AdoMet-dependent enzymes. In addition, the mix-and-measure 96-/384-well format of our assay makes it suitable for automation and high throughput. Our enzyme-coupled luminescence assay, therefore, represents a convenient and ultrasensitive approach to examine methyltransferase activities and identify methyltransferase inhibitors.
- Published
- 2009
16. A phase I trial of a pharmacodynamically-conceived decitabine/thioguanine combination in patients with advanced myeloid malignancies
- Author
-
Katherine Harwood, Martin Santos, J. Gregory Mears, Mark G. Frattini, Joseph M. Scandura, Ruth Santos, Joseph G. Jurcic, Diana Carrillo, Rong Zhang, Anthony Letai, Todd L. Rosenblat, Jacquelyn Gonzales, Azra Raza, Leah Hogdal, Mark L. Heaney, Hakim Djaballah, Glorymar Ibáñez, and Jennifer L. Crombie
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Myeloid ,business.industry ,Screening assay ,Decitabine ,Pharmacology ,medicine.disease ,Leukemia ,medicine.anatomical_structure ,Refractory ,Internal medicine ,medicine ,In patient ,business ,medicine.drug - Abstract
e18025 Background: Using a chemosensitivity screening assay, we showed that combination decitabine and thioguanine was active in primary leukemia cells from patients with relapsed/refractory acute ...
- Published
- 2015
- Full Text
- View/download PDF
17. New Guanidine Alkaloids from the Leaves of Verbesina Peraffinis
- Author
-
Reinaldo S. Compagnone, Anita Israel, Glorymar Ibáñez, María del Rosario Garrido, Alírica I. Suárez, Beth Diaz, and Jhorman Bermudez
- Subjects
Pharmacology ,biology ,Stereochemistry ,Verbesina ,Plant Science ,General Medicine ,Methylamide ,Moraceae ,biology.organism_classification ,chemistry.chemical_compound ,Complementary and alternative medicine ,chemistry ,Drug Discovery ,Caffeic acid ,Methanol ,Guanidine - Abstract
Two new guanidine alkaloids, 1 and 2, were isolated from the methanol extract of the leaf of Verbesina peraffinis, in addition to four known substances: galegine, the methylamide of caffeic acid, galactitol and the flavonoid 3- O-glucosyl-(1–2)-galactoside-5,7,4′ trimethoxy-kaempferol. Their structures were determined on the basis of spectroscopic methods. The antihypertensive activity of the methanolic extract was assayed in rats using a model of hypertension induced by footshock.
- Published
- 2008
- Full Text
- View/download PDF
18. Therapeutic targeting of ATR in alveolar rhabdomyosarcoma
- Author
-
Heathcliff Dorado García, Fabian Pusch, Yi Bei, Jennifer von Stebut, Glorymar Ibáñez, Kristina Guillan, Koshi Imami, Dennis Gürgen, Jana Rolff, Konstantin Helmsauer, Stephanie Meyer-Liesener, Natalie Timme, Victor Bardinet, Rocío Chamorro González, Ian C. MacArthur, Celine Y. Chen, Joachim Schulz, Antje M. Wengner, Christian Furth, Birgit Lala, Angelika Eggert, Georg Seifert, Patrick Hundsoerfer, Marieluise Kirchner, Philipp Mertins, Matthias Selbach, Andrej Lissat, Frank Dubois, David Horst, Johannes H. Schulte, Simone Spuler, Daoqi You, Filemon Dela Cruz, Andrew L. Kung, Kerstin Haase, Michela DiVirgilio, Monika Scheer, Michael V. Ortiz, and Anton G. Henssen
- Subjects
Cancer Research ,Multidisciplinary ,Oncogene Proteins, Fusion ,General Physics and Astronomy ,General Chemistry ,Ataxia Telangiectasia Mutated Proteins ,General Biochemistry, Genetics and Molecular Biology ,Gene Expression Regulation, Neoplastic ,Cardiovascular and Metabolic Diseases ,Cell Line, Tumor ,Rhabdomyosarcoma ,Humans ,Paired Box Transcription Factors ,Rhabdomyosarcoma, Embryonal ,Technology Platforms ,Function and Dysfunction of the Nervous System ,PAX3 Transcription Factor ,Rhabdomyosarcoma, Alveolar - Abstract
Despite advances in multi-modal treatment approaches, clinical outcomes of patients suffering from PAX3-FOXO1 fusion oncogene-expressing alveolar rhabdomyosarcoma (ARMS) remain dismal. Here we show that PAX3-FOXO1-expressing ARMS cells are sensitive to pharmacological ataxia telangiectasia and Rad3 related protein (ATR) inhibition. Expression of PAX3-FOXO1 in muscle progenitor cells is not only sufficient to increase sensitivity to ATR inhibition, but PAX3-FOXO1-expressing rhabdomyosarcoma cells also exhibit increased sensitivity to structurally diverse inhibitors of ATR. Mechanistically, ATR inhibition leads to replication stress exacerbation, decreased BRCA1 phosphorylation and reduced homologous recombination-mediated DNA repair pathway activity. Consequently, ATR inhibitor treatment increases sensitivity of ARMS cells to PARP1 inhibition in vitro, and combined treatment with ATR and PARP1 inhibitors induces complete regression of primary patient-derived ARMS xenografts in vivo. Lastly, a genome-wide CRISPR activation screen (CRISPRa) in combination with transcriptional analyses of ATR inhibitor resistant ARMS cells identifies the RAS-MAPK pathway and its targets, the FOS gene family, as inducers of resistance to ATR inhibition. Our findings provide a rationale for upcoming biomarker-driven clinical trials of ATR inhibitors in patients suffering from ARMS.
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.