15 results on '"Rathi, Komal S."'
Search Results
2. Epigenetic regulator BMI1 promotes alveolar rhabdomyosarcoma proliferation and constitutes a novel therapeutic target.
- Author
-
Shields, Cara E., Potlapalli, Sindhu, Cuya‐Smith, Selma M., Chappell, Sarah K., Chen, Dongdong, Martinez, Daniel, Pogoriler, Jennifer, Rathi, Komal S., Patel, Shiv A., Oristian, Kristianne M., Linardic, Corinne M., Maris, John M., Haynes, Karmella A., and Schnepp, Robert W.
- Published
- 2021
- Full Text
- View/download PDF
3. annoFuse: an R Package to annotate, prioritize, and interactively explore putative oncogenic RNA fusions.
- Author
-
Gaonkar, Krutika S., Marini, Federico, Rathi, Komal S., Jain, Payal, Zhu, Yuankun, Chimicles, Nicholas A., Brown, Miguel A., Naqvi, Ammar S., Zhang, Bo, Storm, Phillip B., Maris, John M., Raman, Pichai, Resnick, Adam C., Strauch, Konstantin, Taroni, Jaclyn N., and Rokita, Jo Lynne
- Abstract
Background: Gene fusion events are significant sources of somatic variation across adult and pediatric cancers and are some of the most clinically-effective therapeutic targets, yet low consensus of RNA-Seq fusion prediction algorithms makes therapeutic prioritization difficult. In addition, events such as polymerase read-throughs, mis-mapping due to gene homology, and fusions occurring in healthy normal tissue require informed filtering, making it difficult for researchers and clinicians to rapidly discern gene fusions that might be true underlying oncogenic drivers of a tumor and in some cases, appropriate targets for therapy. Results: We developed annoFuse, an R package, and shinyFuse, a companion web application, to annotate, prioritize, and explore biologically-relevant expressed gene fusions, downstream of fusion calling. We validated annoFuse using a random cohort of TCGA RNA-Seq samples (N = 160) and achieved a 96% sensitivity for retention of high-confidence fusions (N = 603). annoFuse uses FusionAnnotator annotations to filter non-oncogenic and/or artifactual fusions. Then, fusions are prioritized if previously reported in TCGA and/or fusions containing gene partners that are known oncogenes, tumor suppressor genes, COSMIC genes, and/or transcription factors. We applied annoFuse to fusion calls from pediatric brain tumor RNA-Seq samples (N = 1028) provided as part of the Open Pediatric Brain Tumor Atlas (OpenPBTA) Project to determine recurrent fusions and recurrently-fused genes within different brain tumor histologies. annoFuse annotates protein domains using the PFAM database, assesses reciprocality, and annotates gene partners for kinase domain retention. As a standard function, reportFuse enables generation of a reproducible R Markdown report to summarize filtered fusions, visualize breakpoints and protein domains by transcript, and plot recurrent fusions within cohorts. Finally, we created shinyFuse for algorithm-agnostic interactive exploration and plotting of gene fusions. Conclusions: annoFuse provides standardized filtering and annotation for gene fusion calls from STAR-Fusion and Arriba by merging, filtering, and prioritizing putative oncogenic fusions across large cancer datasets, as demonstrated here with data from the OpenPBTA project. We are expanding the package to be widely-applicable to other fusion algorithms and expect annoFuse to provide researchers a method for rapidly evaluating, prioritizing, and translating fusion findings in patient tumors. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
4. Mitochondrial DNA Haplogroups and Susceptibility to Neuroblastoma.
- Author
-
Chang, Xiao, Bakay, Marina, Liu, Yichuan, Glessner, Joseph, Rathi, Komal S, Hou, Cuiping, Qu, Huiqi, Vaksman, Zalman, Nguyen, Kenny, Sleiman, Patrick M A, Diskin, Sharon J, Maris, John M, and Hakonarson, Hakon
- Subjects
NEUROBLASTOMA ,MITOCHONDRIAL DNA ,SYMPATHETIC nervous system ,CHILDREN'S hospitals ,MITOCHONDRIA ,ETIOLOGY of diseases ,HAPLOGROUPS ,RESEARCH ,DNA ,CELL culture ,RESEARCH methodology ,GENETIC polymorphisms ,CASE-control method ,ACQUISITION of data ,EVALUATION research ,COMPARATIVE studies ,AGE factors in disease ,HAPLOTYPES ,GENES ,DISEASE susceptibility ,RESEARCH funding ,GENETIC techniques ,LONGITUDINAL method - Abstract
Background: Neuroblastoma is a childhood malignancy that arises from the developing sympathetic nervous system. Although mitochondrial dysfunctions have been implicated in the pathophysiology of neuroblastoma, the role of mitochondrial DNA (mtDNA) has not been extensively investigated.Methods: A total of 2404 Caucasian children diagnosed with neuroblastoma and 9310 ancestry-matched controls were recruited at the Children's Hospital of Philadelphia. The mtDNA haplogroups were identified from SNP array data of two independent cohorts. We conducted a case-control study to explore potential associations of mtDNA haplogroups with the susceptibility of neuroblastoma. The genetic effect of neuroblastoma was measured by odds ratios (ORs) of mitochondrial haplogroups. All tests were two-sided.Results: Haplogroup K was statistically significantly associated with reduced risk of neuroblastoma in the discovery cohort consisting of 1474 cases and 5699 controls (OR = 0.72, 95% confidence interval [CI] = 0.57 to 0.90; P = 4.8 × 10-3). The association was replicated in an independent cohort (OR = 0.69, 95% CI = 0.53 to 0.92; P = .01) of 930 cases and 3611 controls. Pooled analysis was performed by combining the two data sets. The association remained highly statistically significant after correction for multiple testing (OR = 0.71, 95% CI = 0.59 to 0.84, P = 1.96 × 10-4, Pcorrected = .002). Further analysis focusing on neuroblastoma subtypes indicated haplogroup K was more associated with high-risk neuroblastoma (OR = 0.57, 95% CI = 0.43 to 0.76; P = 1.46 × 10-4) than low-risk and intermediate-risk neuroblastoma.Conclusions: Haplogroup K is an independent genetic factor associated with reduced risk of developing neuroblastoma in European descents. These findings provide new insights into the genetic basis of neuroblastoma, implicating mitochondrial DNA encoded proteins in the etiology of neuroblastoma. [ABSTRACT FROM AUTHOR]- Published
- 2020
- Full Text
- View/download PDF
5. A transcriptome-based classifier to determine molecular subtypes in medulloblastoma.
- Author
-
Rathi, Komal S., Arif, Sherjeel, Koptyra, Mateusz, Naqvi, Ammar S., Taylor, Deanne M., Storm, Phillip B., Resnick, Adam C., Rokita, Jo Lynne, and Raman, Pichai
- Subjects
MEDULLOBLASTOMA ,RNA sequencing ,BRAIN tumors ,GENE expression ,SCIENTIFIC community - Abstract
Medulloblastoma is a highly heterogeneous pediatric brain tumor with five molecular subtypes, Sonic Hedgehog TP53-mutant, Sonic Hedgehog TP53-wildtype, WNT, Group 3, and Group 4, defined by the World Health Organization. The current mechanism for classification into these molecular subtypes is through the use of immunostaining, methylation, and/or genetics. We surveyed the literature and identified a number of RNA-Seq and microarray datasets in order to develop, train, test, and validate a robust classifier to identify medulloblastoma molecular subtypes through the use of transcriptomic profiling data. We have developed a GPL-3 licensed R package and a Shiny Application to enable users to quickly and robustly classify medulloblastoma samples using transcriptomic data. The classifier utilizes a large composite microarray dataset (15 individual datasets), an individual microarray study, and an RNA-Seq dataset, using gene ratios instead of gene expression measures as features for the model. Discriminating features were identified using the limma R package and samples were classified using an unweighted mean of normalized scores. We utilized two training datasets and applied the classifier in 15 separate datasets. We observed a minimum accuracy of 85.71% in the smallest dataset and a maximum of 100% accuracy in four datasets with an overall median accuracy of 97.8% across the 15 datasets, with the majority of misclassification occurring between the heterogeneous Group 3 and Group 4 subtypes. We anticipate this medulloblastoma transcriptomic subtype classifier will be broadly applicable to the cancer research and clinical communities. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
6. CAMKV Is a Candidate Immunotherapeutic Target in MYCN Amplified Neuroblastoma.
- Author
-
Sussman, Robyn T., Rokita, Jo Lynne, Huang, Kevin, Raman, Pichai, Rathi, Komal S., Martinez, Daniel, Bosse, Kristopher R., Lane, Maria, Hart, Lori S., Bhatti, Tricia, Pawel, Bruce, and Maris, John M.
- Subjects
NEUROBLASTOMA ,GENE expression profiling ,ANTIBODY-drug conjugates ,CENTRAL nervous system ,CELL membranes - Abstract
We developed a computational pipeline designed to use RNA sequencing (n = 136) and gene expression profiling (n = 250) data from neuroblastoma tumors to identify cell surface proteins predicted to be highly expressed in MYCN amplified neuroblastomas and with little or no expression in normal human tissues. We then performed ChIP-seq in the MYCN amplified cell lines KELLY, NB-1643, and NGP to identify gene promoters that are occupied by MYCN protein to define the intersection with the differentially-expressed gene list. We initially identified 116 putative immunotherapy targets with predicted transmembrane domains, with the most significant differentially-expressed of these being the calmodulin kinase-like vesicle-associated gene (CAMKV, p = 2 × 10
−6 ). CAMKV encodes a protein that binds calmodulin in the presence of calcium, but lacks the kinase activity of other calmodulin kinase family members. We confirmed that CAMKV is selectively expressed in 7/7 MYCN amplified neuroblastoma cell lines and showed that the transcription of CAMKV is directly controlled by MYCN. From membrane fractionation and immunohistochemistry, we verified that CAMKV is membranous in MYCN amplified neuroblastoma cell lines and patient-derived xenografts. Finally, immunohistochemistry showed that CAMKV is not expressed on normal tissues outside of the central nervous system. Together, these data demonstrate that CAMKV is a differentially-expressed cell surface protein that is transcriptionally regulated by MYCN, making it a candidate for targeting with antibodies or antibody-drug conjugates that do not cross the blood brain barrier. [ABSTRACT FROM AUTHOR]- Published
- 2020
- Full Text
- View/download PDF
7. The ADP/ATP translocase drives mitophagy independent of nucleotide exchange.
- Author
-
Hoshino, Atsushi, Wang, Wei-jia, Wada, Shogo, McDermott-Roe, Chris, Evans, Chantell S., Gosis, Bridget, Morley, Michael P., Rathi, Komal S., Li, Jian, Li, Kristina, Yang, Steven, McManus, Meagan J., Bowman, Caitlyn, Potluri, Prasanth, Levin, Michael, Damrauer, Scott, Wallace, Douglas C., Holzbaur, Erika L. F., and Arany, Zoltan
- Abstract
Mitochondrial homeostasis depends on mitophagy, the programmed degradation of mitochondria. Only a few proteins are known to participate in mitophagy. Here we develop a multidimensional CRISPR–Cas9 genetic screen, using multiple mitophagy reporter systems and pro-mitophagy triggers, and identify numerous components of parkin-dependent mitophagy1. Unexpectedly, we find that the adenine nucleotide translocator (ANT) complex is required for mitophagy in several cell types. Whereas pharmacological inhibition of ANT-mediated ADP/ATP exchange promotes mitophagy, genetic ablation of ANT paradoxically suppresses mitophagy. Notably, ANT promotes mitophagy independently of its nucleotide translocase catalytic activity. Instead, the ANT complex is required for inhibition of the presequence translocase TIM23, which leads to stabilization of PINK1, in response to bioenergetic collapse. ANT modulates TIM23 indirectly via interaction with TIM44, which regulates peptide import through TIM232. Mice that lack ANT1 show blunted mitophagy and consequent profound accumulation of aberrant mitochondria. Disease-causing human mutations in ANT1 abrogate binding to TIM44 and TIM23 and inhibit mitophagy. Together, our findings show that ANT is an essential and fundamental mediator of mitophagy in health and disease. A CRISPR–Cas9 genetic screen shows that the adenine nucleotide translocator is required for mitophagy and that this role is independent of its nucleotide translocase activity. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
8. Macrophages in SHH subgroup medulloblastoma display dynamic heterogeneity that varies with treatment modality.
- Author
-
Dang, Mai T., Gonzalez, Michael V., Gaonkar, Krutika S., Rathi, Komal S., Young, Patricia, Arif, Sherjeel, Zhai, Li, Alam, Zahidul, Devalaraja, Samir, Jerrick To, Tsun Ki, Folkert, Ian W., Raman, Pichai, Rokita, Jo Lynne, Martinez, Daniel, Taroni, Jaclyn N., Shapiro, Joshua A., Greene, Casey S., Savonen, Candace, Mafra, Fernanda, and Hakonarson, Hakon
- Published
- 2023
- Full Text
- View/download PDF
9. Hedgehog actively maintains adult lung quiescence and regulates repair and regeneration.
- Author
-
Peng, Tien, Frank, David B., Kadzik, Rachel S., Morley, Michael P., Rathi, Komal S., Wang, Tao, Zhou, Su, Cheng, Lan, Lu, Min Min, and Morrisey, Edward E.
- Subjects
HEDGEHOG signaling proteins ,LUNGS ,REGENERATION (Biology) ,MESENCHYME ,PHYSIOLOGICAL control systems ,HOMEOSTASIS ,WOUND healing - Abstract
Postnatal tissue quiescence is thought to be a default state in the absence of a proliferative stimulus such as injury. Although previous studies have demonstrated that certain embryonic developmental programs are reactivated aberrantly in adult organs to drive repair and regeneration, it is not well understood how quiescence is maintained in organs such as the lung, which displays a remarkably low level of cellular turnover. Here we demonstrate that quiescence in the adult lung is an actively maintained state and is regulated by hedgehog signalling. Epithelial-specific deletion of sonic hedgehog (Shh) during postnatal homeostasis in the murine lung results in a proliferative expansion of the adjacent lung mesenchyme. Hedgehog signalling is initially downregulated during the acute phase of epithelial injury as the mesenchyme proliferates in response, but returns to baseline during injury resolution as quiescence is restored. Activation of hedgehog during acute epithelial injury attenuates the proliferative expansion of the lung mesenchyme, whereas inactivation of hedgehog signalling prevents the restoration of quiescence during injury resolution. Finally, we show that hedgehog also regulates epithelial quiescence and regeneration in response to injury via a mesenchymal feedback mechanism. These results demonstrate that epithelial-mesenchymal interactions coordinated by hedgehog actively maintain postnatal tissue homeostasis, and deregulation of hedgehog during injury leads to aberrant repair and regeneration in the lung. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
10. Correcting Transcription Factor Gene Sets for Copy Number and Promoter Methylation Variations.
- Author
-
Rathi, Komal S., Gaykalova, Daria A., Hennessey, Patrick, Califano, Joseph A., and Ochs, Michael F.
- Subjects
METHYLATION ,PROMOTERS (Genetics) ,DNA copy number variations ,TRANSCRIPTION factors ,GENES ,SIMULATION methods & models - Abstract
The article presents a study which introduces a filtering approach that removes genes from consideration if promoter methylation or copy number loss is shown and demonstrates the improvement in transcription factor activity inference in a simulated dataset. A brief overview of gene set analysis (GSA), the simulation method, and the analysis of results are discussed.
- Published
- 2014
- Full Text
- View/download PDF
11. Macrophages in SHH subgroup medulloblastoma display dynamic heterogeneity that varies with treatment modality.
- Author
-
Dang, Mai T., Gonzalez, Michael V., Gaonkar, Krutika S., Rathi, Komal S., Young, Patricia, Arif, Sherjeel, Zhai, Li, Alam, Zahidul, Devalaraja, Samir, To, Tsun Ki Jerrick, Folkert, Ian W., Raman, Pichai, Rokita, Jo Lynne, Martinez, Daniel, Taroni, Jaclyn N., Shapiro, Joshua A., Greene, Casey S., Savonen, Candace, Mafra, Fernanda, and Hakonarson, Hakon
- Abstract
Tumor-associated macrophages (TAMs) play an important role in tumor immunity and comprise of subsets that have distinct phenotype, function, and ontology. Transcriptomic analyses of human medulloblastoma, the most common malignant pediatric brain cancer, showed that medulloblastomas (MBs) with activated sonic hedgehog signaling (SHH-MB) have significantly more TAMs than other MB subtypes. Therefore, we examined MB-associated TAMs by single-cell RNA sequencing of autochthonous murine SHH-MB at steady state and under two distinct treatment modalities: molecular-targeted inhibitor and radiation. Our analyses reveal significant TAM heterogeneity, identify markers of ontologically distinct TAM subsets, and show the impact of brain microenvironment on the differentiation of tumor-infiltrating monocytes. TAM composition undergoes dramatic changes with treatment and differs significantly between molecular-targeted and radiation therapy. We identify an immunosuppressive monocyte-derived TAM subset that emerges with radiation therapy and demonstrate its role in regulating T cell and neutrophil infiltration in MB. [Display omitted] • Sonic Hedgehog (SHH) subgroup of medulloblastoma (MB) recruits diverse macrophages • Radiation or molecular-targeted therapy alters macrophage distribution in SHH-MB • Radiation recruits immunosuppressive monocyte-derived macrophages (TAMoMacs) in SHH-MB • Radiation-induced TAMoMacs regulate CD8 T cell and neutrophil numbers in SHH-MB Dang et al. show that the sonic hedgehog subgroup of medulloblastoma (SHH-MB) contains macrophages derived from microglia and circulating monocytes. Radiation therapy, but not treatment targeting the SHH pathway, led to recruitment of immunosuppressive monocyte-derived macrophages that reduced T cells and neutrophils in the tumor microenvironment. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
12. NASA GeneLab Platform Utilized for Biological Response to Space Radiation in Animal Models.
- Author
-
McDonald, J. Tyson, Stainforth, Robert, Miller, Jack, Cahill, Thomas, Silveira, Willian A. da, Rathi, Komal S., Hardiman, Gary, Taylor, Deanne, Costes, Sylvain V., Chauhan, Vinita, Meller, Robert, and Beheshti, Afshin
- Subjects
MITOCHONDRIAL physiology ,ANIMAL experimentation ,BIOLOGICAL models ,IMMUNITY ,RADIATION ,RADIATION doses ,RADIATION dosimetry ,SPACE flight ,OCCUPATIONAL hazards ,ENVIRONMENTAL exposure ,RIBOSOMAL proteins - Abstract
Background: Ionizing radiation from galactic cosmic rays (GCR) is one of the major risk factors that will impact the health of astronauts on extended missions outside the protective effects of the Earth's magnetic field. The NASA GeneLab project has detailed information on radiation exposure using animal models with curated dosimetry information for spaceflight experiments. Methods: We analyzed multiple GeneLab omics datasets associated with both ground-based and spaceflight radiation studies that included in vivo and in vitro approaches. A range of ions from protons to iron particles with doses from 0.1 to 1.0 Gy for ground studies, as well as samples flown in low Earth orbit (LEO) with total doses of 1.0 mGy to 30 mGy, were utilized. Results: From this analysis, we were able to identify distinct biological signatures associating specific ions with specific biological responses due to radiation exposure in space. For example, we discovered changes in mitochondrial function, ribosomal assembly, and immune pathways as a function of dose. Conclusions: We provided a summary of how the GeneLab's rich database of omics experiments with animal models can be used to generate novel hypotheses to better understand human health risks from GCR exposures. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
13. Genomic Profiling of Childhood Tumor Patient-Derived Xenograft Models to Enable Rational Clinical Trial Design.
- Author
-
Rokita, Jo Lynne, Rathi, Komal S., Cardenas, Maria F., Upton, Kristen A., Jayaseelan, Joy, Cross, Katherine L., Pfeil, Jacob, Egolf, Laura E., Way, Gregory P., Farrel, Alvin, Kendsersky, Nathan M., Patel, Khushbu, Gaonkar, Krutika S., Modi, Apexa, Berko, Esther R., Lopez, Gonzalo, Vaksman, Zalman, Mayoh, Chelsea, Nance, Jonas, and McCoy, Kristyn
- Abstract
Accelerating cures for children with cancer remains an immediate challenge as a result of extensive oncogenic heterogeneity between and within histologies, distinct molecular mechanisms evolving between diagnosis and relapsed disease, and limited therapeutic options. To systematically prioritize and rationally test novel agents in preclinical murine models, researchers within the Pediatric Preclinical Testing Consortium are continuously developing patient-derived xenografts (PDXs)—many of which are refractory to current standard-of-care treatments—from high-risk childhood cancers. Here, we genomically characterize 261 PDX models from 37 unique pediatric cancers; demonstrate faithful recapitulation of histologies and subtypes; and refine our understanding of relapsed disease. In addition, we use expression signatures to classify tumors for TP53 and NF1 pathway inactivation. We anticipate that these data will serve as a resource for pediatric oncology drug development and will guide rational clinical trial design for children with cancer. • Multiplatform analysis facilitates genomic resource of 261 pediatric cancer PDX models • PPTC PDX models are reflective of high-risk and chemotherapy resistant disease • Inferred TP53 pathway inactivation correlates with pediatric cancer copy number burden • Pediatric cancer PDX models will be useful for drug development prioritization Rokita et. al provide an extensively annotated genomic dataset of somatic oncogenic regulation across 37 distinct pediatric malignancies. The 261 patient-derived xenograft models are available to the scientific community, and the genomic annotations will enable rational preclinical agent prioritization and acceleration of therapeutic targets for early-phase pediatric oncology clinical trials. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
14. Clinical utility of custom-designed NGS panel testing in pediatric tumors.
- Author
-
Surrey, Lea F., MacFarland, Suzanne P., Chang, Fengqi, Cao, Kajia, Rathi, Komal S., Akgumus, Gozde T., Gallo, Daniel, Lin, Fumin, Gleason, Adam, Raman, Pichai, Aplenc, Richard, Bagatell, Rochelle, Minturn, Jane, Mosse, Yael, Santi, Mariarita, Tasian, Sarah K., Waanders, Angela J., Sarmady, Mahdi, Maris, John M., and Hunger, Stephen P.
- Subjects
TUMORS in children ,GENETIC testing ,PEDIATRICS ,GENETIC mutation ,DNA copy number variations - Abstract
Background: Somatic genetic testing is rapidly becoming the standard of care in many adult and pediatric cancers. Previously, the standard approach was single-gene or focused multigene testing, but many centers have moved towards broad-based next-generation sequencing (NGS) panels. Here, we report the laboratory validation and clinical utility of a large cohort of clinical NGS somatic sequencing results in diagnosis, prognosis, and treatment of a wide range of pediatric cancers. Methods: Subjects were accrued retrospectively at a single pediatric quaternary-care hospital. Sequence analyses were performed on 367 pediatric cancer samples using custom-designed NGS panels over a 15-month period. Cases were profiled for mutations, copy number variations, and fusions identified through sequencing, and their clinical impact on diagnosis, prognosis, and therapy was assessed. Results: NGS panel testing was incorporated meaningfully into clinical care in 88.7% of leukemia/lymphomas, 90.6% of central nervous system (CNS) tumors, and 62.6% of non-CNS solid tumors included in this cohort. A change in diagnosis as a result of testing occurred in 3.3% of cases. Additionally, 19.4% of all patients had variants requiring further evaluation for potential germline alteration. Conclusions: Use of somatic NGS panel testing resulted in a significant impact on clinical care, including diagnosis, prognosis, and treatment planning in 78.7% of pediatric patients tested in our institution. Somatic NGS tumor testing should be implemented as part of the routine diagnostic workup of newly diagnosed and relapsed pediatric cancer patients. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
15. PRIMA-1MET-induced neuroblastoma cell death is modulated by p53 and mycn through glutathione level.
- Author
-
Mlakar, Vid, Jurkovic Mlakar, Simona, Lesne, Laurence, Marino, Denis, Rathi, Komal S., Maris, John M., Ansari, Marc, and Gumy-Pause, Fabienne
- Subjects
CELL death ,REDUCTASES ,CYSTEINE ,CELL cycle - Abstract
Background: Neuroblastoma is the most common extracranial solid tumor in children. This cancer has a low frequency of TP53 mutations and its downstream pathway is usually intact. This study assessed the efficacy of the p53 activator, PRIMA-1
MET , in inducing neuroblastoma cell death. Methods: CellTiter 2.0 was used to study susceptibility and specificity of NB cell lines to PRIMA-1MET . Real-time PCR and western blot were used to assess the most common p53 transactivation targets. Induction of p53 and Noxa, and inhibition of Cas3/7, were used to assess impact on cell death after PRIMA-1MET treatment. Flow cytometry was used to analyze cell cycle phase and induction of apoptosis, reactive oxygen species, and the collapse of mitochondrial membrane potential. Results: Neuroblastoma cell lines were at least four times more susceptible to PRIMA-1MET than were primary fibroblasts and keratinocyte cell lines. PRIMA-1MET induced cell death rapidly and in all cell cycle phases. Although PRIMA-1MET activated p53 transactivation activity, p53's role is likely limited because its main targets remained unaffected, whereas pan-caspase inhibitor demonstrated no ability to prevent cell death. PRIMA-1MET induced oxidative stress and modulated the methionine/cysteine/glutathione axis. Variations of MYCN and p53 modulated intracellular levels of GSH and resulted in increased/decreased sensitivity of PRIMA-1MET . PRIMA-1MET inhibited thioredoxin reductase, but the effect of PRIMA-1MET was not altered by thioredoxin inhibition. Conclusions: PRIMA-1MET could be a promising new agent to treat neuroblastoma because it demonstrated good anti-tumor action. Although p53 is involved in PRIMA-1MET -mediated cell death, our results suggest that direct interaction with p53 has a limited role in neuroblastoma but rather acts through modulation of GSH levels. [ABSTRACT FROM AUTHOR]- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.