9 results on '"Jasmine L. May"'
Search Results
2. IDH3α regulates one-carbon metabolism in glioblastoma
- Author
-
Jasmine L. May, Serena Tommasini-Ghelfi, Alexander H. Stegh, Peng Gao, Foteini Kouri, Andrew Lee, Andrea E. Calvert, Navdeep S. Chandel, Yanrong Ji, Craig Horbinski, Juan Liu, Jason W. Locasale, Lisa A. Hurley, and Ramana V. Davuluri
- Subjects
IDH1 ,Citric Acid Cycle ,Mice, SCID ,Transfection ,medicine.disease_cause ,IDH2 ,Oxidative Phosphorylation ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,Cytosol ,0302 clinical medicine ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Epigenetics ,neoplasms ,Research Articles ,Cancer ,030304 developmental biology ,Glycine Hydroxymethyltransferase ,0303 health sciences ,Mutation ,Multidisciplinary ,Methionine ,Brain Neoplasms ,DNA replication ,SciAdv r-articles ,Life Sciences ,DNA Methylation ,Isocitrate Dehydrogenase ,nervous system diseases ,3. Good health ,Cell biology ,HEK293 Cells ,chemistry ,030220 oncology & carcinogenesis ,Serine hydroxymethyltransferase ,S Phase Cell Cycle Checkpoints ,DNA methylation ,Heterografts ,Female ,Glioblastoma ,Research Article - Abstract
IDH3α promotes glioblastoma progression and links mitochondrial metabolism to cSHMT-controlled one-carbon metabolism., Mutation or transcriptional up-regulation of isocitrate dehydrogenases 1 and 2 (IDH1 and IDH2) promotes cancer progression through metabolic reprogramming and epigenetic deregulation of gene expression. Here, we demonstrate that IDH3α, a subunit of the IDH3 heterotetramer, is elevated in glioblastoma (GBM) patient samples compared to normal brain tissue and promotes GBM progression in orthotopic glioma mouse models. IDH3α loss of function reduces tricarboxylic acid (TCA) cycle turnover and inhibits oxidative phosphorylation. In addition to its impact on mitochondrial energy metabolism, IDH3α binds to cytosolic serine hydroxymethyltransferase (cSHMT). This interaction enhances nucleotide availability during DNA replication, while the absence of IDH3α promotes methionine cycle activity, S-adenosyl methionine generation, and DNA methylation. Thus, the regulation of one-carbon metabolism via an IDH3α-cSHMT signaling axis represents a novel mechanism of metabolic adaptation in GBM.
- Published
- 2019
- Full Text
- View/download PDF
3. Cancer-associated mutation and beyond: The emerging biology of isocitrate dehydrogenases in human disease
- Author
-
Alexander H. Stegh, Jasmine L. May, Serena Tommasini-Ghelfi, Fotini M. Kouri, Akanksha Mahajan, and Kevin Murnan
- Subjects
Cytoplasm ,IDH1 ,Mutant ,Citric Acid Cycle ,Reviews ,Review ,Biology ,medicine.disease_cause ,IDH2 ,Epigenesis, Genetic ,Glutarates ,03 medical and health sciences ,Inhibitory Concentration 50 ,Mice ,0302 clinical medicine ,Dioxygenase ,Catalytic Domain ,Neoplasms ,medicine ,Animals ,Homeostasis ,Humans ,030304 developmental biology ,chemistry.chemical_classification ,0303 health sciences ,Mutation ,Multidisciplinary ,Neurodegenerative Diseases ,Cell Biology ,DNA Methylation ,Isocitrate Dehydrogenase ,3. Good health ,Cell biology ,Mitochondria ,Citric acid cycle ,Enzyme ,Phenotype ,chemistry ,030220 oncology & carcinogenesis ,Immune System ,NAD+ kinase ,SciAdv reviews ,Oxidation-Reduction ,Allosteric Site ,NADP - Abstract
We review the contribution of altered isocitrate dehydrogenase activity to the pathogenesis of a broad spectrum of human disease., Isocitrate dehydrogenases (IDHs) are critical metabolic enzymes that catalyze the oxidative decarboxylation of isocitrate to α-ketoglutarate (αKG), NAD(P)H, and CO2. IDHs epigenetically control gene expression through effects on αKG-dependent dioxygenases, maintain redox balance and promote anaplerosis by providing cells with NADPH and precursor substrates for macromolecular synthesis, and regulate respiration and energy production through generation of NADH. Cancer-associated mutations in IDH1 and IDH2 represent one of the most comprehensively studied mechanisms of IDH pathogenic effect. Mutant enzymes produce (R)-2-hydroxyglutarate, which in turn inhibits αKG-dependent dioxygenase function, resulting in a global hypermethylation phenotype, increased tumor cell multipotency, and malignancy. Recent studies identified wild-type IDHs as critical regulators of normal organ physiology and, when transcriptionally induced or down-regulated, as contributing to cancer and neurodegeneration, respectively. We describe how mutant and wild-type enzymes contribute on molecular levels to disease pathogenesis, and discuss efforts to pharmacologically target IDH-controlled metabolic rewiring.
- Published
- 2018
4. Cancer-associated IDH1 promotes growth and resistance to targeted therapies in the absence of mutation
- Author
-
Fotini M. Kouri, Yingtao Bi, Alexandra Chalastanis, Ramana V. Davuluri, Elizabeth T. Bartom, Gary E. Schiltz, Youjia Hua, Alexander H. Stegh, Lisa A. Hurley, Andrea E. Calvert, Hongwu Zheng, Marcus E. Peter, Yongfei Wu, Craig Horbinski, Andrew P. Mazar, Rama K. Mishra, Jasmine L. May, C. David James, Maureen Kachman, Charles F. Burant, Navdeep S. Chandel, and Oleksii Dubrovskyi
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Apoptosis ,Mice, SCID ,Targeted therapy ,Histones ,Mice ,Histone methylation ,Molecular Targeted Therapy ,lcsh:QH301-705.5 ,Cell Differentiation ,Forkhead Transcription Factors ,differentiation ,targeted therapy ,Lipids ,Isocitrate Dehydrogenase ,Gene Expression Regulation, Neoplastic ,wild-type IDH1 ,Isocitrate dehydrogenase ,Gene Knockdown Techniques ,Disease Progression ,Neoplastic Stem Cells ,Ketoglutaric Acids ,IDH1 ,EGFR ,Biology ,reactive oxygen species (ROS) ,GBM ,IDH2 ,Methylation ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Erlotinib Hydrochloride ,Glioma ,NADPH ,medicine ,Animals ,RNA, Messenger ,Protein Kinase Inhibitors ,Cell Proliferation ,Oncogene ,Cell growth ,medicine.disease ,Molecular biology ,030104 developmental biology ,lcsh:Biology (General) ,Drug Resistance, Neoplasm ,Mutation ,Cancer research ,Glioblastoma ,Reactive Oxygen Species ,metabolism ,NADP - Abstract
Oncogenic mutations in two isocitrate dehydrogenase (IDH)-encoding genes (IDH1 and IDH2) have been identified in acute myelogenous leukemia, low-grade glioma, and secondary glioblastoma (GBM). Our in silico and wet-bench analyses indicate that non-mutated IDH1 mRNA and protein are commonly overexpressed in primary GBMs. We show that genetic and pharmacologic inactivation of IDH1 decreases GBM cell growth, promotes a more differentiated tumor cell state, increases apoptosis in response to targeted therapies, and prolongs the survival of animal subjects bearing patient-derived xenografts (PDXs). On a molecular level, diminished IDH1 activity results in reduced α-ketoglutarate (αKG) and NADPH production, paralleled by deficient carbon flux from glucose or acetate into lipids, exhaustion of reduced glutathione, increased levels of reactive oxygen species (ROS), and enhanced histone methylation and differentiation marker expression. These findings suggest that IDH1 upregulation represents a common metabolic adaptation by GBMs to support macromolecular synthesis, aggressive growth, and therapy resistance.
- Published
- 2017
5. Dual bioluminescence and near-infrared fluorescence monitoring to evaluate spherical nucleic acid nanoconjugate activity in vivo
- Author
-
Stacey N. Barnaby, Chad A. Mirkin, Jasmine L. May, Anthony J. Sprangers, Nikunjkumar Savalia, Alexander H. Stegh, Serena T. Ghelfi, Lisa E. Cole, Timothy L. Sita, Thomas C. Cayton, Timothy J. Merkel, Lisa A. Hurley, Andrew Lee, Fotini M. Kouri, Alexandra Chalastanis, and Charles David James
- Subjects
0301 basic medicine ,Biodistribution ,Mice, SCID ,Nanoconjugates ,Biology ,Fluorescence ,03 medical and health sciences ,Mice ,0302 clinical medicine ,In vivo ,RNA interference ,medicine ,Temozolomide ,Tumor Cells, Cultured ,Gene silencing ,Animals ,Humans ,RNA, Small Interfering ,neoplasms ,Antineoplastic Agents, Alkylating ,DNA Modification Methylases ,Gene knockdown ,Multidisciplinary ,Brain Neoplasms ,Tumor Suppressor Proteins ,Biological Sciences ,Molecular biology ,Xenograft Model Antitumor Assays ,Dacarbazine ,030104 developmental biology ,DNA Repair Enzymes ,030220 oncology & carcinogenesis ,Spherical nucleic acid ,Female ,RNA Interference ,Glioblastoma ,Ex vivo ,medicine.drug - Abstract
RNA interference (RNAi)-based gene regulation platforms have shown promise as a novel class of therapeutics for the precision treatment of cancer. Techniques in preclinical evaluation of RNAi-based nanoconjugates have yet to allow for optimization of their gene regulatory activity. We have developed spherical nucleic acids (SNAs) as a blood-brain barrier-/blood-tumor barrier-penetrating nanoconjugate to deliver small interfering (si) and micro (mi)RNAs to intracranial glioblastoma (GBM) tumor sites. To identify high-activity SNA conjugates and to determine optimal SNA treatment regimens, we developed a reporter xenograft model to evaluate SNA efficacy in vivo. Engrafted tumors stably coexpress optical reporters for luciferase and a near-infrared (NIR) fluorescent protein (iRFP670), with the latter fused to the DNA repair protein O6-methylguanine-DNA-methyltransferase (MGMT). Using noninvasive imaging of animal subjects bearing reporter-modified intracranial xenografts, we quantitatively assessed MGMT knockdown by SNAs composed of MGMT-targeting siRNA duplexes (siMGMT-SNAs). We show that systemic administration of siMGMT-SNAs via single tail vein injection is capable of robust intratumoral MGMT protein knockdown in vivo, with persistent and SNA dose-dependent MGMT silencing confirmed by Western blotting of tumor tissue ex vivo. Analyses of SNA biodistribution and pharmacokinetics revealed rapid intratumoral uptake and significant intratumoral retention that increased the antitumor activity of coadministered temozolomide (TMZ). Our study demonstrates that dual noninvasive bioluminescence and NIR fluorescence imaging of cancer xenograft models represents a powerful in vivo strategy to identify RNAi-based nanotherapeutics with potent gene silencing activity and will inform additional preclinical and clinical investigations of these constructs.
- Published
- 2017
6. Bioengineering Silicon Quantum Dot Theranostics using a Network Analysis of Metabolomic and Proteomic Data in Cardiac Ischemia
- Author
-
Nikolas Kasabov, Paras N. Prasad, Folarin Erogbogbo, Patrick Gladding, Michelle Jamieson, Seif El Jack, Dariusz Korcyk, Banu Gopalan, Katie Smart, Mark T. Swihart, Mark Webster, Stefan Schliebs, Raphael Hu, Mia Jüllig, Ralph A.H. Stewart, Katherine Bakeev, Linda Liang, Silas G. Villas-Boas, Irene Zeng, and Jasmine L. May
- Subjects
theranostics ,Silicon ,Proteome ,medicine.medical_treatment ,Myocardial Ischemia ,Medicine (miscellaneous) ,Bioengineering ,Bioinformatics ,Gas Chromatography-Mass Spectrometry ,Plasma ,Metabolomics ,Tandem Mass Spectrometry ,Quantum Dots ,medicine ,Humans ,Myocardial infarction ,Biomarker discovery ,Pharmacology, Toxicology and Pharmaceutics (miscellaneous) ,Coronary sinus ,Chemistry ,Percutaneous coronary intervention ,silicon quantum dots ,medicine.disease ,metabolomics ,cardiac ischemia ,Coronary occlusion ,Metabolome ,Ischemic preconditioning ,myocardial infarction ,Reperfusion injury ,Biomarkers ,Research Paper ,Chromatography, Liquid - Abstract
Metabolomic profiling is ideally suited for the analysis of cardiac metabolism in healthy and diseased states. Here, we show that systematic discovery of biomarkers of ischemic preconditioning using metabolomics can be translated to potential nanotheranostics. Thirty-three patients underwent percutaneous coronary intervention (PCI) after myocardial infarction. Blood was sampled from catheters in the coronary sinus, aorta and femoral vein before coronary occlusion and 20 minutes after one minute of coronary occlusion. Plasma was analysed using GC-MS metabolomics and iTRAQ LC-MS/MS proteomics. Proteins and metabolites were mapped into the Metacore network database (GeneGo, MI, USA) to establish functional relevance. Expression of 13 proteins was significantly different (p
- Published
- 2013
7. Distribution of Iron Oxide Core-Titanium Dioxide Shell Nanoparticles in VX2 Tumor Bearing Rabbits Introduced by Two Different Delivery Modalities
- Author
-
Kathleen R. Harris, Reed A. Omary, Tamer Refaat, William Liu, Michael Beau Wanzer, Lydia Finney, Tatjana Paunesku, Derek L. West, Samar El Achy, Daniele Procissi, Stefan Vogt, Jasmine L. May, Lun Xin, Gayle E. Woloschak, Vamsi Parimi, Andrew C. Larson, and Evan Maxey
- Subjects
Materials science ,core–shell nanoparticle ,General Chemical Engineering ,Nanoparticle ,Nanotechnology ,Spleen ,rabbit VX2 liver cancer model ,Article ,030218 nuclear medicine & medical imaging ,lcsh:Chemistry ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,medicine ,Fluorescence microscope ,Distribution (pharmacology) ,General Materials Science ,transarterial intra-catheter delivery ,Kidney ,medicine.disease ,3. Good health ,medicine.anatomical_structure ,lcsh:QD1-999 ,Permeability (electromagnetism) ,030220 oncology & carcinogenesis ,Cancer research ,Liver cancer - Abstract
This work compares intravenous (IV) versus fluoroscopy-guided transarterial intra-catheter (IC) delivery of iron oxide core-titanium dioxide shell nanoparticles (NPs) in vivo in VX2 model of liver cancer in rabbits. NPs coated with glucose and decorated with a peptide sequence from cortactin were administered to animals with developed VX2 liver cancer. Two hours after NPs delivery tumors, normal liver, kidney, lung and spleen tissues were harvested and used for a series on histological and elemental analysis tests. Quantification of NPs in tissues was done both by bulk inductively coupled plasma mass spectrometry (ICP-MS) analysis and by hard X-ray fluorescence microscopy. Both IV and IC NPs injection are feasible modalities for delivering NPs to VX2 liver tumors with comparable tumor accumulation. It is possible that this is an outcome of the fact that VX2 tumors are highly vascularized and hemorrhagic, and therefore enhanced permeability and retention (EPR) plays the most significant role in accumulation of nanoparticles in tumor tissue. It is, however, interesting to note that IV delivery led to increased sequestration of NPs by spleen and normal liver tissue, while IC delivery lead to more NP positive Kupffer cells. This difference is most likely a direct outcome of blood flow dynamics. Armed with this knowledge about nanoparticle delivery, we plan to test them as radiosensitizers in the future.
- Published
- 2016
- Full Text
- View/download PDF
8. Enhancing silicon quantum dot uptake by pancreatic cancer cells via pluronic® encapsulation and antibody targeting
- Author
-
Hong Ding, Ken-Tye Yong, Jasmine L. May, Paras N. Prasad, Wing Cheung Law, Mark T. Swihart, and Folarin Erogbogbo
- Subjects
chemistry.chemical_classification ,Cancer Research ,business.industry ,Quantum yield ,Nanotechnology ,Polymer ,Poloxamer ,Oncology ,chemistry ,Dynamic light scattering ,Zeta potential ,Copolymer ,Medicine ,Surface modification ,Radiology, Nuclear Medicine and imaging ,business ,Luminescence - Abstract
Objectives: Silicon quantum dots (SiQDs) are of great interest for bio - imaging applications due to their tunable luminescence, low toxicity, unique surface chemistry, and high quantum yield. Most synthesis routes produce SiQDs that are not water-dispersible, making them unattractive for biological applications. Here, we show that Pluronic® block copolymers can encapsulate SiQDs to make them water dispersible and suitable for cancer imaging applications. Methods: Transmission electron microscopy (TEM), dynamic light scattering (DLS), zeta potential, and temperature and pH stability measurements were used to evaluate these Pluronic®-encapsulated SiQDs (PSiQDs). The particles were also tested targeted in vitro imaging and in vivo bio - distribution. Results: Encapsulation with Pluronic® polymers renders the SiQDs water dispersible, preserves their optical properties, protects them from oxidation, and prevents aggregation. Surface modification of the PSiQDs with pancreatic cancer targeting moieties, anti-claudin-4 and anti-mesothelin, led to enhanced uptake of these nanoconstructs in comparison to PSiQDs modified with folate as the targeting moeity. Conclusions: The particles are stable at biological conditions, and show promise for targeted diagnostic applications without possessing elementally toxic components.
- Published
- 2012
- Full Text
- View/download PDF
9. The study of in vivo acute effect of two different delivery modalities of iron oxide core with titanium dioxide shell nanoparticles in rabbits liver tumor
- Author
-
Jasmine L. May, Tamer Refaat, Tatjana Paunesku, Reed A. Omary, Andrew C. Larson, Gayle E. Woloschak, Vamsi Parimi, Kathleen R. Harris, Derek L. West, and Samar Elachy
- Subjects
inorganic chemicals ,Cancer Research ,Liver tumor ,business.industry ,technology, industry, and agriculture ,Iron oxide ,Shell (structure) ,Nanoparticle ,Core (manufacturing) ,Acute effect ,respiratory system ,medicine.disease ,chemistry.chemical_compound ,Oncology ,chemistry ,In vivo ,Titanium dioxide ,medicine ,business ,health care economics and organizations ,Nuclear chemistry - Abstract
e22204 Background: To compare intravenous (IV) versus fluoroscopy-guided transarterial intra-catheter (IC) delivery of iron oxide core with titanium dioxide shell nanoparticles (NPs) in vivo in VX2 model of liver cancer in rabbits and detect the NPs distribution and effect of NPs presence on the target tumor and other rabbit’s organs. Methods: After obtaining the IACUC approval, liver tumors were obtained by implantation of tumor tissue obtained from a hind limb VX2 tumor of donor rabbits. NPs were delivered either IV or IC. After rabbit termination, 2 hours post NPs injection, tumor, liver, kidney, lung and spleen were harvested, split in half and a part of it was frozen while the remainder was formalin fixed and paraffin embedded. To assess the NPs distribution in 2D we stained 5um thick paraffin tissue sections using Dopamine-Biotin-DHS histochemical (HC) staining followed by Nanozoomer microscopy analysis. H and E staining, TUNEL assay and Ki67 immunohistochemistry were also done. X-ray Fluorescence Microscopy (XFM) was used to quantify the NPs. Frozen tissue was used for bulk NPs concentration analysis using Inductively Coupled Plasma Mass Spectrometry (ICP-MS). Results: This study included ten rabbits; 3 rabbits had IV NPs injections, 3 had IC NPs injection, 2 control and 2 donors. ICP-MS analysis showed statistically significant higher NPs concentration in tumors of IC arm vs. IV arm (p= 0.0356), while there was higher concentration of NPs in liver (p=0.00077) and spleen (p = 0.01356) of IV vs. IC arms but no difference in kidneys or lungs. These findings were consistent with results from HC and XFM analyses. HC 2D analysis of NPs distribution showed that the RES have taken up the NPs non-specifically. There were no statistically significant differences between the treatment groups regarding the Ki67 proliferation or the TUNEL apoptosis indices or when control rabbits were compared to NPs treated rabbits. Conclusions: Both IV and IC NPs injection are feasible modalities for delivering NPs to tumors with acceptable acute systemic effects and comparable tumor effect. IV delivery increased sequestration of the NPs by RES and their accumulation in spleen and liver.
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.