Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 4862, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38862464

ABSTRACT

As spaceflight becomes more common with commercial crews, blood-based measures of crew health can guide both astronaut biomedicine and countermeasures. By profiling plasma proteins, metabolites, and extracellular vesicles/particles (EVPs) from the SpaceX Inspiration4 crew, we generated "spaceflight secretome profiles," which showed significant differences in coagulation, oxidative stress, and brain-enriched proteins. While >93% of differentially abundant proteins (DAPs) in vesicles and metabolites recovered within six months, the majority (73%) of plasma DAPs were still perturbed post-flight. Moreover, these proteomic alterations correlated better with peripheral blood mononuclear cells than whole blood, suggesting that immune cells contribute more DAPs than erythrocytes. Finally, to discern possible mechanisms leading to brain-enriched protein detection and blood-brain barrier (BBB) disruption, we examined protein changes in dissected brains of spaceflight mice, which showed increases in PECAM-1, a marker of BBB integrity. These data highlight how even short-duration spaceflight can disrupt human and murine physiology and identify spaceflight biomarkers that can guide countermeasure development.


Subject(s)
Blood Coagulation , Blood-Brain Barrier , Brain , Homeostasis , Oxidative Stress , Space Flight , Animals , Humans , Brain/metabolism , Blood-Brain Barrier/metabolism , Mice , Blood Coagulation/physiology , Male , Secretome/metabolism , Mice, Inbred C57BL , Extracellular Vesicles/metabolism , Proteomics/methods , Biomarkers/metabolism , Biomarkers/blood , Female , Adult , Blood Proteins/metabolism , Middle Aged , Leukocytes, Mononuclear/metabolism , Proteome/metabolism
2.
Biofactors ; 49(4): 912-927, 2023.
Article in English | MEDLINE | ID: mdl-37171157

ABSTRACT

The liver is the only solid organ capable of regenerating itself to regain 100% of its mass and function after liver injury and/or partial hepatectomy (PH). This exceptional property represents a therapeutic opportunity for severe liver disease patients. However, liver regeneration (LR) might fail due to poorly understood causes. Here, we have investigated the regulation of liver proteome and phosphoproteome at a short time after PH (9 h), to depict a detailed mechanistic background of the early LR phase. Furthermore, we analyzed the dynamic changes of the serum proteome and metabolome of healthy living donor liver transplant (LDLT) donors at different time points after surgery. The molecular profiles from both analyses were then correlated. Insulin and FXR-FGF15/19 signaling were stimulated in mouse liver after PH, leading to the activation of the main intermediary kinases (AKT and ERK). Besides, inhibition of the hippo pathway led to an increased expression of its target genes and of one of its intermediary proteins (14-3-3 protein), contributing to cell proliferation. In association with these processes, metabolic reprogramming coupled to enhanced mitochondrial activity cope for the energy and biosynthetic requirements of LR. In human serum of LDLT donors, we identified 56 proteins and 13 metabolites statistically differential which recapitulate some of the main cellular processes orchestrating LR in its early phase. These results provide mechanisms and protein mediators of LR that might prove useful for the follow-up of the regenerative process in the liver after PH as well as preventing the occurrence of complications associated with liver resection.


Subject(s)
Liver Regeneration , Liver Transplantation , Mice , Animals , Humans , Liver Regeneration/genetics , Liver Transplantation/methods , Proteome/genetics , Proteome/metabolism , Living Donors , Liver/surgery , Liver/metabolism
3.
Proc Natl Acad Sci U S A ; 118(37)2021 09 14.
Article in English | MEDLINE | ID: mdl-34497125

ABSTRACT

Absolute quantification measurements (copies per cell) of peptide major histocompatibility complex (pMHC) antigens are necessary to inform targeted immunotherapy drug design; however, existing methods for absolute quantification have critical limitations. Here, we present a platform termed SureQuant-IsoMHC, utilizing a series of pMHC isotopologues and internal standard-triggered targeted mass spectrometry to generate an embedded multipoint calibration curve to determine endogenous pMHC concentrations for a panel of 18 tumor antigens. We apply SureQuant-IsoMHC to measure changes in expression of our target panel in a melanoma cell line treated with a MEK inhibitor and translate this approach to estimate antigen concentrations in melanoma tumor biopsies.


Subject(s)
Antigen Presentation/immunology , Antigens, Neoplasm/analysis , Benzimidazoles/pharmacology , Histocompatibility Antigens Class I/immunology , MAP Kinase Kinase 1/antagonists & inhibitors , Melanoma/immunology , Antigen Presentation/drug effects , Antigens, Neoplasm/drug effects , Antigens, Neoplasm/immunology , Histocompatibility Antigens Class I/metabolism , Humans , Immunotherapy , Melanoma/drug therapy , Melanoma/metabolism , Tumor Cells, Cultured
4.
Cancer Res ; 81(9): 2495-2509, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33509940

ABSTRACT

Tyrosine phosphorylation (pTyr) plays a pivotal role in signal transduction and is commonly dysregulated in cancer. As a result, profiling tumor pTyr levels may reveal therapeutic insights critical to combating disease. Existing discovery and targeted mass spectrometry-based methods used to monitor pTyr networks involve a tradeoff between broad coverage of the pTyr network, reproducibility in target identification across analyses, and accurate quantification. To address these limitations, we developed a targeted approach, termed "SureQuant pTyr," coupling low input pTyr enrichment with a panel of isotopically labeled internal standard peptides to guide data acquisition of low-abundance tyrosine phosphopeptides. SureQuant pTyr allowed for reliable quantification of several hundred commonly dysregulated pTyr targets with high quantitative accuracy, improving the robustness and usability of targeted mass spectrometry assays. We established the clinical applicability of SureQuant pTyr by profiling pTyr signaling levels in human colorectal tumors using minimal sample input, characterizing patient-specific oncogenic-driving mechanisms. While in some cases pTyr profiles aligned with previously reported proteomic, genomic, and transcriptomic molecular characterizations, we highlighted instances of new insights gained using pTyr characterization and emphasized the complementary nature of pTyr measurements with traditional biomarkers for improving patient stratification and identifying therapeutic targets. The turn-key nature of this approach opens the door to rapid and reproducible pTyr profiling in research and clinical settings alike and enables pTyr-based measurements for applications in precision medicine. SIGNIFICANCE: SureQuant pTyr is a mass spectrometry-based targeted method that enables sensitive and selective targeted quantitation of several hundred low-abundance tyrosine phosphorylated peptides commonly dysregulated in cancer, including oncogenic signaling networks.


Subject(s)
Colorectal Neoplasms/metabolism , Protein Processing, Post-Translational , Proteome/analysis , Signal Transduction , Tyrosine/metabolism , A549 Cells , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Chromatography, Liquid/methods , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , ErbB Receptors/metabolism , Humans , Mass Spectrometry/methods , Phosphopeptides/analysis , Phosphopeptides/metabolism , Phosphorylation , Protein Interaction Maps , Proteome/metabolism , Proteomics/methods
5.
Sci Signal ; 12(565)2019 01 22.
Article in English | MEDLINE | ID: mdl-30670633

ABSTRACT

Drug-induced compensatory signaling and subsequent rewiring of the signaling pathways that support cell proliferation and survival promote the development of acquired drug resistance in tumors. Here, we sought to analyze the adaptive kinase response in cancer cells after distinct treatment with agents targeting human epidermal growth factor receptor 2 (HER2), specifically those that induce either only temporary cell cycle arrest or, alternatively, apoptosis in HER2-overexpressing cancers. We compared trastuzumab, ARRY380, the combination thereof, and a biparatopic, HER2-targeted designed ankyrin repeat protein (DARPin; specifically, 6L1G) and quantified the phosphoproteome by isobaric tagging using tandem mass tag liquid chromatography/tandem mass spectrometry (TMT LC-MS/MS). We found a specific signature of persistently phosphorylated tyrosine peptides after the nonapoptotic treatments, which we used to distinguish between different treatment-induced cancer cell fates. Next, we analyzed the activation of serine/threonine and tyrosine kinases after treatment using a bait peptide chip array and predicted the corresponding active kinases. Through a combined system-wide analysis, we identified a common adaptive kinase response program that involved the activation of focal adhesion kinase 1 (FAK1), protein kinase C-δ (PRKCD), and Ephrin (EPH) family receptors. These findings reveal potential targets to prevent adaptive resistance to HER2-targeted therapies.


Subject(s)
Breast Neoplasms/metabolism , Protein Kinases/metabolism , Receptor, ErbB-2/antagonists & inhibitors , Signal Transduction/drug effects , Small Molecule Libraries/pharmacology , Trastuzumab/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Breast Neoplasms/pathology , Cell Line, Tumor , Chromatography, Liquid , Drug Resistance, Neoplasm/drug effects , Female , Humans , Phosphoproteins/metabolism , Protein Kinase Inhibitors/pharmacology , Proteome/metabolism , Receptor, ErbB-2/metabolism , Tandem Mass Spectrometry
6.
Proc Natl Acad Sci U S A ; 113(11): 3114-9, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26929352

ABSTRACT

Despite extensive study of the EGF receptor (EGFR) signaling network, the immediate posttranslational changes that occur in response to growth factor stimulation remain poorly characterized; as a result, the biological mechanisms underlying signaling initiation remain obscured. To address this deficiency, we have used a mass spectrometry-based approach to measure system-wide phosphorylation changes throughout the network with 10-s resolution in the 80 s after stimulation in response to a range of eight growth factor concentrations. Significant changes were observed on proteins far downstream in the network as early as 10 s after stimulation, indicating a system capable of transmitting information quickly. Meanwhile, canonical members of the EGFR signaling network fall into clusters with distinct activation patterns. Src homology 2 domain containing transforming protein (Shc) and phosphoinositol 3-kinase (PI3K) phosphorylation levels increase rapidly, but equilibrate within 20 s, whereas proteins such as Grb2-associated binder-1 (Gab1) and SH2-containing tyrosine phosphatase (SHP2) show slower, sustained increases. Proximity ligation assays reveal that Shc and Gab1 phosphorylation patterns are representative of separate timescales for physical association with the receptor. Inhibition of phosphatases with vanadate reveals site-specific regulatory mechanisms and also uncovers primed activating components in the network, including Src family kinases, whose inhibition affects only a subset of proteins within the network. The results presented highlight the complexity of signaling initiation and provide a window into exploring mechanistic hypotheses about receptor tyrosine kinase (RTK) biology.


Subject(s)
Breast/enzymology , ErbB Receptors/physiology , Signal Transduction/physiology , Breast/cytology , Cell Division , Cell Line , Culture Media, Serum-Free/pharmacology , Enzyme Activation/drug effects , Epidermal Growth Factor/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/enzymology , ErbB Receptors/agonists , Female , Humans , Multiprotein Complexes , Phosphoprotein Phosphatases/physiology , Phosphoproteins/analysis , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Time Factors , src-Family Kinases/physiology
7.
Cancer Res ; 75(7): 1495-503, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25670172

ABSTRACT

Tumor protein phosphorylation analysis may provide insight into intracellular signaling networks underlying tumor behavior, revealing diagnostic, prognostic or therapeutic information. Human tumors collected by The Cancer Genome Atlas program potentially offer the opportunity to characterize activated networks driving tumor progression, in parallel with the genetic and transcriptional landscape already documented for these tumors. However, a critical question is whether cellular signaling networks can be reliably analyzed in surgical specimens, where freezing delays and spatial sampling disparities may potentially obscure physiologic signaling. To quantify the extent of these effects, we analyzed the stability of phosphotyrosine (pTyr) sites in ovarian and colon tumors collected under conditions of controlled ischemia and in the context of defined intratumoral sampling. Cold-ischemia produced a rapid, unpredictable, and widespread impact on tumor pTyr networks within 5 minutes of resection, altering up to 50% of pTyr sites by more than 2-fold. Effects on adhesion and migration, inflammatory response, proliferation, and stress response pathways were recapitulated in both ovarian and colon tumors. In addition, sampling of spatially distinct colon tumor biopsies revealed pTyr differences as dramatic as those associated with ischemic times, despite uniform protein expression profiles. Moreover, intratumoral spatial heterogeneity and pTyr dynamic response to ischemia varied dramatically between tumors collected from different patients. Overall, these findings reveal unforeseen phosphorylation complexity, thereby increasing the difficulty of extracting physiologically relevant pTyr signaling networks from archived tissue specimens. In light of this data, prospective tumor pTyr analysis will require appropriate sampling and collection protocols to preserve in vivo signaling features.


Subject(s)
Phosphotyrosine/metabolism , Artifacts , Cell Hypoxia , Colorectal Neoplasms/metabolism , Female , Humans , Ovarian Neoplasms/metabolism , Phosphorylation , Prospective Studies , Protein Processing, Post-Translational , Signal Transduction
8.
Cancer Discov ; 4(10): 1198-213, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25100205

ABSTRACT

UNLABELLED: Alkylating agents are a first-line therapy for the treatment of several aggressive cancers, including pediatric glioblastoma, a lethal tumor in children. Unfortunately, many tumors are resistant to this therapy. We sought to identify ways of sensitizing tumor cells to alkylating agents while leaving normal cells unharmed, increasing therapeutic response while minimizing toxicity. Using an siRNA screen targeting over 240 DNA damage response genes, we identified novel sensitizers to alkylating agents. In particular, the base excision repair (BER) pathway, including 3-methylpurine-DNA glycosylase (MPG), as well as ataxia telangiectasia mutated (ATM), were identified in our screen. Interestingly, we identified MPG as a direct novel substrate of ATM. ATM-mediated phosphorylation of MPG was required for enhanced MPG function. Importantly, combined inhibition or loss of MPG and ATM resulted in increased alkylating agent-induced cytotoxicity in vitro and prolonged survival in vivo. The discovery of the ATM-MPG axis will lead to improved treatment of alkylating agent-resistant tumors. SIGNIFICANCE: Inhibition of ATM and MPG-mediated BER cooperate to sensitize tumor cells to alkylating agents, impairing tumor growth in vitro and in vivo with no toxicity to normal cells, providing an ideal therapeutic window.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Ataxia Telangiectasia Mutated Proteins/metabolism , DNA Glycosylases/metabolism , Drug Resistance, Neoplasm , Age Factors , Animals , Cell Line, Tumor , Cluster Analysis , DNA Copy Number Variations , DNA Repair , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Enzyme Activation , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/mortality , Glioblastoma/pathology , Humans , Models, Biological , Phosphorylation , RNA Interference , RNA, Small Interfering/genetics , Temozolomide , Xenograft Model Antitumor Assays
9.
Curr Opin Biotechnol ; 28: 83-7, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24441143

ABSTRACT

Attempts to characterize cellular behaviors with static, univariate measurements cannot fully capture biological complexity and lead to an inadequate interpretation of cellular processes. Significant biological insight can be gleaned by considering the contribution of dynamic protein post-translational modifications (PTMs) utilizing systems-level quantitative analysis. High-resolution mass spectrometry coupled with computational modeling of dynamic signal-response relationships is a powerful tool to reveal PTM-mediated regulatory networks. Recent advances using this approach have defined network kinetics of growth factor signaling pathways, identified systems level responses to cytotoxic perturbations, elucidated kinase-substrate relationships, and unraveled the dynamics of PTM cross-talk. Innovations in multiplex measurement capacity, PTM annotation accuracy, and computational integration of datasets promise enhanced resolution of dynamic PTM networks and further insight into biological intricacies.


Subject(s)
Protein Processing, Post-Translational , Proteins/metabolism , Animals , Humans , Mass Spectrometry , Phosphorylation , Proteins/chemistry , Signal Transduction
10.
Genes Dev ; 27(19): 2147-63, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-24115771

ABSTRACT

Production of haploid gametes from diploid progenitor cells is mediated by a specialized cell division, meiosis, where two divisions, meiosis I and II, follow a single S phase. Errors in progression from meiosis I to meiosis II lead to aneuploid and polyploid gametes, but the regulatory mechanisms controlling this transition are poorly understood. Here, we demonstrate that the conserved kinase Ime2 regulates the timing and order of the meiotic divisions by controlling translation. Ime2 coordinates translational activation of a cluster of genes at the meiosis I-meiosis II transition, including the critical determinant of the meiotic chromosome segregation pattern CLB3. We further show that Ime2 mediates translational control through the meiosis-specific RNA-binding protein Rim4. Rim4 inhibits translation of CLB3 during meiosis I by interacting with the 5' untranslated region (UTR) of CLB3. At the onset of meiosis II, Ime2 kinase activity rises and triggers a decrease in Rim4 protein levels, thereby alleviating translational repression. Our results elucidate a novel developmentally regulated translational control pathway that establishes the meiotic chromosome segregation pattern.


Subject(s)
Chromosome Segregation/genetics , Gene Expression Regulation, Fungal , Meiosis/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , 5' Untranslated Regions/genetics , Intracellular Signaling Peptides and Proteins , Multigene Family/genetics , Protein Binding , Protein Serine-Threonine Kinases , RNA, Messenger/metabolism , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
11.
Mol Cell Proteomics ; 12(2): 499-514, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23211419

ABSTRACT

Numb is an endocytic adaptor protein that regulates the endocytosis and trafficking of transmembrane receptors including Notch, E-cadherin, and integrins. Vertebrate Numb is alternatively spliced at exons 3 and 9 to give rise to four protein isoforms. Expression of these isoforms varies at different developmental stages, and although the function of Numb isoforms containing exon 3 has been studied, the role of exon 9 inclusion has not been shown. Here we use affinity purification and tandem mass spectrometry to identify Numb associated proteins, including novel interactions with REPS1, BMP2K, and BCR. In vitro binding measurements indicated exon 9-independent Numb interaction with REPS1 and Eps15 EH domains. Selected reaction monitoring mass spectrometry was used to quantitatively compare the proteins associated with the p72 and p66 Numb isoforms, which differ by the exon 9 region. This showed that significantly more EPS15 and three AP-2 subunit proteins bound Numb isoforms containing exon 9. The EPS15 preference for exon 9-containing Numb was confirmed in intact cells by using a proximity ligation assay. Finally, we used multiplexed selected reaction monitoring mass spectrometry to assess the dynamic regulation of Numb association with endocytic proteins. Numb hyper-phosphorylation resulted in disassociation of Numb endocytic complexes, while inhibition of endocytosis did not alter Numb association with the AP-2 complex but altered recruitment of EPS15, REPS1, and BMP2K. Hence, quantitative mass spectrometric analysis of Numb protein-protein interactions has provided new insights into the assembly and regulation of protein complexes important in development and cancer.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Carrier Proteins/genetics , Endocytosis/genetics , Kruppel-Like Transcription Factors/genetics , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Proto-Oncogene Proteins c-bcr/genetics , Adaptor Proteins, Signal Transducing/isolation & purification , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Sequence , Animals , Binding Sites , Calcium-Binding Proteins , Carrier Proteins/isolation & purification , Carrier Proteins/metabolism , Chromatography, Liquid , Exons , Gene Expression Regulation , HEK293 Cells , Humans , Kruppel-Like Transcription Factors/isolation & purification , Kruppel-Like Transcription Factors/metabolism , Membrane Proteins/isolation & purification , Membrane Proteins/metabolism , Mice , Molecular Sequence Data , Nerve Tissue Proteins/isolation & purification , Nerve Tissue Proteins/metabolism , Phosphorylation , Protein Binding , Protein Interaction Mapping , Protein Isoforms/genetics , Protein Isoforms/isolation & purification , Protein Isoforms/metabolism , Proto-Oncogene Proteins c-bcr/isolation & purification , Proto-Oncogene Proteins c-bcr/metabolism , Signal Transduction , Tandem Mass Spectrometry/methods , Transfection
12.
Mol Cancer Res ; 10(3): 428-40, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22232519

ABSTRACT

Aberrations in epidermal growth factor receptor (EGFR/ErbB1) are the most common oncogenic alterations in glioblastoma multiforme (GBM), the most common primary brain tumor. Interactions between wild-type (wt) and mutant EGFRs and their subsequent activation are of biologic and potential therapeutic importance in GBMs. We recently showed that in situ proximity ligation assay (PLA) allows for quantitative evaluation of EGFR dimerization and activation in intact cells. Using this in situ platform, we show the aberrant homo-/heterodimeric properties of EGFRvIII and EGFRc958 mutants, the two most common EGFR mutants in GBMs. In addition, dimer phosphoactivation status could be detected by PLA with superior signal-noise ratio (>17-fold) and sensitivity (>16-fold) than immunofluorescence-based phospho-EGFR measurements. Dimer activation analysis indicated quantitative activation differences of mutant dimers. These aberrant features were not overexpression dependent but appeared independent of cellular expression levels, suggesting inherent properties of the mutant receptors. Moreover, we observed in situ detection of EGFRwt-EGFRvIII heterodimerization in GBM specimens, supporting our cell line observations. Notably, currently used anti-EGFR therapeutics, such as cetuximab, matuzumab, and panitumumab, could effectively block EGFRwt dimerization and activation but did not equally impair EGFRvIII homodimers, EGFRwt-EGFRvIII, or EGFRvIII-EGFRc958 heterodimers. EGFRvIII appears to have intrinsic phosphoactivation independent of dimerization as matuzumab blockade of homodimerization had no effect on receptor phosphorylation levels. These data suggest differences in the dimerization-blocking efficacy of EGFR monoclonal antibodies as mutant EGFR dimer configurations prevalent in GBMs can evade blockade by anti-EGFR treatments. Further studies are warranted to evaluate whether this evasion contributes to poor therapeutic response or resistance.


Subject(s)
ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Glioblastoma/enzymology , Molecular Targeted Therapy , Mutant Proteins/metabolism , Protein Multimerization , Animals , Antibodies, Neoplasm/immunology , Biological Assay , CHO Cells , Cricetinae , Cricetulus , Enzyme Activation , Epitopes/immunology , ErbB Receptors/immunology , Humans , Immunoblotting , Phosphorylation , Signal-To-Noise Ratio
13.
J Clin Invest ; 122(1): 253-66, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22156195

ABSTRACT

Glioblastoma multiforme (GBM) is the most common and lethal of all gliomas. The current standard of care includes surgery followed by concomitant radiation and chemotherapy with the DNA alkylating agent temozolomide (TMZ). O6-methylguanine-DNA methyltransferase (MGMT) repairs the most cytotoxic of lesions generated by TMZ, O6-methylguanine. Methylation of the MGMT promoter in GBM correlates with increased therapeutic sensitivity to alkylating agent therapy. However, several aspects of TMZ sensitivity are not explained by MGMT promoter methylation. Here, we investigated our hypothesis that the base excision repair enzyme alkylpurine-DNA-N-glycosylase (APNG), which repairs the cytotoxic lesions N³-methyladenine and N7-methylguanine, may contribute to TMZ resistance. Silencing of APNG in established and primary TMZ-resistant GBM cell lines endogenously expressing MGMT and APNG attenuated repair of TMZ-induced DNA damage and enhanced apoptosis. Reintroducing expression of APNG in TMZ-sensitive GBM lines conferred resistance to TMZ in vitro and in orthotopic xenograft mouse models. In addition, resistance was enhanced with coexpression of MGMT. Evaluation of APNG protein levels in several clinical datasets demonstrated that in patients, high nuclear APNG expression correlated with poorer overall survival compared with patients lacking APNG expression. Loss of APNG expression in a subset of patients was also associated with increased APNG promoter methylation. Collectively, our data demonstrate that APNG contributes to TMZ resistance in GBM and may be useful in the diagnosis and treatment of the disease.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , DNA Glycosylases/metabolism , Dacarbazine/analogs & derivatives , Glioblastoma/drug therapy , Glioblastoma/enzymology , Animals , Cell Line, Tumor , DNA Glycosylases/antagonists & inhibitors , DNA Glycosylases/genetics , DNA Modification Methylases/antagonists & inhibitors , DNA Modification Methylases/genetics , DNA Modification Methylases/metabolism , DNA Repair , DNA Repair Enzymes/antagonists & inhibitors , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , Dacarbazine/pharmacology , Drug Resistance, Neoplasm/genetics , Gene Expression , Gene Knockdown Techniques , Humans , Mice , Mice, Inbred NOD , Mice, SCID , RNA, Small Interfering/genetics , Temozolomide , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Xenograft Model Antitumor Assays
14.
J Exp Med ; 208(4): 689-702, 2011 Apr 11.
Article in English | MEDLINE | ID: mdl-21464220

ABSTRACT

Glioblastoma Multiforme (GBM), the most common and lethal primary human brain tumor, exhibits multiple molecular aberrations. We report that loss of the transcription factor GATA4, a negative regulator of normal astrocyte proliferation, is a driver in glioma formation and fulfills the hallmarks of a tumor suppressor gene (TSG). Although GATA4 was expressed in normal brain, loss of GATA4 was observed in 94/163 GBM operative samples and was a negative survival prognostic marker. GATA4 loss occurred through promoter hypermethylation or novel somatic mutations. Loss of GATA4 in normal human astrocytes promoted high-grade astrocytoma formation, in cooperation with other relevant genetic alterations such as activated Ras or loss of TP53. Loss of GATA4 with activated Ras in normal astrocytes promoted a progenitor-like phenotype, formation of neurospheres, and the ability to differentiate into astrocytes, neurons, and oligodendrocytes. Re-expression of GATA4 in human GBM cell lines, primary cultures, and brain tumor-initiating cells suppressed tumor growth in vitro and in vivo through direct activation of the cell cycle inhibitor P21(CIP1), independent of TP53. Re-expression of GATA4 also conferred sensitivity of GBM cells to temozolomide, a DNA alkylating agent currently used in GBM therapy. This sensitivity was independent of MGMT (O-6-methylguanine-DNA-methyltransferase), the DNA repair enzyme which is often implicated in temozolomide resistance. Instead, GATA4 reduced expression of APNG (alkylpurine-DNA-N-glycosylase), a DNA repair enzyme which is poorly characterized in GBM-mediated temozolomide resistance. Identification and validation of GATA4 as a TSG and its downstream targets in GBM may yield promising novel therapeutic strategies.


Subject(s)
Brain Neoplasms/prevention & control , GATA4 Transcription Factor/physiology , Glioblastoma/prevention & control , Tumor Suppressor Proteins/physiology , Animals , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p21/physiology , DNA Methylation , DNA Modification Methylases/physiology , DNA Repair Enzymes/physiology , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , GATA4 Transcription Factor/genetics , Glioblastoma/pathology , Humans , Mice , Promoter Regions, Genetic , Temozolomide
15.
Biotechniques ; 48(2): 145-52, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20359299

ABSTRACT

The development of small molecule and antibody inhibitors targeting the interaction of receptor tyrosine kinases (RTKs), such as epidermal growth factor receptor (EGFR), is of high pharmacological and biological interest. Unfortunately, conventional biochemical techniques using cell or tissue lysates and co-immunoprecipitation experiments to investigate EGFR dimerization are not always conclusive. Here we describe a series of technical and biological validation experiments demonstrating the utility of a proximity ligation assay (PLA)-based methodology for in situ visualization and quantification of ligand-dependent EGFR receptor dimerization in intact cells. Using the PLA approach combined with a universally applicable epitope tagging strategy, we detected EGFR dimers in cells transiently co-expressing FLAG-tagged and MYC-tagged human EGFRs. Our data strongly suggest that PLA can be used to detect ligand-dependent EGFR dimerization and this signal is generated in a protein interaction-based manner, rather than solely due to proximity of target proteins. This application represents a generalized RTK expression strategy for protein-interaction analysis in a transient expression system where antibody epitopes are not known or not unique enough to discriminate between interaction partners. This assay also holds promise as a general RTK dimerization screening tool in tissue specimens to identify potential dimerization inhibitors with clinical relevance.


Subject(s)
ErbB Receptors/metabolism , Protein Interaction Mapping/methods , Animals , Blotting, Western , Cell Line , Epitopes/genetics , Epitopes/metabolism , ErbB Receptors/genetics , Humans , Protein Multimerization , Transfection
16.
Neurosurgery ; 62(3): 539-55; discussion 539-55, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18425004

ABSTRACT

PROTEOMICS, IN ITS broadest mandate, is the study of proteins and their functions. As the "workhorses" of the genome, proteins govern normal cellular structure and function. Protein function is not just a reflection of its expression level; it is also the cumulative result of many post-transcriptional (splicing) and post-translational events that together determine cellular localization, interactions, and longevity. The composition and variability of the proteome is vastly more complex than the corresponding genome. It is this proteome variation that helps define an organism and the unique characteristics that separate one individual from another. Aberrations in protein function, which alter normal cellular structure and function, are the ultimate basis of disease, including cancer. Therefore, an understanding of protein networks through a systems biology approach of proteomics is necessary to understand normal and abnormal cellular function, with the goal of performing rational therapeutic interventions. In this review, we focus on two emerging proteomic technologies: mass spectrometry and bioluminescence resonance energy transfer. In addition to reviewing the principles and potential utilization of these two techniques, we highlight their application in neuro-oncology research.


Subject(s)
Brain Neoplasms/metabolism , Brain/metabolism , Neoplasm Proteins/metabolism , Neoplasms/diagnosis , Neoplasms/metabolism , Nerve Tissue Proteins/metabolism , Proteomics/trends , Biomarkers, Tumor/metabolism , Forecasting , Gene Expression Profiling/trends , Humans
17.
J Biol Chem ; 280(33): 29470-8, 2005 Aug 19.
Article in English | MEDLINE | ID: mdl-15955809

ABSTRACT

Ubiquitin-protein ligases (E3s) are implicated in various human disorders and are attractive targets for therapeutic intervention. Although most cellular proteins are ubiquitinated, ubiquitination cannot be linked directly to a specific E3 for a large fraction of these proteins, and the substrates of most E3 enzymes are unknown. We have developed a luminescent assay to detect ubiquitination in vitro, which is more quantitative, effective, and sensitive than conventional ubiquitination assays. By taking advantage of the abundance of purified proteins made available by genomic efforts, we screened hundreds of purified yeast proteins for ubiquitination, and we identified previously reported and novel substrates of the yeast E3 ligase Rsp5. The relevance of these substrates was confirmed in vivo by showing that a number of them interact genetically with Rsp5, and some were ubiquitinated by Rsp5 in vivo. The combination of this sensitive assay and the availability of purified substrates will enable the identification of substrates for any purified E3 enzyme.


Subject(s)
Saccharomyces cerevisiae Proteins/metabolism , Ubiquitin-Protein Ligase Complexes/metabolism , Ubiquitin/metabolism , Endosomal Sorting Complexes Required for Transport , Luminescent Measurements
SELECTION OF CITATIONS
SEARCH DETAIL
...