AML的肿瘤蛋白质组学分析:超越基因组学。

IF 3.8 3区 生物学 Q1 BIOCHEMICAL RESEARCH METHODS
Sunil K Joshi, Cristina E Tognon, Brian J Druker, Karin D Rodland
{"title":"AML的肿瘤蛋白质组学分析:超越基因组学。","authors":"Sunil K Joshi, Cristina E Tognon, Brian J Druker, Karin D Rodland","doi":"10.1080/14789450.2023.2176757","DOIUrl":null,"url":null,"abstract":"Much of what is known about protein-signaling networks in cancer, or ‘oncoproteomics,’ has been indirectly derived from transcriptomic analyses[1],[2]. However, RNA regulation precludes a one-to-one correlation of mRNA abundance to protein abundance or activity. A corollary of this is that evaluation of RNA by itself is insufficient to fully appreciate pathogenic cellular signaling within the tumor ecosystem. Global proteomics and phosphoproteomics have emerged as powerful unbiased methodologies for detailing fundamental signaling networks of cancer cells and perturbations that sustain resistance against targeted therapies, contributing to the discovery of new therapeutic targets[3]. Similar to other cancers, the utility of mass spectrometrybased technologies has augmented our ability to categorize the underlying heterogeneity in acute myeloid leukemia (AML) – expanding our capacity to classify AML beyond genomic features alone. Efforts over the past decade have resulted in the creation of new datasets that have begun to characterize the AML proteome and phosphoproteome. A subset of these studies have been exploratory in nature [4–9] – leading to the generation of new hypotheses, while others have focused on examining particular aspects of a disease state (e.g. drug resistance) to identify new biomarkers. These data provide a rich resource for further investigations aimed at mapping the ‘post-genomic’ landscape of AML (Table 1). Within this editorial, we discuss how integration and aggregation of such data with our current understanding of the AML genome and transcriptome holds the promise of refining our classification of leukemia cells – the genotype and phenotype – and yielding mechanistic insights that can inform the generation of improved therapeutic combinations. Casado et al. profiled the proteome and phosphoproteome of primary AML cells from 30 patients and the aggregation of these datasets with corresponding genomic, immunophenotypic, and pharmacologic analyses was among the first studies to infer that cell differentiation state influences kinase signaling changes and drug sensitivity profiles[4]. The authors also showed that FLT3 mutation status alone was insufficient to predict response to the FDA-approved inhibitor midostaurin and that increased activation of PKCδ and GSK3A in AML cells, as revealed by phosphoproteomics, correlated with midostaurin response[4]. Early attempts to integrate proteomic, genomic, and/or transcriptomic datasets have expanded our ability to categorize the small sub-populations of leukemic stem cells (LSCs) that govern the underlying heterogeneity and complexity of AML[5] and our understanding of the nuclear proteome in the pathogenesis of AML[6]. More recently, Jayavelu et al. identified five AML subtypes with distinct biological features via proteomic characterization of 252 AML patient samples[7]. Integration of these data with corresponding genomic, cytogenetic, and transcriptomic analyses revealed that the mito-AML subtype was only captured within proteomic profiling. This subtype of AML is characterized by high expression of mitochondrial proteins involved with cellular oxidative phosphorylation and confers poor prognosis. While many effectors in AML remain ‘undruggable,’ the work of Jatavelu et al. reveals that proteomics-based technologies have the propensity to identify new members within a signaling network that can be targeted – effectors that otherwise would not be considered from a traditional pharmacologic standpoint but warrant investigation. Specifically, the authors discovered a metabolic vulnerability through proteomics and phosphoproteomics and show that pharmacological inhibition of effector proteins within the mitochondrial network may eradicate AML cells, underscoring that treatment of AML goes beyond traditional regimens established merely by genomic aberrations or transcriptomic changes. Jatavelu et al. are not alone in showing that metabolic changes are regulated post-transcriptionally and require examination of the proteome. Raffel et al. demonstrated that LSCs are dependent upon amino acid metabolism[9]. More broadly, both studies highlight the strength offered by proteomic technologies to survey a large range of effector proteins with great specificity as opposed to a single effector studied through traditional methodologies, which are often hindered by target limitations and cross-reactivity. In line with Jayavelu et al., Kramer et al. developed a proteome and phosphoproteome database using a cohort of 44 AML patients[8]. While their dataset recapitulated many of the well-known features of AML, it refined our conceptualization of the regulatory processes sustaining AML including the importance of post-transcriptional protein modifications.","PeriodicalId":50463,"journal":{"name":"Expert Review of Proteomics","volume":null,"pages":null},"PeriodicalIF":3.8000,"publicationDate":"2022-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10505090/pdf/nihms-1931031.pdf","citationCount":"0","resultStr":"{\"title\":\"Oncoproteomic profiling of AML: moving beyond genomics.\",\"authors\":\"Sunil K Joshi, Cristina E Tognon, Brian J Druker, Karin D Rodland\",\"doi\":\"10.1080/14789450.2023.2176757\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"Much of what is known about protein-signaling networks in cancer, or ‘oncoproteomics,’ has been indirectly derived from transcriptomic analyses[1],[2]. However, RNA regulation precludes a one-to-one correlation of mRNA abundance to protein abundance or activity. A corollary of this is that evaluation of RNA by itself is insufficient to fully appreciate pathogenic cellular signaling within the tumor ecosystem. Global proteomics and phosphoproteomics have emerged as powerful unbiased methodologies for detailing fundamental signaling networks of cancer cells and perturbations that sustain resistance against targeted therapies, contributing to the discovery of new therapeutic targets[3]. Similar to other cancers, the utility of mass spectrometrybased technologies has augmented our ability to categorize the underlying heterogeneity in acute myeloid leukemia (AML) – expanding our capacity to classify AML beyond genomic features alone. Efforts over the past decade have resulted in the creation of new datasets that have begun to characterize the AML proteome and phosphoproteome. A subset of these studies have been exploratory in nature [4–9] – leading to the generation of new hypotheses, while others have focused on examining particular aspects of a disease state (e.g. drug resistance) to identify new biomarkers. These data provide a rich resource for further investigations aimed at mapping the ‘post-genomic’ landscape of AML (Table 1). Within this editorial, we discuss how integration and aggregation of such data with our current understanding of the AML genome and transcriptome holds the promise of refining our classification of leukemia cells – the genotype and phenotype – and yielding mechanistic insights that can inform the generation of improved therapeutic combinations. Casado et al. profiled the proteome and phosphoproteome of primary AML cells from 30 patients and the aggregation of these datasets with corresponding genomic, immunophenotypic, and pharmacologic analyses was among the first studies to infer that cell differentiation state influences kinase signaling changes and drug sensitivity profiles[4]. The authors also showed that FLT3 mutation status alone was insufficient to predict response to the FDA-approved inhibitor midostaurin and that increased activation of PKCδ and GSK3A in AML cells, as revealed by phosphoproteomics, correlated with midostaurin response[4]. Early attempts to integrate proteomic, genomic, and/or transcriptomic datasets have expanded our ability to categorize the small sub-populations of leukemic stem cells (LSCs) that govern the underlying heterogeneity and complexity of AML[5] and our understanding of the nuclear proteome in the pathogenesis of AML[6]. More recently, Jayavelu et al. identified five AML subtypes with distinct biological features via proteomic characterization of 252 AML patient samples[7]. Integration of these data with corresponding genomic, cytogenetic, and transcriptomic analyses revealed that the mito-AML subtype was only captured within proteomic profiling. This subtype of AML is characterized by high expression of mitochondrial proteins involved with cellular oxidative phosphorylation and confers poor prognosis. While many effectors in AML remain ‘undruggable,’ the work of Jatavelu et al. reveals that proteomics-based technologies have the propensity to identify new members within a signaling network that can be targeted – effectors that otherwise would not be considered from a traditional pharmacologic standpoint but warrant investigation. Specifically, the authors discovered a metabolic vulnerability through proteomics and phosphoproteomics and show that pharmacological inhibition of effector proteins within the mitochondrial network may eradicate AML cells, underscoring that treatment of AML goes beyond traditional regimens established merely by genomic aberrations or transcriptomic changes. Jatavelu et al. are not alone in showing that metabolic changes are regulated post-transcriptionally and require examination of the proteome. Raffel et al. demonstrated that LSCs are dependent upon amino acid metabolism[9]. More broadly, both studies highlight the strength offered by proteomic technologies to survey a large range of effector proteins with great specificity as opposed to a single effector studied through traditional methodologies, which are often hindered by target limitations and cross-reactivity. In line with Jayavelu et al., Kramer et al. developed a proteome and phosphoproteome database using a cohort of 44 AML patients[8]. While their dataset recapitulated many of the well-known features of AML, it refined our conceptualization of the regulatory processes sustaining AML including the importance of post-transcriptional protein modifications.\",\"PeriodicalId\":50463,\"journal\":{\"name\":\"Expert Review of Proteomics\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":3.8000,\"publicationDate\":\"2022-07-01\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10505090/pdf/nihms-1931031.pdf\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Expert Review of Proteomics\",\"FirstCategoryId\":\"99\",\"ListUrlMain\":\"https://doi.org/10.1080/14789450.2023.2176757\",\"RegionNum\":3,\"RegionCategory\":\"生物学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q1\",\"JCRName\":\"BIOCHEMICAL RESEARCH METHODS\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Expert Review of Proteomics","FirstCategoryId":"99","ListUrlMain":"https://doi.org/10.1080/14789450.2023.2176757","RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"BIOCHEMICAL RESEARCH METHODS","Score":null,"Total":0}
引用次数: 0

摘要

本文章由计算机程序翻译,如有差异,请以英文原文为准。
Oncoproteomic profiling of AML: moving beyond genomics.
Much of what is known about protein-signaling networks in cancer, or ‘oncoproteomics,’ has been indirectly derived from transcriptomic analyses[1],[2]. However, RNA regulation precludes a one-to-one correlation of mRNA abundance to protein abundance or activity. A corollary of this is that evaluation of RNA by itself is insufficient to fully appreciate pathogenic cellular signaling within the tumor ecosystem. Global proteomics and phosphoproteomics have emerged as powerful unbiased methodologies for detailing fundamental signaling networks of cancer cells and perturbations that sustain resistance against targeted therapies, contributing to the discovery of new therapeutic targets[3]. Similar to other cancers, the utility of mass spectrometrybased technologies has augmented our ability to categorize the underlying heterogeneity in acute myeloid leukemia (AML) – expanding our capacity to classify AML beyond genomic features alone. Efforts over the past decade have resulted in the creation of new datasets that have begun to characterize the AML proteome and phosphoproteome. A subset of these studies have been exploratory in nature [4–9] – leading to the generation of new hypotheses, while others have focused on examining particular aspects of a disease state (e.g. drug resistance) to identify new biomarkers. These data provide a rich resource for further investigations aimed at mapping the ‘post-genomic’ landscape of AML (Table 1). Within this editorial, we discuss how integration and aggregation of such data with our current understanding of the AML genome and transcriptome holds the promise of refining our classification of leukemia cells – the genotype and phenotype – and yielding mechanistic insights that can inform the generation of improved therapeutic combinations. Casado et al. profiled the proteome and phosphoproteome of primary AML cells from 30 patients and the aggregation of these datasets with corresponding genomic, immunophenotypic, and pharmacologic analyses was among the first studies to infer that cell differentiation state influences kinase signaling changes and drug sensitivity profiles[4]. The authors also showed that FLT3 mutation status alone was insufficient to predict response to the FDA-approved inhibitor midostaurin and that increased activation of PKCδ and GSK3A in AML cells, as revealed by phosphoproteomics, correlated with midostaurin response[4]. Early attempts to integrate proteomic, genomic, and/or transcriptomic datasets have expanded our ability to categorize the small sub-populations of leukemic stem cells (LSCs) that govern the underlying heterogeneity and complexity of AML[5] and our understanding of the nuclear proteome in the pathogenesis of AML[6]. More recently, Jayavelu et al. identified five AML subtypes with distinct biological features via proteomic characterization of 252 AML patient samples[7]. Integration of these data with corresponding genomic, cytogenetic, and transcriptomic analyses revealed that the mito-AML subtype was only captured within proteomic profiling. This subtype of AML is characterized by high expression of mitochondrial proteins involved with cellular oxidative phosphorylation and confers poor prognosis. While many effectors in AML remain ‘undruggable,’ the work of Jatavelu et al. reveals that proteomics-based technologies have the propensity to identify new members within a signaling network that can be targeted – effectors that otherwise would not be considered from a traditional pharmacologic standpoint but warrant investigation. Specifically, the authors discovered a metabolic vulnerability through proteomics and phosphoproteomics and show that pharmacological inhibition of effector proteins within the mitochondrial network may eradicate AML cells, underscoring that treatment of AML goes beyond traditional regimens established merely by genomic aberrations or transcriptomic changes. Jatavelu et al. are not alone in showing that metabolic changes are regulated post-transcriptionally and require examination of the proteome. Raffel et al. demonstrated that LSCs are dependent upon amino acid metabolism[9]. More broadly, both studies highlight the strength offered by proteomic technologies to survey a large range of effector proteins with great specificity as opposed to a single effector studied through traditional methodologies, which are often hindered by target limitations and cross-reactivity. In line with Jayavelu et al., Kramer et al. developed a proteome and phosphoproteome database using a cohort of 44 AML patients[8]. While their dataset recapitulated many of the well-known features of AML, it refined our conceptualization of the regulatory processes sustaining AML including the importance of post-transcriptional protein modifications.
求助全文
通过发布文献求助,成功后即可免费获取论文全文。 去求助
来源期刊
Expert Review of Proteomics
Expert Review of Proteomics 生物-生化研究方法
CiteScore
7.60
自引率
0.00%
发文量
20
审稿时长
6-12 weeks
期刊介绍: Expert Review of Proteomics (ISSN 1478-9450) seeks to collect together technologies, methods and discoveries from the field of proteomics to advance scientific understanding of the many varied roles protein expression plays in human health and disease. The journal coverage includes, but is not limited to, overviews of specific technological advances in the development of protein arrays, interaction maps, data archives and biological assays, performance of new technologies and prospects for future drug discovery. The journal adopts the unique Expert Review article format, offering a complete overview of current thinking in a key technology area, research or clinical practice, augmented by the following sections: Expert Opinion - a personal view on the most effective or promising strategies and a clear perspective of future prospects within a realistic timescale Article highlights - an executive summary cutting to the author''s most critical points.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信