首页 > 最新文献

Expert Review of Proteomics最新文献

英文 中文
Rebellion of the deregulated regulators: What is the clinical relevance of studying intrinsically disordered proteins? 解除管制的监管者的反叛:研究内在无序蛋白质的临床相关性是什么?
IF 3.4 3区 生物学 Q2 Biochemistry, Genetics and Molecular Biology Pub Date : 2022-07-01 DOI: 10.1080/14789450.2023.2176755
Vladimir N Uversky
Since at the molecular level, almost all physiological processes are defined by the specific activities of specific proteins or protein groups, dysfunction and deregulation of these proteins are linked to the pathogenesis of various maladies. Therefore, to get to the roots of the pathological processes and find appropriate cure for the related diseases, one should clearly know the connections between protein-centric physiology and pathology. This logic represents premises of the medical protein science, where one is looking for the connections between the ‘right’ protein structure and normal function to understand how dysfunction can be linked back to the ‘wrong’ structure and assuming that fixing such ‘wrong’ structure can serve as a means to restore a normal function and therefore cure a disease. Even though mutations in a gene encoding a culprit protein represent the major reason for this protein to gain ‘wrong’ structure, dysfunctionality can also be caused by the distortion of any means from a very broad arsenal of cellular proteostasis-related mechanisms evolved to control and regulate protein folding, structure, and function. Although for the first time, proteins were described by the Dutch chemist Gerardus Johannes Mulder (1802–1880) as enormous molecules, with empirical formula for fibrin and egg albumin being C400H620N100O120P1S1, in his 1838 paper ‘On the composition of some animal substances’ first published in French [1] and translated to German in 1839 [2], they gained serious attention of researchers only after their polypeptide nature discovered independently in 1902 by a German chemist Hermann Emil Louis Fischer (1852–1919) [3] and an early protein scientist Franz Hofmeister (1850–1922) [4] was connected to the enzymatic activity by an American chemist, James B. Sumner (1887–1955), who, in 1926, showed that the enzyme urease is a protein that can be isolated and crystallized [5]. Curiously, as early as in 1894, enzymatic activity was proposed by Emil Fischer to follow his classical ‘lock-and-key’ model [6]. This concept was eventually elaborated into the famous protein structure-function paradigm, where the amino acid sequence determines a uniquely folded 3D structure that can be visualized in the crystalline state and that, in turn, defines the unique protein function [7]. As a result, in most of the almost 185 years of their history (and definitely since 1894), proteins were equated to enzymes, being considered as biological catalysts, while many other functions of these biological macromolecules and their intriguing potential to be multifunctional were mostly ignored.
{"title":"Rebellion of the deregulated regulators: What is the clinical relevance of studying intrinsically disordered proteins?","authors":"Vladimir N Uversky","doi":"10.1080/14789450.2023.2176755","DOIUrl":"https://doi.org/10.1080/14789450.2023.2176755","url":null,"abstract":"Since at the molecular level, almost all physiological processes are defined by the specific activities of specific proteins or protein groups, dysfunction and deregulation of these proteins are linked to the pathogenesis of various maladies. Therefore, to get to the roots of the pathological processes and find appropriate cure for the related diseases, one should clearly know the connections between protein-centric physiology and pathology. This logic represents premises of the medical protein science, where one is looking for the connections between the ‘right’ protein structure and normal function to understand how dysfunction can be linked back to the ‘wrong’ structure and assuming that fixing such ‘wrong’ structure can serve as a means to restore a normal function and therefore cure a disease. Even though mutations in a gene encoding a culprit protein represent the major reason for this protein to gain ‘wrong’ structure, dysfunctionality can also be caused by the distortion of any means from a very broad arsenal of cellular proteostasis-related mechanisms evolved to control and regulate protein folding, structure, and function. Although for the first time, proteins were described by the Dutch chemist Gerardus Johannes Mulder (1802–1880) as enormous molecules, with empirical formula for fibrin and egg albumin being C400H620N100O120P1S1, in his 1838 paper ‘On the composition of some animal substances’ first published in French [1] and translated to German in 1839 [2], they gained serious attention of researchers only after their polypeptide nature discovered independently in 1902 by a German chemist Hermann Emil Louis Fischer (1852–1919) [3] and an early protein scientist Franz Hofmeister (1850–1922) [4] was connected to the enzymatic activity by an American chemist, James B. Sumner (1887–1955), who, in 1926, showed that the enzyme urease is a protein that can be isolated and crystallized [5]. Curiously, as early as in 1894, enzymatic activity was proposed by Emil Fischer to follow his classical ‘lock-and-key’ model [6]. This concept was eventually elaborated into the famous protein structure-function paradigm, where the amino acid sequence determines a uniquely folded 3D structure that can be visualized in the crystalline state and that, in turn, defines the unique protein function [7]. As a result, in most of the almost 185 years of their history (and definitely since 1894), proteins were equated to enzymes, being considered as biological catalysts, while many other functions of these biological macromolecules and their intriguing potential to be multifunctional were mostly ignored.","PeriodicalId":50463,"journal":{"name":"Expert Review of Proteomics","volume":null,"pages":null},"PeriodicalIF":3.4,"publicationDate":"2022-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9120103","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 1
Advances in phosphoproteomics and its application to COPD. 磷酸蛋白质组学研究进展及其在COPD中的应用。
IF 3.4 3区 生物学 Q2 Biochemistry, Genetics and Molecular Biology Pub Date : 2022-07-01 DOI: 10.1080/14789450.2023.2176756
Xiaoyin Zeng, Yanting Lan, Jing Xiao, Longbo Hu, Long Tan, Mengdi Liang, Xufei Wang, Shaohua Lu, Tao Peng, Fei Long

Introduction: Chronic obstructive pulmonary disease (COPD) was the third leading cause of global death in 2019, causing a huge economic burden to society. Therefore, it is urgent to identify specific phenotypes of COPD patients through early detection, and to promptly treat exacerbations. The field of phosphoproteomics has been a massive advancement, compelled by the developments in mass spectrometry, enrichment strategies, algorithms, and tools. Modern mass spectrometry-based phosphoproteomics allows understanding of disease pathobiology, biomarker discovery, and predicting new therapeutic modalities.

Areas covered: In this article, we present an overview of phosphoproteomic research and strategies for enrichment and fractionation of phosphopeptides, identification of phosphorylation sites, chromatographic separation and mass spectrometry detection strategies, and the potential application of phosphorylated proteomic analysis in the diagnosis, treatment, and prognosis of COPD disease.

Expert opinion: The role of phosphoproteomics in COPD is critical for understanding disease pathobiology, identifying potential biomarkers, and predicting new therapeutic approaches. However, the complexity of COPD requires the more comprehensive understanding that can be achieved through integrated multi-omics studies. Phosphoproteomics, as a part of these multi-omics approaches, can provide valuable insights into the underlying mechanisms of COPD.

慢性阻塞性肺疾病(COPD)是2019年全球第三大死亡原因,给社会造成了巨大的经济负担。因此,通过早期发现来识别COPD患者的特定表型,并及时治疗加重是当务之急。由于质谱、富集策略、算法和工具的发展,磷酸化蛋白质组学领域已经取得了巨大的进步。以现代质谱为基础的磷蛋白质组学可以理解疾病的病理生物学、生物标志物的发现和预测新的治疗方式。涉及领域:在这篇文章中,我们概述了磷酸化蛋白质组学的研究和磷酸化肽的富集和分离策略,磷酸化位点的鉴定,色谱分离和质谱检测策略,以及磷酸化蛋白质组学分析在COPD疾病的诊断、治疗和预后中的潜在应用。专家意见:磷蛋白组学在COPD中的作用对于理解疾病病理生物学、识别潜在的生物标志物和预测新的治疗方法至关重要。然而,COPD的复杂性需要更全面的了解,这可以通过综合多组学研究来实现。磷蛋白组学作为这些多组学方法的一部分,可以为COPD的潜在机制提供有价值的见解。
{"title":"Advances in phosphoproteomics and its application to COPD.","authors":"Xiaoyin Zeng,&nbsp;Yanting Lan,&nbsp;Jing Xiao,&nbsp;Longbo Hu,&nbsp;Long Tan,&nbsp;Mengdi Liang,&nbsp;Xufei Wang,&nbsp;Shaohua Lu,&nbsp;Tao Peng,&nbsp;Fei Long","doi":"10.1080/14789450.2023.2176756","DOIUrl":"https://doi.org/10.1080/14789450.2023.2176756","url":null,"abstract":"<p><strong>Introduction: </strong>Chronic obstructive pulmonary disease (COPD) was the third leading cause of global death in 2019, causing a huge economic burden to society. Therefore, it is urgent to identify specific phenotypes of COPD patients through early detection, and to promptly treat exacerbations. The field of phosphoproteomics has been a massive advancement, compelled by the developments in mass spectrometry, enrichment strategies, algorithms, and tools. Modern mass spectrometry-based phosphoproteomics allows understanding of disease pathobiology, biomarker discovery, and predicting new therapeutic modalities.</p><p><strong>Areas covered: </strong>In this article, we present an overview of phosphoproteomic research and strategies for enrichment and fractionation of phosphopeptides, identification of phosphorylation sites, chromatographic separation and mass spectrometry detection strategies, and the potential application of phosphorylated proteomic analysis in the diagnosis, treatment, and prognosis of COPD disease.</p><p><strong>Expert opinion: </strong>The role of phosphoproteomics in COPD is critical for understanding disease pathobiology, identifying potential biomarkers, and predicting new therapeutic approaches. However, the complexity of COPD requires the more comprehensive understanding that can be achieved through integrated multi-omics studies. Phosphoproteomics, as a part of these multi-omics approaches, can provide valuable insights into the underlying mechanisms of COPD.</p>","PeriodicalId":50463,"journal":{"name":"Expert Review of Proteomics","volume":null,"pages":null},"PeriodicalIF":3.4,"publicationDate":"2022-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9118955","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Oncoproteomic profiling of AML: moving beyond genomics. AML的肿瘤蛋白质组学分析:超越基因组学。
IF 3.4 3区 生物学 Q2 Biochemistry, Genetics and Molecular Biology Pub Date : 2022-07-01 DOI: 10.1080/14789450.2023.2176757
Sunil K Joshi, Cristina E Tognon, Brian J Druker, Karin D Rodland
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 t
{"title":"Oncoproteomic profiling of AML: moving beyond genomics.","authors":"Sunil K Joshi,&nbsp;Cristina E Tognon,&nbsp;Brian J Druker,&nbsp;Karin D Rodland","doi":"10.1080/14789450.2023.2176757","DOIUrl":"https://doi.org/10.1080/14789450.2023.2176757","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 t","PeriodicalId":50463,"journal":{"name":"Expert Review of Proteomics","volume":null,"pages":null},"PeriodicalIF":3.4,"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":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10298600","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
13th annual symposium of the Canadian National Proteomics Network. 第13届加拿大国家蛋白质组学网络年会。
IF 3.4 3区 生物学 Q2 Biochemistry, Genetics and Molecular Biology Pub Date : 2022-04-01 DOI: 10.1080/14789450.2022.2158815
Jennifer Geddes-McAlister, David C Schriemer

The 13th annual Canadian National Proteomics Network was held in May 2022 in Montreal, Quebec, Canada. More than 175 individuals participated in this dynamic and productive meeting either in-person or virtually. A pre-symposium organized by trainees and dedicated to highlighting the best and brightest emerging talent in proteomics across Canada preceded the main symposium, which welcomed plenary and invited speakers from around the world. The presentations covering ground-breaking science were interspersed with critical discussions on improving equity, diversity, and inclusion within the proteomics community across Canada, along with important networking opportunities for early-career researchers.

第13届加拿大国家蛋白质组学网络年会于2022年5月在加拿大魁北克省蒙特利尔举行。超过175人参加了这个充满活力和富有成效的会议,无论是面对面的还是虚拟的。在主要研讨会之前,由学员组织的一个预研讨会,致力于突出加拿大蛋白质组学领域最优秀和最聪明的新兴人才,欢迎来自世界各地的全体会议和特邀演讲者。在涵盖突破性科学的演讲中,穿插着关于提高加拿大蛋白质组学社区的公平性、多样性和包容性的关键讨论,以及为早期职业研究人员提供重要的交流机会。
{"title":"13<sup>th</sup> annual symposium of the Canadian National Proteomics Network.","authors":"Jennifer Geddes-McAlister,&nbsp;David C Schriemer","doi":"10.1080/14789450.2022.2158815","DOIUrl":"https://doi.org/10.1080/14789450.2022.2158815","url":null,"abstract":"<p><p>The 13<sup>th</sup> annual Canadian National Proteomics Network was held in May 2022 in Montreal, Quebec, Canada. More than 175 individuals participated in this dynamic and productive meeting either in-person or virtually. A pre-symposium organized by trainees and dedicated to highlighting the best and brightest emerging talent in proteomics across Canada preceded the main symposium, which welcomed plenary and invited speakers from around the world. The presentations covering ground-breaking science were interspersed with critical discussions on improving equity, diversity, and inclusion within the proteomics community across Canada, along with important networking opportunities for early-career researchers.</p>","PeriodicalId":50463,"journal":{"name":"Expert Review of Proteomics","volume":null,"pages":null},"PeriodicalIF":3.4,"publicationDate":"2022-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9365891","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Can spatially resolved metabolomics uncover weak points in tumors? 空间分解代谢组学能揭示肿瘤的弱点吗?
IF 3.4 3区 生物学 Q2 Biochemistry, Genetics and Molecular Biology Pub Date : 2022-04-01 DOI: 10.1080/14789450.2023.2176754
Zhen Ning, Guowang Xu
The complexity and diversity derived from genetics and evolution lead to tumor heterogeneity. The spatial and temporal evolution of tumor heterogeneity during tumor development results in the dynamic reprogramming of the tumor microenvironment (TME) [1]. Over the last decade, technological developments from bulk genome to single-cell sequencing have provided us with ever-more powerful tool to investigate what happens in TME [2]. Since reprogrammed energy metabolism is one of the hallmarks of cancer, metabolomics may provide a new direction for shedding light on the interactions between small molecules (mainly molecules with molecular weight less than 2000 Da) and other biomolecules in tumors. However, traditional metabolomics cannot give spatiallyrelated information unless combined with spatially resolved sampling, but revealing the metabolic reprogramming characteristics of TME and clarifying the targeting heterogeneity of antitumor drugs rely on the spatial information of metabolites or small molecule drugs. Thus, the advent of spatial metabolomics provides an opportunity to detect molecular localization based on the relative abundance of molecules and to directly correlate changes in small molecules with anatomical features. In other words, spatial metabolomics is oriented to reveal the spatial distribution and variation of metabolites [3]. Most of spatially resolved metabolomics combine ionization techniques with label-free, high-throughput mass spectrometry imaging (MSI) to obtain information on the spatial distribution of metabolites. In addition, laser capture microdissection technique combined with mass spectrometry detection is also one of the research directions in spatial metabolomics, it can select the area of interest for detailed study. Developments in MSI now make it possible to directly observe metabolic changes in tissues, even in single cells. To date, most spatial metabolomics techniques are based on matrix-assisted laser desorption ionization mass spectrometry imaging (MALDI-MSI) or desorption electrospray ionization mass spectrometry imaging (DESI-MSI), both of which are constantly being improved [4]. In recent years, spatially resolved metabolomics has reaped a series of groundbreaking insights in the fields of metabolic heterogeneity of tumors, rapid diagnosis (including tumor boundary determination), metabolic typing, targeting efficiency of antitumor drugs, and efficacy assessment by obtaining information on the distribution of metabolites and smallmolecule drugs in TME (Figure 1). The development of spatially resolved metabolomics technologies will help open the black box of TME and provide new opportunities for precision treatment of tumors.
{"title":"Can spatially resolved metabolomics uncover weak points in tumors?","authors":"Zhen Ning,&nbsp;Guowang Xu","doi":"10.1080/14789450.2023.2176754","DOIUrl":"https://doi.org/10.1080/14789450.2023.2176754","url":null,"abstract":"The complexity and diversity derived from genetics and evolution lead to tumor heterogeneity. The spatial and temporal evolution of tumor heterogeneity during tumor development results in the dynamic reprogramming of the tumor microenvironment (TME) [1]. Over the last decade, technological developments from bulk genome to single-cell sequencing have provided us with ever-more powerful tool to investigate what happens in TME [2]. Since reprogrammed energy metabolism is one of the hallmarks of cancer, metabolomics may provide a new direction for shedding light on the interactions between small molecules (mainly molecules with molecular weight less than 2000 Da) and other biomolecules in tumors. However, traditional metabolomics cannot give spatiallyrelated information unless combined with spatially resolved sampling, but revealing the metabolic reprogramming characteristics of TME and clarifying the targeting heterogeneity of antitumor drugs rely on the spatial information of metabolites or small molecule drugs. Thus, the advent of spatial metabolomics provides an opportunity to detect molecular localization based on the relative abundance of molecules and to directly correlate changes in small molecules with anatomical features. In other words, spatial metabolomics is oriented to reveal the spatial distribution and variation of metabolites [3]. Most of spatially resolved metabolomics combine ionization techniques with label-free, high-throughput mass spectrometry imaging (MSI) to obtain information on the spatial distribution of metabolites. In addition, laser capture microdissection technique combined with mass spectrometry detection is also one of the research directions in spatial metabolomics, it can select the area of interest for detailed study. Developments in MSI now make it possible to directly observe metabolic changes in tissues, even in single cells. To date, most spatial metabolomics techniques are based on matrix-assisted laser desorption ionization mass spectrometry imaging (MALDI-MSI) or desorption electrospray ionization mass spectrometry imaging (DESI-MSI), both of which are constantly being improved [4]. In recent years, spatially resolved metabolomics has reaped a series of groundbreaking insights in the fields of metabolic heterogeneity of tumors, rapid diagnosis (including tumor boundary determination), metabolic typing, targeting efficiency of antitumor drugs, and efficacy assessment by obtaining information on the distribution of metabolites and smallmolecule drugs in TME (Figure 1). The development of spatially resolved metabolomics technologies will help open the black box of TME and provide new opportunities for precision treatment of tumors.","PeriodicalId":50463,"journal":{"name":"Expert Review of Proteomics","volume":null,"pages":null},"PeriodicalIF":3.4,"publicationDate":"2022-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9383269","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Proteomics and phosphoproteomics analysis of tissues for the reoccurrence prediction of colorectal cancer. 组织蛋白质组学和磷酸化蛋白质组学分析预测结直肠癌复发。
IF 3.4 3区 生物学 Q2 Biochemistry, Genetics and Molecular Biology Pub Date : 2022-04-01 DOI: 10.1080/14789450.2022.2142566
Liyun Ji, Zeyuan Wang, Yin Ji, Huiyu Wang, Miao Guo, Lu Zhang, Peng Wang, Hua Xiao

Background: Many stage II/III colorectal cancer (CRC) patients may relapse after routine treatments. Aberrant phosphorylation can regulate pathophysiological processes of tumors, and finding characteristic protein phosphorylation is an efficient approach for the prediction of CRC relapse.

Research design and methods: We compared the tissue proteome and phosphoproteome of stage II/III CRC patients between the relapsed group (n = 5) and the non-relapsed group (n = 5). Phosphopeptides were enriched with Ti4+-IMAC material. We utilized label-free quantification-based proteomics to screen differentially expressed proteins and phosphopeptides between the two groups. Gene Ontology (GO) analysis and Ingenuity Pathway Analysis (IPA) were used for bioinformatics analysis.

Results: The immune response of the relapsed group (Z-score -2.229) was relatively poorer than that of the non-relapsed group (Z-score 1.982), while viability of tumor was more activated (Z-score 2.895) in the relapsed group, which might cause increased relapse risk. The phosphorylation degrees of three phosphosites (phosphosite 1362 of TP53BP1, phosphosite 809 of VCL and phosphosite 438 of STK10) might be reliable prognostic biomarkers.

Conclusions: Some promising proteins and phosphopeptides were discovered to predict the relapse risk in postoperative follow-ups.

背景:许多II/III期结直肠癌(CRC)患者在常规治疗后可能复发。异常磷酸化可以调节肿瘤的病理生理过程,发现特异性磷酸化蛋白是预测结直肠癌复发的有效途径。研究设计和方法:比较复发组(n = 5)和非复发组(n = 5) II/III期CRC患者的组织蛋白质组和磷酸化蛋白质组。磷酸肽富集Ti4+-IMAC物质。我们利用无标记的基于定量的蛋白质组学来筛选两组之间差异表达的蛋白质和磷酸肽。生物信息学分析采用基因本体(GO)分析和独创性途径分析(IPA)。结果:复发组(z评分-2.229)的免疫应答相对于非复发组(z评分1.982)差,而复发组肿瘤活力更活跃(z评分2.895),可能导致复发风险增加。三个磷酸化位点(TP53BP1的1362磷酸化位点、VCL的809磷酸化位点和STK10的438磷酸化位点)的磷酸化程度可能是可靠的预后生物标志物。结论:在术后随访中发现了一些有希望预测复发风险的蛋白和磷酸肽。
{"title":"Proteomics and phosphoproteomics analysis of tissues for the reoccurrence prediction of colorectal cancer.","authors":"Liyun Ji,&nbsp;Zeyuan Wang,&nbsp;Yin Ji,&nbsp;Huiyu Wang,&nbsp;Miao Guo,&nbsp;Lu Zhang,&nbsp;Peng Wang,&nbsp;Hua Xiao","doi":"10.1080/14789450.2022.2142566","DOIUrl":"https://doi.org/10.1080/14789450.2022.2142566","url":null,"abstract":"<p><strong>Background: </strong>Many stage II/III colorectal cancer (CRC) patients may relapse after routine treatments. Aberrant phosphorylation can regulate pathophysiological processes of tumors, and finding characteristic protein phosphorylation is an efficient approach for the prediction of CRC relapse.</p><p><strong>Research design and methods: </strong>We compared the tissue proteome and phosphoproteome of stage II/III CRC patients between the relapsed group (n = 5) and the non-relapsed group (n = 5). Phosphopeptides were enriched with Ti<sup>4+</sup>-IMAC material. We utilized label-free quantification-based proteomics to screen differentially expressed proteins and phosphopeptides between the two groups. Gene Ontology (GO) analysis and Ingenuity Pathway Analysis (IPA) were used for bioinformatics analysis.</p><p><strong>Results: </strong>The immune response of the relapsed group (Z-score -2.229) was relatively poorer than that of the non-relapsed group (Z-score 1.982), while viability of tumor was more activated (Z-score 2.895) in the relapsed group, which might cause increased relapse risk. The phosphorylation degrees of three phosphosites (phosphosite 1362 of TP53BP1, phosphosite 809 of VCL and phosphosite 438 of STK10) might be reliable prognostic biomarkers.</p><p><strong>Conclusions: </strong>Some promising proteins and phosphopeptides were discovered to predict the relapse risk in postoperative follow-ups.</p>","PeriodicalId":50463,"journal":{"name":"Expert Review of Proteomics","volume":null,"pages":null},"PeriodicalIF":3.4,"publicationDate":"2022-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10808081","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The role of E3 ubiquitin ligase in multiple myeloma: potential for cereblon E3 ligase modulators in the treatment of relapsed/refractory disease. E3泛素连接酶在多发性骨髓瘤中的作用:小脑E3连接酶调节剂治疗复发/难治性疾病的潜力
IF 3.4 3区 生物学 Q2 Biochemistry, Genetics and Molecular Biology Pub Date : 2022-04-01 DOI: 10.1080/14789450.2022.2142564
Paul G Richardson, María-Victoria Mateos, Annette J Vangsted, Karthik Ramasamy, Niels Abildgaard, P Joy Ho, Hang Quach, Nizar J Bahlis

Introduction: Insights into the mechanisms of protein homeostasis and proteasomal degradation have led to new strategies of redirecting the ubiquitin-proteasome system (UPS) to reduce or eliminate proteins or survival factors key to malignant pathobiology, multiple myeloma (MM) in particular. These strategies have enabled researchers to target proteins that were previously considered difficult to modulate by pharmacological means.

Areas covered: This review provides a brief overview of UPS biology, particularly the role of the CRL4CRBN E3 ubiquitin ligase complex, and summarizes current strategies for co-opting the UPS, including CELMoD compounds, SNIPERs, PROTACs, and degronimids. A detailed discussion is provided on lead CELMoD compounds iberdomide and mezigdomide, which are currently being evaluated in clinical trials in patients with MM.

Expert opinion: Since a high proportion of patients develop drug resistance, it is vital to have novel therapeutic agents for treating relapsed patients with MM more effectively. It is encouraging that the expanding pathophysiological insight into cellular signaling pathways in MM increasingly translates into the development of novel therapeutic agents such as targeted protein degraders. This holds promise for improving outcomes in MM and beyond.

导读:对蛋白质稳态和蛋白酶体降解机制的深入了解已经导致了重定向泛素-蛋白酶体系统(UPS)以减少或消除恶性病理生物学,特别是多发性骨髓瘤(MM)关键的蛋白质或生存因子的新策略。这些策略使研究人员能够靶向以前被认为难以通过药理学手段调节的蛋白质。涵盖领域:本综述简要概述了UPS生物学,特别是CRL4CRBN E3泛素连接酶复合物的作用,并总结了目前使用UPS的策略,包括CELMoD化合物、SNIPERs、PROTACs和degronimids。详细讨论了CELMoD先导化合物伊伯多胺和美西多胺,目前正在MM患者的临床试验中进行评估。专家意见:由于高比例的患者产生耐药性,因此寻找新的治疗药物更有效地治疗复发MM患者至关重要。令人鼓舞的是,对MM细胞信号通路的病理生理学深入研究越来越多地转化为新型治疗剂的开发,如靶向蛋白质降解剂。这有望改善MM和其他方面的结果。
{"title":"The role of E3 ubiquitin ligase in multiple myeloma: potential for cereblon E3 ligase modulators in the treatment of relapsed/refractory disease.","authors":"Paul G Richardson,&nbsp;María-Victoria Mateos,&nbsp;Annette J Vangsted,&nbsp;Karthik Ramasamy,&nbsp;Niels Abildgaard,&nbsp;P Joy Ho,&nbsp;Hang Quach,&nbsp;Nizar J Bahlis","doi":"10.1080/14789450.2022.2142564","DOIUrl":"https://doi.org/10.1080/14789450.2022.2142564","url":null,"abstract":"<p><strong>Introduction: </strong>Insights into the mechanisms of protein homeostasis and proteasomal degradation have led to new strategies of redirecting the ubiquitin-proteasome system (UPS) to reduce or eliminate proteins or survival factors key to malignant pathobiology, multiple myeloma (MM) in particular. These strategies have enabled researchers to target proteins that were previously considered difficult to modulate by pharmacological means.</p><p><strong>Areas covered: </strong>This review provides a brief overview of UPS biology, particularly the role of the CRL4<sup>CRBN</sup> E3 ubiquitin ligase complex, and summarizes current strategies for co-opting the UPS, including CELMoD compounds, SNIPERs, PROTACs, and degronimids. A detailed discussion is provided on lead CELMoD compounds iberdomide and mezigdomide, which are currently being evaluated in clinical trials in patients with MM.</p><p><strong>Expert opinion: </strong>Since a high proportion of patients develop drug resistance, it is vital to have novel therapeutic agents for treating relapsed patients with MM more effectively. It is encouraging that the expanding pathophysiological insight into cellular signaling pathways in MM increasingly translates into the development of novel therapeutic agents such as targeted protein degraders. This holds promise for improving outcomes in MM and beyond.</p>","PeriodicalId":50463,"journal":{"name":"Expert Review of Proteomics","volume":null,"pages":null},"PeriodicalIF":3.4,"publicationDate":"2022-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10808088","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 1
Schistosome proteomics: updates and clinical implications. 血吸虫蛋白质组学:最新进展及其临床意义。
IF 3.4 3区 生物学 Q2 Biochemistry, Genetics and Molecular Biology Pub Date : 2022-04-01 DOI: 10.1080/14789450.2022.2142565
William Castro-Borges, R Alan Wilson

Introduction: Schistosomes are long-lived blood dwelling helminth parasites using intricate mechanisms to invade, mature, and reproduce inside their vertebrate hosts, whilst simultaneously deploying immune evasion strategies. Their multi-tissue organization and solid body plan presents particular problems for the definition of sub-proteomes.

Areas covered: Here, we focus on the two host-parasite interfaces of the adult worm accessible to the immune system, namely the tegument and the alimentary tract, but also on the secretions of the infective cercaria, the migrating schistosomulum and the mature egg. In parallel, we introduce the concepts of "leakyome' and 'disintegrome' to emphasize the importance of interpreting data in the context of schistosome biology so that misleading conclusions about the distinct proteome compositions are avoided. Lastly, we highlight the possible clinical implications of the reviewed proteomic findings for pathogenesis, vaccine design and diagnostics.

Expert opinion: Proteomics has provided considerable insights into the biology of schistosomes, most importantly for rational selection of novel vaccine candidates that might confer protective immunity, but also into the pathogenesis of schistosomiasis. However, given the increasing sensitivity of mass spectrometric instrumentation, we stress the need for care in data interpretation since schistosomes do not deviate from the fundamental rules of eukaryotic cell biology.

简介:血吸虫是一种长寿的血液寄生蠕虫,利用复杂的机制入侵、成熟和在脊椎动物宿主体内繁殖,同时利用免疫逃避策略。它们的多组织结构和实体结构为亚蛋白质组的定义提出了特殊的问题。涵盖领域:在这里,我们重点关注成虫可进入免疫系统的两个宿主-寄生虫界面,即被皮和消化道,以及感染性尾蚴、迁移血吸虫和成熟卵的分泌物。同时,我们引入了“漏体”和“分解组”的概念,以强调在血吸虫生物学背景下解释数据的重要性,从而避免了关于不同蛋白质组组成的误导性结论。最后,我们强调了蛋白质组学研究结果在发病机制、疫苗设计和诊断方面可能的临床意义。专家意见:蛋白质组学为血吸虫生物学提供了相当多的见解,最重要的是合理选择可能具有保护性免疫的新型候选疫苗,但也为血吸虫病的发病机制提供了见解。然而,鉴于质谱仪器的灵敏度越来越高,我们强调在数据解释中需要注意,因为血吸虫并不偏离真核细胞生物学的基本规则。
{"title":"Schistosome proteomics: updates and clinical implications.","authors":"William Castro-Borges,&nbsp;R Alan Wilson","doi":"10.1080/14789450.2022.2142565","DOIUrl":"https://doi.org/10.1080/14789450.2022.2142565","url":null,"abstract":"<p><strong>Introduction: </strong>Schistosomes are long-lived blood dwelling helminth parasites using intricate mechanisms to invade, mature, and reproduce inside their vertebrate hosts, whilst simultaneously deploying immune evasion strategies. Their multi-tissue organization and solid body plan presents particular problems for the definition of sub-proteomes.</p><p><strong>Areas covered: </strong>Here, we focus on the two host-parasite interfaces of the adult worm accessible to the immune system, namely the tegument and the alimentary tract, but also on the secretions of the infective cercaria, the migrating schistosomulum and the mature egg. In parallel, we introduce the concepts of \"leakyome' and 'disintegrome' to emphasize the importance of interpreting data in the context of schistosome biology so that misleading conclusions about the distinct proteome compositions are avoided. Lastly, we highlight the possible clinical implications of the reviewed proteomic findings for pathogenesis, vaccine design and diagnostics.</p><p><strong>Expert opinion: </strong>Proteomics has provided considerable insights into the biology of schistosomes, most importantly for rational selection of novel vaccine candidates that might confer protective immunity, but also into the pathogenesis of schistosomiasis. However, given the increasing sensitivity of mass spectrometric instrumentation, we stress the need for care in data interpretation since schistosomes do not deviate from the fundamental rules of eukaryotic cell biology.</p>","PeriodicalId":50463,"journal":{"name":"Expert Review of Proteomics","volume":null,"pages":null},"PeriodicalIF":3.4,"publicationDate":"2022-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10826585","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 3
Rise of the SARS-CoV-2 Variants: can proteomics be the silver bullet? SARS-CoV-2变体的兴起:蛋白质组学可以成为银弹吗?
IF 3.4 3区 生物学 Q2 Biochemistry, Genetics and Molecular Biology Pub Date : 2022-03-01 DOI: 10.1080/14789450.2022.2085564
Arup Acharjee, Joshua Stephen Kingsly, Madhura Kamat, Vishakha Kurlawala, Aparajita Chakraborty, Priyanka Vyas, Radhika Vaishnav, Sanjeeva Srivastava

Introduction: The challenges posed by emergent strains of SARS-CoV-2 need to be tackled by contemporary scientific approaches, with proteomics playing a significant role.

Areas covered: In this review, we provide a brief synthesis of the impact of proteomics technologies in elucidating disease pathogenesis and classifiers for the prognosis of COVID-19 and propose proteomics methodologies that could play a crucial role in understanding emerging variants and their altered disease pathology. From aiding the design of novel drug candidates to facilitating the identification of T cell vaccine targets, we have discussed the impact of proteomics methods in COVID-19 research. Techniques varied as mass spectrometry, single-cell proteomics, multiplexed ELISA arrays, high-density proteome arrays, surface plasmon resonance, immunopeptidomics, and in silico docking studies that have helped augment the fight against existing diseases were useful in preparing us to tackle SARS-CoV-2 variants. We also propose an action plan for a pipeline to combat emerging pandemics using proteomics technology by adopting uniform standard operating procedures and unified data analysis paradigms.

Expert opinion: The knowledge about the use of diverse proteomics approaches for COVID-19 investigation will provide a framework for future basic research, better infectious disease prevention strategies, improved diagnostics, and targeted therapeutics.

新出现的SARS-CoV-2毒株所带来的挑战需要通过当代科学方法来解决,蛋白质组学在其中发挥着重要作用。涉及领域:在这篇综述中,我们简要地综合了蛋白质组学技术在阐明疾病发病机制和分类因子对COVID-19预后的影响,并提出了蛋白质组学方法,可以在理解新出现的变异及其改变的疾病病理中发挥关键作用。从帮助设计新的候选药物到促进T细胞疫苗靶点的鉴定,我们讨论了蛋白质组学方法在COVID-19研究中的影响。质谱法、单细胞蛋白质组学、多重ELISA阵列、高密度蛋白质组学阵列、表面等离子体共振、免疫肽组学和硅对接研究等各种技术有助于增强对现有疾病的对抗,有助于我们准备应对SARS-CoV-2变体。我们还提出了一项行动计划,通过采用统一的标准操作程序和统一的数据分析范式,建立一个利用蛋白质组学技术对抗新出现的流行病的管道。专家意见:关于使用多种蛋白质组学方法进行COVID-19调查的知识将为未来的基础研究、更好的传染病预防策略、改进的诊断和靶向治疗提供框架。
{"title":"Rise of the SARS-CoV-2 Variants: can proteomics be the silver bullet?","authors":"Arup Acharjee,&nbsp;Joshua Stephen Kingsly,&nbsp;Madhura Kamat,&nbsp;Vishakha Kurlawala,&nbsp;Aparajita Chakraborty,&nbsp;Priyanka Vyas,&nbsp;Radhika Vaishnav,&nbsp;Sanjeeva Srivastava","doi":"10.1080/14789450.2022.2085564","DOIUrl":"https://doi.org/10.1080/14789450.2022.2085564","url":null,"abstract":"<p><strong>Introduction: </strong>The challenges posed by emergent strains of SARS-CoV-2 need to be tackled by contemporary scientific approaches, with proteomics playing a significant role.</p><p><strong>Areas covered: </strong>In this review, we provide a brief synthesis of the impact of proteomics technologies in elucidating disease pathogenesis and classifiers for the prognosis of COVID-19 and propose proteomics methodologies that could play a crucial role in understanding emerging variants and their altered disease pathology. From aiding the design of novel drug candidates to facilitating the identification of T cell vaccine targets, we have discussed the impact of proteomics methods in COVID-19 research. Techniques varied as mass spectrometry, single-cell proteomics, multiplexed ELISA arrays, high-density proteome arrays, surface plasmon resonance, immunopeptidomics, and <i>in silico</i> docking studies that have helped augment the fight against existing diseases were useful in preparing us to tackle SARS-CoV-2 variants. We also propose an action plan for a pipeline to combat emerging pandemics using proteomics technology by adopting uniform standard operating procedures and unified data analysis paradigms.</p><p><strong>Expert opinion: </strong>The knowledge about the use of diverse proteomics approaches for COVID-19 investigation will provide a framework for future basic research, better infectious disease prevention strategies, improved diagnostics, and targeted therapeutics.</p>","PeriodicalId":50463,"journal":{"name":"Expert Review of Proteomics","volume":null,"pages":null},"PeriodicalIF":3.4,"publicationDate":"2022-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10485452","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 2
An overview of technologies for MS-based proteomics-centric multi-omics. 以ms为中心的蛋白质组学多组学技术综述。
IF 3.4 3区 生物学 Q2 Biochemistry, Genetics and Molecular Biology Pub Date : 2022-03-01 DOI: 10.1080/14789450.2022.2070476
Andrew T Rajczewski, Pratik D Jagtap, Timothy J Griffin

Introduction: Mass spectrometry-based proteomics reveals dynamic molecular signatures underlying phenotypes reflecting normal and perturbed conditions in living systems. Although valuable on its own, the proteome has only one level of moleclar information, with the genome, epigenome, transcriptome, and metabolome, all providing complementary information. Multi-omic analysis integrating information from one or more of these other domains with proteomic information provides a more complete picture of molecular contributors to dynamic biological systems.

Areas covered: Here, we discuss the improvements to mass spectrometry-based technologies, focused on peptide-based, bottom-up approaches that have enabled deep, quantitative characterization of complex proteomes. These advances are facilitating the integration of proteomics data with other 'omic information, providing a more complete picture of living systems. We also describe the current state of bioinformatics software and approaches for integrating proteomics and other 'omics data, critical for enabling new discoveries driven by multi-omics.

Expert commentary: Multi-omics, centered on the integration of proteomics information with other 'omic information, has tremendous promise for biological and biomedical studies. Continued advances in approaches for generating deep, reliable proteomic data and bioinformatics tools aimed at integrating data across 'omic domains will ensure the discoveries offered by these multi-omic studies continue to increase.

简介:基于质谱的蛋白质组学揭示了动态分子特征,这些分子特征反映了生活系统中正常和受干扰的条件。尽管蛋白质组本身很有价值,但它只有一个水平的分子信息,基因组、表观基因组、转录组和代谢组都提供了互补的信息。多组学分析将来自一个或多个其他结构域的信息与蛋白质组学信息相结合,为动态生物系统的分子贡献者提供了更完整的图像。涵盖的领域:在这里,我们讨论了基于质谱技术的改进,重点是基于肽的自下而上的方法,这些方法已经能够对复杂蛋白质组进行深入的定量表征。这些进步促进了蛋白质组学数据与其他组学信息的整合,为生命系统提供了更完整的图像。我们还描述了生物信息学软件的现状以及整合蛋白质组学和其他组学数据的方法,这对于多组学驱动的新发现至关重要。专家评论:多组学以蛋白质组学信息与其他组学信息的整合为中心,在生物学和生物医学研究中具有巨大的前景。在生成深度、可靠的蛋白质组学数据的方法和旨在整合跨基因组域数据的生物信息学工具方面的持续进步,将确保这些多基因组学研究提供的发现继续增加。
{"title":"An overview of technologies for MS-based proteomics-centric multi-omics.","authors":"Andrew T Rajczewski,&nbsp;Pratik D Jagtap,&nbsp;Timothy J Griffin","doi":"10.1080/14789450.2022.2070476","DOIUrl":"https://doi.org/10.1080/14789450.2022.2070476","url":null,"abstract":"<p><strong>Introduction: </strong>Mass spectrometry-based proteomics reveals dynamic molecular signatures underlying phenotypes reflecting normal and perturbed conditions in living systems. Although valuable on its own, the proteome has only one level of moleclar information, with the genome, epigenome, transcriptome, and metabolome, all providing complementary information. Multi-omic analysis integrating information from one or more of these other domains with proteomic information provides a more complete picture of molecular contributors to dynamic biological systems.</p><p><strong>Areas covered: </strong>Here, we discuss the improvements to mass spectrometry-based technologies, focused on peptide-based, bottom-up approaches that have enabled deep, quantitative characterization of complex proteomes. These advances are facilitating the integration of proteomics data with other 'omic information, providing a more complete picture of living systems. We also describe the current state of bioinformatics software and approaches for integrating proteomics and other 'omics data, critical for enabling new discoveries driven by multi-omics.</p><p><strong>Expert commentary: </strong>Multi-omics, centered on the integration of proteomics information with other 'omic information, has tremendous promise for biological and biomedical studies. Continued advances in approaches for generating deep, reliable proteomic data and bioinformatics tools aimed at integrating data across 'omic domains will ensure the discoveries offered by these multi-omic studies continue to increase.</p>","PeriodicalId":50463,"journal":{"name":"Expert Review of Proteomics","volume":null,"pages":null},"PeriodicalIF":3.4,"publicationDate":"2022-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9613604/pdf/nihms-1806996.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9401171","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 7
期刊
Expert Review of Proteomics
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
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
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 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 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1