Comprehensive Physiology Wiley Online Library

Undiscovered Physiology of Transcript and Protein Networks

Full Article on Wiley Online Library



ABSTRACT

The past two decades have witnessed a rapid evolution in our ability to measure RNA and protein from biological systems. As a result, new principles have arisen regarding how information is processed in cells, how decisions are made, and the role of networks in biology. This essay examines this technological evolution, reviewing (and critiquing) the conceptual framework that has emerged to explain how RNA and protein networks control cellular function. We identify how future investigations into transcriptomes, proteomes, and other cellular networks will enable development of more robust, quantitative models of cellular behavior whilst also providing new avenues to use knowledge of biological networks to improve human health. © 2016 American Physiological Society. Compr Physiol 6:1851‐1872, 2016.

Comprehensive Physiology offers downloadable PowerPoint presentations of figures for non-profit, educational use, provided the content is not modified and full credit is given to the author and publication.

Download a PowerPoint presentation of all images


Figure 1. Figure 1. Evolution of RNA quantitation techniques toward a more comprehensive catalogue of RNA species. (A) The first microarray was published in 1995 by Patrick Brown and quantified 45 mRNA species simultaneously using hybridization to DNA probes [reprinted with permission (144)]. (B) Microarrays have since advanced to measure tens of thousands of RNAs, including noncoding RNA. Shown is a heatmap of 768 lncRNAs found by array to exhibit altered abundances in the blood between patients with and without left ventricular remodeling following myocardial infarction [reprinted with permission (89)]. In this case, transcriptome measurements enabled unbiased identification of disease progression biomarkers. (C) Due to the development of RNA‐sequencing and subsequent advances in the library preparation, sequencing and data analysis, quantification a greater diversity of RNA species on a transcriptome‐wide scale is now routine. Shown is a Sashimi plot displaying the relative abundances of different exons in an example measured from the hearts of wild‐type mice (red) and mice with a knockout of a splicing factor. y‐axis represents normalized RNA‐seq reads (expression), x‐axis represents genomic coordinates. The arcs are numbered to indicate the raw number of junction reads. Arcs with greater values bridge two exons that are more often spliced next to each other [reprinted with permission (77)]. While these data were acquired from mice that were experimentally manipulated to disrupt splicing, many studies find exon usage is an important component to the transcriptome regulation of cell‐type specificity, development, and disease.
Figure 2. Figure 2. RNA abundance and protein abundance both correlate better with ribosome occupancy than they do with each other. Expression analysis was performed on lymphoblastoid cell lines of diverse genetic backgrounds taken from the HapMap project. Genes were clustered into modules or neurons (hexagon, right panels) within a self‐organizing map based on similar expression profiles across four different measurements (protein abundance, translation efficiency, RNA abundance, and ribosome occupancy; left panel). The right panel displays the same self‐organizing map colored to portray the mean expression of the genes within the module based on the four different datasets. The authors ask if hexagons with similar mean expression by one measurement (either both colored red or both colored blue) also show similar expression when using an alternate measure of expression. Ribosome occupancy correlates with RNA expression and protein level better than RNA expression and protein level correlate with each other. Note, ribosome occupancy is defined by the total read counts for an RNA after ribosome profiling, while translation efficiency takes into account the total pool of RNA (RNA‐seq) in addition to the ribosome occupancy [reprinted with permission (23)].
Figure 3. Figure 3. Evolution of proteomics toward network analysis. (A) Two‐dimensional protein gels were published in 1975 (top panel, [reprinted with permission (113)]), and remained a common tool for identifying proteome‐level quantitative differences between samples up into the late 1990s (bottom panel [reprinted with permission (29)]). Bottom panel is a computer‐processed image of a silver‐stained 2D gel from a human dilated cardiomyopathy sample. Spots represent protein isoforms identifiable by their position in the gel (number indicates database protein ID). Note that PTM can shift a protein's location in the gel, providing additional information. Red spots indicate isoforms which were less abundant (weaker signal; similar to Western blot analysis) across the dilated cardiomyopathy patients as compared to ischemic cardiomyopathy samples run in a separate gel, and analyzed together using computer software. Note that this analysis reveals on average 1282 spots per sample, in the same general scale as LC/MS/MS analyses; however, the identification of the individual spots, when not coupled to mass spectrometry, remains imprecise. (B) By contrast, advances in mass spectrometry and sample preparation pipeline have enabled quantification of PTMs across entire signaling cascades from multiple conditions. Shown here is the known insulin signaling pathway curated from multiple databases, overlaid with phosphorylation quantitation (expressed as fold‐change) from a mass spectrometry analysis performed on liver samples from mice treated with PBS or insulin at two time points [reprinted with permission (64)]. These techniques are optimized for a focused subproteome, thus enabled thorough, dynamic measurements of the system, which go beyond identifying proteins into the realm of mapping biological processes within a network. (C) Shown is a protein‐protein interaction network from HeLa cells generated through combining coimmunoprecipitation followed by mass spectrometry for 1125 different proteins [reprinted with permission (58)]. Red indicates edges previously annotated in CORUM. On its own, this network represents a database to inform other protein interaction studies. However, the authors took this study a step further to compare their interaction network with the relative abundance of the proteins to infer complex stability. Thus, by comparing across networks, the omics datasets are able to generate new understanding of properties of the proteome.
Figure 4. Figure 4. Mass spectrometry techniques for building protein networks. (A) Peptides (circles; size indicates relative abundance) elute from the LC column into the mass spectrometer. In shotgun/bottom‐up proteomics, peptides are scanned in the MS1 and the most abundant ions selected for fragmentation and identification via multiple MS2 scans. In MRM, both the MS1 and MS2 scan are performed on predetermined m/z ratios set by the user to precisely quantify peptides of interest, including low abundant peptides. SWATH by contrast fragments all ions from the MS1 scan, resulting in many more MS2 scans, each containing spectra from many parent ions. (B) Upstream techniques can be used in conjunction with mass spectrometry to enable protein and PTM identification, quantitation, and spatial localization information used to build protein networks.
Figure 5. Figure 5. The role of genetics in gene expression is organ specific. To test the relationship between genetics, gene expression, and phenotype, we examined data from a panel of 37 genetically diverse, inbred mouse strains with microarray data from multiple organs: Macrophages with and without LPS stimulation (unpublished), striatum (120), hippocampus (120), bone marrow (38), and heart with and without isoproterenol (ISO) stimulation to induce heart failure (128). Strains were clustered based on expression of all genes on the microarray (All) or a class of genes known as the “fetal gene program” (Fetal), whose cardiac expression are considered to be biomarkers of heart failure. The relatedness between each strain‐by‐strain comparison (Euclidean) was compared across organs. If the relative similarity in expression between two strains is similar across two organs, those two organs cluster closer together on the dendrogram. We also incorporated genetic relatedness based on kinship matrix derived from SNPs (Genetics). Macrophages cluster according to genetics, suggesting that strains with similar genetics also show similar expression patterns in macrophages regardless of if we examine all genes, or the cardiac fetal genes, and even when examining expression after LPS stimulation. By contrast, other organs, such as bone marrow, have expression relationships that less closely match genetic relationships. For context, we compared the relationships between genetics versus mRNA expression to that of genetics versus cardiac phenotype [ejection fraction (EF) and heart weight/body weight (HW/BW), two indices which change in heart failure]. In some cases, the genetic relationship more closely matched the phenotype than the expression (basal EF), but in other cases it did not (EF after ISO). We hypothesized that the “fetal gene program” was an intermediate between genetics and phenotype, but found that it no more closely matched the phenotypic relationships than when we examined all genes together. These analyses indicate that the relationship between genetic variation, mRNA expression, and ultimately phenotype is buffered at each level. For example, complex SNP interactions and chromatin features may buffer the relationship between genetic variation and mRNA expression, while posttranscriptional and posttranslational processing as well as compartmentalization may buffer the relationship between mRNA and protein levels, with the relationship between protein and phenotype in turn buffered by protein network properties and interaction with other classes of molecules.
Figure 6. Figure 6. Spectrum of cognitive bias in basic and translational research. Implementation of discovery science and hypothesis‐driven research comprise a spectrum analogous to the “opportunity cost” principle. Points along the curve represent experiments where the opportunity cost is minimized, because some perfect balance between discovery and hypothesis is struck. Point A defines a species of research with very high uncertainty and little or no theoretical underpinning, but with the potential to be very novel. Point B defines another type in which highly focused and inherently biased research reaches full potential by maximizing prior knowledge. Studies that lie under the curve, due to shoddy or underexplored data or an experimental design that builds only incrementally on precedent, fail to meet the ideal balance of discovery and hypothesis [reprinted with permission (106)].


Figure 1. Evolution of RNA quantitation techniques toward a more comprehensive catalogue of RNA species. (A) The first microarray was published in 1995 by Patrick Brown and quantified 45 mRNA species simultaneously using hybridization to DNA probes [reprinted with permission (144)]. (B) Microarrays have since advanced to measure tens of thousands of RNAs, including noncoding RNA. Shown is a heatmap of 768 lncRNAs found by array to exhibit altered abundances in the blood between patients with and without left ventricular remodeling following myocardial infarction [reprinted with permission (89)]. In this case, transcriptome measurements enabled unbiased identification of disease progression biomarkers. (C) Due to the development of RNA‐sequencing and subsequent advances in the library preparation, sequencing and data analysis, quantification a greater diversity of RNA species on a transcriptome‐wide scale is now routine. Shown is a Sashimi plot displaying the relative abundances of different exons in an example measured from the hearts of wild‐type mice (red) and mice with a knockout of a splicing factor. y‐axis represents normalized RNA‐seq reads (expression), x‐axis represents genomic coordinates. The arcs are numbered to indicate the raw number of junction reads. Arcs with greater values bridge two exons that are more often spliced next to each other [reprinted with permission (77)]. While these data were acquired from mice that were experimentally manipulated to disrupt splicing, many studies find exon usage is an important component to the transcriptome regulation of cell‐type specificity, development, and disease.


Figure 2. RNA abundance and protein abundance both correlate better with ribosome occupancy than they do with each other. Expression analysis was performed on lymphoblastoid cell lines of diverse genetic backgrounds taken from the HapMap project. Genes were clustered into modules or neurons (hexagon, right panels) within a self‐organizing map based on similar expression profiles across four different measurements (protein abundance, translation efficiency, RNA abundance, and ribosome occupancy; left panel). The right panel displays the same self‐organizing map colored to portray the mean expression of the genes within the module based on the four different datasets. The authors ask if hexagons with similar mean expression by one measurement (either both colored red or both colored blue) also show similar expression when using an alternate measure of expression. Ribosome occupancy correlates with RNA expression and protein level better than RNA expression and protein level correlate with each other. Note, ribosome occupancy is defined by the total read counts for an RNA after ribosome profiling, while translation efficiency takes into account the total pool of RNA (RNA‐seq) in addition to the ribosome occupancy [reprinted with permission (23)].


Figure 3. Evolution of proteomics toward network analysis. (A) Two‐dimensional protein gels were published in 1975 (top panel, [reprinted with permission (113)]), and remained a common tool for identifying proteome‐level quantitative differences between samples up into the late 1990s (bottom panel [reprinted with permission (29)]). Bottom panel is a computer‐processed image of a silver‐stained 2D gel from a human dilated cardiomyopathy sample. Spots represent protein isoforms identifiable by their position in the gel (number indicates database protein ID). Note that PTM can shift a protein's location in the gel, providing additional information. Red spots indicate isoforms which were less abundant (weaker signal; similar to Western blot analysis) across the dilated cardiomyopathy patients as compared to ischemic cardiomyopathy samples run in a separate gel, and analyzed together using computer software. Note that this analysis reveals on average 1282 spots per sample, in the same general scale as LC/MS/MS analyses; however, the identification of the individual spots, when not coupled to mass spectrometry, remains imprecise. (B) By contrast, advances in mass spectrometry and sample preparation pipeline have enabled quantification of PTMs across entire signaling cascades from multiple conditions. Shown here is the known insulin signaling pathway curated from multiple databases, overlaid with phosphorylation quantitation (expressed as fold‐change) from a mass spectrometry analysis performed on liver samples from mice treated with PBS or insulin at two time points [reprinted with permission (64)]. These techniques are optimized for a focused subproteome, thus enabled thorough, dynamic measurements of the system, which go beyond identifying proteins into the realm of mapping biological processes within a network. (C) Shown is a protein‐protein interaction network from HeLa cells generated through combining coimmunoprecipitation followed by mass spectrometry for 1125 different proteins [reprinted with permission (58)]. Red indicates edges previously annotated in CORUM. On its own, this network represents a database to inform other protein interaction studies. However, the authors took this study a step further to compare their interaction network with the relative abundance of the proteins to infer complex stability. Thus, by comparing across networks, the omics datasets are able to generate new understanding of properties of the proteome.


Figure 4. Mass spectrometry techniques for building protein networks. (A) Peptides (circles; size indicates relative abundance) elute from the LC column into the mass spectrometer. In shotgun/bottom‐up proteomics, peptides are scanned in the MS1 and the most abundant ions selected for fragmentation and identification via multiple MS2 scans. In MRM, both the MS1 and MS2 scan are performed on predetermined m/z ratios set by the user to precisely quantify peptides of interest, including low abundant peptides. SWATH by contrast fragments all ions from the MS1 scan, resulting in many more MS2 scans, each containing spectra from many parent ions. (B) Upstream techniques can be used in conjunction with mass spectrometry to enable protein and PTM identification, quantitation, and spatial localization information used to build protein networks.


Figure 5. The role of genetics in gene expression is organ specific. To test the relationship between genetics, gene expression, and phenotype, we examined data from a panel of 37 genetically diverse, inbred mouse strains with microarray data from multiple organs: Macrophages with and without LPS stimulation (unpublished), striatum (120), hippocampus (120), bone marrow (38), and heart with and without isoproterenol (ISO) stimulation to induce heart failure (128). Strains were clustered based on expression of all genes on the microarray (All) or a class of genes known as the “fetal gene program” (Fetal), whose cardiac expression are considered to be biomarkers of heart failure. The relatedness between each strain‐by‐strain comparison (Euclidean) was compared across organs. If the relative similarity in expression between two strains is similar across two organs, those two organs cluster closer together on the dendrogram. We also incorporated genetic relatedness based on kinship matrix derived from SNPs (Genetics). Macrophages cluster according to genetics, suggesting that strains with similar genetics also show similar expression patterns in macrophages regardless of if we examine all genes, or the cardiac fetal genes, and even when examining expression after LPS stimulation. By contrast, other organs, such as bone marrow, have expression relationships that less closely match genetic relationships. For context, we compared the relationships between genetics versus mRNA expression to that of genetics versus cardiac phenotype [ejection fraction (EF) and heart weight/body weight (HW/BW), two indices which change in heart failure]. In some cases, the genetic relationship more closely matched the phenotype than the expression (basal EF), but in other cases it did not (EF after ISO). We hypothesized that the “fetal gene program” was an intermediate between genetics and phenotype, but found that it no more closely matched the phenotypic relationships than when we examined all genes together. These analyses indicate that the relationship between genetic variation, mRNA expression, and ultimately phenotype is buffered at each level. For example, complex SNP interactions and chromatin features may buffer the relationship between genetic variation and mRNA expression, while posttranscriptional and posttranslational processing as well as compartmentalization may buffer the relationship between mRNA and protein levels, with the relationship between protein and phenotype in turn buffered by protein network properties and interaction with other classes of molecules.


Figure 6. Spectrum of cognitive bias in basic and translational research. Implementation of discovery science and hypothesis‐driven research comprise a spectrum analogous to the “opportunity cost” principle. Points along the curve represent experiments where the opportunity cost is minimized, because some perfect balance between discovery and hypothesis is struck. Point A defines a species of research with very high uncertainty and little or no theoretical underpinning, but with the potential to be very novel. Point B defines another type in which highly focused and inherently biased research reaches full potential by maximizing prior knowledge. Studies that lie under the curve, due to shoddy or underexplored data or an experimental design that builds only incrementally on precedent, fail to meet the ideal balance of discovery and hypothesis [reprinted with permission (106)].
References
 1. An integrated encyclopedia of DNA elements in the human genome. Nature 489: 57‐74, 2012.
 2. Integrated genomic analyses of ovarian carcinoma. Nature 474: 609‐615, 2011.
 3. Addona TA , Shi X , Keshishian H , Mani DR , Burgess M , Gillette MA , Clauser KR , Shen D , Lewis GD , Farrell LA , Fifer MA , Sabatine MS , Gerszten RE , Carr SA . A pipeline that integrates the discovery and verification of plasma protein biomarkers reveals candidate markers for cardiovascular disease. Nat Biotechnol 29: 635‐643, 2011.
 4. Aebersold R , Mann M . Mass spectrometry‐based proteomics. Nature 422: 198‐207, 2003.
 5. Allocco DJ , Kohane IS , Butte AJ . Quantifying the relationship between co‐expression, co‐regulation and gene function. BMC Bioinformatics 5: 18, 2004.
 6. Aten JE , Fuller TF , Lusis AJ , Horvath S . Using genetic markers to orient the edges in quantitative trait networks: The NEO software. BMC Syst Biol 2: 34, 2008.
 7. Au KF , Jiang H , Lin L , Xing Y , Wong WH . Detection of splice junctions from paired‐end RNA‐seq data by SpliceMap. Nucleic Acids Res 38: 4570‐4578, 2010.
 8. Azeloglu EU , Iyengar R . Signaling networks: Information flow, computation, and decision making. Cold Spring Harb Perspect Biol 7: a005934, 2015.
 9. Baboo S , Cook PR . “Dark matter” worlds of unstable RNA and protein. Nucleus 5: 281‐286, 2014.
 10. Ballouz S , Verleyen W , Gillis J . Guidance for RNA‐seq co‐expression network construction and analysis: Safety in numbers. Bioinformatics 31: 2123‐2130, 2015.
 11. Bar‐Joseph Z , Gerber GK , Lee TI , Rinaldi NJ , Yoo JY , Robert F , Gordon DB , Fraenkel E , Jaakkola TS , Young RA , Gifford DK . Computational discovery of gene modules and regulatory networks. Nat Biotechnol 21: 1337‐1342, 2003.
 12. Barabasi AL , Gulbahce N , Loscalzo J . Network medicine: A network‐based approach to human disease. Nat Genet 12: 56‐68, 2010.
 13. Barabasi AL , Oltvai ZN . Network biology: Understanding the cell's functional organization. Nat Rev Genet 5: 101‐113, 2004.
 14. Battle A , Khan Z , Wang SH , Mitrano A , Ford MJ , Pritchard JK , Gilad Y . Genomic variation. Impact of regulatory variation from RNA to protein. Science 347: 664‐667, 2015.
 15. Bendall SC , Simonds EF , Qiu P , Amir el AD , Krutzik PO , Finck R , Bruggner RV , Melamed R , Trejo A , Ornatsky OI , Balderas RS , Plevritis SK , Sachs K , Pe'er D , Tanner SD , Nolan GP . Single‐cell mass cytometry of differential immune and drug responses across a human hematopoietic continuum. Science 332: 687‐696, 2011.
 16. Bengtsson M , Stahlberg A , Rorsman P , Kubista M . Gene expression profiling in single cells from the pancreatic islets of Langerhans reveals lognormal distribution of mRNA levels. Genome Res 15: 1388‐1392, 2005.
 17. Bentley DL . Coupling mRNA processing with transcription in time and space. Nat Rev Genet 15: 163‐175, 2014.
 18. Blagoev B , Ong SE , Kratchmarova I , Mann M . Temporal analysis of phosphotyrosine‐dependent signaling networks by quantitative proteomics. Nat Biotechnol 22: 1139‐1145, 2004.
 19. Boersema PJ , Aye TT , van Veen TA , Heck AJ , Mohammed S . Triplex protein quantification based on stable isotope labeling by peptide dimethylation applied to cell and tissue lysates. Proteomics 8: 4624‐4632, 2008.
 20. Braun P . Reproducibility restored—On toward the human interactome. Nat Methods 10: 301, 303, 2013.
 21. Brown MP , Grundy WN , Lin D , Cristianini N , Sugnet CW , Furey TS , Ares M, Jr. , Haussler D . Knowledge‐based analysis of microarray gene expression data by using support vector machines. Proc Natl Acad Sci U S A 97: 262‐267, 2000.
 22. Carter H , Hofree M , Ideker T . Genotype to phenotype via network analysis. Curr Opin Genet Dev 23: 611‐621, 2013.
 23. Cenik C , Cenik ES , Byeon GW , Grubert F , Candille SI , Spacek D , Alsallakh B , Tilgner H , Araya CL , Tang H , Ricci E , Snyder MP . Integrative analysis of RNA, translation, and protein levels reveals distinct regulatory variation across humans. Genome Res 25: 1610‐1621, 2015.
 24. Chen H , Orozco L , Wang J , Rau CD , Rubbi L , Ren S , Wang Y , Pellegrini M , Lusis AJ , Vondriska TM . DNA methylation indicates susceptibility to isoproterenol‐inducd cardiac pathology and is associated with chromatin states. Circ Res 118: 786‐797, 2016.
 25. Christoforou A , Arias AM , Lilley KS . Determining protein subcellular localization in mammalian cell culture with biochemical fractionation and iTRAQ 8‐plex quantification. Methods Mol Biol 1156: 157‐174, 2014.
 26. Chuang HY , Lee E , Liu YT , Lee D , Ideker T . Network‐based classification of breast cancer metastasis. Mol Syst Biol 3: 140, 2007.
 27. Cloonan N , Forrest AR , Kolle G , Gardiner BB , Faulkner GJ , Brown MK , Taylor DF , Steptoe AL , Wani S , Bethel G , Robertson AJ , Perkins AC , Bruce SJ , Lee CC , Ranade SS , Peckham HE , Manning JM , McKernan KJ , Grimmond SM . Stem cell transcriptome profiling via massive‐scale mRNA sequencing. Nat Methods 5: 613‐619, 2008.
 28. Cohen SM . Everything old is new again: (linc)RNAs make proteins! Embo J 33: 937‐938, 2014.
 29. Corbett JM , Why HJ , Wheeler CH , Richardson PJ , Archard LC , Yacoub MH , Dunn MJ . Cardiac protein abnormalities in dilated cardiomyopathy detected by two‐dimensional polyacrylamide gel electrophoresis. Electrophoresis 19: 2031‐2042, 1998.
 30. Core LJ , Waterfall JJ , Lis JT . Nascent RNA sequencing reveals widespread pausing and divergent initiation at human promoters. Science 322: 1845‐1848, 2008.
 31. Deng Q , Ramskold D , Reinius B , Sandberg R . Single‐cell RNA‐seq reveals dynamic, random monoallelic gene expression in mammalian cells. Science 343: 193‐196, 2014.
 32. Dimon MT , Sorber K , DeRisi JL . HMMSplicer: A tool for efficient and sensitive discovery of known and novel splice junctions in RNA‐Seq data. PLoS One 5: e13875, 2010.
 33. Domon B , Aebersold R . Options and considerations when selecting a quantitative proteomics strategy. Nat Biotechnol 28: 710‐721, 2010.
 34. Eberwine J , Sul JY , Bartfai T , Kim J . The promise of single‐cell sequencing. Nat Methods 11: 25‐27, 2014.
 35. Eisen MB , Spellman PT , Brown PO , Botstein D . Cluster analysis and display of genome‐wide expression patterns. Proc Natl Acad Sci U S A 95: 14863‐14868, 1998.
 36. Eng JK , McCormack AL , Yates JR . An approach to correlate tandem mass spectral data of peptides with amino acid sequences in a protein database. J Am Soc Mass Spectrom 5: 976‐989, 1994.
 37. Evans G , Wheeler CH , Corbett JM , Dunn MJ . Construction of HSC‐2DPAGE: A two‐dimensional gel electrophoresis database of heart proteins. Electrophoresis 18: 471‐479, 1997.
 38. Farber CR , Bennett BJ , Orozco L , Zou W , Lira A , Kostem E , Kang HM , Furlotte N , Berberyan A , Ghazalpour A , Suwanwela J , Drake TA , Eskin E , Wang QT , Teitelbaum SL , Lusis AJ . Mouse genome‐wide association and systems genetics identify Asxl2 as a regulator of bone mineral density and osteoclastogenesis. PLoS genetics 7: e1002038, 2011.
 39. Farh KK , Marson A , Zhu J , Kleinewietfeld M , Housley WJ , Beik S , Shoresh N , Whitton H , Ryan RJ , Shishkin AA , Hatan M , Carrasco‐Alfonso MJ , Mayer D , Luckey CJ , Patsopoulos NA , De Jager PL , Kuchroo VK , Epstein CB , Daly MJ , Hafler DA , Bernstein BE . Genetic and epigenetic fine mapping of causal autoimmune disease variants. Nature 518: 337‐343, 2015.
 40. Fenn JB , Mann M , Meng CK , Wong SF , Whitehouse CM . Electrospray ionization for mass spectrometry of large biomolecules. Science 246: 64‐71, 1989.
 41. Ferrell JE, Jr . Self‐perpetuating states in signal transduction: Positive feedback, double‐negative feedback and bistability. Curr Opin Cell Biol 14: 140‐148, 2002.
 42. Foss EJ , Radulovic D , Shaffer SA , Ruderfer DM , Bedalov A , Goodlett DR , Kruglyak L . Genetic basis of proteome variation in yeast. Nat Genet 39: 1369‐1375, 2007.
 43. Franklin S , Chen H , Mitchell‐Jordan S , Ren S , Wang Y , Vondriska TM . Quantitative analysis of the chromatin proteome in disease reveals remodeling principles and identifies high mobility group protein B2 as a regulator of hypertrophic growth. Mol Cell Proteomics 11: M111, 2012.
 44. Fu J , Keurentjes JJ , Bouwmeester H , America T , Verstappen FW , Ward JL , Beale MH , de Vos RC , Dijkstra M , Scheltema RA , Johannes F , Koornneef M , Vreugdenhil D , Breitling R , Jansen RC . System‐wide molecular evidence for phenotypic buffering in Arabidopsis. Nat Genet 41: 166‐167, 2009.
 45. Fullwood MJ , Wei CL , Liu ET , Ruan Y . Next‐generation DNA sequencing of paired‐end tags (PET) for transcriptome and genome analyses. Genome Res 19: 521‐532, 2009.
 46. Gao X , Wan J , Liu B , Ma M , Shen B , Qian SB . Quantitative profiling of initiating ribosomes in vivo. Nat Methods 12: 147‐153, 2015.
 47. Gavin AC , Aloy P , Grandi P , Krause R , Boesche M , Marzioch M , Rau C , Jensen LJ , Bastuck S , Dumpelfeld B , Edelmann A , Heurtier MA , Hoffman V , Hoefert C , Klein K , Hudak M , Michon AM , Schelder M , Schirle M , Remor M , Rudi T , Hooper S , Bauer A , Bouwmeester T , Casari G , Drewes G , Neubauer G , Rick JM , Kuster B , Bork P , Russell RB , Superti‐Furga G . Proteome survey reveals modularity of the yeast cell machinery. Nature 440: 631‐636, 2006.
 48. Ge H , Liu Z , Church GM , Vidal M . Correlation between transcriptome and interactome mapping data from Saccharomyces cerevisiae . Nat Genet 29: 482‐486, 2001.
 49. Gerstein MB , Kundaje A , Hariharan M , Landt SG , Yan KK , Cheng C , Mu XJ , Khurana E , Rozowsky J , Alexander R , Min R , Alves P , Abyzov A , Addleman N , Bhardwaj N , Boyle AP , Cayting P , Charos A , Chen DZ , Cheng Y , Clarke D , Eastman C , Euskirchen G , Frietze S , Fu Y , Gertz J , Grubert F , Harmanci A , Jain P , Kasowski M , Lacroute P , Leng J , Lian J , Monahan H , O'Geen H , Ouyang Z , Partridge EC , Patacsil D , Pauli F , Raha D , Ramirez L , Reddy TE , Reed B , Shi M , Slifer T , Wang J , Wu L , Yang X , Yip KY , Zilberman‐Schapira G , Batzoglou S , Sidow A , Farnham PJ , Myers RM , Weissman SM , Snyder M . Architecture of the human regulatory network derived from ENCODE data. Nature 489: 91‐100, 2012.
 50. Gibbs DL , Baratt A , Baric RS , Kawaoka Y , Smith RD , Orwoll ES , Katze MG , McWeeney SK . Protein co‐expression network analysis (ProCoNA). J Clin Bioinforma 3: 11, 2013.
 51. Gillet LC , Navarro P , Tate S , Rost H , Selevsek N , Reiter L , Bonner R , Aebersold R . Targeted data extraction of the MS/MS spectra generated by data‐independent acquisition: A new concept for consistent and accurate proteome analysis. Mol Cell Proteomics 11: O111 016717, 2012.
 52. Gonzalez E , Joly S . Impact of RNA‐seq attributes on false positive rates in differential expression analysis of de novo assembled transcriptomes. BMC Res Notes 6: 503, 2013.
 53. Gygi SP , Rochon Y , Franza BR , Aebersold R . Correlation between protein and mRNA abundance in yeast. Mol Cell Biol 19: 1720‐1730, 1999.
 54. Hafner M , Landthaler M , Burger L , Khorshid M , Hausser J , Berninger P , Rothballer A , Ascano M, Jr. , Jungkamp AC , Munschauer M , Ulrich A , Wardle GS , Dewell S , Zavolan M , Tuschl T . Transcriptome‐wide identification of RNA‐binding protein and microRNA target sites by PAR‐CLIP. Cell 141: 129‐141, 2010.
 55. Hafner M , Landthaler M , Burger L , Khorshid M , Hausser J , Berninger P , Rothballer A , Ascano M , Jungkamp AC , Munschauer M , Ulrich A , Wardle GS , Dewell S , Zavolan M , Tuschl T . PAR‐CliP–a method to identify transcriptome‐wide the binding sites of RNA binding proteins. J Vis Exp 41: e2034, 2010.
 56. Hartwell LH , Hopfield JJ , Leibler S , Murray AW . From molecular to modular cell biology. Nature 402: C47‐52, 1999.
 57. Hebenstreit D . Methods, challenges and potentials of single cell RNA‐seq. Biology (Basel) 1: 658‐667, 2012.
 58. Hein MY , Hubner NC , Poser I , Cox J , Nagaraj N , Toyoda Y , Gak IA , Weisswange I , Mansfeld J , Buchholz F , Hyman AA , Mann M . A human interactome in three quantitative dimensions organized by stoichiometries and abundances. Cell 163: 712‐723, 2015.
 59. Hestand MS , Klingenhoff A , Scherf M , Ariyurek Y , Ramos Y , van Workum W , Suzuki M , Werner T , van Ommen GJ , den Dunnen JT , Harbers M , t Hoen PA . Tissue‐specific transcript annotation and expression profiling with complementary next‐generation sequencing technologies. Nucleic Acids Res 38: e165, 2010.
 60. Hindorff LA , Sethupathy P , Junkins HA , Ramos EM , Mehta JP , Collins FS , Manolio TA . Potential etiologic and functional implications of genome‐wide association loci for human diseases and traits. Proc Natl Acad Sci U S A 106: 9362‐9367, 2009.
 61. Holme P , Huss M , Jeong H . Subnetwork hierarchies of biochemical pathways. Bioinformatics 19: 532‐538, 2003.
 62. Hu Z , Hung JH , Wang Y , Chang YC , Huang CL , Huyck M , DeLisi C . VisANT 3.5: Multi‐scale network visualization, analysis and inference based on the gene ontology. Nucleic Acids Res 37: W115‐121, 2009.
 63. Hughes AJ , Spelke DP , Xu Z , Kang CC , Schaffer DV , Herr AE . Single‐cell western blotting. Nat Methods 11: 749‐755, 2014.
 64. Humphrey SJ , Azimifar SB , Mann M . High‐throughput phosphoproteomics reveals in vivo insulin signaling dynamics. Nat Biotechnol 33: 990‐995, 2015.
 65. Huttlin EL , Ting L , Bruckner RJ , Gebreab F , Gygi MP , Szpyt J , Tam S , Zarraga G , Colby G , Baltier K , Dong R , Guarani V , Vaites LP , Ordureau A , Rad R , Erickson BK , Wuhr M , Chick J , Zhai B , Kolippakkam D , Mintseris J , Obar RA , Harris T , Artavanis‐Tsakonas S , Sowa ME , De Camilli P , Paulo JA , Harper JW , Gygi SP . The BioPlex network: A systematic exploration of the human interactome. Cell 162: 425‐440, 2015.
 66. Ibarra RU , Edwards JS , Palsson BO . Escherichia coli K‐12 undergoes adaptive evolution to achieve in silico predicted optimal growth. Nature 420: 186‐189, 2002.
 67. Ingolia NT . Ribosome profiling: New views of translation, from single codons to genome scale. Nat Rev Genet 15: 205‐213, 2014.
 68. Ingolia NT , Ghaemmaghami S , Newman JR , Weissman JS . Genome‐wide analysis in vivo of translation with nucleotide resolution using ribosome profiling. Science 324: 218‐223, 2009.
 69. Issa NT , Kruger J , Byers SW , Dakshanamurthy S . Drug repurposing a reality: From computers to the clinic. Expert Rev Clin Pharmacol 6: 95‐97, 2013.
 70. Jansen R , Greenbaum D , Gerstein M . Relating whole‐genome expression data with protein‐protein interactions. Genome Res 12: 37‐46, 2002.
 71. Jansen R , Yu H , Greenbaum D , Kluger Y , Krogan NJ , Chung S , Emili A , Snyder M , Greenblatt JF , Gerstein M . A Bayesian networks approach for predicting protein‐protein interactions from genomic data. Science 302: 449‐453, 2003.
 72. Jones PA . Functions of DNA methylation: Islands, start sites, gene bodies and beyond. Nat Rev Genet 13: 484‐492, 2012.
 73. Jovanovic M , Rooney MS , Mertins P , Przybylski D , Chevrier N , Satija R , Rodriguez EH , Fields AP , Schwartz S , Raychowdhury R , Mumbach MR , Eisenhaure T , Rabani M , Gennert D , Lu D , Delorey T , Weissman JS , Carr SA , Hacohen N , Regev A . Immunogenetics. Dynamic profiling of the protein life cycle in response to pathogens. Science 347: 1259038, 2015.
 74. Kanehisa M . A database for post‐genome analysis. Trends Genet 13: 375‐376, 1997.
 75. Kanehisa M , Goto S , Furumichi M , Tanabe M , Hirakawa M . KEGG for representation and analysis of molecular networks involving diseases and drugs. Nucleic Acids Res 38: D355‐360, 2010.
 76. Katz Y , Wang ET , Airoldi EM , Burge CB . Analysis and design of RNA sequencing experiments for identifying isoform regulation. Nat Methods 7: 1009‐1015, 2010.
 77. Katz Y , Wang ET , Silterra J , Schwartz S , Wong B , Thorvaldsdottir H , Robinson JT , Mesirov JP , Airoldi EM , Burge CB . Quantitative visualization of alternative exon expression from RNA‐seq data. Bioinformatics 31: 2400‐2402, 2015.
 78. Kim KT , Lee HW , Lee HO , Kim SC , Seo YJ , Chung W , Eum HH , Nam DH , Kim J , Joo KM , Park WY . Single‐cell mRNA sequencing identifies subclonal heterogeneity in anti‐cancer drug responses of lung adenocarcinoma cells. Genome Biol 16: 127, 2015.
 79. Kim MS , Pinto SM , Getnet D , Nirujogi RS , Manda SS , Chaerkady R , Madugundu AK , Kelkar DS , Isserlin R , Jain S , Thomas JK , Muthusamy B , Leal‐Rojas P , Kumar P , Sahasrabuddhe NA , Balakrishnan L , Advani J , George B , Renuse S , Selvan LD , Patil AH , Nanjappa V , Radhakrishnan A , Prasad S , Subbannayya T , Raju R , Kumar M , Sreenivasamurthy SK , Marimuthu A , Sathe GJ , Chavan S , Datta KK , Subbannayya Y , Sahu A , Yelamanchi SD , Jayaram S , Rajagopalan P , Sharma J , Murthy KR , Syed N , Goel R , Khan AA , Ahmad S , Dey G , Mudgal K , Chatterjee A , Huang TC , Zhong J , Wu X , Shaw PG , Freed D , Zahari MS , Mukherjee KK , Shankar S , Mahadevan A , Lam H , Mitchell CJ , Shankar SK , Satishchandra P , Schroeder JT , Sirdeshmukh R , Maitra A , Leach SD , Drake CG , Halushka MK , Prasad TS , Hruban RH , Kerr CL , Bader GD , Iacobuzio‐Donahue CA , Gowda H , Pandey A . A draft map of the human proteome. Nature 509: 575‐581, 2014.
 80. King HA , Gerber AP . Translatome profiling: Methods for genome‐scale analysis of mRNA translation. Brief Funct Genomics 15: 22‐31, 2014.
 81. Kito K , Kawaguchi N , Okada S , Ito T . Discrimination between stable and dynamic components of protein complexes by means of quantitative proteomics. Proteomics 8: 2366‐2370, 2008.
 82. Klijn C , Durinck S , Stawiski EW , Haverty PM , Jiang Z , Liu H , Degenhardt J , Mayba O , Gnad F , Liu J , Pau G , Reeder J , Cao Y , Mukhyala K , Selvaraj SK , Yu M , Zynda GJ , Brauer MJ , Wu TD , Gentleman RC , Manning G , Yauch RL , Bourgon R , Stokoe D , Modrusan Z , Neve RM , de Sauvage FJ , Settleman J , Seshagiri S , Zhang Z . A comprehensive transcriptional portrait of human cancer cell lines. Nat Biotechnol 33: 306‐312, 2015.
 83. Konig J , Zarnack K , Rot G , Curk T , Kayikci M , Zupan B , Turner DJ , Luscombe NM , Ule J . iCLIP reveals the function of hnRNP particles in splicing at individual nucleotide resolution. Nat Struct Mol Biol 17: 909‐915, 2010.
 84. Kratz A , Carninci P . The devil in the details of RNA‐seq. Nat Biotechnol 32: 882‐884, 2014.
 85. Krogan NJ , Cagney G , Yu H , Zhong G , Guo X , Ignatchenko A , Li J , Pu S , Datta N , Tikuisis AP , Punna T , Peregrin‐Alvarez JM , Shales M , Zhang X , Davey M , Robinson MD , Paccanaro A , Bray JE , Sheung A , Beattie B , Richards DP , Canadien V , Lalev A , Mena F , Wong P , Starostine A , Canete MM , Vlasblom J , Wu S , Orsi C , Collins SR , Chandran S , Haw R , Rilstone JJ , Gandi K , Thompson NJ , Musso G , St Onge P , Ghanny S , Lam MH , Butland G , Altaf‐Ul AM , Kanaya S , Shilatifard A , O'Shea E , Weissman JS , Ingles CJ , Hughes TR , Parkinson J , Gerstein M , Wodak SJ , Emili A , Greenblatt JF . Global landscape of protein complexes in the yeast Saccharomyces cerevisiae . Nature 440: 637‐643, 2006.
 86. Krupp M , Marquardt JU , Sahin U , Galle PR , Castle J , Teufel A . RNA‐Seq Atlas—A reference database for gene expression profiling in normal tissue by next‐generation sequencing. Bioinformatics 28: 1184‐1185, 2012.
 87. Krutzik PO , Nolan GP . Fluorescent cell barcoding in flow cytometry allows high‐throughput drug screening and signaling profiling. Nat Methods 3: 361‐368, 2006.
 88. Krutzik PO , Trejo A , Schulz KR , Nolan GP . Phospho flow cytometry methods for the analysis of kinase signaling in cell lines and primary human blood samples. Methods Mol Biol 699: 179‐202, 2011.
 89. Kumarswamy R , Bauters C , Volkmann I , Maury F , Fetisch J , Holzmann A , Lemesle G , de Groote P , Pinet F , Thum T . Circulating long noncoding RNA, LIPCAR, predicts survival in patients with heart failure. Circ Res 114: 1569‐1575, 2014.
 90. Langfelder P , Horvath S . Eigengene networks for studying the relationships between co‐expression modules. BMC Syst Biol 1: 54, 2007.
 91. Langfelder P , Horvath S . WGCNA: An R package for weighted correlation network analysis. BMC Bioinformatics 9: 559, 2008.
 92. Lee S , Liu B , Huang SX , Shen B , Qian SB . Global mapping of translation initiation sites in mammalian cells at single‐nucleotide resolution. Proc Natl Acad Sci U S A 109: E2424‐2432, 2012.
 93. Lee S , Nguyen HM , Kang C . Tiny abortive initiation transcripts exert antitermination activity on an RNA hairpin‐dependent intrinsic terminator. Nucleic Acids Res 38: 6045‐6053, 2010.
 94. Li B , Ruotti V , Stewart RM , Thomson JA , Dewey CN . RNA‐Seq gene expression estimation with read mapping uncertainty. Bioinformatics 26: 493‐500, 2010.
 95. Li JJ , Biggin MD . Gene expression. Statistics requantitates the central dogma. Science 347: 1066‐1067, 2015.
 96. Li W , Dai C , Kang S , Zhou XJ . Integrative analysis of many RNA‐seq datasets to study alternative splicing. Methods 67: 313‐324, 2014.
 97. Lister R , O'Malley RC , Tonti‐Filippini J , Gregory BD , Berry CC , Millar AH , Ecker JR . Highly integrated single‐base resolution maps of the epigenome in Arabidopsis . Cell 133: 523‐536, 2008.
 98. Liu Y , Zhou J , White KP . RNA‐seq differential expression studies: More sequence or more replication? Bioinformatics 30: 301‐304, 2014.
 99. Lu P , Vogel C , Wang R , Yao X , Marcotte EM . Absolute protein expression profiling estimates the relative contributions of transcriptional and translational regulation. Nat Biotechnol 25: 117‐124, 2007.
 100. Marian AJ . Modeling human disease phenotype in model organisms: “It's only a model!”. Circ Res 109: 356‐359, 2011.
 101. Marioni JC , Mason CE , Mane SM , Stephens M , Gilad Y . RNA‐seq: An assessment of technical reproducibility and comparison with gene expression arrays. Genome Res 18: 1509‐1517, 2008.
 102. Mestas J , Hughes CC . Of mice and not men: Differences between mouse and human immunology. J Immunol 172: 2731‐2738, 2004.
 103. Milhorn HT, Jr , Benton R , Ross R , Guyton AC . A Mathematical Model of the Human Respiratory Control System. Biophys J 5: 27‐46, 1965.
 104. Min IM , Waterfall JJ , Core LJ , Munroe RJ , Schimenti J , Lis JT . Regulating RNA polymerase pausing and transcription elongation in embryonic stem cells. Genes Dev 25: 742‐754, 2011.
 105. Molkentin JD , Robbins J . With great power comes great responsibility: Using mouse genetics to study cardiac hypertrophy and failure. J Mol Cell Cardiol 46: 130‐136, 2009.
 106. Monte E , Lopez R , Vondriska TM . Not low hanging but still sweet: Metabolic proteomes in cardiovascular disease. J Mol Cell Cardiol 90: 70‐73, 2015.
 107. Mortazavi A , Williams BA , McCue K , Schaeffer L , Wold B . Mapping and quantifying mammalian transcriptomes by RNA‐Seq. Nat Methods 5: 621‐628, 2008.
 108. Nagalakshmi U , Wang Z , Waern K , Shou C , Raha D , Gerstein M , Snyder M . The transcriptional landscape of the yeast genome defined by RNA sequencing. Science 320: 1344‐1349, 2008.
 109. Neilson KA , Ali NA , Muralidharan S , Mirzaei M , Mariani M , Assadourian G , Lee A , van Sluyter SC , Haynes PA . Less label, more free: Approaches in label‐free quantitative mass spectrometry. Proteomics 11: 535‐553, 2011.
 110. Nesvizhskii AI . Proteogenomics: Concepts, applications and computational strategies. Nat Methods 11: 1114‐1125, 2014.
 111. Newman JR , Ghaemmaghami S , Ihmels J , Breslow DK , Noble M , DeRisi JL , Weissman JS . Single‐cell proteomic analysis of S. cerevisiae reveals the architecture of biological noise. Nature 441: 840‐846, 2006.
 112. Niehrs C , Pollet N . Synexpression groups in eukaryotes. Nature 402: 483‐487, 1999.
 113. O'Farrell PH . High resolution two‐dimensional electrophoresis of proteins. J Biol Chem 250: 4007‐4021, 1975.
 114. O'Farrell PH . High resolution two‐dimensional electrophoresis of proteins. J Biol Chem 250: 4007‐4021, 1975.
 115. Olsen JV , Mann M . Status of large‐scale analysis of post‐translational modifications by mass spectrometry. Mol Cell Proteomics 12: 3444‐3452, 2013.
 116. Ong SE , Blagoev B , Kratchmarova I , Kristensen DB , Steen H , Pandey A , Mann M . Stable isotope labeling by amino acids in cell culture, SILAC, as a simple and accurate approach to expression proteomics. Mol Cell Proteomics 1: 376‐386, 2002.
 117. Orozco LD , Morselli M , Rubbi L , Guo W , Go J , Shi H , Lopez D , Furlotte NA , Bennett BJ , Farber CR , Ghazalpour A , Zhang MQ , Bahous R , Rozen R , Lusis AJ , Pellegrini M . Epigenome‐wide association of liver methylation patterns and complex metabolic traits in mice. Cell Metabolism 21: 905‐917, 2015.
 118. Pagel O , Loroch S , Sickmann A , Zahedi RP . Current strategies and findings in clinically relevant post‐translational modification‐specific proteomics. Expert Rev Proteomics 12: 235‐253, 2015.
 119. Palahniuk C . Fight club. New York, NY: W.W. Norton, 1996.
 120. Park CC , Gale GD , de Jong S , Ghazalpour A , Bennett BJ , Farber CR , Langfelder P , Lin A , Khan AH , Eskin E , Horvath S , Lusis AJ , Ophoff RA , Smith DJ . Gene networks associated with conditional fear in mice identified using a systems genetics approach. BMC Syst Biol 5: 43, 2011.
 121. Pelechano V , Wei W , Steinmetz LM . Extensive transcriptional heterogeneity revealed by isoform profiling. Nature 497: 127‐131, 2013.
 122. Pennington SR , Wilkins MR , Hochstrasser DF , Dunn MJ . Proteome analysis: From protein characterization to biological function. Trends Cell Biol 7: 168‐173, 1997.
 123. Perez‐Riverol Y , Alpi E , Wang R , Hermjakob H , Vizcaino JA . Making proteomics data accessible and reusable: Current state of proteomics databases and repositories. Proteomics 15: 930‐949, 2015.
 124. Pflieger D , Gonnet F , de la Fuente van Bentem S , Hirt H , de la Fuente A . Linking the proteins–elucidation of proteome‐scale networks using mass spectrometry. Mass Spectrom Rev 30: 268‐297, 2011.
 125. Qu Z , Garfinkel A , Weiss JN , Nivala M . Multi‐scale modeling in biology: How to bridge the gaps between scales? Prog Biophys Mol Biol 107: 21‐31, 2011.
 126. Rakyan VK , Down TA , Balding DJ , Beck S . Epigenome‐wide association studies for common human diseases. Nat Rev Genet 12: 529‐541, 2011.
 127. Ramskold D , Luo S , Wang YC , Li R , Deng Q , Faridani OR , Daniels GA , Khrebtukova I , Loring JF , Laurent LC , Schroth GP , Sandberg R . Full‐length mRNA‐Seq from single‐cell levels of RNA and individual circulating tumor cells. Nat Biotechnol 30: 777‐782, 2012.
 128. Rau CD , Wang J , Avetisyan R , Romay M , Ren S , Wang Y , Lusis AJ . Mapping genetic contributions to cardiac pathology induced by beta‐adrenergic stimulation in mice. Circ Cardiovasc Gene 8: 40‐49, 2015.
 129. Rau CD , Wisniewski N , Orozco LD , Bennett B , Weiss J , Lusis AJ . Maximal information component analysis: A novel non‐linear network analysis method. Front Genet 4: 28, 2013.
 130. Ravasz E , Somera AL , Mongru DA , Oltvai ZN , Barabasi AL . Hierarchical organization of modularity in metabolic networks. Science 297: 1551‐1555, 2002.
 131. Rhee HW , Zou P , Udeshi ND , Martell JD , Mootha VK , Carr SA , Ting AY . Proteomic mapping of mitochondria in living cells via spatially restricted enzymatic tagging. Science 339: 1328‐1331, 2013.
 132. Rinner O , Seebacher J , Walzthoeni T , Mueller LN , Beck M , Schmidt A , Mueller M , Aebersold R . Identification of cross‐linked peptides from large sequence databases. Nat Methods 5: 315‐318, 2008.
 133. Rolland T , Tasan M , Charloteaux B , Pevzner SJ , Zhong Q , Sahni N , Yi S , Lemmens I , Fontanillo C , Mosca R , Kamburov A , Ghiassian SD , Yang X , Ghamsari L , Balcha D , Begg BE , Braun P , Brehme M , Broly MP , Carvunis AR , Convery‐Zupan D , Corominas R , Coulombe‐Huntington J , Dann E , Dreze M , Dricot A , Fan C , Franzosa E , Gebreab F , Gutierrez BJ , Hardy MF , Jin M , Kang S , Kiros R , Lin GN , Luck K , MacWilliams A , Menche J , Murray RR , Palagi A , Poulin MM , Rambout X , Rasla J , Reichert P , Romero V , Ruyssinck E , Sahalie JM , Scholz A , Shah AA , Sharma A , Shen Y , Spirohn K , Tam S , Tejeda AO , Trigg SA , Twizere JC , Vega K , Walsh J , Cusick ME , Xia Y , Barabasi AL , Iakoucheva LM , Aloy P , De Las Rivas J , Tavernier J , Calderwood MA , Hill DE , Hao T , Roth FP , Vidal M . A proteome‐scale map of the human interactome network. Cell 159: 1212‐1226, 2014.
 134. Rosa‐Garrido M , Karbassi E , Monte E , Vondriska TM . Regulation of chromatin structure in the cardiovascular system. Circ J 77: 1389‐1398, 2013.
 135. Rost HL , Malmstrom L , Aebersold R . Reproducible quantitative proteotype data matrices for systems biology. Mol Biol Cell 26: 3926‐3931, 2015.
 136. Rost HL , Rosenberger G , Navarro P , Gillet L , Miladinovic SM , Schubert OT , Wolski W , Collins BC , Malmstrom J , Malmstrom L , Aebersold R . OpenSWATH enables automated, targeted analysis of data‐independent acquisition MS data. Nat Biotechnol 32: 219‐223, 2014.
 137. Roux KJ , Kim DI , Burke B . BioID: A screen for protein‐protein interactions. Curr Protoc Protein Sci 74: 23, 2013.
 138. Roux KJ , Kim DI , Raida M , Burke B . A promiscuous biotin ligase fusion protein identifies proximal and interacting proteins in mammalian cells. J Cell Biol 196: 801‐810, 2012.
 139. Ruan X , Ruan Y . Genome wide full‐length transcript analysis using 5′ and 3′ paired‐end‐tag next generation sequencing (RNA‐PET). Methods Mol Biol 809: 535‐562, 2012.
 140. Ruepp A , Waegele B , Lechner M , Brauner B , Dunger‐Kaltenbach I , Fobo G , Frishman G , Montrone C , Mewes HW . CORUM: The comprehensive resource of mammalian protein complexes–2009. Nucleic Acids Res 38: D497‐D501, 2010.
 141. Sahni N , Yi S , Taipale M , Fuxman Bass JI , Coulombe‐Huntington J , Yang F , Peng J , Weile J , Karras GI , Wang Y , Kovacs IA , Kamburov A , Krykbaeva I , Lam MH , Tucker G , Khurana V , Sharma A , Liu YY , Yachie N , Zhong Q , Shen Y , Palagi A , San‐Miguel A , Fan C , Balcha D , Dricot A , Jordan DM , Walsh JM , Shah AA , Yang X , Stoyanova AK , Leighton A , Calderwood MA , Jacob Y , Cusick ME , Salehi‐Ashtiani K , Whitesell LJ , Sunyaev S , Berger B , Barabasi AL , Charloteaux B , Hill DE , Hao T , Roth FP , Xia Y , Walhout AJ , Lindquist S , Vidal M . Widespread macromolecular interaction perturbations in human genetic disorders. Cell 161: 647‐660, 2015.
 142. Sakai H , Naito K , Ogiso‐Tanaka E , Takahashi Y , Iseki K , Muto C , Satou K , Teruya K , Shiroma A , Shimoji M , Hirano T , Itoh T , Kaga A , Tomooka N . The power of single molecule real‐time sequencing technology in the de novo assembly of a eukaryotic genome. Sci Rep 5: 16780, 2015.
 143. Sanchez A , Golding I . Genetic determinants and cellular constraints in noisy gene expression. Science 342: 1188‐1193, 2013.
 144. Schena M , Shalon D , Davis RW , Brown PO . Quantitative monitoring of gene expression patterns with a complementary DNA microarray. Science 270: 467‐470, 1995.
 145. Schmidt C , Urlaub H . iTRAQ‐labeling of in‐gel digested proteins for relative quantification. Methods Mol Biol 564: 207‐226, 2009.
 146. Schwanhausser B , Busse D , Li N , Dittmar G , Schuchhardt J , Wolf J , Chen W , Selbach M . Global quantification of mammalian gene expression control. Nature 473: 337‐342, 2011.
 147. Shalek AK , Satija R , Adiconis X , Gertner RS , Gaublomme JT , Raychowdhury R , Schwartz S , Yosef N , Malboeuf C , Lu D , Trombetta JJ , Gennert D , Gnirke A , Goren A , Hacohen N , Levin JZ , Park H , Regev A . Single‐cell transcriptomics reveals bimodality in expression and splicing in immune cells. Nature 498: 236‐240, 2013.
 148. Shannon P , Markiel A , Ozier O , Baliga NS , Wang JT , Ramage D , Amin N , Schwikowski B , Ideker T . Cytoscape: A software environment for integrated models of biomolecular interaction networks. Genome Res 13: 2498‐2504, 2003.
 149. Shen S , Park JW , Huang J , Dittmar KA , Lu ZX , Zhou Q , Carstens RP , Xing Y . MATS: A Bayesian framework for flexible detection of differential alternative splicing from RNA‐Seq data. Nucleic Acids Res 40: e61, 2012.
 150. Shi Q , Qin L , Wei W , Geng F , Fan R , Shin YS , Guo D , Hood L , Mischel PS , Heath JR . Single‐cell proteomic chip for profiling intracellular signaling pathways in single tumor cells. Proc Natl Acad Sci U S A 109: 419‐424, 2012.
 151. Shi Z , Wang J , Zhang B . NetGestalt: Integrating multidimensional omics data over biological networks. Nat Methods 10: 597‐598, 2013.
 152. Sims D , Sudbery I , Ilott NE , Heger A , Ponting CP . Sequencing depth and coverage: Key considerations in genomic analyses. Nat Rev Genet 15: 121‐132, 2014.
 153. Siuti N , Kelleher NL . Decoding protein modifications using top‐down mass spectrometry. Nat Methods 4: 817‐821, 2007.
 154. Smith LM , Kelleher NL . Proteoform: A single term describing protein complexity. Nat Methods 10: 186‐187, 2013.
 155. Syka JE , Coon JJ , Schroeder MJ , Shabanowitz J , Hunt DF . Peptide and protein sequence analysis by electron transfer dissociation mass spectrometry. Proc Natl Acad Sci U S A 101: 9528‐9533, 2004.
 156. Tessarz P , Kouzarides T . Histone core modifications regulating nucleosome structure and dynamics. Nat Rev Mol Cell Biol 15: 703‐708, 2014.
 157. Tran JC , Zamdborg L , Ahlf DR , Lee JE , Catherman AD , Durbin KR , Tipton JD , Vellaichamy A , Kellie JF , Li M , Wu C , Sweet SM , Early BP , Siuti N , LeDuc RD , Compton PD , Thomas PM , Kelleher NL . Mapping intact protein isoforms in discovery mode using top‐down proteomics. Nature 480: 254‐258, 2011.
 158. Valen E , Pascarella G , Chalk A , Maeda N , Kojima M , Kawazu C , Murata M , Nishiyori H , Lazarevic D , Motti D , Marstrand TT , Tang MH , Zhao X , Krogh A , Winther O , Arakawa T , Kawai J , Wells C , Daub C , Harbers M , Hayashizaki Y , Gustincich S , Sandelin A , Carninci P . Genome‐wide detection and analysis of hippocampus core promoters using DeepCAGE. Genome Res 19: 255‐265, 2009.
 159. van Heesch S , van Iterson M , Jacobi J , Boymans S , Essers PB , de Bruijn E , Hao W , MacInnes AW , Cuppen E , Simonis M . Extensive localization of long noncoding RNAs to the cytosol and mono‐ and polyribosomal complexes. Genome Biol 15: R6, 2014.
 160. Velculescu VE , Zhang L , Vogelstein B , Kinzler KW . Serial analysis of gene expression. Science 270: 484‐487, 1995.
 161. Venkatesan K , Rual JF , Vazquez A , Stelzl U , Lemmens I , Hirozane‐Kishikawa T , Hao T , Zenkner M , Xin X , Goh KI , Yildirim MA , Simonis N , Heinzmann K , Gebreab F , Sahalie JM , Cevik S , Simon C , de Smet AS , Dann E , Smolyar A , Vinayagam A , Yu H , Szeto D , Borick H , Dricot A , Klitgord N , Murray RR , Lin C , Lalowski M , Timm J , Rau K , Boone C , Braun P , Cusick ME , Roth FP , Hill DE , Tavernier J , Wanker EE , Barabasi AL , Vidal M . An empirical framework for binary interactome mapping. Nat Methods 6: 83‐90, 2009.
 162. Vinayagam A , Zirin J , Roesel C , Hu Y , Yilmazel B , Samsonova AA , Neumuller RA , Mohr SE , Perrimon N . Integrating protein‐protein interaction networks with phenotypes reveals signs of interactions. Nat Methods 11: 94‐99, 2014.
 163. Vitting‐Seerup K , Porse BT , Sandelin A , Waage J . spliceR: An R package for classification of alternative splicing and prediction of coding potential from RNA‐seq data. BMC Bioinformatics 15: 81, 2014.
 164. Vogel C , Marcotte EM . Insights into the regulation of protein abundance from proteomic and transcriptomic analyses. Nat Rev Genet 13: 227‐232, 2012.
 165. Vondriska TM , Klein JB , Ping P . Use of functional proteomics to investigate PKC epsilon‐mediated cardioprotection: The signaling module hypothesis. Am J Physiol Heart Circ Physiol 280: H1434‐1441, 2001.
 166. Walker MG , Volkmuth W , Sprinzak E , Hodgson D , Klingler T . Prediction of gene function by genome‐scale expression analysis: Prostate cancer‐associated genes. Genome Res 9: 1198‐1203, 1999.
 167. Wang ET , Sandberg R , Luo S , Khrebtukova I , Zhang L , Mayr C , Kingsmore SF , Schroth GP , Burge CB . Alternative isoform regulation in human tissue transcriptomes. Nature 456: 470‐476, 2008.
 168. Wang K , Singh D , Zeng Z , Coleman SJ , Huang Y , Savich GL , He X , Mieczkowski P , Grimm SA , Perou CM , MacLeod JN , Chiang DY , Prins JF , Liu J . MapSplice: Accurate mapping of RNA‐seq reads for splice junction discovery. Nucleic Acids Res 38: e178, 2010.
 169. Washburn MP , Wolters D , Yates JR, III . Large‐scale analysis of the yeast proteome by multidimensional protein identification technology. Nat Biotechnol 19: 242‐247, 2001.
 170. Weinstein IB . Cancer. Addiction to oncogenes—The Achilles heal of cancer. Science 297: 63‐64, 2002.
 171. Weiss JN , Karma A , MacLellan WR , Deng M , Rau CD , Rees CM , Wang J , Wisniewski N , Eskin E , Horvath S , Qu Z , Wang Y , Lusis AJ . “Good enough solutions” and the genetics of complex diseases. Circ Res 111: 493‐504, 2012.
 172. Weiss JN , Karma A , MacLellan WR , Deng M , Rau CD , Rees CM , Wang J , Wisniewski N , Eskin E , Horvath S , Qu Z , Wang Y , Lusis AJ . “Good enough solutions” and the genetics of complex diseases. Circ Res 111: 493‐504, 2012.
 173. Wilczynska A , Bushell M . The complexity of miRNA‐mediated repression. Cell Death Differ 22: 22‐33, 2015.
 174. Wilhelm M , Schlegl J , Hahne H , Moghaddas Gholami A , Lieberenz M , Savitski MM , Ziegler E , Butzmann L , Gessulat S , Marx H , Mathieson T , Lemeer S , Schnatbaum K , Reimer U , Wenschuh H , Mollenhauer M , Slotta‐Huspenina J , Boese JH , Bantscheff M , Gerstmair A , Faerber F , Kuster B . Mass‐spectrometry‐based draft of the human proteome. Nature 509: 582‐587, 2014.
 175. Witze ES , Old WM , Resing KA , Ahn NG . Mapping protein post‐translational modifications with mass spectrometry. Nat Methods 4: 798‐806, 2007.
 176. Wu AR , Neff NF , Kalisky T , Dalerba P , Treutlein B , Rothenberg ME , Mburu FM , Mantalas GL , Sim S , Clarke MF , Quake SR . Quantitative assessment of single‐cell RNA‐sequencing methods. Nat Methods 11: 41‐46, 2014.
 177. Wu L , Candille SI , Choi Y , Xie D , Jiang L , Li‐Pook‐Than J , Tang H , Snyder M . Variation and genetic control of protein abundance in humans. Nature 499: 79‐82, 2013.
 178. Wu LF , Hughes TR , Davierwala AP , Robinson MD , Stoughton R , Altschuler SJ . Large‐scale prediction of Saccharomyces cerevisiae gene function using overlapping transcriptional clusters. Nat Genet 31: 255‐265, 2002.
 179. Wu Z , Wu H . Experimental Design and Power Calculation for RNA‐seq Experiments. Methods Mol Biol 1418: 379‐390, 2016.
 180. Xiang CC , Chen Y . cDNA microarray technology and its applications. Biotechnol Adv 18: 35‐46, 2000.
 181. Yates J, III . A century of mass spectrometry: From atoms to proteomes. Nat Methods 8: 5, 2011.
 182. Zhang B , Horvath S . A general framework for weighted gene co‐expression network analysis. Stat Appl Genet Mol Biol 4: Article17, 2005.
 183. Zhang Q , Xiao X . Genome sequence‐independent identification of RNA editing sites. Nat Methods 12: 347‐350, 2015.
 184. Zhang Y , Fonslow BR , Shan B , Baek MC , Yates JR, III . Protein analysis by shotgun/bottom‐up proteomics. Chem Rev 113: 2343‐2394, 2013.
 185. Zhao S , Fung‐Leung WP , Bittner A , Ngo K , Liu X . Comparison of RNA‐Seq and microarray in transcriptome profiling of activated T cells. PLoS One 9: e78644, 2014.
 186. Zhao W , He X , Hoadley KA , Parker JS , Hayes DN , Perou CM . Comparison of RNA‐Seq by poly (A) capture, ribosomal RNA depletion, and DNA microarray for expression profiling. BMC Genomics 15: 419, 2014.
 187. Zhernakova DV , de Klerk E , Westra HJ , Mastrokolias A , Amini S , Ariyurek Y , Jansen R , Penninx BW , Hottenga JJ , Willemsen G , de Geus EJ , Boomsma DI , Veldink JH , van den Berg LH , Wijmenga C , den Dunnen JT , van Ommen GJ , t Hoen PA , Franke L . DeepSAGE reveals genetic variants associated with alternative polyadenylation and expression of coding and non‐coding transcripts. PLoS Genet 9: e1003594, 2013.

Contact Editor

Submit a note to the editor about this article by filling in the form below.

* Required Field

How to Cite

Emma Monte, Manuel Rosa‐Garrido, Thomas M. Vondriska, Jessica Wang. Undiscovered Physiology of Transcript and Protein Networks. Compr Physiol 2016, 6: 1851-1872. doi: 10.1002/cphy.c160003