Top Banner
DOI: 10.1634/stemcells.2007-0107 published online Jun 7, 2007; Stem Cells Hossein Baharvand, Ali Fathi, Dennis van Hoof and Ghasem Hosseini Salekdeh Trends in Stem Cell Proteomics This information is current as of July 10, 2007 http://www.StemCells.com the World Wide Web at: The online version of this article, along with updated information and services, is located on 1549-4918. Carolina, 27701. © 2007 by AlphaMed Press, all rights reserved. Print ISSN: 1066-5099. Online ISSN: North owned, published, and trademarked by AlphaMed Press, 318 Blackwell Street, Suite 260, Durham, STEM CELLS® is a monthly publication, it has been published continuously since 1983. The Journal is and genomics; translational and clinical research; technology development. embryonic stem cells; tissue-specific stem cells; cancer stem cells; the stem cell niche; stem cell genetics STEM CELLS®, an international peer-reviewed journal, covers all aspects of stem cell research: by on July 10, 2007 www.StemCells.com Downloaded from
39

Trends in Stem Cell Proteomics

Nov 13, 2014

Download

Documents

Ali Fathi

this review article explain proteomics technologies applied to stem cell untill now
Welcome message from author
This document is posted to help you gain knowledge. Please leave a comment to let me know what you think about it! Share it to your friends and learn new things together.
Transcript
Page 1: Trends in Stem Cell Proteomics

DOI: 10.1634/stemcells.2007-0107 published online Jun 7, 2007; Stem Cells

Hossein Baharvand, Ali Fathi, Dennis van Hoof and Ghasem Hosseini Salekdeh Trends in Stem Cell Proteomics

This information is current as of July 10, 2007

http://www.StemCells.comthe World Wide Web at:

The online version of this article, along with updated information and services, is located on

1549-4918. Carolina, 27701. © 2007 by AlphaMed Press, all rights reserved. Print ISSN: 1066-5099. Online ISSN:

Northowned, published, and trademarked by AlphaMed Press, 318 Blackwell Street, Suite 260, Durham, STEM CELLS® is a monthly publication, it has been published continuously since 1983. The Journal is

and genomics; translational and clinical research; technology development.embryonic stem cells; tissue-specific stem cells; cancer stem cells; the stem cell niche; stem cell genetics STEM CELLS®, an international peer-reviewed journal, covers all aspects of stem cell research:

by on July 10, 2007 w

ww

.StemC

ells.comD

ownloaded from

Page 2: Trends in Stem Cell Proteomics

*Correspondence should be addressed to: Hossein Baharvand, Department of Stem Cells, Royan Institute, P.O. Box: 19395-4644, Tehran, Iran. Tel: +98-21-22172330 ; Fax: +98-21-22414532, Email: [email protected]; Or Ghasem Hosseini Salekdeh, Physiology and Proteomics Department, Agricultural Biotechnology Research Institute of Iran (ABRII), P.O.Box 31535-1897, Karaj, Iran. Tel: 0098-261-2702893; Fax: +98-261-2704539; Email: [email protected]; This work was supported by grants from the Royan Institute. Received February 8, 2007; accepted for publication May 4, 2007; first published online in STEM CELLS Express May 10, 2007; available online without subscription through the open access option. © AlphaMed Press 1066-5099/2007/30.00/0 doi: 10.1634/stemcells.2007-0107

STEM CELLS® STEM CELL GENETICS AND GENOMICS: CONCISE REVIEW Trends in Stem Cell Proteomics Hossein Baharvand 1*, Ali Fathi1, Dennis van Hoof 2,3, Ghasem Hosseini Salekdeh 4*

1. Department of Stem Cells, Royan Institute, Tehran, Iran; 2. Hubrecht Laboratory, Netherlands Institute of Developmental Biology, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands.;3. Department of Biomolecular Mass Spectrometry, Utrecht University, Sorbonnelaan 16, 3584 CA Utrecht, The Netherlands; 4. Department of Physiology and Proteomics, Agricultural Biotechnology Research Institute of Iran, Karaj, Iran. Key words. Stem cells • Heterogeneity • Proteomics • Mass spectrometry • Post-translational modification •

electrophoresis • Quantitative proteomics • Secretomics • Two-dimensional electrophoresis. ABSTRACT Gene expression analyses of stem cells (SCs) will help to uncover or further define signaling pathways and molecular mechanisms involved in the maintenance of self-renewal, pluripotency, and/or multipotency. In recent years, proteomic approaches have produced a wealth of data identifying proteins and mechanisms involved in SC proliferation and differentiation. Although many proteomics techniques have been developed and improved in peptide and protein separation as well as mass spectrometry, several important issues including sample heterogeneity, post-

translational modifications, protein-protein interaction, and high throughput quantification of hydrophobic and low abundant proteins still remain to be addressed and require further technical optimization. The following review summarizes the methodologies used and the information gathered with proteome analyses of SCs, and discusses biological and technical challenges for proteomic study of SCs.

INTRODUCTION Stem cells (SCs) are undifferentiated cells generally characterized by their functional capacity to both self-renew and to generate a large number of differentiated progeny cells [1]. Conventionally, SCs are either classified as those derived from embryo or adult tissues. Embryonic SCs (ESCs), embryonal carcinomal cells (ECCs), and embryonic germ cells are derived from the preimplantation embryo (e.g., inner cell mass of blastocyst, morula (for review see [2]), and single blastomeres [3], teratocarcinomas, and primordial germ cells, respectively. These cells are pluripotent, i.e. they have the ability to

form all embryonic germ layer derivatives, except extracellular tissues (e.g., placenta). SCs found in adult organisms are present in most tissues and are referred to as adult SCs, such as mesenchymal SCs (MSCs), hematopoetic SCs (HSCs), and neural SCs (NSCs) [4]. They are considered multipotent, since they can produce mature cell types of one or more lineages, but cannot reconstitute the organism as a whole. What determines SC potency largely depends on intrinsic properties of SCs as well as extrinsic cues provided by the niche (microenvironment where SCs reside). Because of their exceptional properties, SCs have the potential to be used for developmental biology, drug screening,

Stem Cells Express, published online June 7, 2007; doi:10.1634/stemcells.2007-0107

Copyright © 2007 AlphaMed Press

by on July 10, 2007 w

ww

.StemC

ells.comD

ownloaded from

Page 3: Trends in Stem Cell Proteomics

Trends in Stem Cell Proteomics

2

functional genomics applications, and regenerative medicine. Gene expression analyses of the SCs will help uncover and further define signaling pathways and molecular mechanisms involved in the maintenance of the undifferentiated state and initial loss of pluripotency and/or multipotency. A detailed understanding of these molecular mechanisms will, thus, be essential for the aforementioned SC applications. In contrast to the transcriptome, which is studied with microarrays [5-24], important issues of the proteome, such as protein amount, stability, subcellular localization, post-translational modifications (PTMs), and their interactions can be elucidated at proteome level. Wilkins et al. [25] coined the term “proteome” (PROTEins expressed by a genOME) to refer to the total set of proteins expressed in a cell, tissue or organism. Nowadays, two-dimensional gel electrophoresis (2-DE) and non 2-DE-based approaches are broadly applied to proteomic analyses. Applying proteomics to investigate the programs that control self-renewal, differentiation, and plasticity will provide valuable insight into how the factors involved induce differentiation of SCs to specific lineages. Recent reviews have comprehensively addressed various aspects that are relevant in the context of SC proteomics [26-29]. Here, we review various proteomics methodologies used to study SCs, proteome analyses of SCs, and discuss biological and technical challenges encountered with proteomic studies of SCs, and provide insight into how proteomics-based research is likely to develop. Proteomics: an overview of technology Sample preparation and protein extraction. Although proteomic analysis can be used for qualitative comparisons, it is much more informative when used quantitatively. The isolation of proteins from SCs and derivatives for proteome analyses is complicated. The

human genome harbors 26,000–31,000 protein-encoding genes [30], whereas the total number of human protein products, including splice variants and essential PTMs, has been estimated to be close to one million [31,32]. Another important factor for proteomic analysis is the dynamic range of protein concentrations; one cell can contain between one and more than 100,000 copies of a single protein [33]. A high dynamic range can be partially achieved by fractionation of the proteome into sub-proteomes, e.g. by applying affinity purification [34]. Reduction of a complex sample is also achieved by specific isolation of individual proteins or protein complexes. In general, hydrophobic membrane proteins are much more difficult to handle than hydrophilic proteins; hence, hydrophobic proteins require specific extraction procedures [35,36]. Two dimensional electrophoresis (2-DE). High-resolution 2-DE of proteins is the fundamental tool of proteomics and allows thousands of proteins to be analyzed simultaneously (Fig. 1). 2-DE has been available since 1975 [37]. In 1988, a basic protocol of electrophoresis with immobilized pH gradients (IPGs) was described [38]. The advent of IPGs for the first dimension has produced significant improvements in 2-DE separation with higher resolution, improved reproducibility, and higher loading capacity for preparative gels [39]. Other important technological advances in 2-DE include the development of sensitive protein stains and the use of in-gel sample application in contrast to loading at either anodic or cathodic ends of the gel. Mass spectrometry (MS). The most significant breakthrough in proteomics has been the application of MS. It allows the identification of proteins in the femtomole to picomole range and has superseded classical Edman N-terminal sequencing, which is less automated, less sensitive and requires an unblocked N-terminus [40,41]. The main components of a mass spectrometer are an ion source, one or several mass analyzers that measure the mass-to-charge

by on July 10, 2007 w

ww

.StemC

ells.comD

ownloaded from

Page 4: Trends in Stem Cell Proteomics

Trends in Stem Cell Proteomics

3

ratio (m/z) of the ionized analytes, and a detector that registers the number of ions at each m/z value (Fig 2). Identification of separated proteins from 2-DE gels using MS. This approach is commonly applied for the identification of proteins isolated from 2-DE. It usually begins with peptide mapping, initially suggested by Henzel and co-workers [42]. The separated proteins are digested with an enzyme, for instance trypsin, and the masses of the proteolytic peptides are measured with MS. The mass spectra are obtained with a relatively simple MS instrument, such as matrix-assisted laser desorption/ionization-time of flight (MALDI-TOF). The masses of the measured proteolytic peptides are compared to predicted proteolytic peptides from protein sequence databases. This step can be fully automated, but it requires the complete sequence of the protein or of the coding region of its gene to be present in the database. As more gene sequence data become available, the success rate of this method will increase. The method is now popularly called peptide mass fingerprinting (PMF). In some cases where peptide mapping does not provide sufficient information for confident identification, there is a need for more sophisticated instrumentation such as a MALDI-TOF/TOF or Q-TOF, which provide higher mass accuracy and sensitivity, and include peptide fragmentation and partial peptide sequence determination for several tryptic fragments (Fig. 2). MS-based protein profiling. Although 2-DE has been widely used for proteome analysis, this methodological approach has several limitations. For example, it is inadequate for the analysis of more complex mixtures, and detection as well as identification is strongly biased toward the more abundant proteins [43-45]. Moreover, hydrophobic proteins, such as membrane proteins, which are not readily soluble in aqueous media, are rarely detected with 2-DE [46].

In MS-based protein profiling, the proteins are enzymatically digested and subjected directly to MS. This system also referred to as ‘shotgun proteomics’, features a protein separation step coupled to a mass spectrometer with superior resolving power and dynamic mass range. Most popular at present are 2D (strong cation exchange/reversed phase) [47,48] or 3D (strong cation exchange/avidin/reversed phase) [49] chromatographic separation methods of peptide mixtures. The protein samples can also be pre-fractionated using SDS-PAGE or isoelectric focusing (IEF) prior to analysis. MS-based quantitative analysis. Several MS-based strategies have been developed that allow different samples to be compared quantitatively. In extracted ion current (XIC)-based quantification, the signal intensity of peptides that elute from the chromatographic column is plotted over time and the area under this curve is the ‘XIC’ [35,50]. In this approach, the intensity of the peptide signals between two states can be compared. A major advantage of XIC-based quantification is that no labeling is required and that it can be used with any type of sample. A more versatile approach for precise relative quantification involves the differential labeling of two or more sets of proteins or peptides derived from different cell states with light and heavy isotopes of the same chemical reagent followed by MS analysis (Fig 3). These techniques also allow relative quantification of basic, hydrophobic, or large proteins excluded from analysis using 2-DE or difference in-gel electrophoresis (DIGE). Protein chips. Protein chips will likely be the next major manifestation of the revolution in proteomics and offer another solution to analyze low abundant proteins and have the potential for high throughput applications to identify biomarkers. Protein chips differ from previously described methods: whereas screening by 2-DE or LC MS/MS can potentially detect any protein, protein chips can only provide data on set of proteins selected by the investigator (Fig 4).

by on July 10, 2007 w

ww

.StemC

ells.comD

ownloaded from

Page 5: Trends in Stem Cell Proteomics

Trends in Stem Cell Proteomics

4

Modern surface-enhanced laser desorption and ionization (SELDI) technology uses MS as a read-out system to analyze differential protein expression on spot arrays. Comparison of two mass spectroscopy data sets generated from two different samples immediately identifies the differentially-expressed proteins. Thus, high throughput analysis of crude samples readily and rapidly generates data that can be used for diagnosis or prognosis. The key disadvantage is that the mass spectrum obtained usually does not enable identification of the proteins analyzed, necessitating additional experimental procedures, e.g. enrichment by affinity chromatography and identification by methods such as tandem MS [51]. Stem cell proteomics Profiling and differential expression analysis. Proteome mapping serves as a starting point for building up a comprehensive database of the SC proteome. Several groups have used proteomics to identify SC-specific proteins in mouse ESCs (mESCs) [52-54], hESCs [54,55], human UCB-MSCs [56], human BM-MSCs [57], rat NSCs [58], and human NSCs [27]. A comprehensive list of proteomic investigations including different SC types, practical approaches, and major achievements have been summarized in Table 1. While proteins involved in energy metabolism comprise the largest group of identified proteins in adult SCs [56,58,59], a significant proportion of identified proteins in ESCs are involved in protein synthesis, processing, and transport [53,55], reflecting the potential of hESCs to either maintain an undifferentiated state or quickly change phenotype, as observed in rapid differentiation processes. One of the characteristics of the protein subset identified in ESC lines is that they contain relatively abundant nuclear proteins in terms of both variety and protein content. This might be related to the high nucleus-to-cytoplasm ratio of ESCs.

In total, ninety-two proteins were commonly identified in both mouse and human ESCs (Fig.5). The comparison of rat NSCs [58] with human NSCs [27], and human UCB-MSCs [56] with human AD-MSC [60] revealed 52 and 65 MSC and NSC specific proteins, respectively. NSCs have been shown to be more similar to ESCs than to MSCs (Fig. 5). The global overlap between genes expressed in ESCs and NSCs supports previous results at the transcriptome level [5], and corroborates the observed default differentiation pathway of ESCs to neural lineages. This is in line with observations where embryonic cells of both frogs [61] and mice [62] become neural cells in the absence of cell-to-cell signaling. Possible reasons for discrepancies in proteome analyses of SC will be discussed below. Over the past few years, there has been a growing interest in applying proteomics to study differential expression of SC genes in different developmental stages, thereby specifically aiming at unraveling the regulatory networks active during differentiation of mESCs [63-68], hESCs [54], MSCs [57,60,69,70], NSCs [27,59], HSCs [71]. Interestingly, the nine SC-specific proteins distinguished in this review were among those differentially expressed, i.e. Peroxiredoxin 1 [27,59,60,63], Heat shock cognate 71 kDa protein [27,59], Enolase 1 [59,66], 78 kDa glucose-regulated protein precursor [27,66], T-complex protein 1 [66], TCTP [64], and ATP synthase beta chain [66]. A small number of other differentiation-associated proteins has also been published, which includes proteins involved in stress response and oxidative defence (HSP27 [60,64,65,72]), 60-kDa heat shock protein [59,64,66], and Peroxiredoxin 4 [59,64], cell cytoskeleton (Tubulin alpha) [27,54,59,63,64,72], vimentin [63,65,66], receptor for intracellular transport (Syntaxin 7) [27,60], and multifunctional protein (calreticulin) [64,66,72].

by on July 10, 2007 w

ww

.StemC

ells.comD

ownloaded from

Page 6: Trends in Stem Cell Proteomics

Trends in Stem Cell Proteomics

5

Although these studies have generated a wealth of data, it is rather difficult to create a definitive proteome profile of undifferentiated and differentiated SCs in the different published studies. One of the major hurdles that have to be overcome in large-scale proteomic studies in general is the reduction of sample complexity. As yet, there is no single preparation method that allows identification of all proteins present in a sample. Applying different separation methods will produce varying sample compositions, thus resulting in different data sets after proteome analysis. Also different quantitative analysis methods may show variation in accuracy and sensitivity to samples of various complexities. Wu et al. [73] compared three quantitative methods frequently used in proteomics, 2DE-DIGE, ICAT, and iTRAQ. They reported that there is a limited overlap of differentially expressed proteins identified by the three methods from two closely related HCT-116 cell lines, suggesting the complementary nature of these approaches. Nevertheless, the complementary information obtained through different methods should potentially provide a better portrait of the biological system under investigation. On the other hand, differences in culture methodologies applied in different laboratories are likely to induce variations in protein expression. SCs are notoriously difficult to culture compared with more conventional cell lines, which is mainly due to our lack of knowledge about SCs. Some culture methodologies that work in one laboratory may not work as well in another laboratory because of unknown or less well-defined factors (e.g. serum batches) that affect cell growth and behavior. Additionally, the different methods used to derive SC lines will also contribute to differences in cell line characteristics (see “Heterogeneity of proteome” for more discussion). These forces individual laboratories to empirically adjust and optimize the culture conditions required to grow SCs. Thus, the different protein separation techniques, proteome analysis methods, and culture conditions, all of

which depend on the interest of individual research groups in a specific topic, result in the generation of various proteome data sets that are almost impossible to compare directly. However, the different proteome profiles that have and will become available are usually supplementary, and thereby complement our overall knowledge of SCs and the proteins expressed in different environments as well as under different culture conditions. Membrane proteomics. About 20-30 % of all genes in an organism encode integral membrane proteins, which are involved in numerous cellular processes. The target residues for tryptic cleavage (i.e. lysine and arginine) are mainly absent in transmembrane helices and preferentially found in the hydrophilic part of these lipid bilayer-incorporated proteins. Because of the protein aggregation step of IEF, 2-DE is unsuitable for the separation of integral membrane proteins and is limited to detection of membrane-associated proteins and membrane proteins with a low hydrophobicity (e.g. those having only one or two transmembrane helices). In contrast, the combination of 1D gel separation and LC-MS/MS has been applied with success [74]. Another more successful approach to isolate membrane proteins relies on cell surface-labeling in combination with high resolution two-dimensional (2D) LC-MS/MS. First, cell surface proteins of intact cells are selectively labeled with the membrane-impermeable reagent biotin, and biotinylated plasma membrane proteins are then enriched via affinity capture using immobilized avidin. The biotinylated proteins can be separated by gel electrophoresis and identified with MS [75]. The only record of cell surface proteome characterization of ESCs was made by Nunomura et al. [36]. They studied cell surface proteins using cell surface labeling of undifferentiated mESCs (line D3) coupled to high resolution 2D LC MS/MS. They identified 324 proteins, 235 of which had a putative signal sequence and/or transmembrane segments. Using 1D-gel followed by LC-MS/MS [36], Foster et al. [35] applied an XIC-based quantification method and identified 104 membrane proteins

by on July 10, 2007 w

ww

.StemC

ells.comD

ownloaded from

Page 7: Trends in Stem Cell Proteomics

Trends in Stem Cell Proteomics

6

from human MSCs; they found that expression levels changed during differentiation towards osteoblast cells. In both studies, many of the identified proteins were abundant housekeeping proteins such as ribosomal constituents, structural molecules, histones, and chaperones. Although some of these proteins might be associated with the membrane, it was difficult to distinguish them from the intracellular components released by dead cells. Combining this method with quantitative proteomic approaches, such as stable isotope labeling, will provide valuable information about stage- and lineage-specific expression of SCs. Post-translational modification. Many regulatory steps, especially those involved in cell proliferation, migration, and differentiation depend on protein PTMs rather than protein abundance [76]. Several 2-DE reports have identified large numbers of isoforms or PTMs in SCs (see table 1). By comparing proteomic data to transcriptome analyses, Unwin et al. [77] showed that the shift in proteome from long term reconstituting HSCs (Lin-Sca+Kit+; LSK+) to non-long term reconstituting progenitor cells (Lin-Sca+Kit-; LSK-) were associated with post-transcriptional control of protein levels. Another study, performed by Schrattenholz et al. [78], enabled the enrichment of phosphoproteins of neuronal derivatives of mESCs that were exposed to chemical ischemia in a differential and quantitative proteome analysis. Moreover, in a study that was restricted to a defined set of proteins, Prudhomme et al. [79] used a computational systems biology approach to study phosphorylation states of 31 intracellular signaling network components across 16 different stimuli at three time points. They applied quantitative Western blotting, and partial-least-squares modeling to determine which components showed the strongest correlation with cell proliferation and differentiation rates.

Kratchmarova et al. [80] applied a quantitative phosphoproteomics approach to study the effects of growth factors [epidermal growth factor (EGF) and platelet-derived growth factor (PDGF)] on human MSC. They metabolically labeled the proteins in cell culture using stable isotope labeling by amino acids in cell culture (SILAC) [110] (Fig. 3), combined the cell lysates of the three states and incubated this mixture with antibodies against phosphotyrosine. The precipitated complexes were resolved with 1D SDS–PAGE and proteolytically digested, after which the resulting peptide mixture was analyzed with LC-MS/MS. These results showed that EGF and PDGF modulate ostogenic capability of MSCs through MAPK/ERK, P38 kinase and phosphatidylinositol 3 kinase signaling. Puente et al. [72] sought to characterize the SC state by identifying the phosphoproteome of mESCs and their derivatives formed in embryoid bodies (EBs). Samples were loaded onto phosphoprotein-affinity columns and eluted proteins were separated by 2-DE followed by silver staining. Proteins visualized with silver stain were identified by MALDI MS/MS or LC-MS/MS. The set of proteins that exhibited altered PTM during differentiation included several proteins previously displayed in gene expression arrays as conserved features of the SC phenotype. Proteins related to protein catabolism, protein folding, chromatin remodeling and other functions were found to exhibit altered phosphorylation between the ESC and EB states. As such, these data suggest that kinase activity and the phosphorylation state of target substrates act as critical regulators of SC function. Heterogeneity of proteome. The reproducibility of proteome profiles of individual SC samples or their derivatives generated under similar conditions is a major criterion for large-scale proteomics-based studies. The proteome of a cell is highly dynamic and depends on several parameters including genetic background, the method of derivation, growth condition and the

by on July 10, 2007 w

ww

.StemC

ells.comD

ownloaded from

Page 8: Trends in Stem Cell Proteomics

Trends in Stem Cell Proteomics

7

stage of the cell cycle during sample collection. Therefore, individual samples of cells in the same physiological state should be made for accurate and reliable quantitative proteome comparisons with respect to protein up- or down-regulation. Zenzmaier et al. [81] compared CD34+ preparations from five different umbilical cord samples. Out of hundreds of spots detected on 2D gels, they found only 52 common proteins, 22 of which were identified using nano HPLC MS/MS. Since the purity of the cell samples was >88%, the observed heterogeneity could not be attributed to contaminating cells. Instead, the difference in the protein patterns was interpreted as SC-intrinsic heterogeneity. We analyzed the proteome of three hESC lines in triplicate and identified 54 and 14 proteins showing quantitative (P≤0.01) and qualitative changes, respectively [55]. Moreover, Van Hoof et al. [54] reported that the expression levels of proteins like β-actin and Oct4 were similar, between the hESC lines NL-HESC-01 [82] and HES-2 [83], whereas the expression ratios of several of these proteins were different in another hESC line, HUES-1 [84]. HES-2 and NL-HESC-01 cells were both passaged mechanically by a cut-and-paste method in serum-containing medium [83,85], whereas HUES-1 cells were passaged enzymatically by trypsinization and cultured in serum-replacement with basic fibroblast growth factor [84]. Potential sources of variation among hESC lines including (for review see [86,87]): (I) Differences due to origin of cell lines [(i) genomic diversity, (ii) stage of preimplantation embryo at derivation, (iii) conditions of early culture (feeder layer, culture conditions), (iv) differences in culture [88] and derivation procedures applied in laboratories such as different feeder cell types and densities, culture substrates, culture media, growth factors/other additives, and freezing method, (v) the passage number and method of passaging [2,89,90], and (vi) imprinting and X-inactivation], (2) Differences arising over time in culture [(i)

genetic changes (loss or gain of specific sequences), (ii) general and specific epigenetic changes (DNA methylation, histone acetylation, micoRNAs), for review see [91]], and (III) Differences due to mosaicism in cultures [(i) partial or terminal differentiation of subpopulations within cultures, (ii) variation among epigenetic and genetic changes]. In adult SCs, it was shown that the proliferation and osteogenic capacity of human MSCs decrease during serial subculturing [89]. Moreover, passage-specific proteins were found, which were suggested to be differentially regulated and to play a role in the decrease of osteogenic differentiation potential under serial subculturing. The purification and extraction of specific SC-derived cell types and the consistency and reproducibility of sample generation are thus considered important issues. SC differentiation usually yields mixed and heterogeneous cell populations. Therefore, optimization of protocols for enhancement of differentiation towards a specific cell linage and the following purification should be taken into account (for review see [92]). Feasible methods that may help to achieve this include: (i) addition of specific combinations of growth factors or chemical morphogens, (ii) changing the physical and geometrical microenvironment, (iii) co-culture or transplantation of SCs with inducer tissues or cells, (iv) implantation of SCs into specific organs or tissues and (v) overexpression of transcription factors associated with development of specific cell lineages. However, to date, these strategies have not yielded pure populations of mature progeny and apparently require efficient protocols to purify specific cell populations. Methods like fluorescence-activated cell sorting (FACS) or magnetic-activated cell sorting (MACS) allow purification as such, but depend on the cell to express a surface marker that can be recognized by a fluorescent or magnetic microbead-tagged antibody; to be desirably effective, the marker needs to be cell-type specific. In most cases, these cell markers

by on July 10, 2007 w

ww

.StemC

ells.comD

ownloaded from

Page 9: Trends in Stem Cell Proteomics

Trends in Stem Cell Proteomics

8

are not commercially available; thus, sorting methods rely on e.g. genetic modification of SCs by tagging a lineage-specific promoter to a fluorescent marker. Alternatively, cells could be transduced with a drug-resistance gene instead of a marker, to allow preferential selection of subpopulations. Application of protein array to stem cell proteomics. Protein arrays offer a different solution and have the potential for high throughput applications to identify novel protein markers and molecular pathways. Hayman et al. [93] applied SELDI-TOF to rapidly generate protein peakmap bioprofiles. They demonstrated that this approach can be used with up to 100% accuracy to distinguish human ECCs from differentiated derivatives. It should be noted that this approach does not identify the individual molecules expressed in the cell sample. Yet, if the identification of a particular protein is required, the current approach can be combined with other technology such as SELDI-tandem MS. Using cytokine protein arrays, it has been shown that cytokine induction and signal transduction are important for the differentiation of human UCB-MSCs [94]. Sakaguchi et al. [95] used a ProteinChip system to identify OP9-a BM cell line-conditioned medium molecule responsible for neurosphere formation from NSCs. The application of reverse phase protein arrays for the analysis of primary acute myelogenous leukemia samples as well as leukemic and normal SCs has been demonstrated [96]. Using this strategy, the differences in protein expression in as few as three cell protein equivalents could be detected. Therefore, it was suggested that this approach can be applied as a highly reliable and reproducible high throughput system for rapid large-scale proteomic analyses of protein expression and phosphorylation state in primary acute myelogenous leukemia cells as well as in human SCs.

Secretomics. The secretome is defined as a subset of the proteome that contains all proteins actively exported out of a cell from any origin. The type of proteins secreted by the cells strictly depends on the type of cell and the cellular state; therefore the secretome reveals much about what is going on inside the cell. The proteomics approach was employed to characterize an environment that supports the growth of undifferentiated hESCs, and to identify factors critical for their independent growth. Proteome analysis of conditioned medium (CM) from mouse embryonic fibroblast (MEF) feeder layers (STO cell line) [97] and human neonatal foreskin cell line (HNF02) [98] resulted in identification of several proteins involved in cell growth, differentiation, extracellular matrix formation and remodeling, many intracellular proteins were identified. Zvonic et al. [99] compared the secretomes of CMs obtained from four individual primary AD-SC cultures in uninduced or adipogenic-induced conditions and identified several proteins such as adiponectin, plasminogen activator inhibitor 1 and multiple serine protease inhibitor proteins (serpins). These studies indicate the complexity of the environment formed by the feeder cells and provide a useful starting point for future studies. Secretome studies show a high potential for identification of biomarkers involved in many cellular processes, including growth, division, differentiation, development, and death. Transplantation proteomics. Although considerable progress in human transplantation medicine has been achieved, several major obstacles still restrict more widespread application of cell transplantation and in particular that of SCs. The major clinical obstacle that has to be overcome is demonstrating the safety and feasibility of cell therapy. Proteomic analyses of tissues and body fluids after cell therapy could address these concerns. For example, Kaiser et al. [100]

by on July 10, 2007 w

ww

.StemC

ells.comD

ownloaded from

Page 10: Trends in Stem Cell Proteomics

Trends in Stem Cell Proteomics

9

analyzed urine after HSC transplantation (HSCT) and could clearly distinguish between patients with graft-versus-host disease (GVHD) and those with no problems after HSCT with a high specificity (82%) and a sensitivity of 100%. Wang et al. [101] quantitatively analyzed the human plasma proteome before and after the onset of GVHD, leading to the identification of a large number of proteins that are affected by GVHD after HSCT. They identified 75 proteins that exhibited quantitative changes between the pre- and post-GVHD samples. Some of these proteins were well-known acute-phase reactants including serum amyloid A, apolipoproteins A-I/A-IV, and complement C3. To study salivary protein changes that occur after HSCT, Imanguli et al. [102] analyzed serially collected saliva samples from 41 patients undergoing allo-HSCT using SELDI-MS in conjunction with 2-DE. Significant changes in multiple salivary proteins that lasted at least two months post-transplant were detected including up-regulation of lactoferrin and secretary leukocyte protease inhibitor and down-regulation of secretary IgA. Weissinger et al. [103] could correlate proteomic data with the clinical diagnosis of acute GVHD. From their proteome analysis, a tentatively acute GVHD-specific model consisting of 31 polypeptides was chosen that allowed to distinguish between patients with GVHD and those with no problems after HSCT with high specificity (98%) and a sensitivity of 100%. The subsequent blinded evaluation of 599 samples enabled diagnosis of acute GVHD, even prior to clinical diagnosis, with a sensitivity of 83.1% and a specificity of 75.6 %. These results showed the power of proteomics as an unbiased laboratory-based screening method, enabling diagnosis and pre-emptive therapy. Insight into stem cell protein networks and signaling pathways for pluripotency. Understanding molecular mechanisms underlying SC pluripotency should illuminate fundamental properties of SCs and the process of

cellular reprogramming. Proteomics proved to be a powerful approach to gain insight concerning key intracellular signals governing SC self-renewal and differentiation. In an attempt to analyze cue-signal-response relationship underlying SC self-renewal versus differentiation, the phosphorylation states of 31 intracellular signaling network components were quantitatively studied under fibronectin, laminin, leukemia inhibitory factor, and fibroblast growth factor-4 treatments [79]. Using a multivariate proteomic approach, they identified a set of signaling network components most critically associated with differentiation (Stat3, Raf1, MEK, and ERK), proliferation of undifferentiated mESCs (MEK and ERK), and proliferation of differentiated cells (PKBα_, Stat3, Src, and PKCε). A quantitative MS-based phosphoproteomics approach has been applied to elucidate critical differences in the signaling mechanisms of EGF and PDGF that led to the differential effects on osteoblast differentiation of human MSC (See post-translational modifications section) [80]. By studying tyrosine phosphorylated proteins in response EGF and PDGE, they found that less than 10% of all phosphotyrosine proteins are specific to either the EGF or PDGF activation program in human MSCs, revealing a range of widely shared signaling pathways. Examples include the mitogen activated mitogen activated protein kinase (MAPK) cascades and signal attenuation through receptor ubiquitination followed by endocytic removal from the cell surface. However, based on observation that EGF-treated human MSCs but not PDGF-treated cells undergo osteogenic differentiation, the variation contained in the 10% of differentially activated genes was clearly of crucial significance. The most striking difference was the preferential activation of phosphotidyl-inositol-3-kinase (PI3K) exclusively by PDGF, signifying a possible control point in the osteogenic differentiation process. These results demonstrated that, at least in some cases, decisions can be made by preferentially

by on July 10, 2007 w

ww

.StemC

ells.comD

ownloaded from

Page 11: Trends in Stem Cell Proteomics

Trends in Stem Cell Proteomics

10

activating a small subset of the signaling network. Using a chip-based proteomics approach, factors affecting the proliferation of NSCs have been screened. Sakaguchi et al. [95] used a ProteinChip system to identify molecules present in conditioned medium of OP9-a BM cell line- which induces neurosphere formation from NSCs. In this screen, they identified a soluble carbohydrate-binding protein, Galectin-1, as a candidate. Galectins comprise a family of carbohydrate-binding lectin proteins that are implicated in cell adhesion, growth, differentiation, neoplastic transformation, and metastasis [104]. Galectin-1 has also been identified as one of the relatively abundant proteins in mouse embryonic fibroblast-conditioned medium [97] and human foreskin fibroblast-conditioned medium [98]. Based on results from intraventricular infusion experiments and phenotypic analyses of knockout mice, they suggested that the carbohydrate-binding activity of Galectin-1 is required for its promotion of adult neural progenitor cell proliferation. In a recent investigation, proteomics has been applied to gain insight into the regulatory protein networks in which Nanog operate in mESCs [105]. A construct bearing pluripotency factor Nanog with a Flag tag as well as a peptide tag that serves as a substrate for in vivo biotinylation was expressed in ESCs. The tagged protein was recovered from cellular lysates with streptavidin beads and further purified using anti-Flag antibodies. MS was then applied to identify its interacting partners. Not surprisingly, many of the candidates were other transcription factors, some of which had already been associated with ESCs. The resulting data set was used to generate a complex network of interacting proteins which were depicted in a concise scheme [105]. Most of proteins in this network were shown to be essential for early development and/or ESC properties. The knockout of several network proteins including Prmt1, YY1, Rnf2, BAF155, Rybp, Oct4, Cdk1,

NF45, Sall4, Elys, Tif1β, Pelo, Dax1, and REST resulted in defects in proliferation and /or survival of the inner cell mass or other aspects of early development. The knockout of Err2, Rif1, Nac1, Zfp281 resulted in defects in self-renewal and/or differentiation of ESCs. The coexpression of most of network genes and their roles as both targets and effectors indicate that this interactome may serve as a functional module committed to maintaining ESC pluripotency. This network provided a solid base for further exploration of the signalling pathways involved in ESC maintenance [105]. Sall4 had been found to be involved in these signalling pathways by three other groups independently [106-108]. This protein was also identified in the large-scale proteome study by Van Hoof et al. [54], however, the association with Oct4 and Nanog had not been made. This illustrates the likelihood that numerous proteins specifically identified in SCs play a significant role in SC sustaining processes. Venn diagrams like Fig.5 will narrow down the search for novel ESC-associated proteins, however, the involvement and role of such candidates in SC maintenance needs to be confirmed by additional experiments. Future challenges and outlook Proteomic methods have produced large data sets of proteins involved in mechanisms and pathways that regulate SC proliferation and differentiation. The insights thus obtained in SC biology have also created many opportunities to improve public health. In recent years, numerous proteomics techniques have been developed and are continuously being improved, in both peptide and protein separation (e.g., LC and 2-DE) as well as MS methods and accuracy. However, several important issues that remain to be addressed rely on further technical advances in proteomics analysis. When large proteomes consisting of thousands of proteins are analyzed, the dynamic resolution is restricted and only the most abundant proteins can be detected [109].

by on July 10, 2007 w

ww

.StemC

ells.comD

ownloaded from

Page 12: Trends in Stem Cell Proteomics

Trends in Stem Cell Proteomics

11

Despite advances in non 2-DE based proteomics technologies, 2-DE remains the pivotal and most wide spread method of currently proteomics [73,110]. However, we believe that large-scale MS-based quantification approaches will significantly contribute to our understanding of SCs in the future and will soon become the standard to analyze the SC proteome. To enable proteome-wide quantification, further optimization of chemical-labeling reagents, including chemicals targeting specific protein classes and MS instrument performance are necessary [109]. Proteome-wide quantification of membrane proteins requires methods that solve problems like contamination of intracellular components, protein insolubility, and loss of hydrophobic peptides, which prevent protein identification. Although protein chips are still under development, they have already proven their value to study protein functions and expression patterns. Requiring only small amounts of material makes them exceptionally well-suited to study SC populations. However, further optimization of these techniques is needed before they can be widely used in proteome analyses. The application of several array-based approaches such as phosphorylation or GPCR arrays that are missing from the current SC literature will provide highly valuable contributions. The advantage of MS-based proteomics is its ability to indiscriminately study PTMs that affect activity and binding properties of proteins, thereby altering their roles within the cell. It is likely that the phosphoproteome, protein interactions (interactome), and glycomics will soon become major areas of SC proteomics research. One of the major problems in the SC studies is to obtain consistent results for the same type studies. Proteomics may very well contribute to gaining insight into SC functioning and

behavior, and thus provide clues for how to tackle these problems. Obviously, gaining more insight into how SCs respond to their environment will improve our ways to control their behavior by applying better defined culture methods. Despite increasing conformity in proteomics applications and data storage, it remains difficult to draw consistent conclusions from individual studies because of the use of different cell types, establishment and maintenance, the number of passages and the passaging methods applied. Standardization of proteomic methodologies and strategies between different groups of investigators, and introduction of standard operation procedures would facilitate the comparability of proteomics results. In addition, the establishment of unique databases for the ever-increasing wealth of information generated by proteome-wide and in-depth proteomic studies of SCs will be indispensable (Fig. 6). To this end, several initiatives were set up to characterize numerous existing hESC lines using standardized assay conditions to allow unrestrained comparison of the data sets generated. Such initiatives have been instigated by the International Stem Cell Initiative [111], the International Stem Cell Forum (www.stemcellforum.org), the NIH Stem Cell Unit (http://stemcells.nih.gov/research/nihresearch/scunit/) and the American Type Culture Collection (http://stemcells.atcc.org; [112]). Combined, the various proteomic approaches will continue to revolutionize insights into SC biology.

ACKNOWLEDGMENTS We gratefully thank Dr. Peter Hains (Australia) for their critical reading and helpful comments on the manuscript. This work was supported by grants from the Royan Institute.

by on July 10, 2007 w

ww

.StemC

ells.comD

ownloaded from

Page 13: Trends in Stem Cell Proteomics

Trends in Stem Cell Proteomics

12

REFERENCES 1. Blau HM, Brazelton TR, Weimann JM. The evolving

concept of a stem cell: Entity or function? Cell 2001;105:829-841.

2. Baharvand H. Embryonic stem cells: Establishment, maintenance, and differentiation. In: Grier EV (ed): Embryonic stem cell research. Nova Science Publishers, Inc., 2006, 1-63

3. Klimanskaya I, Chung Y, Becker S, et al. Human embryonic stem cell lines derived from single blastomeres. Nature 2006;444:481-485.

4. Preston SL, Alison MR, Forbes SJ, et al. The new stem cell biology: Something for everyone. Mol Pathol 2003;56:86-96.

5. Ramalho-Santos M, Yoon S, Matsuzaki Y, et al. "Stemness": Transcriptional profiling of embryonic and adult stem cells. Science 2002;298:597-600.

6. Ivanova NB, Dimos JT, Schaniel C, et al. A stem cell molecular signature. Science 2002;298:601-604.

7. Williams C, Wirta V, Meletis K, et al. Catalog of gene expression in adult neural stem cells and their in vivo microenvironment. Exp Cell Res 2006;312:1798-1812.

8. Wei CL, Miura T, Robson P, et al. Transcriptome profiling of human and murine escs identifies divergent paths required to maintain the stem cell state. Stem Cells 2005;23:166-185.

9. Katz AJ, Tholpady A, Tholpady SS, et al. Cell surface and transcriptional characterization of human adipose-derived adherent stromal (hadas) cells. Stem Cells 2005;23:412-423.

10. Jeong JA, Hong SH, Gang EJ, et al. Differential gene expression profiling of human umbilical cord blood-derived mesenchymal stem cells by DNA microarray. Stem Cells 2005;23:584-593.

11. Cimica V, Batusic D, Chen Y, et al. Transcriptome analysis of rat liver regeneration in a model of oval hepatic stem cells. Genomics 2005;86:352-364.

12. Richards M, Tan SP, Tan JH, et al. The transcriptome profile of human embryonic stem cells as defined by sage. Stem Cells 2004;22:51-64.

13. Georgantas RW, 3rd, Tanadve V, Malehorn M, et al. Microarray and serial analysis of gene expression analyses identify known and novel transcripts overexpressed in hematopoietic stem cells. Cancer Res 2004;64:4434-4441.

14. Silva WA, Jr., Covas DT, Panepucci RA, et al. The profile of gene expression of human marrow mesenchymal stem cells. Stem Cells 2003;21:661-669.

15. Anisimov SV, Tarasov KV, Tweedie D, et al. Sage identification of gene transcripts with profiles unique to pluripotent mouse r1 embryonic stem cells. Genomics 2002;79:169-176.

16. Bhattacharya B, Cai J, Luo Y, et al. Comparison of the gene expression profile of undifferentiated human embryonic stem cell lines and differentiating embryoid bodies. BMC Dev Biol 2005;5:22.

17. Bhattacharya B, Miura T, Brandenberger R, et al. Gene expression in human embryonic stem cell lines: Unique molecular signature. Blood 2004;103:2956-2964.

18. Josephson R, Sykes G, Liu Y, et al. A molecular scheme for improved characterization of human embryonic stem cell lines. BMC Biol 2006;4:28.

19. Li H, Liu Y, Shin S, et al. Transcriptome coexpression map of human embryonic stem cells. BMC Genomics 2006;7:103.

20. Player A, Wang Y, Bhattacharya B, et al. Comparisons between transcriptional regulation and rna expression in human embryonic stem cell lines. Stem Cells Dev 2006;15:315-323.

21. Rao RR, Stice SL. Gene expression profiling of embryonic stem cells leads to greater understanding of pluripotency and early developmental events. Biol Reprod 2004.

22. Rao S, Orkin SH. Unraveling the transcriptional network controlling ES cell pluripotency. Genome Biol 2006;7:230.

23. Sun Y, Li H, Liu Y, et al. Cross-species transcriptional profiles establish a functional portrait of embryonic stem cells. Genomics 2007;89:22-35.

24. Sun Y, Li H, Yang H, et al. Mechanisms controlling embryonic stem cell self-renewal and differentiation. Crit Rev Eukaryot Gene Expr 2006;16:211-231.

25. Wilkins MR, Sanchez JC, Gooley AA, et al. Progress with proteome projects: Why all proteins expressed by a genome should be identified and how to do it. Biotechnol Genet Eng Rev 1996;13:19-50.

26. Unwin RD, Gaskell SJ, Evans CA, et al. The potential for proteomic definition of stem cell populations. Exp Hematol 2003;31:1147-1159.

27. Hoffrogge R, Mikkat S, Scharf C, et al. 2-de proteome analysis of a proliferating and differentiating human neuronal stem cell line (rencell vm). Proteomics 2006;6:1833-1847.

28. Roche S, Provansal M, Tiers L, et al. Proteomics of primary mesenchymal stem cells. Regenerative Medicine 2006;1:511-517.

29. Van Hoof D, Mummery CL, Heck AJ, et al. Embryonic stem cell proteomics. Expert Rev Proteomics 2006;3:427-437.

30. Baltimore D. Our genome unveiled. Nature 2001;409:814-816.

31. Wilkins MR, Gasteiger E, Gooley AA, et al. High-throughput mass spectrometric discovery of protein post-translational modifications. J Mol Biol 1999;289:645-657.

by on July 10, 2007 w

ww

.StemC

ells.comD

ownloaded from

Page 14: Trends in Stem Cell Proteomics

Trends in Stem Cell Proteomics

13

32. Godovac-Zimmermann J, Brown LR. Perspectives for mass spectrometry and functional proteomics. Mass Spectrom Rev 2001;20:1-57.

33. Celis JE, Gromov P. 2d protein electrophoresis: Can it be perfected? Curr Opin Biotechnol 1999;10:16-21.

34. Bauer A, Kuster B. Affinity purification-mass spectrometry. Powerful tools for the characterization of protein complexes. Eur J Biochem 2003;270:570-578.

35. Foster LJ, Zeemann PA, Li C, et al. Differential expression profiling of membrane proteins by quantitative proteomics in a human mesenchymal stem cell line undergoing osteoblast differentiation. Stem Cells 2005;23:1367-1377.

36. Nunomura K, Nagano K, Itagaki C, et al. Cell surface labeling and mass spectrometry reveal diversity of cell surface markers and signaling molecules expressed in undifferentiated mouse embryonic stem cells. Mol Cell Proteomics 2005;4:1968-1976.

37. O'Farrell PH. High resolution two-dimensional electrophoresis of proteins. J Biol Chem 1975;250:4007-4021.

38. Gorg A, Postel W, Gunther S. The current state of two-dimensional electrophoresis with immobilized ph gradients. Electrophoresis 1988;9:531-546.

39. Gorg A, Weiss W, Dunn MJ. Current two-dimensional electrophoresis technology for proteomics. Proteomics 2004;4:3665-3685.

40. Aebersold R, Mann M. Mass spectrometry-based proteomics. Nature 2003;422:198-207.

41. Domon B, Aebersold R. Mass spectrometry and protein analysis. Science 2006;312:212-217.

42. Henzel WJ, Billeci TM, Stults JT, et al. Identifying proteins from two-dimensional gels by molecular mass searching of peptide fragments in protein sequence databases. Proc Natl Acad Sci U S A 1993;90:5011-5015.

43. Futcher B, Latter GI, Monardo P, et al. A sampling of the yeast proteome. Mol Cell Biol 1999;19:7357-7368.

44. Perrot M, Sagliocco F, Mini T, et al. Two-dimensional gel protein database of saccharomyces cerevisiae (update 1999). Electrophoresis 1999;20:2280-2298.

45. Gygi SP, Corthals GL, Zhang Y, et al. Evaluation of two-dimensional gel electrophoresis-based proteome analysis technology. Proc Natl Acad Sci U S A 2000;97:9390-9395.

46. Molloy MP. Two-dimensional electrophoresis of membrane proteins using immobilized ph gradients. Anal Biochem 2000;280:1-10.

47. Link AJ, Eng J, Schieltz DM, et al. Direct analysis of protein complexes using mass spectrometry. Nat Biotechnol 1999;17:676-682.

48. Wolters DA, Washburn MP, Yates JR, 3rd. An automated multidimensional protein identification technology for shotgun proteomics. Anal Chem 2001;73:5683-5690.

49. Han DK, Eng J, Zhou H, et al. Quantitative profiling of differentiation-induced microsomal proteins using

isotope-coded affinity tags and mass spectrometry. Nat Biotechnol 2001;19:946-951.

50. Andersen JS, Wilkinson CJ, Mayor T, et al. Proteomic characterization of the human centrosome by protein correlation profiling. Nature 2003;426:570-574.

51. Issaq HJ, Veenstra TD, Conrads TP, et al. The seldi-tof ms approach to proteomics: Protein profiling and biomarker identification. Biochem Biophys Res Commun 2002;292:587-592.

52. Elliott ST, Crider DG, Garnham CP, et al. Two-dimensional gel electrophoresis database of murine r1 embryonic stem cells. Proteomics 2004;4:3813-3832.

53. Nagano K, Taoka M, Yamauchi Y, et al. Large-scale identification of proteins expressed in mouse embryonic stem cells. Proteomics 2005;5:1346-1361.

54. Van Hoof D, Passier R, Ward-Van Oostwaard D, et al. A quest for human and mouse embryonic stem cell-specific proteins. Mol Cell Proteomics 2006;5:1261-1273.

55. Baharvand H, Hajheidari M, Kazemi Ashtiani S, et al. Proteomic signature of human embryonic stem cells. Proteomics 2006;6:3544-3549.

56. Feldmann RE, Jr., Bieback K, Maurer MH, et al. Stem cell proteomes: A profile of human mesenchymal stem cells derived from umbilical cord blood. Electrophoresis 2005;26:2749-2758.

57. Salasznyk RM, Westcott AM, Klees RF, et al. Comparing the protein expression profiles of human mesenchymal stem cells and human osteoblasts using gene ontologies. Stem Cells Dev 2005;14:354-366.

58. Maurer MH, Feldmann RE, Jr., Futterer CD, et al. The proteome of neural stem cells from adult rat hippocampus. Proteome Sci 2003;1:4.

59. Maurer MH, Feldmann RE, Jr., Futterer CD, et al. Comprehensive proteome expression profiling of undifferentiated versus differentiated neural stem cells from adult rat hippocampus. Neurochem Res 2004;29:1129-1144.

60. DeLany JP, Floyd ZE, Zvonic S, et al. Proteomic analysis of primary cultures of human adipose-derived stem cells: Modulation by adipogenesis. Mol Cell Proteomics 2005;4:731-740.

61. Hemmati-Brivanlou A, Melton D. Vertebrate embryonic cells will become nerve cells unless told otherwise. Cell 1997;88:13-17.

62. Tropepe V, Hitoshi S, Sirard C, et al. Direct neural fate specification from embryonic stem cells: A primitive mammalian neural stem cell stage acquired through a default mechanism. Neuron 2001;30:65-78.

63. Guo X, Ying W, Wan J, et al. Proteomic characterization of early-stage differentiation of mouse embryonic stem cells into neural cells induced by all-trans retinoic acid in vitro. Electrophoresis 2001;22:3067-3075.

64. Wang D, Gao L. Proteomic analysis of neural differentiation of mouse embryonic stem cells. Proteomics 2005;5:4414-4426.

by on July 10, 2007 w

ww

.StemC

ells.comD

ownloaded from

Page 15: Trends in Stem Cell Proteomics

Trends in Stem Cell Proteomics

14

65. Battersby A, Jones RD, Lilley KS, et al. Comparative proteomic analysis reveals differential expression of hsp25 following the directed differentiation of mouse embryonic stem cells. Biochim Biophys Acta 2006.

66. Kurisaki A, Hamazaki TS, Okabayashi K, et al. Chromatin-related proteins in pluripotent mouse embryonic stem cells are downregulated after removal of leukemia inhibitory factor. Biochem Biophys Res Commun 2005;335:667-675.

67. Kadota M, Nishigaki R, Wang CC, et al. Proteomic signatures and aberrations of mouse embryonic stem cells containing a single human chromosome 21 in neuronal differentiation: An in vitro model of down syndrome. Neuroscience 2004;129:325-335.

68. An J, Yuan Q, Wang C, et al. Differential display of proteins involved in the neural differentiation of mouse embryonic carcinoma p19 cells by comparative proteomic analysis. Proteomics 2005;5:1656-1668.

69. Wang D, Park JS, Chu JS, et al. Proteomic profiling of bone marrow mesenchymal stem cells upon transforming growth factor beta1 stimulation. J Biol Chem 2004;279:43725-43734.

70. Lee HK, Lee BH, Park SA, et al. The proteomic analysis of an adipocyte differentiated from human mesenchymal stem cells using two-dimensional gel electrophoresis. Proteomics 2006;6:1223-1229.

71. Tao W, Wang M, Voss ED, et al. Comparative proteomic analysis of human cd34+ stem/progenitor cells and mature cd15+ myeloid cells. Stem Cells 2004;22:1003-1014.

72. Puente LG, Borris DJ, Carriere JF, et al. Identification of candidate regulators of embryonic stem cell differentiation by comparative phosphoprotein affinity profiling. Mol Cell Proteomics 2006;5:57-67.

73. Wu WW, Wang G, Baek SJ, et al. Comparative study of three proteomic quantitative methods, dige, cicat, and itraq, using 2d gel- or lc-maldi tof/tof. J Proteome Res 2006;5:651-658.

74. Gu S, Chen J, Dobos KM, et al. Comprehensive proteomic profiling of the membrane constituents of a mycobacterium tuberculosis strain. Mol Cell Proteomics 2003;2:1284-1296.

75. Zhao Y, Zhang W, Kho Y, et al. Proteomic analysis of integral plasma membrane proteins. Anal Chem 2004;76:1817-1823.

76. Levchenko A. Proteomics takes stem cell analyses to another level. Nat Biotechnol 2005;23:828-830.

77. Unwin RD, Smith DL, Blinco D, et al. Quantitative proteomics reveals posttranslational control as a regulatory factor in primary hematopoietic stem cells. Blood 2006;107:4687-4694.

78. Schrattenholz A, Wozny W, Klemm M, et al. Differential and quantitative molecular analysis of ischemia complexity reduction by isotopic labeling of proteins using a neural embryonic stem cell model. J Neurol Sci 2005;229-230:261-267.

79. Prudhomme W, Daley GQ, Zandstra P, et al. Multivariate proteomic analysis of murine embryonic

stem cell self-renewal versus differentiation signaling. Proc Natl Acad Sci U S A 2004;101:2900-2905.

80. Kratchmarova I, Blagoev B, Haack-Sorensen M, et al. Mechanism of divergent growth factor effects in mesenchymal stem cell differentiation. Science 2005;308:1472-1477.

81. Zenzmaier C, Gesslbauer B, Grobuschek N, et al. Proteomic profiling of human stem cells derived from umbilical cord blood. Biochem Biophys Res Commun 2005;328:968-972.

82. van de Stolpe A, van den Brink S, van Rooijen M, et al. Human embryonic stem cells: Towards therapies for cardiac disease. Derivation of a dutch human embryonic stem cell line. Reprod Biomed Online 2005;11:476-485.

83. Reubinoff BE, Pera MF, Fong CY, et al. Embryonic stem cell lines from human blastocysts: Somatic differentiation in vitro. Nat Biotechnol 2000;18:399-404.

84. Cowan CA, Klimanskaya I, McMahon J, et al. Derivation of embryonic stem-cell lines from human blastocysts. N Engl J Med 2004;350:1353-1356.

85. Mummery C, Ward-van Oostwaard D, Doevendans P, et al. Differentiation of human embryonic stem cells to cardiomyocytes: Role of coculture with visceral endoderm-like cells. Circulation 2003;107:2733-2740.

86. Allegrucci C, Young LE. Differences between human embryonic stem cell lines. Hum Reprod Update 2006.

87. Loring JF, Rao MS. Establishing standards for the characterization of human embryonic stem cell lines. Stem Cells 2006;24:145-150.

88. Maitra A, Arking DE, Shivapurkar N, et al. Genomic alterations in cultured human embryonic stem cells. Nat Genet 2005;37:1099-1103.

89. Sun HJ, Bahk YY, Choi YR, et al. A proteomic analysis during serial subculture and osteogenic differentiation of human mesenchymal stem cell. J Orthop Res 2006;24:2059-2071.

90. Rosler ES, Fisk GJ, Ares X, et al. Long-term culture of human embryonic stem cells in feeder- free conditions. Developmental Dynamics 2004;229:259-274.

91. Wu H, Sun YE. Epigenetic regulation of stem cell differentiation. Pediatr Res 2006;59:21R-25R.

92. Odorico JS, Kaufman DS, Thomson JA. Multilineage differentiation from human embryonic stem cell lines. Stem Cells 2001;19:193-204.

93. Hayman MW, Przyborski SA. Proteomic identification of biomarkers expressed by human pluripotent stem cells. Biochem Biophys Res Commun 2004;316:918-923.

94. Liu CH, Hwang SM. Cytokine interactions in mesenchymal stem cells from cord blood. Cytokine 2005;32:270-279.

95. Sakaguchi M, Shingo T, Shimazaki T, et al. A carbohydrate-binding protein, galectin-1, promotes proliferation of adult neural stem cells. Proc Natl Acad Sci U S A 2006;103:7112-7117.

by on July 10, 2007 w

ww

.StemC

ells.comD

ownloaded from

Page 16: Trends in Stem Cell Proteomics

Trends in Stem Cell Proteomics

15

96. Tibes R, Qiu Y, Lu Y, et al. Reverse phase protein array: Validation of a novel proteomic technology and utility for analysis of primary leukemia specimens and hematopoietic stem cells. Mol Cancer Ther 2006;5:2512-2521.

97. Lim JW, Bodnar A. Proteome analysis of conditioned medium from mouse embryonic fibroblast feeder layers which support the growth of human embryonic stem cells. Proteomics 2002;2:1187-1203.

98. Prowse AB, McQuade LR, Bryant KJ, et al. A proteome analysis of conditioned media from human neonatal fibroblasts used in the maintenance of human embryonic stem cells. Proteomics 2005;5:978-989.

99. Zvonic S, Lefevre M, Kilroy G, et al. Secretome of primary cultures of human adipose-derived stem cells (ascs): Modulation of serpins by adipogenesis. Mol Cell Proteomics 2006.

100. Kaiser T, Kamal H, Rank A, et al. Proteomics applied to the clinical follow-up of patients after allogeneic hematopoietic stem cell transplantation. Blood 2004;104:340-349.

101. Wang H, Clouthier SG, Galchev V, et al. Intact-protein-based high-resolution three-dimensional quantitative analysis system for proteome profiling of biological fluids. Mol Cell Proteomics 2005;4:618-625.

102. Imanguli MM, Atkinson JC, Harvey KE, et al. Changes in salivary proteome following allogeneic hematopoietic stem cell transplantation. Exp Hematol 2007;35:184-192.

103. Weissinger EM, Schiffer E, Hertenstein B, et al. Proteomic patterns predict acute graft-versus-host disease after allogeneic hematopoietic stem cell transplantation. Blood 2007.

104. Perillo NL, Marcus ME, Baum LG. Galectins: Versatile modulators of cell adhesion, cell proliferation, and cell death. J Mol Med 1998;76:402-412.

105. Wang J, Rao S, Chu J, et al. A protein interaction network for pluripotency of embryonic stem cells. Nature 2006;444:364-368.

106. Zhang J, Tam WL, Tong GQ, et al. Sall4 modulates embryonic stem cell pluripotency and early embryonic development by the transcriptional regulation of pou5f1. Nat Cell Biol 2006;8:1114-1123.

107. Elling U, Klasen C, Eisenberger T, et al. Murine inner cell mass-derived lineages depend on sall4 function. Proc Natl Acad Sci U S A 2006;103:16319-16324.

108. Wu Q, Chen X, Zhang J, et al. Sall4 interacts with nanog and co-occupies nanog genomic sites in embryonic stem cells. J Biol Chem 2006;281:24090-24094.

109. Ong SE, Mann M. Mass spectrometry-based proteomics turns quantitative. Nat Chem Biol 2005;1:252-262.

110. Carrette O, Burkhard PR, Sanchez JC, et al. State-of-the-art two-dimensional gel electrophoresis: A key tool of proteomics research. Nat Protoc 2006;1:812-823.

111. Andrews PW, Benvenisty N, McKay R, et al. The international stem cell initiative: Toward benchmarks for human embryonic stem cell research. Nat Biotechnol 2005;23:795-797.

112. Rao MS, Civin CI. Translational research: Toward better characterization of human embryonic stem cell lines. Stem Cells 2005;23:1453.

113. Tonge R, Shaw J, Middleton B, et al. Validation and development of fluorescence two-dimensional differential gel electrophoresis proteomics technology. Proteomics 2001;1:377-396.

114. Berggren K, Steinberg TH, Lauber WM, et al. A luminescent ruthenium complex for ultrasensitive detection of proteins immobilized on membrane supports. Anal Biochem 1999;276:129-143.

115. Hu Q, Noll RJ, Li H, et al. The orbitrap: A new mass spectrometer. J Mass Spectrom 2005;40:430-443.

116. Gygi SP, Rist B, Gerber SA, et al. Quantitative analysis of complex protein mixtures using isotope-coded affinity tags. Nat Biotechnol 1999;17:994-999.

117. Ross PL, Huang YN, Marchese JN, et al. Multiplexed protein quantitation in saccharomyces cerevisiae using amine-reactive isobaric tagging reagents. Mol Cell Proteomics 2004;3:1154-1169.

118. Mirgorodskaya OA, Kozmin YP, Titov MI, et al. Quantitation of peptides and proteins by matrix-assisted laser desorption/ionization mass spectrometry using (18)o-labeled internal standards. Rapid Commun Mass Spectrom 2000;14:1226-1232.

119. Yao X, Freas A, Ramirez J, et al. Proteolytic 18o labeling for comparative proteomics: Model studies with two serotypes of adenovirus. Anal Chem 2001;73:2836-2842.

120. Baharvand H, Matthaei KI. Culture condition difference for establishment of new embryonic stem cell linesfrom the c57bl/6 and balb/c mouse strains. In Vitro Cell Dev Biol Anim 2004;40:76-81.

121. Baharvand H, Kazemi Ashtiani S, Taee A, et al. Generation of new human embryonic stem cell lines with diploid and triploid karyotypes. Dev. Growth. Differ. 2006;48:117-128.

122. Baharvand H, Hajheidari M, Zonouzi R, et al. Comparative proteomic analysis of mouse embryonic stem cells and neonatal-derived cardiomyocytes. Biochem Biophys Res Commun 2006;349:1041-1049.

123. Yin X, Mayr M, Xiao Q, et al. Proteomic analysis reveals higher demand for antioxidant protection in embryonic stem cell-derived smooth muscle cells. Proteomics 2006;6:6437-6446.

124. Colter DC, Sekiya I, Prockop DJ. Identification of a subpopulation of rapidly self-renewing and multipotential adult stem cells in colonies of human marrow stromal cells. Proc Natl Acad Sci U S A 2001;98:7841-7845.

125. Wagner W, Feldmann RE, Jr., Seckinger A, et al. The heterogeneity of human mesenchymal stem cell preparations--evidence from simultaneous analysis of

by on July 10, 2007 w

ww

.StemC

ells.comD

ownloaded from

Page 17: Trends in Stem Cell Proteomics

Trends in Stem Cell Proteomics

16

proteomes and transcriptomes. Exp Hematol 2006;34:536-548.

126. Choi CB, Cho YK, Prakash KV, et al. Analysis of neuron-like differentiation of human bone marrow mesenchymal stem cells. Biochem Biophys Res Commun 2006;350:138-146.

127. Ye NS, Zhang RL, Zhao YF, et al. Effect of 5-azacytidine on the protein expression of porcine bone marrow mesenchymal stem cells in vitro. Genomics Proteomics Bioinformatics 2006;4:18-25.

128. Ye NS, Chen J, Luo GA, et al. Proteomic profiling of rat bone marrow mesenchymal stem cells induced by 5-azacytidine. Stem Cells Dev 2006;15:665-676.

129. Osterhues A, Liebmann S, Schmid M, et al. Stem cells and experimental leukemia can be distinguished

by lipid raft protein composition. Stem Cells Dev 2006;15:677-686.

130. Jeong JA, Lee Y, Lee W, et al. Proteomic analysis of the hydrophobic fraction of mesenchymal stem cells derived from human umbilical cord blood. Mol Cells 2006;22:36-43.

131. Unwin RD, Whetton AD. Systematic proteome and transcriptome analysis of stem cell populations. Cell Cycle 2006;5:1587-1591.

132. Gevaert K, Pinxteren J, Demol H, et al. Four stage liquid chromatographic selection of methionyl peptides for peptide-centric proteome analysis: The proteome of human multipotent adult progenitor cells. J Proteome Res 2006;5:1415-1428.

by on July 10, 2007 w

ww

.StemC

ells.comD

ownloaded from

Page 18: Trends in Stem Cell Proteomics

Figure 1. A schematic representation of differential protein display using 2-DE and

difference in-gel electrophoresis (DIGE). The number in the center of panel represents

the ‘stage’ in the legend. (A) Cell samples are grown under different

conditions/treatments (stage 1 or 1), total proteins are extracted (2) and subjected to

isoelectric focusing (IEF) (first dimension electrophoresis) (3). The IEF gels are reduced

with DTT and alkylated with iodoacetamide prior to SDS-PAGE (second dimension

electrophoresis) (4). The first dimension separates proteins according to isoelectric point

(pI), whereas the second dimension separates them approximately according to molecular

weight (Mr). Proteins are then visualized using silver, Coomassie Brilliant Blue (CBB),

or SYPRO ruby staining methods (5), and the protein pattern is captured by a high-

resolution camera or densitometer and analyzed by software (6). The Mr and pI of each

protein are estimated by comparison with the mobility of standard proteins, and changes

in staining intensity between replicate gels or between treatments are measured. For

protein identification, gels are stained with mass spectrometry-compatible stains such as

SYPRO ruby or CBB. Protein spots are excised from the gel and analyzed by MS (7). (B)

DIGE system includes steps common to 2-DE. However, in DIGE, two protein samples

are differentially labeled by Cy3 and Cy5, respectively, and then mixed (8) and run on

the same IEF and SDS-PAGE gel. This allows co-separation of different labeled samples

in the same gel and ensures that all samples will be subject to exactly the same first- and

second-dimension electrophoresis running conditions, limiting experimental variation and

resulting in accurate within-gel matching [113]. Following 2D separation, the gel is

imaged at the excitation wavelengths of each of fluorescent dye using a scanner, after

17

by on July 10, 2007 w

ww

.StemC

ells.comD

ownloaded from

Page 19: Trends in Stem Cell Proteomics

which an overlay image can be generated (9). Differences in protein abundance are then

accurately quantified using software. Coomassie stain is the most economical and easy-

to–use, but having a detection limit of 50-100 ng per spot, also the least sensitive. Apart

from radioactive gel visualization, silver staining is the most sensitive method with a

detection limit of 1-2ng. SYPRO Ruby is very easy to use and almost as sensitive as

silver stain but more expensive than CBB and silver staining [114]. Cy3/Cy5 is the most

expensive method and requires costly imaging systems but shows very small variability

with different gels and operators. Unlike silver staining, CBB, SYPRO Ruby and

Cy3/Cy5 show a good compatibility with MS. When the aim of the study is relative

quantification between samples, a wide linear dynamic range is required. CBB and silver

stain have a low dynamic range (around 10-fold) , whereas SYPRO Ruby and Cy3/Cy5

represent a much higher dynamic range (1000-fold) and show better correlation between

spot density and protein content compared with silver staining.

18

by on July 10, 2007 w

ww

.StemC

ells.comD

ownloaded from

Page 20: Trends in Stem Cell Proteomics

19

by on July 10, 2007 w

ww

.StemC

ells.comD

ownloaded from

Page 21: Trends in Stem Cell Proteomics

Figure 2. Schematic overview of peptide sequencing by tandem mass spectrometry. The

main components of a mass spectrometer are an ion source, one or more mass analyzers

that measure the mass-to-charge ratio (m/z) of the ionized analytes, and a detector that

registers the number of ions at each m/z value. The gas-phase ions are produced in the

ion source, after which they enter the mass analyzer and are separated according to their

mass/charge (m/z) ratios in the mass analyzer. Matrix-assisted laser desorption/ionization

(MALDI) (A and B) and electrospray ionization (ESI) (C, D, and E) are two techniques

most commonly used to volatize and ionize the variety of biomolecules, including

peptides, proteins, metabolites, and oligonucleotides to enable their measurement by MS.

The mass analyzer is the central component behind the technology. Examples of mass

analyzers currently used in proteomics research are: time-of-flight (TOF), quadrupole

(Q), triple quadrupole or linear ion trap (LIT), ion trap (IT) Fourier transform ion

cyclotron resonance (FTICR), and Orbitrap. The TOF analyzers separate ions based on

the differences in transit time from the ion source to the detector in flight tubes under

vacuum (A, B, and C). The quadrupole analyzer transmits only ions within a narrow m/z

range and uses the stability of the trajectory to separate these ions according to their m/z

ratio on 4 parallel cylindrical metal rods (C). In triple quadrupole or LIT, ions of a

particular m/z are selected in a first section (Q1), fragmented in a collision cell (q2), after

which the fragments are separated in Q3. In the linear ion trap, ions are captured in Q3.

They are then excited via resonant electric field and the fragments are scanned out,

creating the tandem mass spectrum (D). The ion trap analyzer captures or traps the ions

which are then subjected to MS or MS/MS analysis (E). In ion traps, selection,

20

by on July 10, 2007 w

ww

.StemC

ells.comD

ownloaded from

Page 22: Trends in Stem Cell Proteomics

fragmentation, and analysis of ions takes place in the same space. Fourier transform ion

cyclotron resonance mass spectrometer (FT-ICR-MS) is also a trapping device which

operates under high vacuum in a high magnetic field. Recently, Orbitrap has been

introduced as powerful MS which provide high resolution, high-mass accuracy, and good

dynamic range [115]. Its principles are based on injection of ions for storage into an

electrostatic field. The ions are detected using Fourier transform similar to the detection

method used in FT-ICR. Due to the high resolving power of the Orbitrap and the

sensitivity that can be gained using Fourier transform for ion detection, the Orbitrap will

become a very significant MS technology. Several configurations of mass spectrometers

that combine ESI and MALDI with a variety of mass analyzers are routinely used.

MALDI is usually coupled to TOF analyzers that measure the mass of intact peptides.

The MALDI has also been implemented in TOF-TOF mass spectrometers to provide true

MS/MS capabilities (B). The two TOF sections are separated by a collision cell. An ion

of a particular m/z is selected in a first mass analyzer, fragmented in a collision cell and

the fragment ion masses are analyzed by second TOF analyzer. Fragmentation occurs

mainly at the amide bonds of the peptide, resulting in a nested set of peptides that differ

in mass of one amino acid. The measured fragment masses can be compared to

theoretical mass spectra calculated from the protein sequences in the database. ESI has

mostly been coupled to ion traps and quadrupole instruments. Q-TOF instruments exhibit

high resolution and mass accuracy in MS and MS/MS mode (C). The precursor ions are

selected in the quadrupole (Q1) and undergo fragmentation through collision-induced

dissociation (q2). The product ions are analyzed in the TOF device.

21

by on July 10, 2007 w

ww

.StemC

ells.comD

ownloaded from

Page 23: Trends in Stem Cell Proteomics

22

by on July 10, 2007 w

ww

.StemC

ells.comD

ownloaded from

Page 24: Trends in Stem Cell Proteomics

23

Figure 3. Schematic representation of methods for isotopic labeling for relative

quantification using MS. This strategy makes use of the facts that chemically identical

analytes of different stable-isotope composition can be distinguished in a mass

spectrometer owing to their mass difference, and that the ratio of signal intensities for

such analytes indicates their abundance ratio (for review see [109]. (A) Proteins or

peptides derived from two different cell states are derivatized with light and heavy

isotopes of the same chemical reagent. The samples are then combined and analyzed by

MS. The relative abundance levels of the proteins are calculated by comparing the peak

heights of the light- and heavy-labeled peptides. In the chemical labeling approach (A),

stable isotope-bearing chemical reagents are targeted towards reactive sites on a protein.

A method that has gained popularity is the isotope-coded affinity tag (ICAT) approach

[116]. The label contains a thiol group (which reacts with cysteine residues), eight

hydrogens (light) or deuteriums (heavy), used for relative quantification, and a biotin

group that is selectively recognized during the affinity extraction step by an avidin

moiety attached to the chromatographic column. Another method is the iTRAQ which

uses the same approach, but adds an innovative concept, namely a tag that generates a

specific reporter ion in fragmentation spectra [117]. There are four tags that allow the

analysis of four separately labeled protein pools in a single experiment, increasing

analytical throughput. In metabolic labeling (B), the incorporation is achieved by using

media that include isototopic labels, such as 15N or 14N. SILAC has proven to be a simple

and powerful approach for quantitative proteomics [109]. In this method, one cell state is

metabolically labeled by, for example, 13C-containing arginine, which is incorporated

by on July 10, 2007 w

ww

.StemC

ells.comD

ownloaded from

Page 25: Trends in Stem Cell Proteomics

into newly synthesized polypeptides in a sequence-specific fashion. Thus, all arginine-

containing peptides can be labeled. Both the high labelings-efficiency and the absence of

additional chemical labeling steps make the method easy to apply. In labeling via enzyme

reaction (C), an 18O isotope is coupled to the peptides by the protease that splits the

proteins into peptides. During this proteolytic step, which takes place in H218O, 18O from

the water molecules is incorporated into the resulting peptides [118,119].

24

by on July 10, 2007 w

ww

.StemC

ells.comD

ownloaded from

Page 26: Trends in Stem Cell Proteomics

25

by on July 10, 2007 www.StemCells.com Downloaded from

Page 27: Trends in Stem Cell Proteomics

Figure 4. Schematic representation of protein microarray. Protein chips are made in

much the same way as DNA microarrays. A glass or plastic surface is spotted with an

array of molecules designed to capture specific proteins; these capturing molecules can

be other proteins such as antibodies, antigens, enzymes, peptides, or even snippets of

DNA or small organic ligands. Fluorescent markers or other detection schemes reveal

which spots have formed a complex with their targets. Because the identity of each

protein-binding molecule on the chip is known, a particular spot on the chip that emits

light upon excitation-specific stimulation indicates that a target protein has been captured

or that an enzyme has been activated. However, this technique is not as straight forward

as that for conventional DNA chips. (A) Analytical protein microarrays contain.

antibodies with high affinity and specificity that are spotted on an appropriate surface.

These chips are used for proteome profiling and monitoring proteome expression pattern

and clinical diagnostics. In the example shown, all of the proteins in two biological

samples that one would like to compare are labeled with distinguishable markers (e.g.

labeled with red or green fluorescent dyes). Then, protein samples are mixed and this

mixture is used to incubate the array. Spots that show up as green or red have an excess

of protein from one sample over the other. Spots that appear yellow approximate an equal

amount of protein from each sample. This experimental procedure is analogous to gene

expression profiling with DNA microarrays, and allows the effect of various

physiological stimuli or genetic alterations on proteins levels to be examined

simultaneously. In analytical protein array, different types of ligands, including

antibodies, antigens, DNA or RNA, aptamers, carbohydrates or small molecules, with

high affinity and specificity, are spotted onto a derivatized surface. (B) Functional protein

26

by on July 10, 2007 w

ww

.StemC

ells.comD

ownloaded from

Page 28: Trends in Stem Cell Proteomics

microarrays require native proteins or peptides that are individually purified or

synthesized from cDNAs by in vitro transcription/translation or other high throughput

techniques, after which they are spotted onto a suitable surface to form the functional

protein microarrays. These chips are used to probe protein activities, binding properties

and post-translational modifications. In the example shown, proteins are labeled with

distinguishable markers (e.g. labeled with green fluorescent dye). The spots that `light up'

are candidate binding partners. With the proper detection method, functional protein

microarrays can be used to identify the substrates of enzymes of interest. This class of

chips is particularly useful in drug and drug-target identification and in ubraveling

biological networks.

27

by on July 10, 2007 w

ww

.StemC

ells.comD

ownloaded from

Page 29: Trends in Stem Cell Proteomics

28

Figure 5. Venn diagram of shared and unique protein expression in ESCs, MSCs, and

NSCs. Regions of overlap between circles indicate common gene expression. Regions

that do not overlap between circles indicate unique gene expression signatures of

particular cell types. (A) Venn diagram showing unique and common proteins in human

(left) and mouse (right) ESCs. The overlapping region of hESCs and mESCs contains 92

proteins identified in both ESCs. (B) Venn diagram showing unique and common

proteins in human BM-MSCs (left) and AD-MSCs (right). The overlapping circles

indicate 52 proteins identified in both MSCs. (C) Venn diagram showing unique and

common proteins in rat NSCs (left) and human NSCs (right). The overlapping circles

indicate 65 proteins identified in both NSCs. (D) Venn diagram showing unique and

common proteins in ESCs, MSCs, and NSCs. The overlapping circles indicate nine

proteins expressed in all three SCs. The gene symbols of SCs specific and common

proteins are indicated in boxes.

The three SC types (ESCs, MSCs, and NSCs) shared nine proteins identified in

proteomics screens, including proteins involved in energy production and metabolisms

(Atp5b: ATP synthase beta chain, Eno1: Enolase 1, and Tpi1: Triosephosphate

isomerase), disease and stress (Stip1: stress-induced-phosphoprotein 1 and Prdx1:

Peroxiredoxin 1), protein folding (Cct2: chaperonin containing TCP1- subunit 2β, Hspa5:

78 kDa glucose-regulated protein precursor, Hspa8: beta subunit and Heat shock cognate

71 kDa protein), and an unclassified protein (Tpt1: Translationally controlled tumor

protein, or TCTP).

by on July 10, 2007 w

ww

.StemC

ells.comD

ownloaded from

Page 30: Trends in Stem Cell Proteomics

29

by on July 10, 2007 www.StemCells.com Downloaded from

Page 31: Trends in Stem Cell Proteomics

Figure 6. Morphology and corresponding 2-DE gel of ESC colonies of different species

cultured on mouse embryonic fibroblast feeder cells. Phase contrast microscopy of (A)

mESC colonies (Royan B1, [120]) derived from C57BL/6 strain, (B) monkey ESC

colonies (Macaca fascicularis) (a gift from professor Taru Kita, Kyoto University) (C)

human ESC colonies (Royan H5, [121]). (D) We developed a 2-DE database of hESCs

that contains hyperlinked 2-DE gel images and descriptive textual information such as

protein name, Mr/pI values, MS score, sequence coverage and other information that is

publicly available at http://www.RoyanProteomics.ir. It also provides facilities to search

protein spots on 2D gels and retrieve information related to experimental design, MS

analysis, and Mascot search results. The information available in this database will be

linked to the results of other SC proteome studies, aiming to provide a comprehensive,

comparable, and expandable resource for SC research. 2-DE gels from mouse and

monkey ESCs have been incorporated recently in this database.

30

by on July 10, 2007 w

ww

.StemC

ells.comD

ownloaded from

Page 32: Trends in Stem Cell Proteomics

31

by on July 10, 2007 w

ww

.StemC

ells.comD

ownloaded from

Page 33: Trends in Stem Cell Proteomics

Table 1: A summary of published papers about stem cell proteomics.

Identified proteins

Cell type/source cell line passage Differentiated

cells Treatment Proteomic approach Total Unique

Differentially expressed proteins

Major result Ref.

PKU ND Neurons RA 2DE/MALDI TOF 12 12 24

As a first proteomic analysis of ESC differentiation, it showed the application of proteomics to unravel the regulatory network of differentiation.

[63]

R1 4-5 NA NA 2DE/MALDI TOF 241 218 NA The first 2-DE map of mESC [52]

TT2F and TT2F/hChr 21

ND Neurons Coculture with PA6

2DE/MALDI TOF 18 18 18 Candidate proteins involved in developmental

abnormalities in down syndrome [67]

CCE ND Mixed cells

Combination of fibronectin, laminin, LIF, FGF4

Western blotting NA NA

Phosphorylation states of 31 defined proteins were studied

A set of seven phosphoproteins (Stat3, Raf1, MEK, ERK, Src, PKCα, and PKBε) most critically associated with cell differentiation rates and or proliferation rates

[79]

E14-1 ND NA NA 2D LC MS/MS 1706 1706 NA Comprehensive proteome map of mESC using

2D LC MS/MS [53]

E14 ND Neurons RA 2DE/Q TOF MS/MS

23 23 23 Potential roles of TCTP in modulating neural differentiation [64]

mESC

D3 ND Mixed cells Feeder-free culture

DIGE/ MALDI TOF

100 93 100

First nuclear proteome analysis of ESC showing that specific chromatin-related proteins may be involved in maintaining the unique properties of pluripotent ESCs

[66]

Derived from Balb/c and 129

ND Neurons Growth factor mediated

2DE/MALDI TOF 7 6 7

Studied the effect of chemical ischemia on neural ESC model indicating a particular role of an isoform of the acidic calcium-independent phospholipase A2 in this type of insult

[78]

32

by on July 10, 2007 www.StemCells.com Downloaded from

Page 34: Trends in Stem Cell Proteomics

Identified

proteins

Cell type/source cell line passage Differentiated

cells Treatment Proteomic approach Total Unique

Differentially expressed proteins

Major result Ref.

mESC (continued)

LC FTICR-MS/MS

The largest proteome map of mESCs/identification of 743 SC specific proteins

D3 20 Mixed cells Feeder-free culture 1775 1775 743 [54]

2DE/ Q-TOF and MALDI MS/MS

Altered PTM of proteins related to Hsp90 chaperone function, to protein catabolism, and to chromatin remodeling may be highly relevant to SC fate

J1 ND Mixed cells Feeder-free culture 183 108

39 phosphorylated

[72]

2DE/MALDI TOF/TOF

Comparative proteome analysis of mESC- and neonatal-derived cardiomyocyte showed evidence for a close relation between them

Feeder-free culture Royan B1 20-25 Cardiomyocyte 60 58 109 [122]

Feeder- and serum-free culture

DIGE/Q-TOF MS/MS

Hsp25 was suggested as a potentially marker of early ESC neural cell differentiation E14 ND Neurons 6 6 10 [65]

Sca+ D3, CRL-1934 and ATCC

selection and Growth factor mediated

DIGE/ MALDI TOF

ESC-derived smooth muscle cells showed higher demand for antioxidant protection compared to aortic smooth muscle cells

Smooth muscle cell 128 75 146 [123] ND

mECC Two of the identified proteins, cofilin and peroxiredoxin 6, had the same expression patterns as ESCs [63]

2DE/MALDI TOF P19C6 ND Neurons RA 28 28 28 [68]

hESCs Royan H2, Royan H3, Royan H5

2DE/MALDI TOF/TOF

The first 2-DE map of hESCs and the largest 2-DE map of SCs NA NA 685 434 NA [55]

The largest proteome map of hESCs/identification of 639 SC specific proteins

LC FTICR-MS/MS

Feeder-free culture HES-2 47 Mixed cells 1532 1532 639 [54]

33

by on July 10, 2007 www.StemCells.com Downloaded from

Page 35: Trends in Stem Cell Proteomics

Identified

proteins

Cell type/source cell line passage Differentiated

cells Treatment Proteomic approach Total Unique

Differentially expressed proteins

Major result Ref.

hBM-MSCs The importance of distinguishing the major subpopulation of MSCs, due to their difference in subpupolations of hBM-MSCs with rapidly self renewing and multipotential lineages

2DE/MALDI TOF & MS/MS

NA ND Very small round MSCs

Collected not differentiated 40 37 40 [124]

TGF-β coordinates the increase of α-actin and the decrease of gelsolin to promote MSC differentiation

2DE/Q-TOF NA Up to 10 Mixed cells TGF-beta 30 27 60 [69]

2D LC MS/MS

Comparative proteome analysis of hMSC and osteoblast NA ND Osteoblast NA 597 597 NA [57]

Using osteogenic medium

Identifying reveal novel markers for hMSCs using MS-based proteomics/ weak correlation between transcriptome and proteome results

hMSCs - hTERT

2D LC MS/MS ND Osteoblast 463 463 104 [35]

Using a quantitative phosphoproteomics approach, it was shown that EGF and PDGF modulate osteogenic capability of MSCs through MAPK/ERK. P38 kinase and phosphatidylinositol 3 kinase signaling

Grwoth factors (PDGF & EGF)

SILAC/Q TOF & LIT-FT MS/MS

NA NA NA 113 113 113 [80]

Different culture media

culture conditions exert a prominent impact on transcriptome, proteome, and cellular organization of hBM-MSC

2DE/MALDI TOF NA 3-6 NA 134 81 241 [125]

NA after 4 Neuron-like

Serum-free culture and induction medium

Neuron-like cells derived from chemical induction of hMSCs are not the genuine neurons

2DE/MALDI TOF

20 18 80 [126] Differentially regulated passage-specific

proteins may play a role in the decrease of osteogenic differentiation potential under serial subculturing

Using osteogenic medium

2DE/MALDI TOF NA 1 to 7 Osteoblast 12 12 12 [89]

34

by on July 10, 2007 www.StemCells.com Downloaded from

Page 36: Trends in Stem Cell Proteomics

Identified

proteins

Cell type/source cell line passage Differentiated

cells Treatment Proteomic approach Total Unique

Differentially expressed proteins

Major result Ref.

Using adipogenic medium

hBM-MSCs 2DE/MALDI TOF

Eight adipocyte-differentiation associated proteins [70] NA 6 Adipocyte 8 8 32 (continued)

Porcine BM-MSCs The expression level of proteins involved in

cell proliferation, differentiation, and cell defense changed in response to 5-aza

NA 2 Cardiomyocyte 5-azacytidine 2DE/MALDI TOF 21 18 36 [127]

Rat BM-MSCs The expression level of proteins involved in

cell proliferation, differentiation, and cellular defense was found to change

NA 3-5 Cardiomyocyte 5-azacytidine 2DE/MALDI TOF 34 29 9 [128]

DO64 (CD34

Canine BM-SCs Comparison of lipid raft fraction of DO64 with

its transformed lines showed that lipid raft proteins may be useful for identification of molecular markers of transformation form.

- and plastic-adherent cells)

2DE/MALDI TOF/TOF

ND NA NA 5 5 7 [129]

hAD-MSCs Proteome mapping and differentiation analysis/ the importance of heat shock protein family with respect to the etiology of obesity and type 2 diabetes

Adipogenic Differentiation Medium

2DE/Q TOF MS/MS

NA 1 Adipocyte 424 286 53 [60]

hUCB-MSCs 2DE/MAL

DI TOF/TOF

NA 3 NA NA 205 145 NA Proteome mapping of hUCB-MSCs [56]

2DE/MALDI TOF NA up to 8 NA NA 35 32 NA Identification of hydrophobic proteins [130]

HSCs Growth factor mediated

iTRAQ/LC MS-MS

validation and use of iTRAQ reagents on the FDCP-Mix TEL/PDGFR system FDCP-Mix ND myeloid cells 347 347 13 [131]

35

by on July 10, 2007 www.StemCells.com Downloaded from

Page 37: Trends in Stem Cell Proteomics

36

Identified

proteins

Cell type/source cell line passage Differentiated

cells Treatment Proteomic approach Total Unique

Differentially expressed proteins

Major result Ref.

mBM-HSC

NA NA NA NA iTRAQ/LC MS/MS 948 948 145

A comparision between long term reconstituting HSCs (Lin-Sca+Kit+; LSK+) and non-long term reconstituting progenitor cells (Lin-Sca+Kit-; LSK-) revealed that LSK+ cells are adapted for anaerobic environments/weak correlation between transcriptome and proteome results

[77]

NA NA NA NA 2DE/IT MS/MS 22 19 NA stem cell-intrinsic heterogeneity of individual

samples [81]

hUCB-HSC

NA NA myeloid cells (CD34+ to CD15+ cells)

Growth factor mediated

2DE/MALDI TOF 127 127 127

The first proteomic approach for global cellular protein expression patterns in human HSC differentiation to myeloid cells

[71]

hNSCs ReNcell VM ND Neural tissue

Growth factor mediated

MALDI TOF and MALDI TOF/TOF

402 318 49 The large rearrangement of the proteome during the differentiation [27]

hMAPCs

NA ND NA NA 2D LC MS/MS 2151 2151 NA

The first proteomic analysis of hMAPC. Unlike differentiated cells, the ASCs express a patchwork of genes which is uniformly distributed among the 22 autosomal chromosomes.

[132]

NA P1 NA NA 2DE/MALDI TOF 266 109 NA The first proteome map of hippocampal NSC [58]

Rat NSCs

NA ND Neural cells Growth factor mediated

2DE/MALDI TOF 128 87 367 Massive reorganization of the stem cell

proteomic profile upon differentiation [59]

by on July 10, 2007 www.StemCells.com Downloaded from

Page 38: Trends in Stem Cell Proteomics

2-DE: two-dimensional electrophoresis, ASCs: adult stem cells, DIGE: difference in-gel

electrophoresis, EGF: Epidermal growth factor, ESCs: embryonic stem cells, FGF:

fibroblast growth factor, FTICR: Fourier transform ion cyclotron resonance, hAD-MSCs:

human adipose derived-mesenchymal stem cells, hBM: human bone marrow, hChr:

human chromosome, hMAPC: human multipotent adult progenitor cells, hNSCs: human

neural stem cells, HSCs: hematopoeitic stem cells, Hsp: heat shock protein, hTERT:

human telomerase reverse transcription, hUCB: human umbilical cord blood, iTRAQ:

isobaric Tag for relative and absolute quantitation, LC: liquid chromatography, LIF:

leulemia inhibitory factor, MS: mass spectrometry, MALDI: matrix-assisted laser

desorption/ionization, mESCs: mouse ESCs, MSCs: mesenchymal stem cells, NA: not

available, ND: not determined, NSCs: neural stem cells, PDGF: platelet-derived growth

factor, PTM: post translational modification, Q: quadrupole, RA: retinoic acid, SCs: stem

cells, SILAC: Stable isotope labeling by amino acids in cell culture, TCTP:

translationally controlled tumor protein, TGF: transforming growth factor, TOF: time-of-

flight. IT: Ion trap

37

by on July 10, 2007 w

ww

.StemC

ells.comD

ownloaded from

Page 39: Trends in Stem Cell Proteomics

DOI: 10.1634/stemcells.2007-0107 published online Jun 7, 2007; Stem Cells

Hossein Baharvand, Ali Fathi, Dennis van Hoof and Ghasem Hosseini Salekdeh Trends in Stem Cell Proteomics

This information is current as of July 10, 2007

& ServicesUpdated Information

http://www.StemCells.comincluding high-resolution figures, can be found at:

by on July 10, 2007 w

ww

.StemC

ells.comD

ownloaded from