Mutagenesis vol. 27 no. 3 pp. 257–266, 2012 Advance Access Publication 6 December 2011 doi:10.1093/mutage/ger086 REVIEW The Syrian hamster embryo (SHE) assay (pH 6.7): mechanisms of cell transformation and application of vibrational spectroscopy to objectively score endpoint alterations Abdullah A. Ahmadzai, Júlio Trevisan, Nigel J. Fullwood1, Paul L. Carmichael2, Andrew D. Scott2 and Francis L. Martin* Centre for Biophotonics, Lancaster Environment Centre and 1Division of Biomedical and Life Sciences, School of Health and Medicine, Lancaster University, Lancaster LA1 4YQ, UK, 2Safety and Environmental Assurance Centre, Unilever Colworth Science Park, Bedfordshire MK44 1LQ, UK. * To whom correspondence should be addressed. Centre for Biophotonics, Lancaster Environment Centre, Lancaster University, Lancaster LA1 4YQ, UK. Tel: þ44 (0) 1524 510206; Fax: þ44 (0) 1524 510217; Email: f.martin@ lancaster.ac.uk Received on August 18, 2011; revised on October 6, 2011; accepted on October 19, 2011 Using morphological transformation as an endpoint, the Syrian hamster embryo (SHE) cell transformation assay (pH 6.7) is an in vitro system with a high sensitivity and specificity for testing the carcinogenic potential of test agents. Advantages of the assay are that SHE cells are metabolically competent, genetically stable and acquire spontaneous transformation with a low frequency; additionally, it detects both genotoxic and non-genotoxic carcinogens. However, in comparison with other shortterm mammalian cell assays, it is time consuming, laborious and, most importantly, the visual scoring of morphological transformation might be subjective. In this review, we examine the background to the test and why it has the potential for use in safety risk assessment. Additionally, we propose a novel approach to objectively interrogate and classify SHE colonies using vibrational spectroscopy coupled to a mathematical framework for high-throughput screening. It is our view that this alternative approach has the potential to improve the sensitivity and specificity of the in vitro SHE assay. Introduction Initially described by Berwald and Sachs in 1963, the Syrian hamster embryo (SHE) cell transformation assay is an in vitro cell culture system that has been used to test the carcinogenic potential of biological, physical and chemical agents (1–3). To date, SHE cell neoplastic transformation is the most relevant and truest in vitro simulation of the in vivo neoplastic process (3,4). SHE cells are primary diploid cells, isolated from 13day-old embryos, with a finite lifespan (4). Following test agent exposure, SHE cells may undergo morphological transformation as a result of a block in cellular differentiation (Figure 1) (3,5). Additional mutations in morphologically transformed SHE cells lead to immortal, tumorigenic and then malignant cells (3,6,7). The absolute goal of short-term genotoxicity testing is the risk detection of human carcinogens rather than identifying positives in rodents (8). Such chemicals cannot be tested in humans, so the rodent bioassay is the current standard by which the potential human carcinogenicity of different test agents is established (8). The SHE cell transformation assay is a unique in vitro assay that detects both genotoxic and nongenotoxic rodent carcinogens (2,9); as suggested later, this uniqueness critically underpins the rationale for harnessing this assay to spectroscopy interrogation with computational analysis to allow one to discriminate test agent effects based on mechanisms of action. SHE cells, under standard culture conditions, like human cells and unlike mouse cells, are genetically very stable and acquire spontaneous transformation with a low frequency (4,10,11). However, when treated with carcinogenic agents, the immortality rate increases within SHE cell populations with a frequency dependant on the particular test agent being investigated (4,12,13), thus making this assay a useful tool to study the multistage carcinogenic process (4). Since its first introduction in 1963, many modifications have been made to the SHE assay in order to improve its performance. The reduction of SHE cell culture media from pH 7.3 to pH 6.7 by LeBoeuf and Kerckaert (13) improved the assay sensitivity (ability to detect true positives) to 87% from 82% and specificity (ability to classify true negatives) to 83% from 69% (2,8). Thus far, in the pH 6.7 version of the SHE cell transformation assay, .200 physical and chemical agents have been tested (5,8). With an overall concordance of 85% with rodent bioassays, the SHE assay has been reported to outperform the standard battery of in vitro genotoxicity tests (Salmonella typhimurium bacterial mutagenicity assay, mouse lymphoma assay and in vitro chromosome aberrations test) in terms of their ability to predict carcinogenic potential (8). However, the SHE assay has limitations including the subjectivity of scoring morphologically transformed colonies and its low rate of increased transformation in response to carcinogenic agents (2,14). In this review, we examine the background to the assay and why it still has potential as an important test for safety risk assessment. Additionally, we propose a novel approach to objectively interrogate and classify SHE colonies based on infrared (IR) spectroscopy coupled to a mathematical framework for high-throughput screening. Such an approach offers the possibility to objectively discriminate agents based on mechanism of action according to the post-spectroscopy classification of altered IR spectra (14). With IR spectroscopy, staining is not necessary and results are produced computationally, based on a previously built knowledge base. This knowledge base consists basically of data and classification models trained on these data. Because of the non-destructive nature of this approach, SHE colonies can be subsequently stained, and conventionally visually scored for comparative purposes and towards the long-term validation of this novel approach. Ó The Author 2011. Published by Oxford University Press on behalf of the UK Environmental Mutagen Society. All rights reserved. For permissions, please e-mail: [email protected]. 257 A. A. Ahmadzai et al. Fig. 1. Schematic overview of the SHE cell transformation assay. A cytotoxicity assay is carried out prior to the SHE cell transformation assay to determine the appropriate test agent exposure, e.g. concentration. The exposure that causes 50% cytotoxicity is taken as the highest in the main study. The assay includes solvent (VC) and positive control treatment groups in addition to test agent treatments. The SHE cells are exposed for 24 h or 7 days before being fixed, stained and microscopically assessed for cell/colony morphology. Why the SHE assay remains relevant The SHE cells are metabolically competent and can activate many chemicals to their ultimate carcinogenic form without the addition of an exogenous activation system (2,5). As they are embryo-derived, the SHE cell population is composed of multiple cell types that are at different stages of the differentiation process, thus providing a broad spectrum of cellular targets for the transformation response (2,15–18); this attribute is believed to contribute towards the overall sensitivity of the assay. Under standard culture conditions (pH 7.3), .472 agents have been tested in the SHE cell transformation assay (15,19). Only 213 (of 472) agents have in vivo rodent carcinogenicity data available; the pH 7.3 version of the assay classified 177 (of 213) agents as rodent carcinogens and 36 as non-carcinogens (19). For these 213 physical and chemical agents, the SHE assay at pH 7.3 has a sensitivity of 82% (146/ 177), a specificity of 69% (25/36) and an overall concordance with rodent bioassay of 80% (171/213) (19). In 1987, the pH 7.3 of the Dulbecco’s Modified Eagle Medium (DMEM) cell culture medium was reduced to pH 6.7 by LeBoeuf and Kerckaert (13). This resulted in better SHE cell clonal growth, improved morphological transformation performance and less assay-to-assay variability (13,15). Lowering the culture pH had direct effects on the SHE cell phenotype including a decrease in levels of mRNA transcripts for gap junction protein leading to reduced intercellular gap junction communication, changes in protein charge, changes in protein phosphorylation at serine/ 258 threonine and tyrosine residues, and changes in some gene expression (4,16,19). Under the reduced pH methodology, the SHE assay has a sensitivity of 87%, specificity of 83% and an overall concordance with rodent carcinogenicity data of 85% (2,8,15). These improved accuracy and performance outcomes of the SHE assay are consistent with a database of .200 chemicals (2,8,15). In comparison, a battery of S. typhimurium bacterial mutagenicity, in vitro chromosome aberrations and mouse lymphoma tests positively detected 47 of 60 carcinogens; the SHE assay alone was sensitive to 52 of these test agents (2,8,20). Additionally, the three-test battery had a specificity of 32% (13/40), whereas the SHE assay had a specificity of 83% (33/40) (2,8,20). The SHE assay was also shown to be able to detect S. typhimurium-positive carcinogens and S. typhimurium-negative carcinogens with a sensitivity of 100% and 78%, respectively (2,8,20). Limitations of the assay in current practice Although the SHE cell transformation assay is an attractive model, several challenges have made it problematic for routine use in risk assessment. Within the SHE cell population, 1/3 of the clonable cells are capable of undergoing morphological transformation following carcinogen exposure (15). These sensitive-to-morphological-transformation cells are relatively undifferentiated and, based on their responsiveness to growth differentiation factor, they can be divided into three populations: an undifferentiated cell population, a committed progenitor cell Biospectroscopy to objectively score the SHE assay population with partial epithelial cell-like characteristics and a committed progenitor cell population possessing mesenchymal cell-like characteristics (15). While this composition of different cell types gives the SHE assay the advantage of capability to detect a wide range of carcinogens, including those with tissuespecific modes of action, it is also the cause of variation in sensitivity to chemical transformation when different aliquots of cells from the same culture are used (15,16,18). The ability of an agent to induce pre-neoplastic changes is measured by visually scoring the morphologically transformed SHE colonies. At present, there is currently no objective molecular marker to identify transformed SHE cells (21); thus, a trained eye is required to score the colonies under the microscope (Figures 1 and 2). Large sample sizes are required in order to derive appropriate statistical analysis because of the low frequency of induced morphological transformation following chemical treatment (2,17). In vitro simulation of in vivo multistage neoplastic transformation Neoplastic transformation is a multistage process in which cells acquire multiple genetic alterations resulting in loss of the organism-determined growth control of individual cells (4). A number of in vitro cell transformation assays have been used to explore the mechanistic basis of both in vivo and in vitro multistage neoplastic transformation. Different in vitro assays model different stages of the multistage carcinogenesis process (22,23). For instance, the BALB/c 3T3 and C3H/10T½ cell assays have been used to predict the conversion of immortal cells to malignant cells, which is the final stage of neoplastic transformation. In contrast, the SHE cell transformation assay is the only in vitro assay which models multistage carcinogenesis from a normal diploid cell to a fully malignant one (4,10,12). When treated with carcinogen, the SHE cells undergo morphological transformation due to genetic alter- ation. Morphologically transformed SHE cells are more susceptible to further genetic alterations compared to normal SHE cells. Thus, further genetic changes in morphologically transformed SHE cells results in an immortal cell line. Immortal SHE cells acquire additional genetic alterations en route to becoming tumourigenic and eventually malignant. Each stage of this transformation process is examined below. Morphological transformation The SHE cells can acquire morphological transformation following exposure to genotoxic or non-genotoxic carcinogenic agents (1,24). Morphologically transformed SHE cells display a high nuclear-to-cytoplasmic ratio with criss-crossed, randomly oriented and three-dimensional colony growth characteristics (4,15,24) (Figure 2). Morphological transformation is believed to arise from a block in the normal cellular differentiation of stem cells within the SHE cell population (25). It has been shown that 75% of the morphologically transformed SHE cells exhibit reduced expression of h19 gene compared to normal SHE cells; this is believed to be responsible for the blockade in cellular differentiation (15,25). In addition, re-establishment of wild-type h19 in tumourigenic SHE cells results in the suppression of tumourigenicity in the cell population (25). The h19 gene is believed to be involved in the regulation of cellular differentiation in embryogenesis. Expression of h19 is low in undifferentiated embryonic stem cells but increases upon the induction of embryonic stem cell differentiation into embryoid bodies (25–28). Different agents induce morphological transformation in SHE cells by targeting different genes. Zinc has anti-apoptotic properties and can induce morphological transformation of normal SHE cells (29). Zinc-induced morphologically transformed SHE cells show up-regulation of anti-apoptotic bcl-2 and down-regulation of pro-apoptotic bax expression (30,31). Studies of the anti-apoptotic properties of zinc suggest that Fig. 2. The multistage SHE neoplasm. Normal SHE cells (A) may acquire a morphologically transformed phenotype (B) after exposure to carcinogenic test agent. Morphologically transformed SHE cells senesce unless they acquire further genetic alteration en route to immortalisation and tumorigenicity (C). When tumourigenic cells are injected into nude mice, cancer sites may develop (D), which through further mutation transform into malignant cells and metastasise. 259 A. A. Ahmadzai et al. inhibition of apoptosis occurs by an epigenetic mechanism and contributes to the early stage of transformation by extending the viability of altered cells (29,30). However, over expression of c-myc is not observed in zinc-induced morphologically transformed SHE cells. Benzo[a]pyrene (B[a]P) also has no effect on c-myc expression at transforming concentrations in SHE cells, whereas 2,4-diaminotoluene (2,4-D) induces over expression of this gene (32). The proto-oncogene c-myc is believed to play an important role in cell apoptosis and is closely linked to bcl-2 (30,33–35). These contrasting observations suggest that different carcinogens mediate their transforming effects via different genes and proteins in SHE cells. The rate of spontaneous morphological transformation in the SHE assay (pH 6.7) approximates 0.1–0.4% (4). Morphological transformation of SHE cells can result in either a stable or a reversible phenotype depending upon the carcinogen being tested and different test agents transform SHE cells with different frequencies (4). For instance, the frequency of morphological transformation caused by B[a]P is 1–3% whereas 12-O-tetradecanoylphorbol 13-acetate, a cancer promoter, induces a level of 10% in SHE cells. However, the window of opportunity within which morphological transformation can occur is believed to be transient and is lost at higher passages as the number of differentiated daughter cells (in which a block in differentiation is impossible) continually increases, whereas the stem cell population (in which differentiation could be blocked) decreases in number (15). Immortality Morphologically transformed cells acquire immortality with a frequency of 10–100% depending on the test agent being used to induce morphological transformation (4,11,36). For instance, the frequency for B[a]P-induced morphologically transformed cells going on to establish an immortal cell line is 30% whereas the level derived from 3-methylcholanthrene (MCA)-induced morphological transformation is 14% (11,36). However, control SHE cells achieve immortality with a frequency of 0–3% (11,36). Immortality is the result of further genetic alterations in morphologically transformed SHE cells; otherwise, the cells senesce (4,37). Cellular senescence appears to be primarily controlled by four genetic complementation groups involving at least eight chromosomal loci, including the tp53 and rbi (19,38). Multiple biochemical factors including mortalin, DNA methylation, ceramide, telomerase activity and prohibitin are also considered to be pivotal in the control of cellular senescence (19,39–41). Although the exact nature and number of mutations required to give rise to immortal SHE cells remains unknown, the majority that do acquire immortalisation are aneuploid (11,42). The karyotype of chemically induced SHE cell lines often exhibits double non-disjunctions resulting in trisomy of chromosome 11 and duplication of a translocated chromosome (43). Cytogenetic studies on early-passage SHE cell lines that were exposed to asbestos showed that six of the eight cell lines had trisomy chromosome 11, whereas the remaining two cell lines had other chromosomal abnormalities, including X or Y, þ3, 8p-, -13 and t(13;21) (44). Four of the eight cell lines had a gain of chromosome 8 (44). Endo and Hieber (45) reported that four of six radiation-induced immortal SHE cell lines exhibited non-random rearrangements in the long arms of chromosome 6 (6qþ) and chromosome 3 (3qþ); however, spontaneously transformed SHE cells did not show the same 260 genetic alterations. B[a]P-induced immortal SHE cell lines exhibit trisomy of chromosome 8, monosomy of chromosome 16, monosomy of chromosome 10 and deletion of 2p. Other chromosomal alterations that have been reported in immortal SHE cell lines include trisomy of chromosomes 3 and 7, and monosomy of chromosomes 13 and 15 (4,46–50). All the available studies to date on immortal SHE cells suggest that different agents cause different chromosomal alterations. Untransformed SHE cells are stimulated in response to epidermal growth factor (EGF), to some members of fibroblast growth factor family proteins, to certain cytokines [including interleukin (IL)-4, IL-9 and erythropoietin] and the plateletderived growth factor (PDGF) family, while being inhibited by other cytokines (including IL-1a and IL-1b), transforming growth factor (TGF)-b and nerve growth factor (18). However, immortal SHE cell lines are unresponsive to EGF, PDGF and TGF-b1 (18). Furthermore, immortal SHE cells express retinoblastoma (rb) protein, which is normally in its phosphorylated form (51). Such immortal SHE cells can be induced to undergo senescence by introducing a normal copy of human chromosome 1 into them, suggesting that some genes located on this chromosome are involved in the regulation of cellular senescence (4,52). Towards acquisition of tumourigenicity Immortal SHE cells may then go on to acquire tumourigenicforming capacity following further genetic alterations, which mostly occur spontaneously as a result of genetic instability (4). Transformed SHE cells cycle at twice the rate of untransformed SHE cells and exhibit anchorage-independent growth in soft agar (53,54). The resultant tumourigenic SHE cells express ornithine decarboxylase at twice the levels of those found in untransformed SHE cells and high levels of cytosolic NAD(P)H: quinone oxidoreductase (DT diaphorase); after 24 passages, they are nearly 100% effective at inducing cancer in newborn hamsters within 6 weeks (53). Several proto-oncogene and tumour suppresser genes have been identified that are believed to contribute towards neoplastic transformation. The c-srv is a non-receptor thymidine kinase; mutations in the c-srv gene leading to increased expression of altered forms of pp60c-src protein have been noted in asbestos-induced tumourigenic SHE cell lines (55). Cells that did not translate the correct pp60c-src protein failed to grow in soft agar (55,56). In another study, 20% of SHE cell lines transformed by 3-MCA were shown to contain an inactive tp53 gene (57), whereas the asbestos- or diesthylstilbestrol (DES)transformed SHE cell lines carried wild-type tp53 (58). Thus, transformed cells that carry wild-type tp53 probably contain mutations in other apoptosis-regulating genes. Arsenic-exposed tumourigenic SHE cells over express c-myc and c-Ha-ras oncogenes (59). Nevertheless, the SHE cells that were treated with DES also expressed elevated levels of c-Haras oncogene mRNA transcripts at the neoplastic stage, whereas trenbolone induced high transcriptional levels of c-myc and cHa-ras at the pre-neoplastic (immortal) stage (60). However, asbestos, N-methylnitrosourea and aflatoxin B1 had no apparent effect on the transcriptional levels of these oncogenes (60). Other mutated oncogenes that have been identified in neoplastic SHE cells include c-Nras, cph and c-fos (4,60–63). The proliferation of normal cells is strictly controlled by cell cycle checkpoints that have been shown to be impaired in cancer cells (64). These checkpoints are in place to best ensure Biospectroscopy to objectively score the SHE assay the fidelity of the DNA sequence and, to this end, regulate the timing of cell cycle events (64,65). Any damage in DNA should lead to a blockade of mitosis at the G2/M-phase checkpoint, which provides the cell enough time to repair or activate apoptosis (64). This control is bypassed in SHE cells that have been transformed by malachite green; these have abrogated G2/M-phase checkpoint control resulting in uncontrolled cell division (64). Malignancy and metastasis in vivo Tumourigenic SHE cells undergo further genetic alteration in vivo to acquire a malignant state and an ability to metastasise (Figure 2) (4). Metastasis, the spread of malignant cells from the primary site, is a non-random multistep process that is dependent on the properties of the tumour cells and the host response (66,67). Acquisition of the metastatic phenotype is not well understood. However, the general process involves the production of proteolytic enzymes by tumour cells to enable detachment from the primary site so that they may invade the surrounding tissues and pass through the blood stream to lodge at distant (or secondary) sites (67). Such malignant cells exhibit loss of cell adhesion molecules and acquire the ability to escape from host immune surveillance (67,68). Tumour cells in vivo are surrounded by extracellular matrix (ECM), which is mainly composed of proteoglycans, heparan sulfate, collagen and keratan sulfate. ECM plays a key role in providing protection for tumour cells against chemotherapy-induced cell death (69). Tumour cells also express increased levels of plasminogen activators (PAs), which convert plasminogen to plasmin, conferring the property of enhanced fibrinolytic activity (70– 72). Although PAs are involved in metastasis, increased activity is not always correlated with malignancy (73–77). When introduced into nude mice or neonatal hamster, transformed SHE cells give rise to tumours with high frequency (10,12) (Figure 2). Malignant SHE cells are capable of metastasizing as they produce ECM proteins and t-PA and show resistance towards macrophages (4,10,77–80). Mutations leading to activated Ras have been reported in numerous human tumours, a process that is involved in both tumourigenicity and metastasis in rodent cells in vitro (66,81–84). In SHE cells, activated Ha-ras oncogene-induced metastasis goes through the Ras/Ra1GDS/Ra1 signalling pathway in vivo (66). Fourier-transform infrared (IR) spectroscopy as an approach to score SHE colonies Using metabolomic profiling, thousands of molecules can be identified and quantified within biological samples (85). Different tools can be used for metabolomic assessment, including mass spectroscopy, nuclear magnetic resonance (86), Fourier-transform IR (FTIR) spectroscopy, metabolite arrays and Raman spectroscopy. These tools generate complex datasets, which need to be reduced, using visualisation software, to derive interpretable datasets for analysis (85). Visualisation software packages often apply principal component analysis (PCA), hierarchical cluster analysis, Fisher discriminant analysis, linear discriminant analysis (LDA) or artificial neural networks (85,87,88). Cancer cells have altered metabolic activities associated with increased rates of lipolysis, protein turnover and glycolysis (89–94). Of huge benefit would be a method capable of fingerprinting the complexity of any cell (Figure 3) (95,96). Fig. 3. Low-temperature scanning electron microscopy images of cells after cryo-planing to expose cell interior on the cold stage of a JEOL 840 using a Hexland CT1000 cryo-trans system. (A) Cells appear vitrified, but cellular features evident include nucleus (Nu), cell membrane (CM; identified by arrow), nuclear envelope (NuEnv; identified by arrow), mitochondrion (Mit; example identified by arrow) and cytoplasm (Cy). Scale bar 5 3.0 lm. (B) Higher magnification with cellular features evident including nucleus (Nu), nuclear envelope (NuEnv; identified by arrow) and mitochondrion (Mit; example identified by arrow). Scale bar 5 1.0 lm. Such complexity is based on structural and functional characteristics and, as such, will lend itself to a particular chemical composition. Thus, if one can fingerprint the normal complexity, deviations from this can be associated with various pathological states, such as morphological transformation (97). The emerging field of biospectroscopy offers a range of technologies capable of interrogating the signatures of cell populations down to individual cell organelles (87). This approach has been applied for cancer screening (98), stem cell characterisation (99) and monitoring effects of contaminants or mixtures thereof in mammalian cells (100–103). The application of biospectroscopy tools generates spectra containing hundreds of variables; as such, mathematical models for exploratory (e.g. PCA) or classification (e.g. LDA) purposes are required (104) (Figure 4). The SHE assay has conventionally relied on subjective visual scoring of stained foci; however, there is now evidence that FTIR spectroscopy can be applied as an objective tool to perform such predictions. Using FTIR in attenuated total reflection (ATR) mode, Walsh et al. (14) and Trevisan et al. (105) demonstrated that that SHE foci exhibited IR spectral alterations associated with test agent treatment or morphological transformation. In ATR-FTIR spectroscopy, the IR beam undergoes total internal reflection within a crystal (made of germanium, zinc selenide or diamond) generating an evanescent wave that penetrates beyond a surface of the crystal by a few microns to interact with a sample (in this case, biological) placed in close contact (87). The beam exiting the crystal is received by a detector that generates a spectrum with a number of absorbance intensity peaks based on the sample-attenuated signal, each characteristic of a particular biomolecule. The derived spectral range of 1800–900 cm1 has been termed the ‘biochemical-cell fingerprint’ as it contains the fundamental vibrational modes of structures present in biological systems. Figure 4 shows the work flow that Walsh et al. (14) and Trevisan et al. (105) employed; the SHE cells were treated with test agents and then fixed with methanol on low-E reflective slides. Each SHE 261 A. A. Ahmadzai et al. Fig. 4. Identifying the classifying biomarkers for transformed versus non-transformed SHE cells using IR spectroscopy. The SHE cells are treated with test agent, cultured and then fixed on low-E reflective slides. Each colony on the slides is interrogated using IR spectroscopy. Multivariate computational analysis is applied to identify key biomarkers. colony was located on the slides, referenced and interrogated using ATR-FTIR spectroscopy. The applicability of this approach to the cell transformation assay in terms of how one might manage the derived dataset in order to extract meaningful information is illustrated in Figures 5 and 6. The IR spectra derived from transformed colonies exhibit significant alterations when compared to those classed as non-transformed SHE colonies, e.g. Nnitroso-N-methylnitroguanidine (MNNG)-treated transformed versus MNNG-treated non-transformed category spectral points in an LDA scores plot significantly segregate with a P-value ,10100; the same is the case for B[a]P treatment (Figure 5, left panels). When a per-wave number t-test was carried out for MNNG-treated or B[a]P-treated transformed versus non-transformed colony categories, significant spectral alterations are noted in the 1750–1200 cm1 (associated with protein and nucleic acids) and 1205–900 cm1 (associated with nucleic acids) regions. In addition, IR spectra derived from a particular test agent treatment exhibit significant differences compared to those of the vehicle control (VC) category. The derived spectra from a test agent treatment could be used to classify that particular test agent against other treatments, e.g. MNNG versus VC or B[a]P versus VC (Figure 6). The potential to identify biochemical effects related to test agent treatment or morphological transformation in SHE foci represents an important move towards the development of a novel risk assessment approach that can be applied to new or unknown entities. Because of the non-destructive nature of interrogation by FTIR spectroscopy, this novel scoring 262 system can be correlated for validation purposes with visual screening subsequent to staining. Conclusions The SHE cell transformation assay has recently acquired renewed focus as a tool for chemical safety risk assessment and the identification of potential chemical carcinogens (106). However, despite the fact that it exhibits good correlation with rodent bioassay data, progressing its application has been limited primarily because of the complexity in setting up the assay and more importantly the lack of an objective scoring methodology. Ideally, such a scoring methodology would allow the derivation of a phenotypic measure of SHE colonies post-treatment with test agent that would then allow one to assign a class label of ‘no risk’, ‘DNA-reactive transforming agent’ or ‘non-reactive DNA transforming agent’. The advantage of the SHE cell transformation assay is that it has a high sensitivity and specificity for rodent carcinogens and non-carcinogens, respectively (15). This makes it a potential replacement for in vivo testing strategies (106). An objective measure of morphological transformation would allow risk assessors to make informed quantitative decisions regarding the categorisation of a particular test agent. We propose that by harnessing the availability of new physical sciences sensing tools in combination with new computational approaches, the SHE cell transformation assay has the potential to gain prominence as a tool for chemical safety risk assessment and as an alternative to in vivo testing. Biospectroscopy to objectively score the SHE assay Fig. 5. Techniques for distinguishing between non-transformed versus transformed colonies using IR spectroscopy. Left panels: LDA scores plots showing segregation between non-transformed and transformed IR spectra for two different chemical treatments [MNNG (12.5 lg/ml) and B[a]P (5 lg/ml)]. A statistical hypothesis test (‘two-sample t-test’) found in both cases a P-value , 10100. Right panels: plots showing [wavenumber] [log10(P-value)] for the same chemicals as in the left panels (note that plotting the logarithm of the P-values allows for more detail than plotting the P-values themselves). The significance threshold is marked in both plots (significance level of 5%; log10(0.05) 1.3). Significant differences between non-transformed versus transformed spectra are observed throughout the spectral region (non-significant wavenumbers are hachured out in gray). The MNNG treatment exhibits its highest significance levels between 1130 and 930 cm1, whereas B[a]P shows highest significance between 1620 and 1220 cm1. The gaps in the plots correspond to wavenumbers where log10(p) 5 N or equivalently, P 5 0. Fig. 6. Techniques for distinguishing between test agent treatments using IR spectroscopy. Left panels: LDA scores plots showing segregation between different chemical (MNNG or B[a]P) versus their respective VC category (dimethyl sulfoxide; DMSO). A statistical hypothesis test (‘two-sample t-test’) found in both cases a P-value , 10100. Right panels: two plots showing [wavenumber] [log10(P-value)] for the same chemicals (note that plotting the logarithm of the P-values allows for more detail than plotting the P-values themselves). The significance threshold is marked in both plots (significance level of 5%; log10(0.05) 1.3). Significant differences between treatment versus VC is observed throughout the spectral region (non-significant wavenumbers are hachured out in gray). The gaps in the plots correspond to wavenumbers where log10(p) 5 N or equivalently, P 5 0. 263 A. A. Ahmadzai et al. Funding Unilever as ‘‘part of Unilever’s ongoing effort to develop novel ways of delivering consumer safety’’. Acknowledgements Conflict of interest statement: None declared. References 1. Berwald, Y. and Sachs, L. (1963) In vitro transformation of normal cells to tumor cells by carcinogenic hydrocarbons. J. Natl. Cancer Inst., 35, 641–661. 2. LeBoeuf, R. A., Kerckaert, G. A., Aardema, M. J., Gibson, D. P., Brauninger, R. and Isfort, R. J. (1996) The pH 6.7 Syrian hamster embryo cell transformation SHE assay for assessing the carcinogenic potential of chemicals. Mutat. Res., 356, 85–127. 3. Ridder, G. M., Stuard, S. B., Kerckaert, G. A., Cody, D. B., LeBoeuf, R. A. and Isfort, R. J. (1997) Computerized image analysis of morphologically transformed and nontransformed Syrian hamster embryo (SHE) cell colonies: application to objective SHE cell transformation assay scoring. Carcinogenesis, 18, 1965–1972. 4. Isfort, R. J. and LeBoeuf, R. A. (1995) The Syrian hamster embryo (SHE) cell transformation system: a biologically relevant in vitro model—with carcinogen predicting capabilities—of in vivo multistage neoplastic transformation. Crit. Rev. Oncog., 6, 251–260. 5. Isfort, R. J., Kerckaert, G. A. and LeBoeuf, R. A. (1996) Comparison of the standard and reduced pH Syrian hamster embryo (SHE) cell in vitro transformation assays in predicting the carcinogenic potential of chemicals. Mutat. Res., 356, 11–63. 6. Barrett, J. C. (1979) The progressive nature of neoplastic transformation of Syrian hamster embryo cells in culture. Prog. Exp. Tumor Res., 24, 17–27. 7. Gilmer, T. M., Annab, L. A. and Barrett, J. C. (1988) Characterization of activated proto-oncogenes in chemically transformed Syrian hamster embryo cells. Mol. Carcinog., 1, 180–188. 8. Aardema, M. J., Crosby, L. L., Gibson, D. P., Kerckaert, G. A. and LeBoeuf, R. A. (1996) Aneuploidy and consistent structural chromosome changes associated with transformation of Syrian hamster embryo cells. Cancer Genet. Cytogenet., 96, 140–150. 9. Harvey, J. S., Howe, J. R., Lynch, A. M. and Rees, R. W. (2005) The results of five coded compounds: genistein, metaproterenol, rotenone, p-anisidine and resorcinol tested in the pH 6.7 Syrian hamster embryo cell morphological transformation assay. Mutagenesis, 20, 51–56. 10. Barrett, J. C. and Ts’o, P. O. (1978) Evidence for the progressive nature of neoplastic transformation in vitro. Proc. Natl. Acad. Sci. U S A, 75, 3761–3765. 11. LeBoeuf, R. A., Kerckaert, G. A., Aardema, M. J. and Gibson, D. P. (1990) Multistage neoplastic transformation of Syrian hamster embryo cells cultured at pH 6.70. Cancer Res., 50, 3722–3729. 12. Isfort, R. J., Cody, D. B., Doersen, C. J., Kerckaert, G. A. and Leboeuf, R. A. (1994) Alterations in cellular differentiation, mitogenesis, cytoskeleton and growth characteristics during Syrian hamster embryo cell multistep in vitro transformation. Int. J. Cancer, 59, 114–125. 13. LeBoeuf, R. A. and Kerchaert, G. A. (1987) Enhanced morphological transformation of early passage Syrian hamster embryo cells cultured in medium with a reduced bicarbonate concentration and pH. Carcinogenesis, 8, 689–697. 14. Walsh, M. J., Bruce, S. W., Pant, K., Carmichael, P. L., Scott, A. D. and Martin, F. L. (2009) Discrimination of a transformation phenotype in Syrian golden hamster embryo (SHE) cells using ATR-FTIR spectroscopy. Toxicology, 258, 33–38. 15. Isfort, R. J., Kerckaert, G. A., Cody, D. B., Carter, J., Driscoll, K. E. and LeBoeuf, R. A. (1996) Isolation and biological characterization of morphological transformation-sensitive Syrian hamster embryo cells. Carcinogenesis, 17, 997–1005. 16. Isfort, R. J., Kerckaert, G., Anderson, N. L. and LeBoeuf, R. A. (1992) Two-dimensional gel electrophoresis analysis of Syrian hamster embryo cells: morphological transformation is not cell type specific. Electrophoresis, 13, 855–861. 17. Pienta, R. J., Poiley, J. A. and Lebhertz, W. B., III (1977) Morphological transformation of early passage golden Syrian hamster embryo cells 264 derived from cryopreserved cultures as a reliable in vitro bioassay for identifying diverse carcinogens. Int. J. Cancer, 19, 642–655. 18. Isfort, R. J., Cody, D. B., Kerckaert, G. A. and LeBoeuf, R. A. (1994) Growth factor responsiveness and alterations in growth factor homeostasis in Syrian hamster embryo cells during in vitro transformation. Carcinogenesis, 15, 1203–1209. 19. Kerckaert, G. A., Brauninger, R., LeBoeuf, R. A. and Isfort, R. J. (1996) Use of the Syrian hamster embryo cell transformation assay for carcinogenicity prediction of chemicals currently being tested by the National Toxicology Program in rodent bioassays. Environ. Health Perspect., 104, 1075–1084. 20. Haseman, J. K. and Clark, A. M. (1990) Carcinogenicity results for 114 laboratory animal studies used to assess the predictivity of four in vitro genetic toxicity assays for rodent carcinogenicity. Environ. Mol. Mutagen., 18, 15–31. 21. Farmer, P. B. (2002) ILSI/HESI research programme on alternative cancer models: results of Syrian hamster embryo cell transformation assay. Toxicol. Pathol., 30, 536–538. 22. Matthews, E. J., Spalding, J. W. and Tennant, R. W. (1993) Transformation of BALB/c-3T3 Cells: transformation responses of 168 chemicals compared with mutagenicity in Salmonella and carcinogenicity in rodent bioassays. Environ. Health Perspect., 101 (Suppl. 2), 347–482. 23. Barrett, J. C., Hesterberg, T. W. and Thomassen, D. G. (1984) Use of cell transformation systems for carcinogenicity testing and mechanistic studies of carcinogenesis. Pharmacol. Rev., 36, 53–70. 24. DiPaolo, J. A. and Donovan, P. J. (1967) Properties of Syrian hamster cells transformed in the presence of carcinogenic hydrocarbons. Exp. Cell Res., 48, 361–377. 25. Isfort, R. J., Cody, D. B., Kerckaert, G. A., Tycko, B. and LeBoeuf, R. A. (1997) Role of the H19 gene in Syrian hamster embryo cell tumorigenicity. Mol. Carcinog., 20, 189–193. 26. Bartolomei, M. S., Zemel, S. and Tilghman, S. M. (1991) Parental imprinting of the mouse H19 gene. Nature, 351, 153–155. 27. Leighton, P. A., Ingram, R. S., Eggenschwiler, J., Efstratiadis, A. and Tilghman, S. M. (1995) Disruption of imprinting caused by deletion of the H19 gene region in mice. Nature, 375, 34–39. 28. Blythe, N. L., Senior, P. V. and Beck, F. (1996) Expression of insulin-like growth factor II (IGF-II) and H19 in murine teratocarcinomas derived from embryonic stem (ES) cells. J. Anat., 188, 65–74. 29. Alexandre, S., Rast, C., Maire, M. A., Orfila, L. and Vasseur, P. (2003) ZnCl2 induces Syrian hamster embryo (SHE) cell transformation. Toxicol. Lett., 142, 77–87. 30. Maire, M. A., Rast, C., Pagnout, C. and Vasseur, P. (2005) Changes in expression of bcl-2 and bax in Syrian hamster embryo (SHE) cells exposed to ZnCl2. Arch. Toxicol., 79, 90–101. 31. Adams, J. M. and Cory, S. (1998) The Bcl-2 protein family: arbiters of cell survival. Science, 281, 1322–1326. 32. Maire, M. A., Rast, C., Landkocz, Y. and Vasseur, P. (2007) 2,4Dichlorophenoxyacetic acid: effects on Syrian hamster embryo (SHE) cell transformation, c-Myc expression, DNA damage and apoptosis. Mutat. Res., 631, 124–136. 33. Evan, G. I., Wyllie, A. H., Gilbert, C. S., Littlewood, T. D., Brooks, M., Waters, C. M., Penn, L. Z. and Hancock, D. C. (1992) Induction of apoptosis in fibroblasts by c-myc protein. Cell, 69, 119–128. 34. Soucie, E. L., Annis, M. G., Sedivy, J., Filmus, J., Leber, B., Andrews, D. W. and Penn, L. Z. (2001) Myc potentiates apoptosis by stimulating bax activity at the mitochondria. Mol. Cell Biol., 21, 4725–4736. 35. Prendergast, G. C. (1999) Mechanisms of apoptosis by c-Myc. Oncogene, 18, 2967–2987. 36. Watanabe, M. and Suzuki, K. (1991) Expression dynamics of transforming phenotypes in X-irradiated Syrian golden hamster embryo cells. Mutat. Res., 249, 71–80. 37. Hayflick, L. (1976) The cell biology of human aging. N. Engl. J. Med., 295, 1302–1308. 38. Sasaki, M., Honda, T., Yamada, H., Wake, N., Barrett, J. C. and Oshimura, M. (1994) Evidence for multiple pathways to cellular senescence. Cancer Res., 54, 6090–6093. 39. Greider, C. W. (1994) Mammalian telomere dynamics: healing, fragmentation shortening and stabilization. Curr. Opin. Genet. Dev., 4, 203–211. 40. Laderoute, M. P. (1994) A new perspective on the nature of the cancer problem: anti-cellular senescence. Mol. Carcinog., 10, 125–133. 41. Venable, M. E., Blobe, G. C. and Obeid, L. M. (1994) Identification of a defect in the phospholipase D/diacylglycerol pathway in cellular senescence. J. Biol. Chem., 269, 26040–26044. Biospectroscopy to objectively score the SHE assay 42. Koi, M. and Barrett, J. C. (1986) Loss of tumor-suppressive function during chemically induced neoplastic progression of Syrian hamster embryo cells. Proc. Natl. Acad. Sci. U S A, 83, 5992–5996. 43. Oshimura, M. and Barrett, J. C. (1985) Double nondisjunction during karyotypic progression of chemically induced Syrian hamster cell lines. Cancer Genet. Cytogenet., 18, 131–139. 44. Oshimura, M., Hesterberg, T. W. and Barrett, J. C. (1986) An early, nonrandom karyotypic change in immortal Syrian hamster cell lines transformed by asbestos: trisomy of chromosome 11. Cancer Genet. Cytogenet., 22, 225–237. 45. Endo, S. and Hieber, L. (1995) Chromosome alterations that correlate with progression to immortality in Syrian hamster embryo cells transformed by gamma-irradiation. Int. J. Radiat. Biol., 67, 177–186. 46. Suzuki, K., Suzuki, F., Nikaido, O. and Watanabe, M. (1991) Suppression of differentiation phenotypes in myogenic cells: association of aneuploidy and altered regulation of c-myc gene expression. Exp. Cell Res., 195, 416–422. 47. Afshari, C. A., Kodama, S., Bivins, H. M., Willard, T. B., Fujiki, H. and Barrett, J. C. (1993) Induction of neoplastic progression in Syrian hamster embryo cells treated with protein phosphatase inhibitors. Cancer Res., 53, 1777–1782. 48. Watanabe, M., Suzuki, K. and Kodama, S. (1990) Karyotypic changes with neoplastic conversion in morphologically transformed golden hamster embryo cells induced by X-rays. Cancer Res., 50, 760–765. 49. Barrett, J. C. and Wiseman, R. W. (1987) Cellular and molecular mechanisms of multistep carcinogenesis: relevance to carcinogen risk assessment. Environ. Health Perspect., 76, 65–70. 50. Oshimura, M., Gilmer, T. M. and Barrett, J. C. (1985) Nonrandom loss of chromosome 15 in Syrian hamster tumours induced by v-Ha-ras plus v-myc oncogenes. Nature, 316, 636–639. 51. Futreal, P. A. and Barrett, J. C. (1991) Failure of senescent cells to phosphorylate the RB protein. Oncogene, 6, 1109–1113. 52. Sugawara, O., Oshimura, M., Koi, M., Annab, L. A. and Barrett, J. C. (1990) Induction of cellular senescence in immortalized cells by human chromosome 1. Science, 247, 707–710. 53. Nguyen-Ba, G. and Vasseur, P. (1999) Epigenetic events during the process of cell transformation induced by carcinogens (review). Oncol. Rep., 6, 925–932. 54. Nguyen-Ba, G., Dhalluin, S., Elias, Z., Tapiero, H., Poirot, O. and Hornebeck, W. (1993) Overinduction of ornithine decarboxylase and intracellular proteinase in SHE transformed cells. In Li F. P. and Montesano R. (eds.), Interaction of Cancer Susceptibility Genes and Environmental Carcinogens. Joint meeting AACR and IARC, Lyon, France, a19. 55. Lansing, T. J., Turk, B. F., Kanner, S. B. and Gilmer, T. M. (1997) Mutational activation of pp.60(c-src) leads to a tumorigenic phenotype in a preneoplastic Syrian hamster embryo cell line. Cancer Res., 57, 1962–1969. 56. Gilmer, T. M., Lamb, P. W., Oshimura, M. and Barrett, J. C. (1987) Correlation of V-src gene amplification with the tumorigenic phenotype in a Syrian hamster embryo cell line. Cancer Res., 47, 4663–4666. 57. Albor, A., Flessate, D. M., Soussi, T. and Notario, V. (1994) 3Methylcholanthrene inactivates the p53 gene in Syrian hamster embryo fibroblasts by inducing a specific intronic point mutation. Cancer Res., 54, 4502–4507. 58. Preston, G. A., Lang, J. E., Maronpot, R. R. and Barrett, J. C. (1994) Regulation of apoptosis by low serum in cells of different stages of neoplastic progression: enhanced susceptibility after loss of a senescence gene and decreased susceptibility after loss of a tumor suppressor gene. Cancer Res., 54, 4214–4223. 59. Takahashi, M., Barrett, J. C. and Tsutsui, T. (2002) Transformation by inorganic arsenic compounds of normal Syrian hamster embryo cells into a neoplastic state in which they become anchorage-independent and cause tumors in newborn hamsters. Int. J. Cancer, 99, 629–634. 60. Ebert, R., Barrett, J. C., Wiseman, R. W., Pechan, R., Reiss, E., Röllich, G. and Schiffmann, D. (1990) Activation of cellular oncogenes by chemical carcinogens in Syrian hamster embryo fibroblasts. Environ. Health Perspect., 88, 175–178. 61. Velasco, J. A., Castro, R., Avila, M. A., Laborda, J., DiPaolo, J. A., Cansado, J. and Notario, V. (1994) cph, a novel oncogene which cooperates with H-ras in the transformation of NIH3T3 fibroblasts. Oncogene, 9, 2065–2069. 62. Notario, V., Castro, R., Flessate, D. M., Doniger, J. and Di Paolo, J. A. (1990) Frequent activation of non-ras transforming sequences in neoplastic Syrian hamster cells initiated with chemical carcinogens. Oncogene, 5, 1425–1430. 63. Ebert, R., Wiseman, R. W., Barrett, J. C., Reiss, E., Rollich, G. and Schiffmann, D. (1992) Characterization of the Syrian hamster c-Ha-ras gene and intron-D-exon transcript. Mol. Carcinog., 5, 254–258. 64. Ashra, H. and Rao, K. V. (2006) Elevated phosphorylation of Chk1 and decreased phosphorylation of Chk2 are associated with abrogation of G2/M checkpoint control during transformation of Syrian hamster embryo (SHE) cells by malachite green. Cancer Lett., 237, 188–198. 65. Hartwell, L. H. and Weinert, T. A. (1989) Checkpoints: controls that ensure the order of cell cycle events. Science, 246, 629–634. 66. Tchevkina, E., Agapova, L., Dyakova, N., Martinjuk, A., Komelkov, A. and Tatosyan, A. (2005) The small G-protein RalA stimulates metastasis of transformed cells. Oncogene, 24, 329–335. 67. Mundy, G. R. (1997) Mechanisms of bone metastasis. Cancer, 80, 1546–1556. 68. Nannuru, K. C. and Singh, R. K. (2010) Tumor-stromal interactions in bone metastasis. Curr. Osteoporos. Rep., 8, 105–113. 69. Sethi, T., Rintoul, R. C., Moore, S. M. et al. (1999) Extracellular matrix proteins protect small cell lung cancer cells against apoptosis: a mechanism for small cell lung cancer growth and drug resistance in vivo. Nat. Med., 5, 662–668. 70. Zacharski, L. R., Memoli, V. A., Orstein, D. L. et al. (1993) Tumor cell procoagulant and urokinase expression in carcinoma of the ovary. J. Natl. Cancer Inst., 85, 1225. 71. Rao, V. M. (1992) Tissue factor as a tumor procoagulant. Cancer Metastasis Rev., 11, 249. 72. Koh, S. C., Tham, K. F., Razvi, K., Oei, P. L., Lim, F. K., Roy, A. C. and Prasad, R. N. (2001) Hemostatic and fibrinolytic status in patients with ovarian cancer and benign ovarian cysts: could D-dimer and antithrombin III levels be included as prognostic markers for survival outcome? Clin. Appl. Thromb. Hemost., 7, 141–148. 73. Ossowski, L. and Reich, E. (1983) Antibodies to plasminogen activator inhibit tumor metastasis. Cell, 35, 611–619. 74. Quigley, J. P. (1979) Proteolytic enzymes of normal and malignant cells. In Heynes, R. O. (ed.), Surfaces of Normal and Malignant Cells. Wiley & Sons, London, UK, pp. 247–285. 75. Mullins, D. E. and Rohrlich, S. T. (1983) The role of proteinases in cellular invasiveness. Biochim. Biophys. Acta, 695, 177–214. 76. Markus, G. (1984) The role of homeostasis and fibrinolysis in the metastatic spread of cancer. Semin. Thromb. Hemost., 10, 61–70. 77. Kok, K., Osinga, J., Schotanus, D. C., Berendsen, H. H., de Leij, L. F. and Buys, C. H. (1989) Amplification and expression of different myc-family genes in a tumor specimen and 3 cell lines derived from one small-cell lung cancer patient during longitudinal follow-up. Int. J. Cancer, 44, 75–78. 78. Peterkofsky, B. and Prather, W. (1992) A post-translational modification, unrelated to hydroxylation, in the collagenous domain of nonhelical proalpha 2(I) procollagen chains secreted by chemically transformed hamster fibroblasts. J. Biol. Chem., 267, 5388–5395. 79. Lavnikova, N. A. and Burdelia, L. G. (1990) Susceptibility of cells with different stage of malignancy to cytostatic action of resident peritoneal macrophages of Syrian hamsters. Biull. Eksp. Biol. Med., 110, 83–85. 80. Volpe, E. A. (1992) Resistance of in vivo-selected spontaneously transformed cells and Rous sarcoma virus-transformed cells to macrophage-mediated cytotoxicity. Experientia, 48, 500–503. 81. Bos, J. L. (1989) Ras oncogenes in human cancer: a review. Cancer Res., 49, 4682–4689. 82. Malumbres, M. and Barbacid, M. (2003) RAS oncogenes: the first 30 years. Nat. Rev. Cancer, 3, 459–465. 83. Chambers, A. F., Groom, A. C. and MacDonald, I. C. (2002) Dissemination and growth of cancer cells in metastatic sites. Nat. Rev. Cancer, 2, 563–572. 84. Chambers, A. F. and Tuck, A. B. (1993) Ras-responsive genes and tumor metastasis. Crit. Rev. Oncog., 4, 95–114. 85. Oakman, C., Tenori, L., Biganzoli, L., Santarpia, L., Cappadona, S., Luchinat, C. and Di Leo, A. (2010) Uncovering the metabolomic fingerprint of breast cancer. Int. J. Biochem. Cell Biol., 43, 1010–1020. 86. Beckonert, O., Coen, M., Keun, H. C., Wang, Y., Ebbels, T. M., Holmes, E., Lindon, J. C. and Nicholson, J. K. (2010) High-resolution magic-angle spinning NMR spectroscopy for metabolic profiling of intact tissues. Nat. Protoc., 5, 1019–1032. 87. Martin, F. L., Kelly, J. G., Llabjani, V., Martin-Hirsch, P. L., Patel, I. I., Trevisan, J., Fullwood, N. J. and Walsh, M. J. (2010) Distinguishing cell types or populations based on the computational analysis of their infrared spectra. Nat. Protoc., 5, 1748–1760. 88. Kelly, J. G., Cheung, K. T., Martin, C., O’Leary, J. J., Prendiville, W., Martin-Hirsch, P. L. and Martin, F. L. (2010) A spectral phenotype of 265 A. A. Ahmadzai et al. oncogenic human papillomavirus-infected exfoliative cervical cytology distinguishes women based on age. Clin. Chim. Acta, 411, 1027–1033. 89. Al-Majid, S. and Waters, H. (2008) The biological mechanisms of cancerrelated skeletal muscle wasting: the role of progressive resistance exercise. Biol. Res. Nurs., 10, 7–20. 90. Argilés, J. M., López-Soriano, F. J. and Busquets, S. (2007) Mechanisms to explain wasting of muscle and fat in cancer cachexia. Curr. Opin. Support. Palliat. Care, 1, 293–298. 91. Gatenby, R. A. and Gillies, R. J. (2004) Why do cancers have high aerobic glycolysis? Nat. Rev. Cancer, 4, 891–899. 92. Nam, H., Chung, B. C., Kim, Y., Lee, K. and Lee, D. (2009) Combining tissue transcriptomics and urine metabolomics for breast cancer biomarker identification. Bioinformatics, 25, 3151–3157. 93. Kim, J. W. and Dang, C. V. (2006) Cancer’s molecular sweet tooth and the Warburg effect. Cancer Res., 66, 8927–8930. 94. Yeh, C. S., Cheng, T. L., Juan, C. H., Wu, C. H. and Lin, S. R. (2006) Fatty acid metabolism pathway plays an important role in carcinogenesis of human colorectal cancers by Microarray-Bioinformatics analysis. Cancer Lett., 233, 297–308. 95. Fullwood, N. J., Martin, F. L., Bentley, A. J., Lee, J. P. and Lee, S. J. (2011) Imaging sclera with hard X-ray microscopy. Micron, 42, 506–511. 96. Fullwood, N. J. and Meek, K. M. (1993) A synchroton X-ray study of the changes occurring in the corneal stroma during processing for electron microscopy. J. Microsc., 169, 53–60. 97. Martin, F. L. (2011) Shining a light into molecular workings. Nat. Methods, 8, 385–387. 98. German, M. J., Hammiche, A., Ragavan, N. et al. (2006) Infrared spectroscopy with multivariate analysis potentially facilitates the segregation of different types of prostate cell. Biophys. J., 90, 3783–3795. 99. Walsh, M. J., Fellous, T. G., Hammiche, A. et al. (2008) Fourier transform infrared microspectroscopy identifies PO2 modifications as a marker of the putative stem cell region of human intestinal crypts. Stem Cells, 26, 108–118. 100. Bi, X., Walsh, M. J., Wei, X., Sheng, G., Fu, J., Martin-Hirsch, P. L., Thomas, G. O., Jones, K. C. and Martin, F. L. (2007) Infrared spectral analysis of MCF-7 cells treated with serum-lipid extracts segregates predominantly brominated flame retardant-exposed subjects from those with mainly organochlorine exposures. Environ. Sci. Technol., 41, 5915–5922. 101. Llabjani, V., Jones, K. C., Thomas, G. O., Walker, L. A., Shore, R. F. and Martin, F. L. (2009) Polybrominated diphenyl ether-associated alterations in cell biochemistry as determined by attenuated total reflection Fouriertransform infrared spectroscopy: a comparison with DNA-reactive and/or endocrine-disrupting agents. Environ. Sci. Technol., 43, 3356–3364. 102. Barber, J. L., Walsh, M. J., Hewitt, R., Jones, K. C. and Martin, F. L. (2006) Low-dose treatment with polybrominated diphenyl ethers (PBDEs) induce altered characteristics in MCF-7 cells. Mutagenesis, 21, 351–360. 103. Llabjani, V., Trevisan, J., Jones, K. C., Shore, R. F. and Martin, F. L. (2010) Binary mixture effects of PBDE congeners (47, 153, 183, or 209) and PCB congeners (126 or 153) in MCF-7 cells: biochemical alterations assessed by IR spectroscopy and multivariate analysis. Environ. Sci. Technol., 44, 3992–3998. 104. Kelly, J. G., Trevisan, J., Scott, A. D., Carmichael, P. L., Pollock, H. M., Martin-Hirsch, P. L. and Martin, F. L. (2011) Biospectroscopy to metabolically profile biomolecular structure: a multistage approach linking computational analysis with biomarkers. J. Proteome Res., 10, 1437–1448. 105. Trevisan, J., Angelov, P. P., Patel, I. I. et al. (2010) Syrian hamster embryo (SHE) assay (pH 6.7) coupled with infrared spectroscopy and chemometrics towards toxicological assessment. Analyst, 135, 3266–3272. 106. Creton, S., Aardema, M. J., Carmichael, P. L. et al. Cell transformation assays for prediction of carcinogenic potential: state of the science and future research needs. Mutagenesis, 27, 93–101. 266
© Copyright 2025 Paperzz