High content analysis of cytotoxic effects of pDMAEMA on human intestinal epithelial and monocyte cultures Lee-Anne B. Rawlinson, Peter J. O’Brien, David J. Brayden* UCD School of Veterinary Medicine and UCD Conway Institute, University College Dublin, Dublin 4, Ireland. * For correspondence: David Brayden, Ph.D. Room 214, Veterinary Sciences Building, School of Agriculture, Food Science and Veterinary Medicine, UCD, Belfield, Dublin 4, Ireland Tel: +353 1 716 6013 Fax: +353 1 716 6219 Email: david.brayden@ucd.ie 1 Abstract Poly(2-(dimethylamino ethyl)methacrylate) (pDMAEMA) is a cationic polymer with potential as an antimicrobial agent and as a non-viral gene delivery vector. The aim was to further elucidate the cytotoxicity of a selected pDMAEMA low molecular weight (MW) polymer against human U937 monocytes and Caco-2 intestinal epithelial cells using a novel multi-parameter high content analysis (HCA) assay and to investigate histological effects on isolated rat intestinal mucosae. Seven parameters of cytotoxicity were measured: nuclear intensity (NI), nuclear area (NA), intracellular calcium ([Ca2+]i), mitochondrial membrane potential (MMP), plasma membrane permeability (PMP), cell number (CN) and phospholipidosis. Histological effects of pDMAEMA on excised rat ileal and colonic mucosae were investigated in Ussing chambers. Following 24-72 hours exposure, 25-50 µg/ml pDMAEMA induced necrosis in U937 cells, while 100-250 µg/ml induced apoptosis in Caco-2. pDMAEMA increased NA and NI and decreased [Ca2+]i, PMP, MMP and CN in U937 cells. In Caco-2, it increased NI and [Ca2+]i, but decreased NA, PMP, MMP and CN. Phospholipidosis was not observed in either cell line. pDMAEMA (10 mg/ml) did not induce any histological damage on rat colonic tissue and only mild damage to ileal tissue following exposure for 60 min. In conclusion, HCA reveals that pDMAEMA induces cytotoxicity in different ways on different cell types at different concentrations. HCA potential for high throughput toxicity screening in drug formulation programmes. Key words: pDMAEMA, high content analysis, cytotoxicity assays, Caco-2 intestinal epithelia, antimicrobial polymers 2 1. Introduction Poly(2-(dimethylamino ethyl)methacrylate) (pDMAEMA) is a mucoadhesive polymer, that is cationic in acidified media or when quaternised with an alkylating agent [1, 2]. It is being investigated as a potential antimicrobial coating on medical devices [35] and as a non-viral gene delivery vector [6, 7]. It induces cytotoxicity in U937 human monocyte-like cells, but less cytotoxicity in human Caco-2 intestinal epithelial cells [3]. Intravenous delivery of 5.1 mg/kg pDMAEMA was fatal to rat, but 2.1 mg/kg was well tolerated [8]. In order to eventually use pDMAEMA in a clinical setting, its cytotoxic potential should therefore be further investigated with improved assays since in vitro cytotoxicity data from standard assays is equivocal and lacks predictive power. High Content Analysis (HCA) is a novel technology that allows quantitative analysis of each cell at sub-cellular microscopic resolution using a selection of multicoloured fluorescence-based non-toxic dyes [9]. It allows combinations of numerous fluorescent probes to be used concurrently to investigate different parameters of cytotoxicity following acute and chronic exposure at varying concentrations [10, 11]. The information obtained and throughput is more than an order of magnitude higher than conventional cytotoxicity assays. For example, a 96-well plate is read in 30 min with a large proportion of cells in each well analysed. Multiple biochemical, functional and morphological parameters are measured on individual cells and there is a higher sensitivity of cytotoxicity detection as indicated by the lower IC50 values for HCA compared to the methylthiazolyldiphenyl-tetrazolium bromide (MTT) assay. HCA has recently been validated against in vivo human toxicity data for hundreds of marketed drugs (10-13). Additional advantages of HCA over conventional cell-based cytotoxicity 3 assays include detailed data on numerous cell physiological and morphological parameters measured kinetically up to 72 hours, measurements on individual cells, and mechanistic information on elucidating the induced pathophysiology. HCA has also been used to investigate the influence of siRNA on cell cycling [14], to screen for activators of DNA damage [15], as well as in the study of visionrelated diseases to investigate neurite outgrowth in retinal ganglion cells [16]. The assay used in this study was based on the multi-parameter cytotoxicity assay protocol developed by O’Brien et al. [11]. This assay allows concurrent analysis of 6 parameters of cell cytotoxicity (nuclear intensity (NI), nuclear area (NA), intracellular calcium ([Ca2+]i), mitochondrial membrane potential (MMP), plasma membrane permeability (PMP) and cell number (CN)). HCA therefore provides high sensitivity (predictive detection of known cytotoxic agents) and high specificity (predictive correlation with human toxicity) compared to conventional cytotoxicity end-point assays [11]. By combining flow cytometry, intracellular fluorescence probes and image analysis of multiple parameters, extensive data is provided to examine sub-lethal effects, which can relate to in vivo toxicological outcomes. HCA was also used here to investigate the potential of pDMAEMA to induce phospholipidosis. Drug-induced phospholipidosis is a lipid storage disorder that results in the accumulation of phospholipids in lysosomes in tissues throughout the body due to inhibition of phospholipid breakdown [17]. Evidence of drug-induced phospholipidosis in pre-clinical studies can lead to delays or removal of drugs from development [18]. Despite this, there are over 50 marketed pharmaceuticals that are known to induce phospholipidosis [18]. Many of these molecules are cationic amphiphilic drugs, which 4 are generally characterised by a structure that is comprised of a hydrophobic region and a hydrophilic side chain with a charged amine group [19]. As pDMAEMA has a similar structure with charged amine groups, it is possible that it may also induce phospholipidosis. Due to the variation in pDMAEMA cytotoxicity between red blood cells and human cell lines [3], it is possible that the range of different cell types in intact tissue may afford differential protection. In addition, the mucus layer that is present in many epithelial tissues may reduce toxicity [20]. To investigate this, freshly excised rat ileal and colonic intestinal epithelial tissue was incubated for 60 min with pDMAEMA in Ussing-type chambers. This allowed for a fast and simple assessment of the histological effects of pDMAEMA on whole tissue. Using HCA, we report that high concentrations of a relatively low molecular weight pDMAEMA polymer induced necrosis in U937 cells, while apoptosis was induced in Caco-2 cells. pDMAEMA did not induce phospholipidosis in either cell line. The cytotoxicity induced in U937 cells was seen at lower concentrations than in Caco-2 cells. pDMAEMA induced only minimal histological damage to rat intestinal tissue in vitro, and isolated intestinal tissue mucosae was clearly more resilient to the polymer than cell cultures. 5 2. Materials and methods 2.1. Materials All cell culture and HCA reagents were obtained from Invitrogen Corporation (CA, USA), unless otherwise stated. Cell lines were obtained from the American Tissue Type Culture Collection (ATCC). Adult male Wistar rats (210–300 g) were obtained from the Biomedical Facility, UCD. All other general chemicals and reagents used were of analytical grade and were obtained from Sigma-Aldrich Company Ltd. (Dorset, UK), unless otherwise stated. 2.2. Synthesis of pDMAEMA pDMAEMA was synthesized by atom transfer living radical polymerisation (ATRP) in accordance with our previous descriptions [2, 3]. The molecular weight and polydispersity index of pDMAEMA were 12.8 kDa and 1.16, respectively, as determined by size exclusion chromatography (SEC-HPLC) [3]. pDMAEMA had high purity and minimal levels of catalyst residue and we used the specific polymer that was described and characterised in [3] 2.3. HCA assay of pDMAEMA against U937 and Caco-2 cells The HCA cytotoxicity assay was based on the methods of O’Brien et al. [11]. For HCA, either phenol red-free supplemented DMEM or RPMI media was used. Caco-2 epithelial cells (ATCC: HTB-37, passage numbers 65-69) were cultured in DMEM containing GlutaMAXTM, supplemented with 10 % foetal bovine serum (FBS), 1 % nonessential amino acids (NEAA) and 1 % penicillin/streptomycin (Pen-Strep). U937 6 monocytes (ATCC: CRL-1593.2, passage numbers 12-14) were cultured using RPMI medium supplemented with FBS, NEAA, Pen-Strep and 1 % L-glutamine. All cells were grown in a humidified 37°C incubator with 5 % CO2 in air. The Caco-2 cells were seeded in 96-well cell culture plates at a density of 500 cells/well and incubated for 24 hours at 37°C with 5 % CO2. After incubation, Caco-2 cell media was replaced with 50 µl fresh media in each well. U937 cells were seeded in 96-well plates by adding 50 µl cells at a density of 5000 cells/well; these were incubated for 60 min to allow cells to settle. 100 µl pDMAEMA–containing solutions were added to all wells (at 1.5 times final concentration) and plates were further incubated for 24, 48 or 72 hours. Plate lids were left on during acquisition to prevent evaporation. Media alone was added as untreated controls for the 24 to 72 hour incubations. Dye cocktails were made up (Table 1). Positive control treatments were mixed with the dye cocktails (Table 2) and added to selected wells after incubations, 30 min prior to data acquisition. After 24-72 hour incubations with or without pDMAEMA, 50 µl of dye cocktail was added to wells. Plates were then incubated for 30 to 40 min and images were acquired on the In Cell® 1000 High Content Analyzer (GE Healthcare, UK), using a 10 X objective. Ten random fields were viewed per well and concentrations of pDMAEMA were repeated in triplicate. Experiments were repeated on at least three separate occasions. Fluorescence of the dyes were monitored at excitation and emission wavelengths respectively of: (1) 360 nm and 460 nm for Hoechst 33342; (2) 480 nm and 535 nm for Fluo 4-AM; (3) 535 nm and 600 nm for TMRM; and (4) 620 nm and 700 nm for 7 TOTO®-3. Exposure times were varied between experiments to optimise image quality. However, typical exposure and hardware autofocus (HWAF) values were: (1) 100 ms and 11.1 µm for Hoechst 33342; (2) 250 ms and -1.1 µm for Fluo 4-AM; (3) 300 ms and -2.1 µm for TMRM; and (4) 200 ms and -0.6 µm for TOTO®-3. After acquisition of the images, the data was analysed using In Cell® 1000 Workstation software (GE Healthcare, UK) using multi-target analysis. Table 3 summarises the analysis settings with a glossary of In Cell® 1000 analysis terms. For [Ca2+]i and MMP analyses, values were calculated minus background readings. 2.4. HCA phospholipidosis assay of pDMAEMA Evidence for phospholipidosis includes intracellular accumulation of phospholipids and lamellar bodies in cells. To accurately view lamellar bodies, electron microscopy is normally used [27]. The accumulation of phospholipids also correlates to induction of phospholipidosis as visualised by Nile red [28]. The HCA phospholipidosis assay used was based on the method of Halstead et al. [29]. U937 cells were selected to test for phospholipidosis as they have been found to be sensitive for evaluating this parameter [30]. In addition, lamellar bodies that are associated with phospholipidosis can be found in intestinal epithelial cells [31], so Caco-2 cells were also used. 100 µl of Caco-2 cells were seeded in 96-well cell culture plates at a density of 5 x 104 cells/ml. Plates were incubated at 37°C in 5 % CO2 for 24 hours. After incubation, Caco-2 cell media was replaced with 100 µl fresh media. For U937 cells, wells were seeded with 100 µl of cells at a density of 2 x 105 cells/ml and incubated for 60 min to allow cells to settle. 100 µl pDMAEMA (2 times final concentration) was added and the plates further 8 incubated for 24, 48 or 72 hours. Erythromycin (403 µM (250 µg/ml)) was added as a positive control [32] and Triton X-100 (0.01%) was added as a negative control [33]. After the 24 to 72 hour incubations the plates were centrifuged at 1000 x g for 5 min and the media removed. 150 µl dye solution, containing 1 µg/ml Nile red (Sigma) and 0.8 µM Hoechst 33342, was added to wells. The plates were further incubated for 30 min and images were acquired using a 40 x objective as described above. Fluorescence was monitored at respective excitation and emission wavelengths of: (1) 360 nm and 460 nm for Hoechst 33342; (2) 535 nm and 600 nm for Nile red. Exposure and HWAF values were set at, respectively: (1) 1500 ms and 5 µm for Hoechst 33342; (2) 100 ms and -0.1 µm for Nile red. After acquisition of the images, the data was analysed as described in Table 4. 2.4. Gross histological effects of pDMAEMA on rat intestinal tissue Maintenance and welfare of animals were in accordance with the “Principles of Laboratory Animal Care” (National Institutes of Health publication number 85-23, revised in 1985). Rats were sacrificed by cervical dislocation. Intestinal tissue was exposed in each species via a mid-ventral abdominal incision. Segments were excised and immediately immersed in freshly prepared oxygenated Krebs–Henseleit solution (KH) at room temperature. Proximal ileal and colonic tissue were immediately excised, dissected along the line of the mesentery and washed with warm oxygenated buffer. Four-inch tissue segments were then pinned mucosal side up on a cork-board. Mucosal sheets were mounted in modified Ussing chambers within 15 min of euthanasia, as previously described [34]. The area of tissue exposed in the Ussing chamber window 9 was 0.63 cm2 and the apical and basolateral compartments contained 1 and 5 ml KH, respectively. KH was gassed via a flexible polyethylene tube with 95 % O2:5 % CO2 on the basolateral side only, as apical gassing led to evaporation. Temperature was maintained at 37°C by immersing the rack of chambers in a heated water bath. Immediately after mounting tissue, pre-warmed KH with or without pDMAEMA at selected concentrations or with Triton X-100 (0.1 %, as a positive cytotoxic control), was added to the apical compartment and the tissue incubated for 60 min. 2.5. Preparation of intestinal tissue for microscopic examination Tissue for histology was fixed in 10 % buffered formalin and routinely processed into paraffin wax. 4 µm sections were cut and stained in Harris's haematoxylin and eosin (H & E) for general morphological assessment. Slides were viewed using a Nikon (Japan) light microscope and images were captured using a Micropublisher 3.3 RTV camera (QImaging, Surrey, BC, Canada) with Image-Pro 6.2 software (Media Cybernetics, Inc., Silver Spring, MD, USA), using a 10 x objective. 2.6. Statistical analysis Statistical analyses were carried out using one-way ANOVA with Bonferroni post-hoc tests. The significance level was set at α = 0.05 (95 % confidence intervals). 10 3. Results 3.1. HCA: Cytotoxicity measurements of pDMAEMA in U937 and Caco-2 cells pDMAEMA is more cytotoxic to U937 monocytes than to Caco-2 intestinal epithelial cells by MTT analysis [3]. To investigate this further, HCA was utilised here to concurrently measure 6 parameters from 4 different fluors in the presence and absence of pDMAEMA over time. The parameters tested were: (1) nuclear area (NA), (2) nuclear intensity (NI), (3) intracellular calcium ([Ca2+]i,) (4) mitochondrial membrane potential (MMP), (5) plasma membrane permeability (PMP) and (6) cell number (CN). For illustration, representative fused images of the separate U937 and Caco-2 cell HCA assays are shown in response to response to the calcium ionophore, ionomycin (20μM) (Fig. 1). The In Cell® software allows for three colours to be fused. Blue is the cell nucleus (stained with Hoechst 33342). Red is the MMP (stained with TMRM). Green is [Ca2+]i (stained with Fluo-4-AM). pDMAEMA was then assessed in both cell types using concentrations from 0.01 to 5000 µg/ml for 24, 48 and 72 hours. Results are presented as percent values compared to 100 % negative control. The concentrations of pDMAEMA that induced significant effects compared to untreated cells for the cytotoxicity parameters are summarised in Table 5. pDMAEMA induced disruption to both PMP and MMP at concentrations of 25-50 µg/ml in U937 cells, while 50-100 µg/ml decreased [Ca2+]i following 24-72 hours exposure. PMP and [Ca2+]i were disrupted at 100-250 µg/ml concentrations of pDMAEMA at 24, 48 and 72 hours in Caco-2, while NI was increased (100-250 µg/ml) and MMP was disrupted with an IC50 of 550 µg/ml. In general, most parameters show increased changes in parameters with increasing time of exposure to pDMAEMA and with increasing concentrations. 11 Fig. 2 shows concentration-response curves of the effect of pDMAEMA on the cell lines after 72 hours in respect of the cytotoxicity parameters. IC50 values from the cytotoxicity parameters are also described (Table 6A). The IC50 values for pDMAEMA against U937 cells were generally lower than those against Caco-2 cells for the cytotoxicity parameters. When cells were treated with increasing concentrations of pDMAEMA, NA decreased in Caco-2, while in U937 it increased. NI and PMP were increased in both cell lines with pDMAEMA treatment. [Ca2+]i greatly increased in Caco-2 cells when treated with pDMAEMA, whereas U937 cells showed only a slight increase followed by a decrease with increasing pDMAEMA concentration. As pDMAEMA concentration increased, MMP and CN increased and then decreased. Table 6B gives the IC50 values of pDMAEMA against U937 and Caco-2 cells over time. The concentrations shown represent the lowest IC50 values of the 6 cytotoxicity parameters tested. Against U937 cells, pDMAEMA demonstrated cytotoxic effects at concentrations of 25-50 µg/ml. In Caco-2 cells, pDMAEMA demonstrated cytotoxic effects at concentrations of 100-250 µg/ml. In Caco-2 the IC50 values decreased with time, while in U937 there was a small increase in the IC50 values at 72 hours. This increase may be due to an increase in the number of cells and closer packing due to faster growth of U937, which may hinder polymer access. The positive control agents used in the HCA assay induced the expected activity in each cell type, i.e. ionomycin increased intracellular calcium, FCCP decreased the 12 MMP and Triton X-100 induced increased the PMP (data not shown). Some of these agents however caused effects on parameters other than their intended effect. For example, in addition to induction of increased [Ca2+]i, ionomycin (20µM) increased MMP in both cell lines (data not shown). This may be due to a mild stimulatory effect of the drug on protective biochemical processes, similar to what was observed with pDMAEMA. In addition to its effect on increasing PMP, Triton X-100 (0.2 %) decreased [Ca2+]i (data not shown). This may be related to the increased membrane permeability allowing dye leakage. 3.2. HCA: Phospholipidosis induction Representative fused images of the U937 and Caco-2 cell phospholipidosis assays are shown (Fig. 3). The nucleus was stained red by Hoechst 33342 and the phospholipids were stained by green by Nile red. Compared to untreated U937 and Caco-2 cells in Fig. 3A and C, there was an increase in green fluorescence when U937 and Caco-2 were exposed to the positive control, erythromycin (250 μg/ml) at all time points. Erythromycin induces an increase in lysosomal phospholipids by inhibiting the activity of lysosomal phospholipases [32]. There were occasional cells in control images that showed increased green/phospholipids fluorescence that represent naturally occurring dying cells. There was no significant increase in phospholipids in either the U937 or the Caco-2 cells exposed to pDMAEMA (1-100 µg/ml) for 24 to 72 hours (Fig. 4). Concentrations of pDMAEMA that did not cause cytotoxicity were used in this assay because higher concentrations would have lead to permeabilisation of the cell membranes and dye/phospholipid loss. The negative control, Triton X-100, did not induce a 13 significant increase in phospholipid accumulation. There was some decrease in fluorescence with Triton X-100 and pDMAEMA (10 µg/ml), however this may be related to the ability of these molecules to permeabilise plasma membranes or lysosomes, therefore allowing dye leakage. 3.3. Histological effects of pDMAEMA on rat ileum and colon After 60 min incubation in the horizontal diffusion chambers, control rat ileal tissue sections showed an intact mucosal epithelium with homogeneous distribution of villi. pDMAEMA did not cause any disruption to the epithelial surface of rat ileum at concentrations up to 0.1 mg/ml after 60 min (data not shown). At a concentration of 1 mg/ml, there was some evidence of mucosal sloughing (Fig. 5B) and at 10 mg/ml, the intestinal villi were somewhat disrupted (Fig. 5C). However, the lower layer of cells was unaffected and maintained a normal epithelial surface. None of the concentrations of pDMAEMA tested induced toxicity in comparison to the complete aberration caused by 0.1 % Triton X-100 (Fig. 5D). With treatments of 1 mg/ml (Fig. 6B) and 10 mg/ml pDMAEMA (Fig. 6C) for 60 min, there was no disruption of the rat colonic epithelial layer, nor sloughing of cells, compared with control tissue (Fig. 6A). Treatment of rat colonic tissue with 0.1 % Triton X-100 caused complete aberration of the epithelial surface (Fig. 6D). There was some oedema visible in all tissue samples, which may be due to the stretching of the tissue in the horizontal chambers. 14 4. Discussion HCA analysis allows the combination of fluorescent probes to be used concurrently to investigate different parameters of cytotoxicity [9-13]. Separate fluorescence signals can be detected simultaneously using HCA and dyes that have overlapping fluorescence may be differentiated if their cellular locations do not overlap [35]. By incorporating three to four fluorescent dyes, up to eight markers of cytotoxicity have been successfully investigated concurrently [10]. These include nuclear morphology, plasma membrane integrity, mitochondrial function, intracellular calcium and cell proliferation [10, 11]. Use of HCA in the development of novel antimicrobial agents may increase the speed by which these molecules are assessed and processed to the clinical trials stage of development. To date, in vitro assays including MTT and lactate dehydrogenase have been commonly used to assess cytotoxicity of pDMAEMA [3, 36, 37, 38]. Although useful indicators, they do not give much information about the processes of cytotoxicity nor early stage events. Other fluorescent-based tests using flow cytometry and confocal microscopy may provide additional information about the gradual process of cytotoxicity induced by a polymer, nor their mechanism. They are also time consuming, costly and labour intensive. HCA overcomes these problems by providing a high throughput process that allows testing of numerous cytotoxicity markers concurrently. Data from MTT analysis has suggested variable cytotoxic effects of pDMAEMA in different cell types [3]. Therefore, the cytotoxic-inducing aspects of this polymer were investigated by HCA. NA was found to increase in U937 cells and decrease in Caco-2 cells upon exposure to pDMAEMA. An increase in NA may be associated with nuclear 15 swelling as a result of premature senescence [39-41]. Cellular shrinkage and condensation of the chromatin are markers of apoptosis [42]. The decrease in NA in Caco-2 cells may be an indication of apoptosis and the swelling of the U937 nuclei may indicate necrosis. The increased [Ca2+]i observed in Caco-2 cells after treatment with pDMAEMA may also be an indicator of apoptosis [43]. In U937 cells, [Ca2+]i decreased upon treatment with pDMAEMA. This may be related to the increased plasma membrane permeability (PMP), which may cause dye release from the cell thereby masking an increase in calcium. In addition, the nuclear intensity (NI) of both cell types increased with increasing concentrations of pDMAEMA. This may also be due to an increase in the PMP allowing more dye into the cell. pDMAEMA appeared to induce apoptosis in Caco-2 cells but not in U937 cells, and it has previously been hypothesised to be inducing cell death in these cells through a necrotic process as detected by flow cytometry [38]. Our data supports this conclusion, although caution should be taken when discussing the cytotoxicity of this polymer. Jones et al. [38] found a late-stage decrease in MMP in U937 cells. They hypothesized that, due to the fact that disruption of MMP is an early indicator of apoptosis and also a late stage event, the reduction in MMP was an indicator of necrosis. The data presented here supports this conclusion, as the concentrations that induced a decrease in MMP in U937 cells were similar to those that produced other cytotoxic effects. This indicates a late-stage necrotic event. However, in Caco-2, the MMP was decreased at concentrations significantly lower than those that induced other cytotoxicity effects, including PMP and this is evidence further supporting a process of apoptosis in Caco-2. 16 Hormesis is a process produced by some drugs where a biphasic effect is seen at high versus low concentrations of drugs [11, 44]. This process is characterised by low concentration stimulation and high concentration inhibition and has been found to occur with parameters such as MMP and CN [11]. In U937, both parameters were increased at low pDMAEMA concentrations. At higher concentrations however, there was a decrease in MMP, but no change in CN. Hormesis may be a reaction of the cells to subcytotoxic concentrations of drug whereby the cells try to overcompensate for its presence by increasing their biochemical protective mechanisms [45]. In Caco-2 cells, hormesis was also evident with CN increasing at low concentrations of pDMAEMA, then decreasing at high concentrations. For MMP, a multiphasic reaction to pDMAEMA was seen in Caco-2 cells. At all time points there was a decrease in MMP at lower concentrations, followed by recovery, then a decrease again as the concentration of pDMAEMA increases. The reason this occurs is unclear, but it may be that a dual mechanism of action is occurring, i.e., at low concentrations pDMAEMA induces an apoptotic process of cell death, while at high concentrations a necrotic process is induced. Modifications of pDMAEMA have also been found to reduce cytotoxicity. These include PEGylation [6], reduction of terminal thiol groups [46] and complex formation with n-carboxyethylchitosan [47]. Complexation of pDMAEMA with DNA also improves transfection efficiency, as did copolymer formation with methyl methacrylate [48]. It is known that altering the MW of pDMAEMA impacts on cytotoxicity: low MW pDMAEMA (43 kDa) is considered less toxic than high MW pDMAEMA (915 kDa) [49]. The MW of the pDMAEMA used here was also quite low (12.8 kDa). Deciphering these relationships using HCA will be the subject of future investigations of 17 pDMAEMA polymers of different MW, in co-polymer formats and in complexes with DNA and siRNA. pDMAEMA itself has not been tested by oral administration in vivo. However, methyl methacrylate, a component of pDMAEMA, has been found to be non-toxic by oral administration in vivo in rats [50]. It was rapidly hydrolysed by serum carboxylesterases resulting in rapid degradation of methacrylic acid to less toxic compounds. Due to the variation in effect of pDMAEMA on different cell lines, its effect on whole tissue was assessed. Against rat colonic tissue, pDMAEMA did not cause any histopathology, while against rat ileal tissue there was a degree of sloughing of cells at high concentrations. Fluorescent hostasol-conjugated pDMAEMA has previously been used to visualise interactions of the polymer with rat intestinal tissue [2]. In stomach, caecum and colon tissue, pDMAEMA was retained in the surface mucus layer of the tissue, while in ileum, it penetrated into the intestinal crypts. This varying distribution may explain the reason for the increased histological damage found here in ileal versus colonic tissue. It is possible that the small amount of damage seen with pDMAEMA in vitro on rat intestinal ileal tissue may be diminished in vivo. Absorption enhancers have typically been found to be cytotoxic in vitro, while they produce reversible damage in vivo, possibly due to the presence of mucus, or to the large capacity for the epithelium to repair itself [51]. 18 5. Conclusions In summary, using HCA analysis, pDMAEMA induced cytotoxicity with pooled IC50 values of 25-50 µg/ml against U937 cells and 100-250 µg/ml against Caco-2 cells over 72 hours. The cytotoxicity induced by lower concentrations of pDMAEMA against U937 compared with Caco-2 cells reflects similar results found by MTT assay [3], but the HCA assay detected cytotoxicity at lower concentrations than MTT analysis. It also provided more information about the state of the cells and the mechanism of cell death induced by the polymer. Against isolated rat intestinal mucosa tissue, in vitro, however, pDMAEMA caused no histopathology on rat colon and only limited disruption of the ileal villi at high concentrations. Therefore, although pDMAEMA may not ultimately be suitable for intravenous use, it may have potential as an oral or topical mucoadhesive antibacterial formulation, bearing in mind the high concentrations required for such an effect [3]. Acknowledgements This study was funded by Science Foundation Ireland Strategic Research Cluster Grant 07/SRC/B1154. We thank Eamonn Fitzpatrick from the UCD School of Agriculture, Food Science and Veterinary Medicine for processing and staining histology samples. We also thank David Haddleton at the University of Warwick for synthesis and characterisation of pDMAEMA [3]. 19 References [1] V. Butun, S.P. Armes, N.C. Billingham, Synthesis and aqueous solution properties of near-monodisperse tertiary amine methacrylate homopolymers and diblock copolymers, Polymer 42 (2001) 5993-6008. [2] A.J. Limer, A.K. Rullay, V.S. Miguel, C. Peinado, S. Keely, E. Fitzpatrick, S.D. Carrington, D.J. Brayden, D.M. Haddleton, Fluorescently tagged star polymers by living radical polymerisation for mucoadhesion and bioadhesion, Functional Polymers 66 (2006) 51-64. [3] L.-A. Rawlinson, S.M. Ryan, G. Mantovani, J.A. Syrett, D.M. Haddleton, D.J. Brayden, Antibacterial effects of poly(2-(dimethylamino ethyl)methacrylate) against selected Gram-positive and Gram-negative bacteria, Biomacromolecules 11(2010) 443-453. [4] A. Yousefi Rad, H. Ayhan, U. Kisa, E. Piskin, Adhesion of different bacterial strains to low-temperature plasma treated biomedical PVC catheter surfaces, J. Biomater. Sci. Polym. Ed. 9 (1998) 915-929. [5] S.B. Lee, R.R. Koepsel, S.W. Morley, K. Matyjaszewski, Y. Sun, A.J. Russell, Permanent, nonleaching antibacterial surfaces. 1. Synthesis by atom transfer radical polymerization, Biomacromolecules. 5 (2004) 877-882. [6] Y. Qiao, Y. Huang, C. Qiu, X. Yue, L. Deng, Y. Wan, J. Xing, C. Zhang, S. Yuan, A. Dong, J. Xu, The use of PEGylated poly [2-(N,N-dimethylamino) ethyl methacrylate] as a mucosal DNA delivery vector and the activation of innate immunity and improvement of HIV-1-specific immune responses, Biomaterials 31 (2009) 115-123. [7] F.J. Verbaan, C. Oussoren, C.J. Snel, D.J. Crommelin, W.E. Hennink, G. Storm, Steric stabilization of poly(2-(dimethylamino)ethyl methacrylate)-based polyplexes mediates prolonged circulation and tumor targeting in mice, J. Gene Med. 6 (2004) 64-75. [8] E. Moreau, M. Domurado, P. Chapon, M. Vert, D. Domurad, Biocompatibility of polycations: in vitro agglutination and lysis of red blood cells and in vivo toxicity, J. Drug Target. 10 (2002) 161-173. [9] D.L. Taylor, In: D. L. Taylor, J. R. Haskins and K. A. Guiliano (Eds.), High content screening: A powerful approach to systems cell biology and drug discovery, Humana Press Inc, Totowa, NJ, USA, 2007, pp. 3-18. [10] V.C. Abraham, D.L. Towne, J.F. Waring, U. Warrior, D.J. Burns, Application of a high-content multi-parameter cytotoxicity assay to prioritize compounds based on toxicity potential in humans, J. Biomol. Screen. 13 (2008) 527-537. 20 [11] P.J. O'Brien, W. Irwin, D. Diaz, E. Howard-Cofield, C.M. Krejsa, M.R. Slaughter, B. Gao, N. Kaludercic, A. Angeline, P. Bernardi, P. Brain, C. Hougham, High concordance of drug-induced human hepatotoxicity with in vitro cytotoxicity measured in a novel cell-based model using high content screening, Arch. Toxicol. 80 (2006) 580-604. [12] P.J O’Brien, M. Canelas-Domingos, Use of high content analysis in toxicologic clinical pathology for identification and monitoring of translational safety biomarkers. Int. Drug Discovery 1 (2009) 17-21. [13] J.J. Xu, P. V. Henstock, M. C. Dunn, A. R. Smith, J. R. Chabot, D. de Graaf, Cellular imaging predictions of clinical drug-induced liver injury, Tox. Sci. 105 (2008) 97-105. [14] J.L. Stilwell, Y. Guan, R.M. Neve, J.W. Gray, In D. L. Taylor, J. R. Haskins and K. A. Giuliano (Eds.), High content screening: A powerful approach to systems cell biology and drug discovery, Humana Press Inc, Totowa, NJ, USA, 2007, pp. 353-365. [15] B. Zhang, X. Gu, U. Uppalapati, M.A. Ashwell, D.S. Leggett, C.J. Li, High-content fluorescent-based assay for screening activators of DNA damage checkpoint pathways, J. Biomol. Screen. 13 (2008) 538-543. [16] J.B. Kerrison, D.J. Zack, In D. L. Taylor, J. R. Haskins and K. A. Giuliano (Eds.), High content screening: A powerful approach to systems cell biology and drug discovery, Humana Press Inc, Totowa, NJ, USA, 2007, pp. 427-434. [17] N. Anderson, J. Borlak, Drug-induced phospholipidosis, FEBS Lett. 580 (2006) 5533-5540. [18] M.J. Reasor, S. Kacew, Drug-induced phospholipidosis: are there functional consequences?, Exp. Biol. Med. (Maywood). 226 (2001) 825-830. [19] P. Nioi, I.D. Pardo, R.D. Snyder, Monitoring the accumulation of fluorescently labeled phospholipids in cell cultures provides an accurate screen for drugs that induce phospholipidosis, Drug Chem. Toxicol. 31 (2008) 515-528. [20] C. Laboisse, A. Jarry, J.E. Branka, D. Merlin, C. Bou-Hanna, G. Vallette, Recent aspects of the regulation of intestinal mucus secretion, Proc. Nutr. Soc. 55 (1996) 259-264. [21] B. Maro, M.C. Marty, M. Bornens, In vivo and in vitro effects of the mitochondrial uncoupler FCCP on microtubules, EMBO J. 1 (1982) 1347-1352. [22] A.H. Guse, E. Roth, F. Emmrich, Intracellular Ca2+ pools in Jurkat T-lymphocytes, Biochem. J. 291 ( Pt 2) (1993) 447-451. 21 [23] R.M. Martin, H. Leonhardt, M.C. Cardoso, DNA labeling in living cells, Cytom. Part A. 67 (2005) 45-52. [24] K.R. Gee, K.A. Brown, W.N. Chen, J. Bishop-Stewart, D. Gray, I. Johnson, Chemical and physiological characterization of fluo-4 Ca(2+)-indicator dyes, Cell Calcium 27 (2000) 97-106. [25] D. Floryk, J. Houstek, Tetramethyl rhodamine methyl ester (TMRM) is suitable for cytofluorometric measurements of mitochondrial membrane potential in cells treated with digitonin, Biosci. Rep. 19 (1999) 27-34. [26] T. Zuliani, R. Duval, C. Jayat, S. Schnebert, P. Andre, M. Dumas, M.H. Ratinaud, Sensitive and reliable JC-1 and TOTO-3 double staining to assess mitochondrial transmembrane potential and plasma membrane integrity: interest for cell death investigations, Cytom. Part A. 54 (2003) 100-108. [27] T. Nonoyama, R. Fukuda, Drug-induced phospholipidosis - pathological aspects and its prediction, J. Toxicol. Pathol. 21 (2008) 9-24. [28] P. Greenspan, E.P. Mayer, S.D. Fowler, Nile red: a selective fluorescent stain for intracellular lipid droplets, J. Cell Biol. 100 (1985) 965-973. [29] B.W. Halstead, C.M. Zwickl, R.E. Morgan, D.K. Monteith, C.E. Thomas, R.K. Bowers, B.R. Berridge, A clinical flow cytometric biomarker strategy: validation of peripheral leukocyte phospholipidosis using Nile red, J. Appl. Toxicol. 26 (2006) 169-177. [30] Z. Xia, E.L. Appelkvist, J.W. DePierre, L. Nassberger, Tricyclic antidepressantinduced lipidosis in human peripheral monocytes in vitro, as well as in a monocytederived cell line, as monitored by spectrofluorimetry and flow cytometry after staining with Nile red, Biochem. Pharmacol. 53 (1997) 1521-1532. [31] G. Schmitz, G. Muller, Structure and function of lamellar bodies, lipid-protein complexes involved in storage and secretion of cellular lipids, J. Lipid Res. 32 (1991) 1539-1570. [32] J.P. Montenez, F. Van Bambeke, J. Piret, R. Brasseur, P.M. Tulkens, M.P. MingeotLeclercq, Interactions of macrolide antibiotics (Erythromycin A, roxithromycin, erythromycylamine [Dirithromycin], and azithromycin) with phospholipids: computer-aided conformational analysis and studies on acellular and cell culture models, Toxicol. Appl. Pharmacol. 156 (1999) 129-140. [33] J.K. Morelli, M. Buehrle, F. Pognan, L.R. Barone, W. Fieles, P.J. Ciaccio, Validation of an in vitro screen for phospholipidosis using a high-content biology platform, Cell Biol Toxicol. 22 (2006) 15-27. 22 [34] A.B. Cox, L.A. Rawlinson, A.W. Baird, V. Bzik, D.J. Brayden, In vitro interactions between the oral absorption promoter, sodium caprate (C(10)) and S. typhimurium in rat intestinal ileal mucosae, Pharm. Res. 25 (2008) 114-122. [35] P.J. O'Brien, J.R. Haskins, In D. L. Taylor, J. R. Haskins and K. A. Giuliano (Eds.), High content screening: A powerful approach to systems cell biology and drug discovery, Humana Press Inc, Totowa, NJ, USA, 2007, pp. 415-425. [36] S. Keely, A. Rullay, C. Wilson, A. Carmichael, S. Carrington, A. Corfield, D.M. Haddleton, D.J. Brayden, In vitro and ex vivo intestinal tissue models to measure mucoadhesion of poly (methacrylate) and N-trimethylated chitosan polymers, Pharm. Res. 22 (2005) 38-49. [37] Y.Z. You, D.S. Manickam, Q.H. Zhou, D. Oupicky, Reducible poly(2dimethylaminoethyl methacrylate): synthesis, cytotoxicity, and gene delivery activity, J. Control Release. 122 (2007) 217-225. [38] R.A. Jones, M.H. Poniris, M.R. Wilson, pDMAEMA is internalised by endocytosis but does not physically disrupt endosomes, J. Control Release. 96 (2004) 379-391. [39] L. Hayflick, The limited in vitro lifetime of human diploid cell strains, Exp. Cell Res. 37 (1965) 614-636. [40] Y. Kobayashi, R. Sakemura, A. Kumagai, E. Sumikawa, M. Fujii, D. Ayusawa, Nuclear swelling occurs during premature senescence mediated by MAP kinases in normal human fibroblasts, Biosci. Biotechnol. Biochem. 72 (2008) 1122-1125. [41] D.H. Young, H. Kauss, Release of calcium from suspension-cultured glycine max cells by chitosan, other polycations, and polyamines in relation to effects on membrane permeability, Plant Physiol. 73 (1983) 698-702. [42] S. van Cruchten, W. van den Broeck, Morphological and biochemical aspects of apoptosis, oncosis and necrosis, Anat. Histol. Embryol. 31 (2002) 214-223. [43] D.Y. Yu, Q.L. Zhao, Z.L. Wei, T. Nomura, I. Kashiwakura, T.V. Kagiya, T. Kondo, Enhancement of radiation-induced apoptosis of human lymphoma U937 cells by sanazole, Apoptosis. 14 (2009) 655-664. [44] E.J. Calabrese, L.A. Baldwin, A quantitatively-based methodology for the evaluation of chemical hormesis, Hum. Ecol. Risk Assess. 3 (1997) 545-554. [45] A.R. Stebbing, Interpreting 'dose-response' curves using homeodynamic data: with an improved explanation for hormesis, Dose-Response 7 (2009) 221-233. 23 [46] Y. Z. You, D. S. Manickam, Q. H. Zhou, D. Oupicky, Reducible poly(2dimethylaminoethyl methacrylate): synthesis, cytotoxicity, and gene delivery activity, J. Control. Release. 122 (2007) 217-225. [47] E. Yancheva, D. Paneva, D. Danchev, L. Mespouille, P. Dubois, N. Manolova, I. Rashkov, Polyelectrolyte complexes based on (quaternized) poly[(2dimethylamino)ethyl methacrylate]: behavior in contact with blood, Macromol. Biosci. 7 (2007) 940-954. [49] J. M. Layman, S. M. Ramirez, M. D. Green, T. E. Long, Influence of polycation molecular weight on poly(2-dimethylaminoethyl methacrylate)-mediated DNA delivery in vitro, Biomacromolecules. 10 (2009) 1244-1252. [48] P. van de Wetering, J. Y. Cherng, H. Talsma, D, J. Crommelin, W. E. Hennink, 2(Dimethylamino)ethyl methacrylate based (co)polymers as gene transfer agents. J. Control. Release 53 (1998) 145-153. [50] Z. Bereznowski, In vivo assessment of methyl methacrylate metabolism and toxicity, Int. J Biochem. Cell B. 27 (1995) 1311-1316. [51] S. Maher, T. W. Leonard,J. Jacobsen, D. J. Brayden, Safety and efficacy of sodium caprate in promoting oral drug absorption: from in vitro to the clinic. Adv. Drug Delivery Rev. 61 (2009):1427-1449. 24 Table 1 Dye cocktails for HCA assay. Dye Hoechst 33342 Fluo 4-AM TOTO®-3 iodide (642/660) TMRM Volume [Stock] 32 µl 40 µl 1 mM, in dH20 1 mM, in DMSO 40 µl 8 µl 1 mM, in DMSO 100 µM in dH20, from 100 mM stock (DMSO) TMRM: tetramethyl rhodamine methyl ester; TOTO®-3: TOTO®-3 iodide (642/660); total volume=10 ml. Table 2 Positive controls and dyes for HCA assay. Control/Dye Target [Conc] Ref FCCP (mitochondrial uncoupler) Ionomycin (calcium ionophore) Triton X-100 (membranepreturbing detergent) Hoechst 33342* Fluo 4-AM* TMRM* TOTO®-3* Decrease MMP 10 µM [21] Increase [Ca2+]i 20 µM [22] Decrease PMP 0.2 % [11] Nuclear DNA [Ca2+]i MMP PMP 0.8 µM 1 µM 20 nM 1 µM [23] [24] [25] [26] FCCP: carbonylcyanide-p-trifluoromethyphenylhydrazone; MMP: mitochondrial membrane potential; [Ca 2+]i: intracellular calcium; PMP: plasma membrane permeability. * Dye. 25 Table 3 Settings for In Cell® 1000 Workstation analysis for U937 cells. Bracketed numbers indicate units used for Caco-2. Feature Source Segmentation Nuclei Wave 1 (NA, NI, CN) Wave 2 ([Ca2+]i) Wave 4 (PMP) Wave 3 (MMP) Top-hat Cells Reference 1 Reference 2 Min. Area Sensitivity Collar 30 (50) µm2 10 (100) % - Collar - - 1 (5) µm Pseudo Nuclei Pseudo - Cells - - - - - - Glossary. Feature: the parameter that the analysis package is analysing; Source: the wavelength channel used for the feature; Segmentation: how the analysis package analyses the data, e.g. based on nucleus, cytoplasm or whole cell; ‘Top-hat’ is the method used to identify nuclei, ‘Collar’ is the method used to identify cell cytoplasm and ‘Pseudo’ is the method used to identify other dyes within specific areas of the cell; Min.(Minimum) area: area of the smallest cell to be tested; Sensitivity: sensitivity for picking cells from background noise; Collar (size): radius around nucleus for analysis. Table 4 Settings for In Cell® 1000 Workstation analysis for phospholipidosis in cells. Feature Source Segmentation Min. area Sensitivity Nuclei Cells Wave 1 Wave 2 Top-hat Collar 40 µm2 100 % Collar 5 µm 26 Table 5 Summary of pDMAEMA effects on cells. Concentrations (µg/ml) of pDMAEMA that produce significant changes from controls (P < 0.05 compared to 100 % controls containing no drug at all time points). A. U937 Time (Hours) 24 NA NI [Ca2+]i PMP MMP CN ↑100 ↑50 ↓50 25 ↑0.01-1 48 ↑100 ↑50 ↓100 25 72 ↑100 ↑50 ↓100 50 ↑0.01-0.1, ↓50 ↑0.01-0.1, ↓50 ↓50 NA NI [Ca2+]i PMP MMP CN ↓100 ↓100 ↑250 ↑100 ↑250 ↑250 250 250 ↓10 ↓500 ↓0.1 ↑100 ↑100 250 ↓500 ↓5000 ↑10, ↓1000 ↓0.1 B. Caco-2 Time (Hours) 24 48 72 ↑0.1-1 >1000 Upward arrows indicate increase compared to control, downward arrows indicate decrease compared to controls. NA: nuclear area; NI: nuclear intensity; [Ca2+]i: intracellular calcium; PMP: plasma membrane permeability; MMP: mitochondrial membrane potential; CN: cell number. 27 Table 6 IC50 values for pDMAEMA on Caco-2 and U937 cells after exposure to pDMAEMA. A: 72 hours treatment, B: Time course. Notation as for Table 5. A. pDMAEMA IC50 values after 72 hours Parameter Caco-2 U937 NA NI [Ca2+]i MMP PMP CN >5000 250 100 140 550 100 >1000 >1000 100 50 45 >1000 B. pDMAEMA IC50 values over time Time Hours Caco-2 24 250 48 150 72 100 Values given in µg/ml A U937 25 25 50 B 28 Fig. 1. Caco-2 U937 29 % Intracellular Ca % Nuclear Intensity % Nuclear Area % Mem Permeability % Mito Potential % Cell Number 120 *** NA 100 ** *** 80 60 250 200 NI 150 *** *** 100 50 0 600 *** [Ca2+]i 400 * 200 0 200 150 *** MMP ** ** 100 ** 50 0 500 400 *** *** *** *** PMP 300 200 *** *** 100 0 300 CN *** 200 100 *** ** *** *** *** 1 10 10 0 10 00 10 00 0 0.1 1 10 10 0 10 00 10 00 0.0 0 01 0.0 1 0.1 0.0 01 0.0 1 0 [pDMAEMA] (g/ml) Fig. 2. 30 A B C D Fig. 3. 31 % Fluorescence 140 A *** 120 *** 100 ** ** Ery TTX 50 10 1 Control 80 [pDMAEMA] (g/ml) 180 B *** % Fluorescence 160 140 * 120 100 Ery TTX 100 50 10 Control 80 [pDMAEMA] (g/ml) Fig. 4. 32 A C B D Fig. 5. 33 A B C D Fig. 6. Figure Legends Fig. 1. Representative fused images of the HCA assay. A: U937, B: Caco-2. Cells were grown for 24 hours and then exposed to 20µM ionomycin for 30 min. Blue/green and red cells indicate healthy cells with active MMPs. Green cells indicate cells with increased [Ca2+]i. Blue cells indicated cells with no MMP and an increased PMP. Horizontal bars = 50 µm. Fig. 2. Concentration-response curves of effects of pDMAEMA on Caco-2 and U937 cells after 72 hours. Readings were compared to 100 % negative controls. * P < 0.05, ** 34 P < 0.01, *** P < 0.001 compared to controls containing no drug. Notation as for Table 5 legend. Fig. 3. Representative fused images of the phospholipidosis HCA assay. A: U937 cells, B: U937 cells treated with erythromycin (250 µg/ml), C: Caco-2 cells, D: Caco-2 cells treated with erythromycin (250 µg/ml). Exposure time was 24 hours. Nuclei were stained red by Hoechst 33342. The phospholipids were stained green by Nile red. Horizontal bars = 10 µm. Fig. 4. Effect of pDMAEMA on phospholipidosis. A: U937, B: Caco-2. ■ = 24 hours, ▲ = 48 hours, □ = 72 hours. * P < 0.05, ** P < 0.01, *** P < 0.001 compared to 100 % controls containing no drug at all time points. Ery = erythromycin, TTX = Triton X-100. Ery: 250 µg/ml, TTX: 0.01 %. Fig. 5. Representative images from 3 independent experiments of H & E staining of Ussing chamber-mounted rat ileal mucosae. A: Untreated, B: 1 mg/ml pDMAEMA, C: 10 mg/ml pDMAEMA, D: 0.1 % Triton X-100. Horizontal bars = 50 µm. Fig. 6. Representative images from 3 independent experiments H & E staining of Ussing chamber-mounted rat colonic mucosae. A: Untreated, B: 1 mg/ml pDMAEMA, C: 10 mg/ml pDMAEMA, D: 0.1 % Triton X-100. Horizontal bars = 50 µm. 35 36