Báo cáo khoa học: Tumor suppressor p16INK4a: Downregulation of galectin-3, an endogenous competitor of the pro-anoikis effector galectin-1, in a pancreatic carcinoma model

pdf
Số trang Báo cáo khoa học: Tumor suppressor p16INK4a: Downregulation of galectin-3, an endogenous competitor of the pro-anoikis effector galectin-1, in a pancreatic carcinoma model 12 Cỡ tệp Báo cáo khoa học: Tumor suppressor p16INK4a: Downregulation of galectin-3, an endogenous competitor of the pro-anoikis effector galectin-1, in a pancreatic carcinoma model 494 KB Lượt tải Báo cáo khoa học: Tumor suppressor p16INK4a: Downregulation of galectin-3, an endogenous competitor of the pro-anoikis effector galectin-1, in a pancreatic carcinoma model 0 Lượt đọc Báo cáo khoa học: Tumor suppressor p16INK4a: Downregulation of galectin-3, an endogenous competitor of the pro-anoikis effector galectin-1, in a pancreatic carcinoma model 0
Đánh giá Báo cáo khoa học: Tumor suppressor p16INK4a: Downregulation of galectin-3, an endogenous competitor of the pro-anoikis effector galectin-1, in a pancreatic carcinoma model
4.4 ( 17 lượt)
Nhấn vào bên dưới để tải tài liệu
Đang xem trước 10 trên tổng 12 trang, để tải xuống xem đầy đủ hãy nhấn vào bên trên
Chủ đề liên quan

Nội dung

Tumor suppressor p16INK4a: Downregulation of galectin-3, an endogenous competitor of the pro-anoikis effector galectin-1, in a pancreatic carcinoma model Hugo Sanchez-Ruderisch1,*, Christian Fischer1, Katharina M. Detjen1, Martina Welzel1, Anja Wimmel1, Joachim C. Manning2, Sabine André2 and Hans-Joachim Gabius2 1 Medizinische Klinik m.S. Hepatologie und Gastroenterologie, Charité-Universitätsmedizin Berlin, Germany 2 Institut für Physiologische Chemie, Ludwig-Maximilians-Universität München, Germany Keywords anoikis; galectin; glycosylation; integrin; pancreatic carcinoma Correspondence K. M. Detjen, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Med. Klinik m.S. Hepatologie und Gastroenterologie, Augustenburger Platz 1, D-13353 Berlin, Germany Fax: +49 30 450 559939 Tel: +49 30 450 559679 E-mail: katharina.detjen@charite.de *Present address Center for Cardiovascular Research, CharitéUniversitätsmedizin Berlin, Germany (Received 20 May 2010, revised 1 July 2010, accepted 5 July 2010) doi:10.1111/j.1742-4658.2010.07764.x The tumor suppressor p16INK4a has functions beyond cell-cycle control via cyclin-dependent kinases. A coordinated remodeling of N- and O-glycosylation, and an increase in the presentation of the endogenous lectin galectin1 sensing these changes on the surface of p16INK4a-expressing pancreatic carcinoma cells (Capan-1), lead to potent pro-anoikis signals. We show that the p16INK4a-dependent impact on growth-regulatory lectins is not limited to galectin-1, but also concerns galectin-3. By monitoring its expression in relation to p16INK4a status, as well as running anoikis assays with galectin-3 and cell transfectants with up- or downregulated lectin expression, a negative correlation between anoikis and the presence of this lectin was established. Nuclear run-off and northern blotting experiments revealed an effect of the presence of p16INK4a on steady-state levels of galectin-3-specific mRNA that differed from decreasing the transcriptional rate. On the cell surface, galectin-3 interferes with galectin-1, which initiates signaling toward its pro-anoikis activity via caspase-8 activation. The detected opposite effects of p16INK4a at the levels of growth-regulatory galectins-1 and -3 shift the status markedly towards the galectin-1-dependent pro-anoikis activity. A previously undescribed orchestrated fine-tuning of this effector system by a tumor suppressor is discovered. Introduction The tumor suppressor p16INK4a, a frequent target for deletion mutations underlying carcinogenesis, is known as a binding partner of cyclin-dependent kinases CDK4 and CDK6, interfering with their association with D-type cyclins [1,2]. Emerging evidence broadens the spectrum of p16INK4a functionality beyond cellcycle control. In fact, recent experiments have unraveled an effect on distinct aspects of gene expression. One functional consequence is to restore the susceptibility of tumor cells to anoikis (the category of apoptosis caused by inadequate or inappropriate cell–matrix contacts). In detail, enhanced production of the a5-integrin subunit and also an increased cell-surface presence of a5b1-integrin (the fibronectin receptor) were detected in p16INK4a-restituted Capan-1 pancreatic carcinoma cells [3]. Routing of this glycoprotein and the levels of its cell-surface presentation, binding activity and capacity for downstream signaling may, in principle, depend not only on the protein part, but also on its glycosylation. Because the fibronectin receptor is heavily glycosylated with 26 sites for N-glycosylation [4] and variations in the structures of glycan Abbreviations Gal-1, galectin-1; Gal-3, galectin-3; PCNA, proliferating cell nuclear antigen; poly-HEMA, poly(2-hydroxyethyl methacrylate). 3552 FEBS Journal 277 (2010) 3552–3563 ª 2010 The Authors Journal compilation ª 2010 FEBS p16INK4a downregulates anti-anoikis effector Gal-3 H. Sanchez-Ruderisch et al. chains, such as status of sialylation, have a bearing on protein functions in general and on integrins in particular [5–8], glycan remodeling affords an attractive level of regulation of integrin functionality. The assumption of a modulatory impact of p16INK4a at the level of glycosylation was a reasonable and testable hypothesis. This hypothesis was verified by combining glycogene microarray analysis, chromatographic glycan profiling and lectin binding [9]. Influencing N- and O-glycan galactosylation and sialylation, salient biochemical signals in protein–carbohydrate ⁄ protein interplay [10–12], have thus become a new aspect of p16INK4a functionality. Hereby, the tumor suppressor can affect integrin processing and routing, as well as its affinity for protein ligands. In addition, modified glycan chains can be engaged in recognitive interactions at the cell surface with endogenous lectins, which translate sugar-encoded messages into cellular responses [13–15]. Even seemingly small modifications can act as potent switches of affinity. In fact, altering branch-end positions, the status of core substitutions and glycan density strongly affects lectin reactivity, as measured for example for members of the family of adhesion ⁄ growth-regulatory galectins [16–19]. Fittingly for a functional implication, microarray analysis on a set of 1996 cancer-associated genes, proteomic profiling and cytofluorometry and anoikis assays revealed the upregulation of homodimeric galectin-1 (Gal-1) as a carbohydrate-binding effector for anoikis induction under the control of p16INK4a [9]. Thus, this tumor suppressor sensitizes cells for the onset of anoikis by increasing the complementary sides of pro-anoikis protein–carbohydrate interactions. As the term Gal-1 implies, the functionality of this protein might be embedded in a network with other family members [20]. This raises the possibility of additive or antagonistic effects. Exploring this issue, model studies on SK-N-MC neuroblastoma cells illustrated the potential of other galectins, especially galectin-3 (Gal-3), to factor in growth inhibition by Gal-1 at the level of cell-surface ligand binding [21,22]. Having recently delineated a direct connection between p16INK4a and Gal-1 [9], the question arises as to whether the influence of p16INK4a is restricted to this family member. The following four lines of evidence direct the focus of attention on Gal-3. First, it is functionally antagonistic to Gal-1 in the neuroblastoma system by blocking access to ganglioside GM1, the common galectin ligand on the cell surface in these cells [21]. Second, activating K-ras mutations are common in pancreatic cancer, and Gal-3 (but not Gal-1) interacts with oncogenic K-ras–GTP to promote Raf-1 and phosphoinositide 3-kinase activities, as well as a signal attenuating extracellular signal-regulated kinase [23]. This is in accordance with the lack of aberrantly enhanced extracellular signal-regulated kinase signaling in pancreatic tumors which harbor the mentioned gene defect [24]. Third, Gal-3 protects BT549 breast cancer cells from anoikis [25]. Fourth, Capan-1 wild-type cells are known to express Gal-3 [26]. On this basis, we investigated the influence of the presence of p16INK4a on the level of Gal-3 production and possible functional competition between galectins-1 and -3. The reported results document the relevance of Gal-3 in interfering with anoikis induction and, more importantly, shed light on the intriguing capacity of this tumor suppressor to coordinately exploit this aspect of the galectin network. Results Expression of p16INK4a decreases the level of Gal-3 In order to delineate any impact of the tumor suppressor p16INK4a on Gal-3 expression we worked with Capan-1 pancreatic carcinoma wild-type and vectortransfected cells (mock) as well as three independently generated clones stably transfected with p16INK4a tumor suppressor cDNA (p16 1-3). Of note, these clones had been studied previously, revealing increased expression and cell-surface presentation of the a5-integrin subunit and Gal-1 [3,9]. Western blot analyses first resulted in the expected pattern for the p16INK4a protein, then confirmed the reported presence of Gal-3 in Capan-1 wildtype cells [26] and excluded an effect of the control transfection on Gal-3 and general protein synthesis (Fig. 1A). In contrast to the mock process, the presence of p16INK4a protein had an effect. A clear decrease in the intensity of signals for Gal-3 was invariably observed, although proliferating cell nuclear antigen (PCNA) staining as a control for loading remained rather constant (Fig. 1A,B). To examine whether such a negative correlation between Gal-3 and p16INK4a can also be seen in vivo, we processed routinely fixed sections from normal pancreas tissue and pancreatic cancer immunohistochemically. The resulting staining profiles, shown in Fig. S1, confirmed an inverse expression pattern in accordance with the in vitro data. In principle, Gal-3 can act as an anti-anoikis effector in the cell and ⁄ or at the cell surface, for example, blocking Gal-1 functionally as seen previously in a neuroblastoma model [21]. In view of the proven pro-anoikis cell-surface activity of Gal-1, the cultured cells provided the opportunity to test the hypothesis of reduced FEBS Journal 277 (2010) 3552–3563 ª 2010 The Authors Journal compilation ª 2010 FEBS 3553 p16INK4a downregulates anti-anoikis effector Gal-3 H. Sanchez-Ruderisch et al. A cell-surface expression of Gal-3. Indeed, cytofluorimetric monitoring of mock-transfected and p16INK4a-restituted Capan-1 cells revealed that cell-surface Gal-3 was downregulated (Fig. 1C). As reported previously [9], Gal-1 cell-surface presentation determined in the control increased (not shown). The presence of the tumor suppressor thus reduced the level of Gal-3 and its cellsurface presentation. To further test whether Gal-3 protein abundance was regulated by prolonged culture in suspension, we maintained cells for up to 24 h under this condition, with the percentage of cells undergoing anoikis increasing as expected in p16INK4a-expresssing cells (Fig. 2A). Loss of anchorage did not notably change the level of Gal-3 detected in western blots (Fig. 2B). These results document a significant decrease in the presence of Gal-3 in tumor-suppressor-positive cells, which is not further enhanced in suspension culture. This suggests regulation at the level of transcription, as previously detected for Gal-1 [9]. To test this, we determined the steady-state mRNA concentration by northern blotting and the de novo transcription rate by run-off assays. B p16INK4a negatively affects Gal-3 mRNA availability 100 100 0 0 100 C 101 102 103 104 100 101 102 103 104 Fig. 1. p16INK4a restitution inhibits Gal-3 expression. (A) Western blot after 1D SDS ⁄ PAGE [15% gel, proteins blotted onto poly(vinylidene fluoride)] with protein extracts from Capan-1 wild-type (wt), mock-transfected and three p16INK4a-expressing clones incubated with antibodies to p16INK4a, Gal-3 and PCNA, respectively. The level of Gal-3 is reduced in p16INK4a-expressing clones. (B) Western blot after 2D gel electrophoresis with protein extract from mock-transfected (1,2: 200, 400 lg) and p16INK4a-transfected cells (3,4: 200, 400 lg). Residual protein in gels was visualized by silver staining (lower part of each panel) and each blotting procedure included a positive control with Gal-3 (top right). (C) Quantitation of cell-surface presentation of Gal-3 in mock-transfected (left) and p16INK4aexpressing Capan-1 pancreatic cancer cells (right). The control for antigen-independent staining by omitting the incubation step with the lectin-specific antibody from the protocol is given in each panel (gray area), as are the percentage of positive cells and the mean fluorescence intensity of staining when incubating with 20 lgÆmL)1 non-cross-reactive anti-galectin-3 Ig. Standard deviation did not exceed 11% in experimental series with four different experiments run in triplicates. 3554 Northern blotting using samples from p16INK4a-positive cells revealed a conspicuous decrease in the availability of Gal-3-specific mRNA, when compared with mocktransfected controls, whereas similar mRNA levels for the housekeeping gene GAPDH were present irrespective of cell status (Fig. 2C, left). This diminished steadystate amount may be because of alterations in de novo production or availability. Nuclear run-off assays, which report on de novo synthesis, revealed rather similar signal intensities in samples from mock- and p16INK4a-transfected cells for the two tested housekeeping genes and for Gal-3 (Fig. 2C, right). De novo Gal-1 gene transcription, by contrast, increased significantly in the presence of the tumor suppressor (not shown). Thus, the transcriptional rate is a major factor in increasing the production of Gal-1, whereas post-transcriptional processes decrease Gal-3 production. We propose a functional relevance for this alteration. In order to prove an effect of Gal-3 on anoikis in this cell system, especially regarding a functional antagonism with Gal-1 at the level of the cell surface, we followed three routes of investigation to test the validity of this assumption. Gal-3 is an inhibitor of anoikis In the first set of experiments, we tested the capacity of Gal-3 to block anoikis in p16INK4a-expressing cells FEBS Journal 277 (2010) 3552–3563 ª 2010 The Authors Journal compilation ª 2010 FEBS p16INK4a downregulates anti-anoikis effector Gal-3 Time (h) 200 400 FL2-H 0 600 200 400 FL2-H 600 12 0 20 40 60 80 5 0 0 600 0 200 400 FL2-H 600 30 60 90 120 6 0 0 200 400 FL2-H 600 12 40 80 120 160 400 FL2-H 0 200 6 0 8 200 0 400 FL2-H 16 600 200 400 FL2-H 600 31 0 10 20 30 40 50 60 5 0 0 Counts p16-3 0 0 40 80 120160 200 Mock 5 0 30 60 90120 150 50 100 150 200 A 50 100 150 200 H. Sanchez-Ruderisch et al. 0 200 400 FL2-H 600 24 B C Fig. 2. The level of Gal-3 is diminished in p16INK4a-expressing clones but remains constant over the 24 h anoikis induction period. (A) Representative FACS histograms illustrating the increased extent of anoikis induction in Capan-1 ⁄ p16INK4a-positive cells compared with a mocktransfected clone. Cells were harvested following incubation on poly-HEMA to preclude attachment and trigger anoikis for the indicated time. The given numbers indicate the percentage of cells with subdiploid DNA content (pre-G1 fraction). (B) Western blot analysis from extracts obtained under the conditions described above and analyzed with anti-Gal-3 or anti-PCNA Ig as indicated. (C) Poly-(A+) RNAs were isolated from mock-transfected and p16INK4a-expressing clones and northern blots were probed for the presence of mRNA specific for Gal-3 or GAPDH (left). In vitro elongation of de novo RNA transcripts was performed in isolated nuclei from mock-transfected and p16INK4aexpressing clones in the presence of [32P]UTP[aP], and the radioactively labeled RNA was hybridized to immobilized cDNA for Gal-3, GAPDH and b-actin (right). One representative of three experiments that yielded similar results is shown. when added to the medium. As shown in Fig. 3, exposure of cells to Gal-3 was inhibitory in this experimental setting. The percentage of p16INK4a-positive cells undergoing anoikis after 20 h in suspension was reduced when kept in the presence of suitable concentrations of Gal-3. To further strengthen the link between Gal-3 and anoikis, the availability of this lectin was upregulated deliberately by generating p16INK4-positive cells, which additionally express Gal-3 at a high level via a second transfection. Thus, the presence of Gal-3 was increased, counteracting the p16INK4a-dependent downregulation. A series of five clones was obtained with notably enhanced Gal-3 concentration, an unspecific influence of the second transfection step rendered unlikely by a mock control (Fig. 4A). The level of anoikis in these control cells was used as a reference, and four Gal-3-overexpressing clones revealed a trend towards reduced anoikis susceptibility. This reached statistical significance in clone Gal-3 ⁄ 2 (Fig. 4B). Results showing that exogenous addition and vector-directed overexpression of Gal-3 can reduce the level of anoikis shaped the notion that Gal-3 physically hampers the induction of this cell death program. In this case, even p16INK4a-negative cells should become sensitized toward anoikis if their Gal-3 production is downregulated. This reasoning led to the third set of experiments. To test the given hypothesis, we generated clones harboring an antisense vector for Gal-3. Because of this engineering, the clones contained a lower level of Gal-3 than wild-type and mock-transfected cell populations (Fig. 4C). Measurements of the corresponding cell-cycle profiles and percentages of cells undergoing anoikis revealed a Gal-3-dependent increase in this FEBS Journal 277 (2010) 3552–3563 ª 2010 The Authors Journal compilation ª 2010 FEBS 3555 p16INK4a downregulates anti-anoikis effector Gal-3 H. Sanchez-Ruderisch et al. fibronectin receptor as had been shown in this and other carcinoma cell systems [9,27]. If Gal-3 interferes with Gal-1 binding to and cross-linking the a5-subunit, then Gal-3 should also negatively affect post-binding signaling by the integrin. We tested and established the involvement of caspase-8 activation for Gal-1-dependent anoikis induction (Fig. 6A) and revealed a negative impact of Gal-3 at this level (Fig. 6B,C). A Discussion 120 12 40 80 60 39 0 0 40 20 Counts 80 160 B 0 200 400 0 200 DNA No Gal-3 + 125 DNA µg·mL–1 400 Gal-3 Fig. 3. Addition of Gal-3 to the culture medium inhibits anoikis induction. (A) Gal-3 was added to p16INK4a-expressing cells (clone p16-3) in the concentrations given. Following 20 h incubation on poly-HEMA, anoikis rates were determined from DNA histograms based on the fraction of cells with subdiploid DNA content. The data are expressed as percentage of the control without the addition of Gal-3 (n = 3, **P < 0.01, ***P < 0.005). (B) Representative DNA histograms in the absence (no Gal-3) or in the presence of 125 lgÆmL)1 Gal-3. The given numbers indicate the percentage of cells in the pre-G1 fraction. parameter (Fig. 4D,E). Evidently, the level of Gal-3 is important to protect wild-type cells from anoikis induction, and the forced expression of Gal-3 in p16INK4a-positive cells reduces their susceptibility to anoikis. Because this pattern is inverse to the Gal-1-induced effects reported previously [9], it is reasonable to propose functional competition between galectins-1 and -3 at the cell surface. To probe for such an effect, we stimulated anoikis by addition of Gal-1 to the culture medium. Should Gal-3 be an inhibitor, the extent of Gal-1-dependent anoikis induction via glycan binding will decrease. Stepwise increases in the Gal-3 concentration progressively diminished the Gal-1-dependent effect (Fig. 5A), and galectin binding, shown to be carbohydrate dependent [9], was reduced in cross-competition assays (Fig. 5B,C). The presence of Gal-3 can thus impair the pro-anoikis effect of Gal-1 at the level of the cell surface, here most likely targeting the 3556 The term ‘tumor suppressor’ summarizes an obvious function of a protein on malignancy. Naturally, a suppressor engages secondary effectors at different levels, which turn its presence as a master organizer into renormalization of phenotypic characteristics. Because of growing insights into the role of glycosylation in cellular communication, including growth control [6,28], we hypothesized that p16INK4a is capable of taking advantage of this network, explicitly by modulating the lectin reactivity of glycans and ⁄ or lectin expression. As a consequence, further study provided a novel explanation for why this tumor suppressor restores susceptibility to anoikis in Capan-1 pancreatic carcinoma cells [9]. Having detected coordinated upregulation of both Gal-1 and suitable cell-surface ligands, the results provoked a question regarding orchestration of effects on expression in the lectin family beyond Gal-1. Evidently, the chimera-type Gal-3, known as a functional antagonist of Gal-1 in a tumor model, offered a prime target for study. Cumulatively, the experiments reported herein revealed p16INK4a-dependent regulation of the presence of Gal-3 in this cell system. They also delineated a strong influence of Gal-3 on anoikis induction. Of special note, we detected functional competition at the cell surface with the recently proven pro-anoikis activity of Gal-1, which involves caspase-8 activation. Combining previous results on Gal-1 ⁄ a5b1-integrin co-immunoprecipitation, and the effects of p16INK4a on a5b1integrin, cell-surface glycosylation and Gal-1 [3,9] with the presented data enabled us to set up a scheme of p16INK4a-orchestrated changes that favor Gal-1-dependent anoikis (Fig. 7). Whether and how a reduction in the intracellular activities of Gal-3 documented in other tumor cell types (e.g. interaction with oncogenic K-ras and transcriptional modulation of cell-cycle regulators such as cyclins A, D1 and E as well as p21 ⁄ p27 [23,25,29]) will cooperate with the functional interference of cell-surface binding of homodimeric Gal-1, is not clear. At the cell surface, the particular profile of the chimera-type galectin for ligand cross-linking shown recently [30] will definitely not elicit pro-anoikis FEBS Journal 277 (2010) 3552–3563 ª 2010 The Authors Journal compilation ª 2010 FEBS p16INK4a downregulates anti-anoikis effector Gal-3 H. Sanchez-Ruderisch et al. C D 0 200 400 DNA Wt 0 200 400 DNA 22 60 17 0 0 0 0 10 30 20 40 60 80 100 120 60 90 9 30 Counts 120 E 90 120 150 B 20 40 60 80 100 120 A 0 Mock 200 400 DNA asG3-A 0 200 400 DNA asG3-B Fig. 4. Upregulation of Gal-3 in clones changes anoikis susceptibility. (A,B) Overexpression of Gal-3 protects p16INK4a-expressing clones from anoikis. (A) Increased Gal-3 content was confirmed via detection of Gal-3 in western blots of clones that were stably transfected with a Gal3 expression construct. Blots were conducted on whole-cell lysates of Capan-1 wild-type cells (wt), a p16INK4a-expressing clone (p16) and p16INK4a-positive clones following mock transfection and with additional overexpression of Gal-3 (p16 ⁄ Mock and Gal-3 ⁄ 1-5). Blots were probed with anti-Gal-3 or anti-PCNA Ig. (B) Determination of anoikis level in five p16INK4a-positive clones with overexpression of Gal-3 and a respective mock control. Extent of anoikis is expressed as a percentage relative to the mock-transfected control clone (n = 3, *P < 0.05). (C–E) Reduction in the level of endogenous Gal-3 stimulates anoikis induction. (C) Downregulation of cellular Gal-3 was ascertained by detection of Gal-3 in western blot analyses of protein extracts from wild-type (wt) and mock-transfected cells, as well as from two clones transfected with an antisense (as) Gal-3 cDNA construct (asG3A ⁄ B). Additional immunoblotting for PCNA (lower) was conducted to control for unspecific effects on protein synthesis. (D) Determination of level of anoikis in clones with reduced Gal-3. Summary of anoikis rates obtained in clones with decreased Gal-3 and mock controls. Anoikis rates are given as a percentage of the total number of cells (n = 3, *P < 0.05, **P < 0.01). (E) Representative cell-cycle histograms from cultures kept on poly-HEMA for 20 h. The given numbers indicate the percentage of cells in the pre-G1 fraction. signaling of Gal-1 in this cell system. This functional competition between galectins, described initially for SK-N-MC neuroblastoma cells and ganglioside GM1 [21], may well play a more general role in tumor growth control. Its detection gives a clear direction to further research. In this respect, such a study is warranted on the p27-dependent downregulation of carcinoma cell growth triggered by Gal-1 and most likely the a5b1-integrin [27], as well as the Gal-1 ⁄ GM1- dependent communication between effector T and regulatory T cells, which involves a4 ⁄ a5b1-integrins and Ca2+ influx via TRPC5 channels [31]. Eventually, these investigations will unravel the mostly unexplored intricacies of the galectin network monitored in tumors by RT-PCR and by immunohistochemistry [32–34]. Such cell biological studies will then shed light on additive ⁄ synergistic, as opposed to antagonistic, activities in malignancy and immune regulation. FEBS Journal 277 (2010) 3552–3563 ª 2010 The Authors Journal compilation ª 2010 FEBS 3557 p16INK4a downregulates anti-anoikis effector Gal-3 ** ** # * 150 ## 100 50 * 30 20 10 0 0 ) –1 l tro on C L 00 -1 al -1 al G + G · µg l-3 (2 0 l-3 -1 + G al G L -1 al G L -3 50 -3 16 24 B (1 al al + µ 0 (1 · µg 6 Time (h) L m m g· 0 (5 ) –1 ) –1 m · µg a a G L m 5 (1 ) –1 ) –1 m · µg FAM-LETD-FMK 40 200 Anoikis (% control) Control A * Apoptotic cells (%) A H. Sanchez-Ruderisch et al. G -1 + G al G 80 C 0 40 Counts 120 B 100 101 102 Gal-3 binding (log fluorescence) 80 0 40 Counts 120 C 100 101 102 Gal-1 binding (log fluorescence) Fig. 5. Gal-3 inhibits Gal-1-stimulated anoikis and Gal-1 binding. (A) Determination of Gal-1-stimulated (100 lgÆmL)1) anoikis in p16INK4a-restituted cells in the presence of increasing concentrations of Gal-3. Anoikis rates were determined following 20 h of culture on poly-HEMA and were calculated based on the pre-G1 fraction from cell-cycle analyses. Data are expressed as the percentage of vehicle-treated mock control cells (n = 4, *P < 0.05, **P < 0.01, compared with control without Gal-1; #P < 0.05, ##P < 0.01 compared with data for Gal-1). (B) Binding of biotinylated Gal-3 in the presence (light line) or absence (bold line) of 100 lgÆmL)1 Gal-1. The dashed line signifies the control with fluorescent reagent, without prior incubation with biotinylated Gal-3. (C) Binding of biotinylated Gal-1 in the presence (light line) or absence (bold line) of 150 lgÆmL)1 Gal-3. The dashed line signifies the control. 3558 Fig. 6. Gal-3 affects the onset of anoikis by inhibiting Gal-1-dependent caspase-8 activation. (A) Anoikis after 6, 12 or 24 h incubation on poly-HEMA in the presence or absence of the caspase 8 inhibitor FAM-LETD-FMK. (B) Caspase 8 activation expressed as the percentage of cells with active caspase 8 in the absence (control, filled circles) and presence (open circles) of 100 lgÆmL)1 Gal-3 (**P < 0.01, ***P < 0.001, compared with control). (C) Anoikis (percentage of cells in the pre-G1 fraction) in the absence (control, filled circles) and presence (open circles) of 100 lgÆmL)1 Gal-3 (**P < 0.01, compared with control). Equally important, our study broadens the basis for galectin involvement in tumor suppressor activity. Here, the key reference point to date had been p53. In detail, SAGE screening on DLD-1 colon carcinoma cells expressing p53 identified galectin-7 as a p53-induced gene 1 in a group of 14 genes markedly upregulated from a total of 7202 tested transcripts [35]. Along this line, genotoxic stress by UVB irradiation of human keratinocytes afforded a second system in which a connection between p53 and galectin-7 was FEBS Journal 277 (2010) 3552–3563 ª 2010 The Authors Journal compilation ª 2010 FEBS p16INK4a downregulates anti-anoikis effector Gal-3 H. Sanchez-Ruderisch et al. Fig. 7. Glycobiology of p16INK4a functionality in Capan-1 pancreatic carcinoma cells in vitro. The tumor suppressor orchestrates: an increase in the cell-surface presentation of the fibronectin receptor (involving transcriptional upregulation of a5-subunit gene expression), regulation of glycogenes enabling increased Gal-1 cell-surface reactivity, and an increase in the cell-surface presentation of Gal-1 (involving transcriptional upregulation of Gal-1 gene expression) [3,9]. Formation of Gal-1 ⁄ a5b1-integrin complexes with ensuing cross-linking appears to lead to anoikis induction via caspase-8 activation. Gal-3 can interfere with Gal-1 binding and ⁄ or Gal-1dependent cross-linking at the level of the cell surface, decreasing the pro-anoikis activity of Gal-1. p16INK4a-dependent Gal-3 downregulation, with potential bearing also on intracellular anti-anoikis activity of Gal-3, favors the pro-anoikis effect of cell surface Gal-1. made likely [36]. When expressed in HeLa cell transfectants, the intracellular function of galectin-7 appeared to relate to affecting gene expression profiles. Conspicuous changes were attributed to pro-apoptosis signaling upstream of c-Jun N-terminal kinase activation and cytochrome c release [37]. When tested as an extracellular effector, carbohydrate-dependent binding led to growth inhibition of activated T cells or neuroblastoma cells via caspase-dependent or -independent pathways [38,39]. Of further relevance, the recently documented effect of compensation of loss of supressor genes in microsatellite instability on glycosylation, including a2,6-sialylation of N-glycans, broadens the scope for co-regulation [40]. Further work to strengthen this connection between a tumor suppressor and lectin ⁄ glycan remodeling as an effector pathway, especially by examining clinical samples, is clearly justified. Examining galectin expression in clinical samples of pancreatic cancer by gene-expression profiling uncovered a consistent, albeit quantitatively variable, upreg- ulation of Gal-3 expression [41–45]. The similarly consistent upregulation of Gal-1 gene expression and protein production may at first seem puzzling. Looking at its immunohistochemical pattern, it could mostly be accounted for by a desmoplastic reaction around the tumor cells, with a Gal-1 ⁄ tissue plasminogen activator interaction being involved [44–50]. Taking our data literally, an absence or low level of Gal-1, combined with an abundance of Gal-3 in the tumor cells, seems to reflect impairment of p16INK4a. As shown in the Supporting Information, preliminary immunohistochemical analysis on a limited number of cases focusing on the presence of p16INK4a and Gal-3 indicated a tendency for a negative correlation, in accordance with the in vitro data. These results encourage thorough investigation of this aspect to strengthen clinical relevance. By following this line of research, the orchestration of galectin expression described here may become instrumental in devising a novel therapeutic strategy to rationally shift the balance between resistance and susceptibility to favor the anoikis process. Materials and methods Galectins Human galectins were produced by recombinant expression, isolated by affinity chromatography on lactosylated Sepharose 4B as crucial step followed by gel filtration, and tested for purity using 1D and 2D gel electrophoresis and nanoelectrospray ionization mass spectrometry, as well as for activity by hemagglutination [9,38,51,52]. Biotinylation labeling was performed under activity-preserving conditions using the commercial N-hydroxysuccinimide ester derivative of biotin (Sigma, Munich, Germany). Its incorporation into the galectins was determined using a proteomics protocol, and the activity of the labeled proteins was checked using carbohydrate-dependent solid-phase and cell binding assays [52,53]. Polyclonal antibodies were raised in rabbits and rigorously checked for lack of cross-reactivity using enzymelinked immunosorbent assays and western blotting [54,55]. No experimental animals were used in this study. Monitoring the cell-surface presentation of galectins was carried out by flow cytofluorometry using fluorescent goat anti-rabbit IgG with 20 lgÆmL)1 galectin-type-specific IgG fractions and FACS equipment (Becton-Dickinson, Heidelberg, Germany) [9]. Cell culture Human Capan-1 pancreatic carcinoma cells and clones with stable vector-directed p16INK4a expression were established and cultured as described previously [3]. FEBS Journal 277 (2010) 3552–3563 ª 2010 The Authors Journal compilation ª 2010 FEBS 3559 p16INK4a downregulates anti-anoikis effector Gal-3 H. Sanchez-Ruderisch et al. Antibodies INK4a Antibodies to PCNA and p16 were obtained from Santa Cruz Biotechnology (Santa Cruz, CA, USA) and NeoMarkers (Fremont, CA, USA), respectively. Protein extraction and western blotting Cells were lyzed in radioimmunoprecipitation assay buffer (50 mm Tris ⁄ HCl at pH 7.5, 0.15 m NaCl, 0.25% SDS, 0.05% sodium deoxycholate, 1% NP-40, 1 mm dithiothreitol, 1 lgÆmL)1 aprotinin, 2 mm leupeptin, 1 mm Na3VO4, 1 mm NaF, 1 mm phenylmethylsulfonyl fluoride), and sonicated on ice. Aliquots (5–10 lg) were subjected to SDS ⁄ PAGE and electroblotted onto poly(vinylidene fluoride) membranes (NEN, Cologne, Germany). Blots were incubated overnight at 4 C with the respective antibodies (diluted 1 : 1000 in 5% non-fat dried milk in phosphate buffered saline with 0.5% Tween-20). Immunoreactive bands were visualized by enhanced chemoluminescence (NEN). Cell processing, blotting and signal generation when using 2D gel electrophoresis followed the procedure previously used to detect Gal-1 [9]. Immunohistochemistry Immunohistochemistry was performed on cryosections with antibodies controlled for specificity and lack of intergalectin cross-reactivity, as described previously [3,56,57]. Briefly, sections were fixed in 4% paraformaldehyde and primary antibodies were applied at a dilution of 10 lgÆmL)1 (Gal-3) or 1.25 lgÆmL)1 (p16INK4a). Immunoreactivity was detected with biotinylated secondary antibodies and avidin–biotin– peroxidase complex, with 3-amino-9-ethylcarbazole as the chromogenic substrate. Sections were counterstained with hemalaun. gency of 0.1· NaCl ⁄ Cit, 0.1% SDS and exposed to X-ray films at –70 C. Nuclear run-off Blots were prepared using isolated and denatured cDNA fragments immobilized on Hybond N+ nylon membranes (Amersham Pharmacia). The amount of denatured cDNA for Gal-1, GAPDH and b-actin blotted was 5, 2 and 1 lgÆslot)1, respectively. Nuclear RNA preparation, labeling and hybridization were performed as described previously [3]. Blots were washed twice for 10 min at 42 C with 40 mm NaH2PO4 ⁄ Na2HPO4 pH 7.2, 1% SDS. Labeled de novo mRNA transcripts were detected on autoradiographs. Induction and detection of anoikis For determination of anoikis, 2 · 105 cells were cultured as suspension cultures in plates coated with poly(2-hydroxyethyl methacrylate) (poly-HEMA) (Sigma, Deisenhofen, Germany) for the indicated times. Apoptotic cells were then quantitated from the pre-G1 fraction in cell cycle analyses as described [3]. Stable transfection of Gal-3 sense/antisense cDNA Full-length cDNA obtained from amplification of human Gal-3-specific mRNA of human DLD-1 colon carcinoma cells in either the sense (pcDNA–Gal-3S) or antisense (pcDNA–Gal-3AS) orientation was subcloned into the pcDNA3.1 vector, and the Effectene Transfection Reagent (Quiagen, Hilden, Germany) was used to generate stably transfected cells following the manufacturer’s protocol. Determination of binding of labeled galectins Northern blot analysis Total RNA from 108 cells was isolated using RNAzolB (WAK-Chemie Medicals GmbH, Bad Homburg, Germany), and the poly-(A+) fraction was purified using the PolyATrack System 1000 (Promega, Mannheim, Germany) according to the manufacturer’s protocol. Aliquots were separated on a 1% agarose ⁄ 3-(N-morpholino)propanesulfonic acid ⁄ formaldehyde gel, blotted to Hybond N+ filters (Amersham Pharmacia, Freiburg, Germany) and linked to the membrane using UV light. Full-length cDNA preparations for human Gal-3 and glyceraldehyde-3-phosphate dehydrogenase (GAPDH) were labeled with [32P]dCTP[aP] by random priming (Megaprime DNA labeling kit; Amersham Pharmacia). Residual nucleotides were removed and hybridization was carried out in Quick-Hyb buffer (Stratagene, La Jolla, CA, USA) at 65 C for 2 h. Following hybridization, membranes were washed at 65 C to a strin- 3560 Cells were incubated with 100 lgÆmL)1 of biotinylated Gal-1 and -3, washed and surface-bound probe was detected by flow cytometry using an indocarbocyanine– streptavidin conjugate. Fluorescence intensity was recorded on a FACSCalibur (Becton Dickinson) and analyzed with cellquest software [9,58]. Cells incubated with the fluorescent indicator only were used to determine the background fluorescence. Determination of caspase-8 activity Cells with activated caspase-8 were detected using the carboxyfluorescein-labeled derivative of the caspase-8 inhibitor Z-LETD-FMK (FAM-LETD-FMK) (Biocarta, Hamburg, Germany), which irreversibly binds to activated caspase-8. Fluorescence intensity was evaluated by flow cytometry. FEBS Journal 277 (2010) 3552–3563 ª 2010 The Authors Journal compilation ª 2010 FEBS p16INK4a downregulates anti-anoikis effector Gal-3 H. Sanchez-Ruderisch et al. Statistical analysis Unless indicated, unpaired Student’s t-test analyses (twotailed distribution, two-sample unequal variance) were performed using prism software (Prism, San Diego, CA, USA). Data were considered significant at P-values < 0.05. 10 Acknowledgements This work is dedicated to Prof. Dr Stefan Rosewicz (1960–2004), who was crucial to start this project line. We are grateful to Drs B. Friday, G. Ippans and S. Namirha for helpful comments, to L. Mantel for excellent technical assistance as well as to the Dr Mildred Scheel Stiftung, Sonnenfeld Stiftung, the Wilhelm-Sander-Stiftung, LMUexcellent program, the Verein zur Förderung des biologisch-technologischen Fortschritts in der Medizin e.V. and the EC program on Marie Curie Research Training Networks (contract no. MRTNCT-2005-019561) for generous financial support. 11 12 13 14 References 1 Sherr CJ (2001) The INK4a ⁄ ARF network in tumour suppression. Nat Rev Mol Cell Biol 2, 731–737. 2 Kim WY & Sharpless NE (2006) The regulation of INK4 ⁄ ARF in cancer and aging. Cell 127, 265–275. 3 Plath T, Detjen K, Welzel M, von Marschall Z, Murphy D, Schirner M, Wiedenmann B & Rosewicz S (2000) A novel function for the tumor suppressor p16INK4a: induction of anoikis via upregulation of the a5b1 fibronectin receptor. J Cell Biol 150, 1467–1478. 4 Nakagawa H, Zheng M, Hakomori SI, Tsukamoto Y, Kawamura Y & Takahashi N (1996) Detailed oligosaccharide structures of human integrin a5b1 analyzed by a three-dimensional mapping technique. Eur J Biochem 237, 76–85. 5 Sharon N & Lis H (1997) Glycoproteins: structure and function. In Glycosciences: Status and Perspectives (Gabius H-J & Gabius S eds), pp 133–162. Chapman & Hall, Weinheim, Germany. 6 Reuter G & Gabius H-J (1999) Eukaryotic glycosylation: whim of nature or multipurpose tool? Cell Mol Life Sci 55, 368–422. 7 Bellis SL (2004) Variant glycosylation: an underappreciated regulatory mechanism for b1 integrins. Biochim Biophys Acta 1663, 52–60. 8 Honke K & Taniguchi N (2009) Animal models to delineate glycan functionality. In The Sugar Code. Fundamentals of Glycosciences (Gabius H-J ed), pp 385– 401. Wiley-VCH, Weinheim, Germany. 9 André S, Sanchez-Ruderisch H, Nakagawa H, Buchholz M, Kopitz J, Forberich P, Kemmner W, Böck C, 15 16 17 18 19 20 Deguchi K, Detjen KM et al. (2007) Tumor suppressor p16INK4a: modulator of glycomic profile and galectin-1 expression to increase susceptibility to carbohydratedependent induction of anoikis in pancreatic carcinoma cells. FEBS J 274, 3233–3256. Reuter G & Gabius H-J (1996) Sialic acids: structure– analysis–metabolism–occurrence–recognition. Biol Chem Hoppe Seyler 377, 325–342. Patsos G & Corfield A (2009) O-Glycosylation: structural diversity and functions. In The Sugar Code. Fundamentals of Glycosciences (Gabius H-J ed), pp 111–137. Wiley-VCH, Weinheim, Germany. Zuber C & Roth J (2009) N-Glycosylation. In The Sugar Code. Fundamentals of Glycosciences (Gabius H-J ed), pp. 87–110. Wiley-VCH, Weinheim, Germany. Gabius H-J (2006) Cell surface glycans: the why and how of their functionality as biochemical signals in lectin-mediated information transfer. Crit Rev Immunol 26, 43–79. Villalobo A, Nogales-González A & Gabius H-J (2006) A guide to signaling pathways connecting protein–glycan interaction with the emerging versatile effector functionality of mammalian lectins. Trends Glycosci Glycotechnol 18, 1–37. Gabius H-J, ed. (2009) The Sugar Code. Fundamentals of Glycosciences. Weinheim: Wiley-VCH, Germany. Ahmad N, Gabius H-J, Kaltner H, André S, Kuwabara I, Liu F-T, Oscarson S, Norberg T & Brewer CF (2002) Thermodynamic binding studies of cell surface carbohydrate epitopes to galectins-1, -3, and -7: evidence for differential binding specificities. Can J Chem 80, 1096– 1104. Wu AM, Singh T, Wu JH, Lensch M, André S & Gabius H-J (2006) Interaction profile of galectin-5 with free saccharides and mammalian glycoproteins: probing its fine specificity and the effect of naturally clustered ligand presentation. Glycobiology 16, 524–537. André S, Sansone F, Kaltner H, Casnati A, Kopitz J, Gabius H-J & Ungaro R (2008) Calix[n]arene-based glycoclusters: bioactivity of thiourea-linked galactose ⁄ lactose moieties as inhibitors of binding of medically relevant lectins to a glycoprotein and cell-surface glycoconjugates and selectivity among human adhesion ⁄ growth-regulatory galectins. ChemBioChem 9, 1649–1661. André S, Kožár T, Kojima S, Unverzagt C & Gabius H-J (2009) From structural to functional glycomics: core substitutions as molecular switches for shape and lectin affinity of N-glycans. Biol Chem 390, 557–565. Lahm H, André S, Hoeflich A, Kaltner H, Siebert H-C, Sordat B, von der Lieth C-W, Wolf E & Gabius H-J (2004) Tumor galectinology: insights into the complex network of a family of endogenous lectins. Glycoconj J 20, 227–238. FEBS Journal 277 (2010) 3552–3563 ª 2010 The Authors Journal compilation ª 2010 FEBS 3561
This site is protected by reCAPTCHA and the Google Privacy Policy and Terms of Service apply.