Skip to main content
Erschienen in: BMC Cancer 1/2011

Open Access 01.12.2011 | Research article

Nck2 promotes human melanoma cell proliferation, migration and invasion in vitro and primary melanoma-derived tumor growth in vivo

verfasst von: Mélissa Labelle-Côté, Julie Dusseault, Salma Ismaïl, Aude Picard-Cloutier, Peter M Siegel, Louise Larose

Erschienen in: BMC Cancer | Ausgabe 1/2011

Abstract

Background

Nck1 and Nck2 adaptor proteins are involved in signaling pathways mediating proliferation, cytoskeleton organization and integrated stress response. Overexpression of Nck1 in fibroblasts has been shown to be oncogenic. Through the years this concept has been challenged and the consensus is now that overexpression of either Nck cooperates with strong oncogenes to transform cells. Therefore, variations in Nck expression levels in transformed cells could endorse cancer progression.

Methods

Expression of Nck1 and Nck2 proteins in various cancer cell lines at different stages of progression were analyzed by western blots. We created human primary melanoma cell lines overexpressing GFP-Nck2 and investigated their ability to proliferate along with metastatic characteristics such as migration and invasion. By western blot analysis, we compared levels of proteins phosphorylated on tyrosine as well as cadherins and integrins in human melanoma cells overexpressing or not Nck2. Finally, in mice we assessed tumor growth rate of human melanoma cells expressing increasing levels of Nck2.

Results

We found that expression of Nck2 is consistently increased in various metastatic cancer cell lines compared with primary counterparts. Particularly, we observed significant higher levels of Nck2 protein and mRNA, as opposed to no change in Nck1, in human metastatic melanoma cell lines compared with non-metastatic melanoma and normal melanocytes. We demonstrated the involvement of Nck2 in proliferation, migration and invasion in human melanoma cells. Moreover, we discovered that Nck2 overexpression in human primary melanoma cells correlates with higher levels of proteins phosphorylated on tyrosine residues, assembly of Nck2-dependent pY-proteins-containing molecular complexes and downregulation of cadherins and integrins. Importantly, we uncovered that injection of Nck2-overexpressing human primary melanoma cells into mice increases melanoma-derived tumor growth rate.

Conclusions

Collectively, our data indicate that Nck2 effectively influences human melanoma phenotype progression. At the molecular level, we propose that Nck2 in human primary melanoma promotes the formation of molecular complexes regulating proliferation and actin cytoskeleton dynamics by modulating kinases or phosphatases activities that results in increased levels of proteins phosphorylated on tyrosine residues. This study provides new insights regarding cancer progression that could impact on the therapeutic strategies targeting cancer.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1471-2407-11-443) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

MLC participated to most experiments and contributed to the analysis of the data. JD performed experiments to generate Figure 8 and has been essential to the study design and the draft the manuscript. SI participated in the cellular and biochemical assays. APC carried out wound healing assays experiments reported in Additional file 3. PMS contributed to the conception and achievement of in vivo experiments. LL conceived the study, participated in its design and coordination, and to the preparation of the final manuscript. All authors read and approved the final manuscript.

Background

Melanoma skin cancer is one of the most devastating types of cancer, extremely aggressive with high metastatic potential. Melanoma metastasis to distant organs is the primary cause of human cancer-related deaths. Worldwide, the incidence of cutaneous malignant melanoma is increasing faster than any other type of cancer. Cutaneous melanoma originates from pigment-producing melanocytes localized at the epidermal-dermal junction in human skin and develops through different steps [1]. Among various hypotheses, it is proposed that these involve radial (RGP) and vertical (VGP) aberrant growth phases of preexisting nevi or at new site. Then to metastasize at distant sites, melanoma detach from a primary lesion, acquire motility and proteolytic activities to reach lymphatic and blood circulation and undergo growth to distinct organs, all this according to stepwise molecular changes involving defined genetic events [2, 3]. However, the exact mechanisms underlying this devastating process are complex and somehow still poorly understood. From a molecular point of view, oncogenic activation of the mitogen-activated protein kinase (MAPK) pathway, due to somatic mutations in B-RAF (V600E), is frequently observed in melanoma (70%) [4].
In mammals, the family of Nck (non-catalytic region of tyrosine kinase) proteins is represented by two highly conserved members, Nck1 and Nck2, composed of three N-terminal SH3 (Src homology 3) domains followed by a unique C-terminal SH2 (Src homology 2) domain and devoid of any catalytic activity [5, 6]. Like other SH2/SH3 domain-containing proteins, Nck1 and Nck2 behave as adaptor proteins by physically coupling activated membrane receptors to specific downstream effectors [7]. In mice, individual Nck knockout resulted in no phenotype, confirming redundancy of Nck proteins, while early embryonic lethality of the double Nck knockout mice revealed their crucial role in embryonic development [8]. However, regardless that Nck1 and Nck2 share high amino acid identity, and common cellular functions and binding partners, increasing evidence support specific roles and proteins interactions, as well as tissue expression patterns for these adaptors [7, 915]. Previous studies have reported that overexpression of Nck1 in fibroblasts induces cellular transformation and that these cells form tumors in mice [16, 17]. Furthermore, either Nck has been shown to cooperate with potent oncogenes (v-Abl and Ras) to transform cells, influence cell morphology and anchorage-independent growth [6]. Although, these studies strongly suggest a role for Nck in cancer development, the mechanism by which Nck oncogenic potential is achieved still remains to be established.
Originally the Nck1 cDNA was isolated from a human melanoma cDNA expression library using a monoclonal antibody produced against the human melanoma-associated antigen [5], which has no similarity with Nck1. This suggests that the Nck1 mRNA might be abundant in human melanoma. Most recently, the Nck2 gene was found as being overexpressed in human metastatic melanoma compared with non-metastatic melanoma lesions [18]. In agreement, the cancer microarray database Oncomine (https://​www.​oncomine.​org/​) reports Nck2 as a gene upregulated in several human cancer cell lines, including human melanoma. Therefore, the concept that deregulated expression of Nck adaptor proteins could contribute to promote melanoma development and/or progression deserves further investigation. In the present study, using human melanoma cell lines harboring the activating B-RAF (V600E) mutation, that are well defined for stage of cancer progression [19, 20], we demonstrate that Nck2 protein and mRNA levels are increased in human metastatic melanoma cells compared with human primary melanoma cells that rarely metastasis. We show that Nck2 promotes cell proliferation, migration and invasion in human melanoma cells. In addition, using an in vivo xenograft model, we provide evidence that increased Nck2 expression in human primary melanoma cells promotes melanoma-derived tumor growth rate. Collectively, our findings indicate that Nck2 plays a role in human melanoma progression.

Methods

Cell lines

The Wistar melanoma cell lines (WM278, WM1232, WM115, 1205Lu, WM164, WM1617 and 451Lu) were obtained from Dr Meenhard Herlyn (PA, USA). Human Epidermal Melanocytes (HEM) cell line was purchased from Cell Applications Inc. Murine colon carcinoma cell (CT26, CT36 and CT51) were obtained from Dr. Nicole Beauchemin (McGill University, Montreal, QC). Breast cancer cell lines (MCF10, MCF7, T47D, MDA-MB-231) were kindly provided by Dr. Morag Park (McGill University, Montreal, Qc).

Cell culture

Unless specified, all chemicals used in this study are from regular commercial sources. Cells were maintained at 37°C in 5% CO2-95%O2 atmosphere. HEK293, colon and breast cancer cell lines were grown in DMEM (Dulbecco's modified Eagle's medium) containing 10% FBS and supplemented with 100 Units/ml of penicillin, 100 μg/ml of streptomycin and 0.25 μg/ml of Amphotericin B. Melanoma cell lines were grown in RPMI 1640 supplemented with 2 mg/ml NaHCO3 and 0.3 mg/ml glutamine. MCF10 cells were grown in DMEM containing 5% Horse Serum (Invitrogen), 20 μg/ml of mouse epidermal growth factor (mEGF, Collaborative Biomedical Products), 10 μg/ml of insulin and 0.1 μg/ml of cholera toxin. To induce MCF10 cell differentiation, cells were grown in media in absence of mEGF but supplemented with 0.5 μg/ml of hydrocortisone for two days. HEM (human epidermal melanocyte) cells were grown in HEM media (Cell Applications Inc.) and cultured according to the manufacturer's instructions.
To analyze phospho-tyrosine proteins, cells were exposed to protein phosphotyrosine phosphatase inhibitor (pervanadate (Na3VO4) or bpVPhen, 100 μM, 15 min at 37°C) prior to be harvested and total cell lysates processed for anti-phosphotyrosine western blot as reported below. Alternatively, total cell lysates (2 mg protein) were incubated with indicated antibodies (4 μg) for 2 hours at 4°C and 40 μl of 50% slurry solution of Protein-A immobilized on Sepharose beads (Santa Cruz Biotech.) were added for an additional 2 hours of incubation at 4°C. Immunoprecipitated samples were washed 3X with lysis buffer before to be finally recovered in Laemmli buffer and processed for anti-phosphotyrosine western blot as reposted below.

Antibodies

Nck polyclonal antibodies were raised by immunizing rabbits with GST-Nck fusion proteins as antigens. Crude serum samples were Protein-A-purified (ProChem, MA) and further tested for Nck specificity as described below. A pan-Nck antibody (1793), which recognizes both Nck isoforms was raised against residues 1-251 containing the three SH3 domains of human Nck1 as previously reported [21]. Nck1 antibody (2383) and Nck2 antibody (3313) were raised against isoform specific amino acid sequence in between the last SH3 and the SH2 domain of each Nck as reported earlier [15]. Other antibodies used are: CrkII (C-18), Integrin β3 (H-96), phospho-tyrosine (clone PY99), HA (Y-11) and GFP (B-2) from Santa Cruz Biotech. Antibodies against Integrin β1 (anti-CD29, clone 18), E-Cadherin (clone 34) and N-Cadherin (clone 32) were purchased from BD (ON, Canada). Antibodies to detect vinculin (clone h-VIN-1) and Tubulin (TUB2.1) were from Sigma-Aldrich, USA. Secondary antibodies coupled to HRP were from Bio-Rad Inc. Rhodamine-coupled to mouse anti-IgG was bought at Jackson ImmunoResearch Inc. Phalloidin-coupled to AlexaFluor®555 and 488 were purchased from Molecular Probes (Invitrogen, CA, USA)

Cell lysis and western blots

Cell lysates were prepared in lysis buffer (50 mM HEPES pH 7.4, 150 mM NaCl, 10% Glycerol, 1% Triton X-100, 1.5 mM MgCl2, 1 mM EGTA, 10 mM Sodium Pyrophosphate, 100 mM Sodium Fluoride, supplemented with the protease inhibitors Aprotinin and Leupeptin at 1 μg/ml and PMSF at 1 mM. Lysates normalized for protein content (Bradford protein assay, Bio-Rad) were prepared in Laemmli buffer, heated, subjected to SDS-PAGE on 10% acrylamide gels and transferred onto nitrocellulose membranes. For western blot analyses, membranes were blocked in TBS (Tris-buffered saline) containing 10% dry milk and 0.1% Tween-20, and then incubated overnight at 4°C with indicated primary antibodies appropriately diluted in the blocking solution. For pY western blot, blocking and primary antibody solution was TBS containing 5% bovine serum albumin (BSA, Sigma) and 0.1% Tween-20. Next morning, the membranes were washed twice with TBS for 5 minutes followed by two 5 minutes washes using TBS-T (TBS-0.1% Tween-20) and two 5 minutes washes with TBS. The membranes were then incubated with secondary antibody appropriately diluted in milk-blocking solution for 1 hour and washed as above. Finally, signal was detected using ECL Plus Western Blotting Detection System (GE Healthcare, UK) and XR film exposure.

RNA isolation and RT-PCR

Total RNA was isolated from melanoma cells using the TRIZOL (Invitrogen) according to the manufacturer's protocol. Briefly, cells from 100-mm dishes (1 × 106 cells) were suspended in 7.8 ml of TRIZOL. The aqueous and organic phases were separated after addition of chloroform. Precipitated RNA by isopropyl alcohol addition was washed in 70% ethanol and dissolved in RNase-free water. RNA concentration and purity (OD260/280) was measured using an Ultrospec 2100 Pro UV/visible Spectrophotometer (Fisher Scientific, ON). First-strand cDNA synthesis was generated by reverse transcriptase reaction in a final volume of 50 μl. For this, 2.0 μg of total RNA were mixed in a total reaction volume of 20 μl of RNAse free water containing 1 μM Oligo d(T)20 for Nck amplifications or 6 μg of Random Primers for 18S amplification. The reactions were incubated at 65°C for 5 min and quenched on ice. Then, the RT reaction was assembled by adding 10 μl of the 5X 1st strand buffer (Invitrogen), 5 μl of 0.1 M DTT (Invitrogen), 2.5 μl of RNase Inhibitor (40 U/μl) (Invitrogen), 2.5 μl of 10 mM dNTPs, 5 μl of 50 mM MgSO4 and 2.5 μl of Superscript III (200 U/μl) (Invitrogen). Samples were incubated at 37°C for 50 min and deactivated at 70°C for 15 min. PCR amplification was performed using 0.5 μl of cDNA template in a final volume of 50 μl containing 5 μl of 10X PCR Enhancer buffer (Invitrogen), 1.5 mM MgCl2, 0.2 mM dNTPs, 50 pM of specific forward and reverse primers, 10 μl of Amplification buffer (Invitrogen) and 1 U of Taq DNA polymerase (Invitrogen) and DEPC water. Primers used were: Nck1 forward 5'-GCCAGATTCTGCATCTCCTG-3', Nck1 reverse 5'-ACACTTGCCCAGTATTTAGG-3', Nck2 forward 5'-CGAGTACCCCGCCAATGG-3' and Nck2 reverse 5'-CCCGTCACTGAGGACCACC-3'. Reactions were carried out on PTC-100 Programmable Thermal Controller (MJ Research Inc.) according to the following program conditions: initial denaturation at 94°C for 1 min, followed by 1 min at 94°C, 30 seconds of annealing (47°C for Nck 1, 53°C for Nck 2 and 55°C for 18S) and 1 min at 72°C. The final elongation step was 10 minutes at 72°C and the samples were kept at 4°C until analysis. PCR products were separated on a 1% agarose gels and imaged using an NIH Image J 1.30 system. Fifteen, 20, 25 and 30 cycles of PCR amplification products were analyzed to confirm that the amplification was in the linear range for each gene. Ratios of Nck1 and Nck2 over 18S were calculated from similar assays performed in triplicate and repeated three times.

Cell transfection

Human HA-tagged Nck2 cDNA generously provided by Dr. Wei Li (University of Southern California, LA, CA) was subcloned into the retroviral vector pLXSN (Clonetech Laboratories Inc., CA, USA) and the viral particles produced using the GP2-293 cell line according to the manufacturers' instructions. Human Nck2 cDNA was also subcloned into the pEGFP-C1 plasmid (BD, NJ, USA). To establish stable clones of human WM278 primary melanoma overexpressing GFP or GFP-Nck2, cells plated in 100-mm dishes (1 × 106 cells) were transiently transfected with 10 μg of plasmid DNA (pEGFP or pEGFP-Nck2) using calcium phosphate and following selection with neomycin, clones were isolated, amplified and analyzed for GFP or GFP-Nck2 by western blot. 451Lu cells plated at 40-60% confluence were transfected with either 100 nM Nck2 or control siRNA 13379 (Ambion, Austin, TX) using Lipofectamine Plus reagent according to the manufacturer's protocol and analyzed for protein expression after 24 or 48 h.

Proliferation assays

Briefly, cells (4 × 103) were seeded in 96-well plates and 24, 48, 72 or 96 h after, cells were fixed by adding glutaraldehyde (20 min, final concentration 1%). Then, fixed cells were washed twice with deionised water and stained with Crystal Violet (20 min, 0.4% in 10% ethanol, Sigma). The excess of Crystal Violet was removed by washing the cells three times with water and finally, incorporated Crystal Violet was dissolved in 10% acetic acid and read at 570 nm using a spectrophotometer (Beckman Coulter). Wells without cells, but containing medium were used as blank value that was subtracted from all values. Data were expresses a raw OD at 570 nm or as ratio of OD at specific time point over initial OD at day 1.

Wound healing assays

Cell migration was assessed in classical wound healing assays. Confluent monolayer cells in a 6-well plate were wounded using a plastic pipette tip (P200) and rinsed with PBS before to add back culture medium. The bottoms of the wells were marked to indicate where the initial pictures of the wound area were taken. After 8 h incubation at 37°C, pictures (10X) of the same areas were recorded using an Axiovert 200 M microscope (Zeiss) equipped with a CoolSnap™ES camera (Photometric®, Roper Scientific) and closure of the wound evaluated using Metamorph® (V6.3, Molecular Devices Corp.).

Cell invasion assays using Transwells

Melanoma cells (1 × 105) resuspended in 10% serum containing medium were added to the top chamber of a Transwell (8 μm, DD Biosciences, NJ, USA) pre-coated with matrigel™ (BD Biosciences, NJ, USA) diluted in ice-cold PBS (175 μg/ml) at a total of 35 μg per well and allowed to migrate for 24 h. To evaluate the amount of cells that had invaded through each transwell, excess of media was discarded and the transwells were washed once with PBS and then placed in trypsin solution (0.025%) to release the invaded cells underside of the transwells and in the bottom chamber. Total invaded cells were estimated using Calcein AM (BD Biosciences, NJ, USA) as recommended by the manufacturer. Data were normalized according to the respective total amount of cells for each line plated at the same time in adjacent wells devoid of transwells to take into account variations in cell number between cell lines.

Spheroid formation assays

Spheroid formation and culture in 3D were performed according to the hanging drop method [22]. Briefly, 2 × 104 cells in 20 μl of culture medium were suspended on the lid of tissue culture dishes containing 10 ml of culture medium for 48 h to form spheroids. Then spheroids were transferred in culture dishes containing culture medium and on 2% agar (Agar Select, Invitrogen, CA, USA) at the bottom. After 72 h of growth in suspension, individual spheroid has been transferred in 4-well plate containing 80% collagen type IV (PureCol®, Advanced BioMatrix) in RPMI without FBS. Following 30 min at 37°C to allow collagen polymerization, 500 μl of RPMI containing 10% FBS was added to each well. Images were recorded initially and at 12-24 hr intervals as reported above for wound healing assays. Spheroid invasion was determined qualitatively as either positive or negative comparing sequential images.

Actin and focal adhesions

Cells were plated at 3 × 104 cells/well on glass coverslips pre-coated or not with various extracellular matrices and incubated in culture medium for 24 h. All steps were carried at room temperature and coverslips were rinsed with PBS between each step. Cells were fixed in freshly prepared 3.7% formaldehyde for 10 min, permeabilized in 0.2% Triton-X-100 for 5 min and blocked in 0.1% BSA for 30 min. For vinculin staining, cells were incubated with primary monoclonal anti-vinculin antibody (1:400) for 1 h and with a mixture of secondary tetramethylrhodamine isothiocyanate-conjugated phalloidin-conjugated goat mouse antibody (TRITC-GAM, Sigma) for 30 min. Actin staining was performed by incubating the coverslips for 30 min with Phalloidin-AlexaFluor®555. Coverslips were mounted by inverting them on glass slides using Prolong anti-fade mounting media (Molecular Probes). Coverslips were examined on a Zeiss Axiovert 200 M microscope (Zeiss) using 40X or oil immersion 63× objective lens. Fluorescent images were captured using a CoolSnap™ES camera (Photometric®, Roper Scientific) and analyzed using Metamorph® (V6.3, Molecular Devices Corp.).

Tumor growth in vivo

WM278 primary melanoma cells either parental, overexpressing GFP (C2) or GFP-Nck2 at low (N7) or high (N14) levels were grown in RPMI medium supplemented with 10% FBS to 80% confluency. Cells (5 × 106) resuspended in 500 μl at 50% Matrigel™ were injected subcutaneously in the right flank of 6-week-old CD-1 Nude mice (Charles River, Qc, Canada) (n = 5 for each group). Tumors development was followed for 20 weeks. Tumor size was measured every week with calipers to assess tumor volume ([πlength × width2]/6). Mice were housed in McGill University Animal facilities at the Genome building. Mice experiments were conducted under a McGill University-approved animal use protocol (Dr. P.M. Siegel) in accordance with guidelines established by the Canadian Council on Animal Care.

Data analysis and statistics

Densitometry analysis results are expressed as means ± S.E.M. Student's t test was used to evaluate the statistical significance of the results. A p ≤value 0.05 is assumed to be significant.

Results

Nck2 protein and mRNA levels are increased in human metastatic melanoma cell lines

To investigate the potential involvement of Nck proteins in human melanoma development and progression, we first analyzed total Nck protein levels in human melanoma cell lines at different stages of cancer progression and compared with normal human melanocytes. The human melanoma cell lines used in this study were provided by the laboratory of Dr. Meenhard Herlyn at the Wistar Institute (PA, USA) and already used in vivo for tumorigenicity and experimental metastasis [23, 24]. Mainly, these include the WM278, a melanoma cell line derived from a human primary tumor in vertical growth phase that rarely metastasis; WM1617, a WM278 sister melanoma cell line derived from lymph nodes metastasis in the same patient few years later; 451Lu, a melanoma cell line isolated from lung metastasis in mice injected with the WM164 cell line, which is a human melanoma cell line isolated from lymph nodes metastasis similar to WM1617, but from a different patient.
From western blots performed using a rabbit polyclonal antibody that equally recognizes both Nck isotypes (Pan-Nck, Additional file 1) [21], we observed higher levels of Nck proteins in highly metastatic melanoma (WM164, 451Lu, 1205Lu and WM1232) compared with weakly metastatic primary melanoma (WM115 and WM278) and normal melanocyte (FW2294) cell lines (Figure 1A). Further analyses using in-house generated Nck isoforms specific antibodies (Additional file 1) [15] revealed that increased expression of Nck in metastatic melanoma cells is mainly due to drastic higher expression levels of Nck2 (Figure 1B, compare WM164 and 451Lu with WM278). Additional comparison of highly metastatic (WM1617) and weakly metastatic (WM278) human melanoma cell lines isolated from the same patient (Figure 1B), further confirmed increased expression of Nck2 in human metastatic melanoma. Interestingly, Nck1 protein levels normalized according to actin or tubulin loading control were comparable among the human melanoma cell lines investigated (Figure 1B and Additional file 2). In addition, we also found no change in expression levels of other SH2/SH3 domain-containing signaling proteins, such as PLC-γ1, p85 of PI3K, Grb2 and Crk, as normalized for actin or tubulin loading control (Additional file 2 and data not shown). Altogether, these results suggest a specific role for Nck2 in human melanoma progression.
To assess whether increased expression of Nck2 protein levels in human metastatic melanoma cells correlates with upregulated transcription of Nck2 gene, we compared Nck1 and Nck2 mRNA levels in three human melanoma cell lines at different stages of progression and in human primary melanocytes (HEM) by performing RT-PCR using Nck isoforms specific primers. In the linear range of PCR amplification, no significant change in Nck1 mRNA levels was detected in all human melanoma cell lines and compared with HEM (Figure 2A). In contrast, we observed a strong increase in Nck2 mRNA levels in all human melanoma cell lines compared with HEM. In addition, compared with primary melanoma cells (WM278), metastatic melanoma cell lines (WM164 and 451LU) showed significant increased Nck2 mRNA levels (Figure 2B). Altogether, our results reveal that Nck2 protein and mRNA levels are significantly increased in different human metastatic melanoma cells compared with human weakly metastatic primary melanoma and melanocyte cells, suggesting Nck2 as a biological marker of human melanoma metastasis that could contribute to melanoma progression.

Nck2 promotes human melanoma cell proliferation

To demonstrate a role for Nck2 in melanoma progression, we first determined whether Nck2 regulates cell proliferation in WM278 human primary melanoma cells, which endogenously express low levels of Nck2 protein and rarely metastasis compared with its metastatic counterpart WM1617 melanoma cells. For this, we created WM278 cell lines stably overexpressing increasing levels of GFP-Nck2 (N15 < N7 < N14) or GFP as control (C2) (Figure 3A). Using these WM278 stable cell lines, we found that overexpressing high levels of Nck2 significantly enhanced cell proliferation (Figure 3B, left panel, compare clone N14 with C2). It is interesting to note though that the effect of Nck2 on WM278 primary melanoma cells proliferation seems to parallel the levels of Nck2 overexpressed (Figure 3A and 3B, compared N15, N7 and N14). In agreement, the WM1617 human metastatic melanoma cells that endogenously express higher levels of Nck2 compared with human primary melanoma cells, also show higher proliferative abilities than its paired WM278 primary melanoma cells that rarely metastasis (Figure 3C). As expected, we found no change in the protein levels of Nck1 or CrkII, a SH2-SH3 domain-containing adaptor protein previously identify as an oncogene [25] and recently reported to regulate sarcoma cell proliferation [26] (Figure 3C).
To confirm a role for Nck2 in melanoma cell proliferation, we assessed whether siRNA-mediated downregulation of Nck2 in human metastatic melanoma cells affects cell proliferation. As shown in Figure 4A, Nck2 siRNA transfection of 451Lu metastatic melanoma cells resulted in decreased Nck2 protein and mRNA levels, while Nck1 protein and mRNA levels were not altered. More importantly, we found that cell proliferation was significantly decreased in Nck2 depleted metastatic melanoma cells compared with control siRNA treated cells (Figure 4B). To rule out that this effect was due to increased cell death in Nck2 depleted melanoma cells, 2 days post transfection we evaluated the percentage of cells with nuclei stained by Trypan Blue and observed no difference between metastatic melanoma cells transfected with control (8.7% ± 2.5) and Nck2 (7.7% ± 1.7) siRNA. Altogether, these results indicate that Nck2 contributes to the control of proliferation in human melanoma cells.

Nck2 modulates migration and invasion of human melanoma cells

Cancer progression involves that transformed cells must acquire motility and invasive activities. Therefore, we next determined whether Nck2 was critical to melanoma cell migration and invasion. We compared migration of WM278 primary melanoma cells overexpressing GFP (C2) or increasing levels of GFP-Nck2 (N15 < N7 < N14) in wound healing assays. As shown in Figure 5, increasing levels of GFP-Nck2 in WM278 melanoma cells promoted migration and this was significant in cell line N14, which expresses higher levels of Nck2 proteins compared with N5 and N7 cell lines (Figure 3A). To exclude that a clonal effect is responsible of increased migration of WM278 melanoma cells overexpressing GFP-Nck2, we transiently overexpressed HA-Nck2 in WM278 primary melanoma cells using retroviral infection (Additional file 3). In this context, we still observed a significant increase in migration of WM278 human primary melanoma cells overexpressing HA-Nck2 compared to control infected WM278 melanoma cells in wound healing assays. It is interesting to note though that the effect of Nck2 on migration was already observed 8 hours after wounding, suggesting that cell proliferation is not involved.
To determine whether overexpression of Nck2 in primary melanoma cells promotes invasion in a tumor-like context, we evaluated migration of melanoma cells at the edge of multicellular spheroids embedded into a collagen type I matrix (Figure 6A). As expected, spheroids of human primary melanoma cells overexpressing or not GFP (WM278, C2) grew as compact units devoid of cells migrating outward after 48 h in culture. In contrast, WM1617 human metastatic melanoma cells after 48 h in culture formed open-wave fragile spheroids with cells in periphery sending long projections invading the surrounding collagen. These observations are in agreement with the established invasive phenotype of WM1617 melanoma cells. Interestingly, WM278 human primary melanoma cells overexpressing GFP-Nck2 (N14) apparently did not show similar extensions from spheroid border cells, but form less compact spheroids than WM278-GFP with individual cells that detached form the mother spheroid after 48 h of culture into collagen gel. This observation reveals that overexpression of Nck2 in primary melanoma cells may contribute to invasiveness by promoting cell detachment and migration from primary melanoma lesion in vivo. In agreement, we found that Nck2 overexpression significantly promoted primary melanoma cells invasion through Matrigel™ matrix in transwells assays (Figure 6B). Altogether, these results suggest that Nck2 promotes cell migration and invasion in human melanoma cells.

Nck2 modulates focal adhesions in human melanoma cells

Because Nck adaptor proteins play an important role in regulating actin cytoskeleton reorganization, we then compared actin staining in WM278 human primary melanoma cells expressing either GFP (C2) or increasing levels of GFP-Nck2 (N15 < N7 < N14) (Figure 7A). Regardless of Nck2 expression levels, we found no apparent change in actin staining in these cells. This suggests that overexpression of Nck2 has no major effect on actin polymerization and organization, as well as on overall cell morphology in human primary melanoma. In contrast, vinculin staining, which shows that GFP-Nck2 colocalizes with vinculin at focal adhesions (Figure 7B), revealed significant reduced number of focal adhesions in human primary melanoma cells overexpressing Nck2 (N14) compared with control melanoma cells (C2) (Figure 7C). Therefore, these data suggest that increased expression of Nck2 in human primary melanoma cells might facilitate melanoma migration by decreasing focal adhesions.

Nck2 promotes phosphorylation of proteins on tyrosine and downregulation of cell surface adhesion proteins in human primary melanoma cells

Protein tyrosine phosphorylation is a critical mechanism regulating focal adhesion dynamics [27, 28]. Substantial evidence support a role for protein tyrosine kinases in focal adhesions assembly/disassembly toward the formation of invasive adhesion structures called invadopodia during cancer progression [2933]. To investigate the mechanism by which Nck2 overexpression impinges on the phenotype of primary melanoma cells, we compared the levels of tyrosine phosphorylated proteins between human melanoma cells expressing different levels of Nck2 protein. To evaluate protein tyrosine phosphorylation, we exposed melanoma cells to pervanadate, a potent protein tyrosine phosphatase inhibitor [3436] that allows tyrosine phosphorylated proteins to accumulate before harvesting the cells and performing immunoprecipitation. In these conditions, as observed in human metastatic (WM1617, WM164) compared with (WM278) melanoma cells (Figure 8A), WM278 cells overexpressing GFP-Nck2 presented increasing levels of proteins phosphorylated on tyrosine residues than WM278 control cells overexpressing GFP (Figure 8A). In addition, we found that tyrosine phosphorylated proteins coimmunoprecipitated with Nck2 (Nck2 IP) or total Nck (pan-Nck IP) were more abundant in human metastatic melanoma WM1617 cells compared with the counterpart WM278 primary melanoma cells (Figure 8B). We discovered also that like the metastatic WM1617 melanoma cells, the WM278 primary melanoma cells overexpressing high levels GFP-Nck2 (N14) displayed low levels of Integrin β1 and β3, as well as E- and N-Cadherin in Triton X-100 soluble extracts compared either with parental WM278 cells, WM278 cells overexpressing GFP (C2) or low levels of GFP-Nck2 (N7) (Figure 9). Collectively, these results reveal that increased expression of Nck2 in human primary melanoma cells promotes phosphorylation of proteins on tyrosine, concomitant with the assembly of Nck2-dependent pY-proteins containing molecular complexes and downregulation of cell surface adhesion proteins.

Nck2 promotes primary melanoma-derived tumor growth in vivo

To establish the biological relevance of our findings, we examined whether overexpression of Nck2 in human primary melanoma cells confers some tumorigenic advantage in a xenograft mouse model. To test this, WM278 human primary melanoma cells overexpressing GFP-Nck2 at low (N7) or high (N14) levels, along with parental WM278 cells, WM278 cells overexpressing GFP (C2) and WM1617 human metastatic melanoma cells were injected subcutaneously into CD-1 Nude mice. Two out of 5 mice injected with WM278 human primary melanoma cells overexpressing high levels of GFP-Nck2 (N14) developed tumor at the site of injection between 13-16 weeks post injection (Figure 10A). At the same time, 1 out of 5 mice that received either parental WM278 cells, WM278 melanoma cells overexpressing GFP (C2) or overexpressing low levels of GFP-Nck2 (N7) presented a subcutaneous tumor at the site of injection. In contrast, all mice injected with the metastatic WM1617 cells developed tumors at the site of injection 2-7 weeks post-injection (Figure 10A). As expected, tumors derived from metastatic WM1617 cells grew rapidly and reached maximal volume allowed between 5-11 weeks post-injection. Altogether, these observations suggest that increased expression of Nck2 in human melanoma cells is not sufficient to promote the appearance of subcutaneous tumor derived from melanoma. However, melanoma-derived tumor growth rate in mice injected with WM278 cells overexpressing GFP-Nck2 (N7 and N14) was greatly enhanced compared with tumor found in mice injected with WM278 cells parental or overexpressing GFP (C2) (Figure 10B), suggesting that increased expression level of Nck2 promotes primary melanoma-derived tumor growth rate. Subcutaneous tumors derived from WM278 cells overexpressing GFP-Nck2 (N7 and N14) could not be further monitored than few weeks after their occurrence due to the appearance of important spontaneous tumor necrosis core that required mice to be euthanized. Nevertheless, in line with our in vitro studies (Figure 3B), these results strongly support a role for Nck2 in melanoma-derived tumor growth rate in vivo.

Nck2 expression is upregulated in invasive colon and breast cancer cell lines

To find out whether increased expression of Nck2 during cancer progression is observed in other cancer types than in melanoma skin cancer, we assessed Nck isoforms protein levels in murine colon (CT) and human breast cancer cell lines at different stages of progression (Figure 11). The CT represent mouse tumorigenic colonic carcinoma cell lines established in culture from three transplantable murine tumors of colonic origin at different stages of progression [37]. Based on growth rate and metastatic spreading, CT26 is the most aggressive, while CT51 is intermediary and CT36 is the least and rarely metastasizes. Using these cells lines, we observed that Nck1 protein levels are increased in the metastatic CT26 and CT51, while just barely detected in CT36. However, when Nck1 expression levels were normalized according to actin, these variations were not statistically significant. In contrast, Nck2 that was below detection level in CT36 and CT51, was distinctly detected in CT26, revealing increased expression of Nck2 in aggressive metastatic colon cancer cells. To determine whether expression of Nck isoforms vary during breast cancer progression, we selected few of the widely used human breast cancer cell lines (MCF-7, T-47D and MDA-MB-231) and an immortalized normal human breast epithelial cell line (MCF10A). MCF-7, T-47D and MDA-MB-231 are invasive ductal carcinoma of similar origin with metastatic properties [38]. Independently of estrogen receptor expression (ER+: MCF-7 and T-47D; ER-: MDA-MB-231) or epithelial/mesenchymal phenotype (epithelial-like low invasion: MCF-7 and T-47D; mesenchymal-like high invasion: MDA-MB-231), Nck1 protein levels in breast cancer cells were not consistent with the cancer stage. Considering actin as loading control, Nck1 protein levels were significantly increased in T-47D cells, decreased in MDA-MB-231 cells and not change in MCF7 cells compared to MCF-10A cells, excluding a potential correlation between Nck1 expression levels and breast cancer progression. In contrast, Nck2 was only detected in the most aggressive MDA-MB-231 cells, which are mesenchymal-like ER+ breast cancer cells with strong migratory and metastatic abilities [38, 39]. Together these results provide the first evidence that the expression of Nck2 is increased in metastatic cancer cells of various origins and argue for a role of Nck2 in cancer progression.

Discussion

Nck1 and Nck2 SH2-SH3 domain-containing proteins have been reported to be differently expressed in numerous mouse tissues [8, 15]. In agreement with the ability of both Nck to collaborate with strong oncogenes to transform cells [6], Nck1 and Nck2 genes were found upregulated in several human cancer cell lines, including melanoma (https://​www.​oncomine.​org/​). However, Nck proteins expression levels in cancer tissues and possible mechanism(s) by which these adaptors contribute to cancer development have been poorly investigated to date. In this study, we provide evidence that Nck2 plays a role in promoting proliferation, migration and invasion of human melanoma cells in vitro and growth of melanoma-derived tumors in vivo, while its expression is upregulated in metastatic cancer cells, including colon, breast and melanoma.
Our investigation revealing that Nck2 overexpression in human primary melanoma cells induces metastatic characteristics point towards Nck2 sufficiency to promote metastasis phenotype. In this study, we did not address whether Nck2 is necessary for melanoma metastasis. However, we provided some insights suggesting that Nck2 could play such function. In fact, we found higher levels of Nck2 expression in metastatic compared to non metastatic cell lines in three different types of cancer. In addition, we demonstrate that depletion of Nck2 in metastatic melanoma reduces cell proliferation. This does not exclude that other yet identified players could be required to fully promote metastasis in melanoma overexpressing Nck2. None the less, our findings clearly demonstrate that overexpression of Nck2 in human primary melanoma correlates with upregulation of the total phospho-tyrosine proteins content, assembly of novel Nck2-dependent pY-protein complexes and downregulation of E- and N-cadherins, and β-1 and -3 integrins. E-cadherin, found at adherens junctions, is the principal effector of cell-cell adhesion [40]. Loss of E-cadherin expression in cancer cells weakens cell-cell adhesion and is associated with cancer progression, invasion and metastasis [4143]. At the present time, there is no evidence for a direct link between E-cadherin and Nck2. Further investigation is required to elucidate the molecular events responsible for E-cadherin downregulation associated with overexpression of Nck2 in human primary melanoma cells and whether downregulation of Nck2 in metastatic human melanoma cells would restore E-cadherin expression remains to be determined. On the other hand, the degree of cancer cells cohesion in primary tumor also depends on the strength of cell-ECM contacts mediated by integrins [44]. Alteration in integrins expression has been also implicated in cancer progression, invasion and metastasis [45, 46]. Integrins signaling associated with regulation of the actin cytoskeleton leading to adhesive attachment involves the activation of the focal adhesion kinase (FAK) and the integrin-like kinase (ILK) (reviewed in [47]). Interestingly, Nck2 has been shown to affect cell motility through its direct interaction with FAK [48]. Moreover, increasing evidence support a close relationship between integrins and growth factor receptor tyrosine kinases to activate signaling pathways that promote proliferation and metastatic activity (reviewed in [49]). Nck2 has been reported to function as a molecular link connecting integrins and growth factor receptor tyrosine kinases signaling pathways. In fact, Nck2 associates with numerous receptor tyrosine kinases [17, 5054] through its SH2 domain and using its third SH3 domain, it binds to a LIM domain in PINCH (Particularly Interesting Cys-His-rich Protein) [55]. PINCH, a binding protein for ILK, plays an important role in mediating integrins-induced cell-ECM interaction by directing ILK to focal adhesions [56]. It is recognized that the ILK-PINCH complex participates to signaling pathways regulating fundamental cellular processes (reviewed in [57], including cell shape and migration [58]. A crucial role for Nck in regulating these processes was particularly illustrated by the findings showing that fibroblasts derived from Nck double knockout mice embryos display major defects in cell attachment, cell motility and actin remodeling [8]. Therefore, increased expression of Nck2 in human primary melanoma cells may elicit protein interactions that re-wire signaling pathways in a fashion that alters focal adhesions and promotes cell motility by interacting with FAK and PINCH. Alternatively, increased expression of Nck2 could passively destroy proper stoichiometry of molecular complexes and in this manner, indirectly contributes to cancer progression by altering signaling pathways regulating the actin cytoskeleton network supporting cell migration.
Nck proteins are known to couple activated receptor tyrosine kinases, as well as non receptor tyrosine kinases, to effectors involved in signaling pathways regulating proliferation and actin cytoskeleton dynamics [7, 14, 5961]. Non-receptor protein kinases of the Src and Abl families are often overexpressed or aberrantly activated in a wide variety of human cancers and their roles in cancer progression, including proliferation, survival, motility, invasiveness, metastasis and angiogenesis, is significant. Of note, Nck directly binds to and promotes Abl activation and signaling [62, 63], and associates with p60v-src in vitro [16]. c-Src has been recently reported to be overexpressed in human metastatic melanoma tumors [64]. Interestingly, Src-dependent phosphorylation of Tks5 and cortactin recruits Nck to invadopodia, where it regulates actin assembly and ECM degradation [6567]. Invadopodia, exclusive invasive cancer cell membrane actin-based protrusions enriched in signaling and proteolytic activities, are used by invasive cancer cells to degrade the ECM and invade surrounding tissues [6871]. It is then possible that upregulation of tyrosine phosphorylated proteins and downregulation of cadherins and integrins in human primary melanoma cells that overexpress Nck2 may endow melanoma cells with altered adhesive properties and spatial relationships that favor uncontrolled proliferation, migration and invasion.
Nck1 and Nck2 proteins are highly identical, but despite high homology, redundant functions and common binding partners, increasing evidence suggest specific roles and protein interactions [7, 912, 14], as well as specific tissue expression patterns for Nck proteins [8, 15]. In this study, the effect of Nck1 overexpression on melanoma phenotype was not addressed. However, our results demonstrate that increased endogenous expression of Nck2 in human metastatic melanoma cells relative to primary melanoma cells and melanocytes results from increased Nck2 transcription, suggesting that Nck1 and Nck2 promoters are under different regulatory controls.

Conclusions

In conclusion, in this study we provide evidence for a role of the adaptor protein Nck2 in melanoma proliferation, migration and invasion in vitro and melanoma-derived tumor growth in vivo. Collectively, our data support Nck2 as a cornerstone governing the aspects that promote melanoma progression. Given other common metastatic cancer cell lines also overexpress Nck2, a general paradigm could make Nck2 a potential molecular marker of cancer progression and a novel target for anti-cancer drug therapy.

Acknowledgements

This work was supported by a grant to LL from the Canadian Health Research Institutes (CIHR). MLC was supported by a studentship from the CIHR. JD is postdoctoral fellow supported by the Fond de la Recherche en Santé du Québec (FRSQ). SI received support from AstraZeneca. LL is FRSQ Chercheur National. We thank Caterina Russo and Dr. Josée-France Villemure for expert experimental assistance. We also thank Lama Yamani and Dr. Nathalie Lamarche-Vane for critical reading of the manuscript.
Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://​creativecommons.​org/​licenses/​by/​2.​0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

MLC participated to most experiments and contributed to the analysis of the data. JD performed experiments to generate Figure 8 and has been essential to the study design and the draft the manuscript. SI participated in the cellular and biochemical assays. APC carried out wound healing assays experiments reported in Additional file 3. PMS contributed to the conception and achievement of in vivo experiments. LL conceived the study, participated in its design and coordination, and to the preparation of the final manuscript. All authors read and approved the final manuscript.
Anhänge
Literatur
1.
Zurück zum Zitat Larue L, Beermann F: Cutaneous melanoma in genetically modified animals. Pigment cell research/sponsored by the European Society for Pigment Cell Research and the International Pigment Cell Society. 2007, 20 (6): 485-497.CrossRef Larue L, Beermann F: Cutaneous melanoma in genetically modified animals. Pigment cell research/sponsored by the European Society for Pigment Cell Research and the International Pigment Cell Society. 2007, 20 (6): 485-497.CrossRef
2.
Zurück zum Zitat Gupta PB, Kuperwasser C, Brunet JP, Ramaswamy S, Kuo WL, Gray JW, Naber SP, Weinberg RA: The melanocyte differentiation program predisposes to metastasis after neoplastic transformation. Nature genetics. 2005, 37 (10): 1047-1054. 10.1038/ng1634.CrossRefPubMedPubMedCentral Gupta PB, Kuperwasser C, Brunet JP, Ramaswamy S, Kuo WL, Gray JW, Naber SP, Weinberg RA: The melanocyte differentiation program predisposes to metastasis after neoplastic transformation. Nature genetics. 2005, 37 (10): 1047-1054. 10.1038/ng1634.CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat Miller AJ, Mihm MC: Melanoma. The New England journal of medicine. 2006, 355 (1): 51-65. 10.1056/NEJMra052166.CrossRefPubMed Miller AJ, Mihm MC: Melanoma. The New England journal of medicine. 2006, 355 (1): 51-65. 10.1056/NEJMra052166.CrossRefPubMed
4.
Zurück zum Zitat Pollock PM, Harper UL, Hansen KS, Yudt LM, Stark M, Robbins CM, Moses TY, Hostetter G, Wagner U, Kakareka J, et al: High frequency of BRAF mutations in nevi. Nature genetics. 2003, 33 (1): 19-20. 10.1038/ng1054.CrossRefPubMed Pollock PM, Harper UL, Hansen KS, Yudt LM, Stark M, Robbins CM, Moses TY, Hostetter G, Wagner U, Kakareka J, et al: High frequency of BRAF mutations in nevi. Nature genetics. 2003, 33 (1): 19-20. 10.1038/ng1054.CrossRefPubMed
5.
Zurück zum Zitat Lehmann JM, Riethmuller G, Johnson JP: Nck, a melanoma cDNA encoding a cytoplasmic protein consisting of the src homology units SH2 and SH3. Nucleic Acids Res. 1990, 18 (4): 1048-10.1093/nar/18.4.1048.CrossRefPubMedPubMedCentral Lehmann JM, Riethmuller G, Johnson JP: Nck, a melanoma cDNA encoding a cytoplasmic protein consisting of the src homology units SH2 and SH3. Nucleic Acids Res. 1990, 18 (4): 1048-10.1093/nar/18.4.1048.CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat Braverman LE, Quilliam LA: Identification of Grb4/Nck beta, a src homology 2 and 3 domain-containing adapter protein having similar binding and biological properties to Nck. J Biol Chem. 1999, 274: 5542-5549. 10.1074/jbc.274.9.5542.CrossRefPubMed Braverman LE, Quilliam LA: Identification of Grb4/Nck beta, a src homology 2 and 3 domain-containing adapter protein having similar binding and biological properties to Nck. J Biol Chem. 1999, 274: 5542-5549. 10.1074/jbc.274.9.5542.CrossRefPubMed
7.
Zurück zum Zitat McCarty JH: The Nck SH2/SH3 adaptor protein: a regulator of multiple intracellular signal transduction events. BioEssays. 1998, 20: 913-921. 10.1002/(SICI)1521-1878(199811)20:11<913::AID-BIES6>3.0.CO;2-T.CrossRefPubMed McCarty JH: The Nck SH2/SH3 adaptor protein: a regulator of multiple intracellular signal transduction events. BioEssays. 1998, 20: 913-921. 10.1002/(SICI)1521-1878(199811)20:11<913::AID-BIES6>3.0.CO;2-T.CrossRefPubMed
8.
Zurück zum Zitat Bladt F, Aippersbach E, Gelkop S, Strasser GA, Nash P, Tafuri A, Gertler FB, Pawson T: The murine Nck SH2/SH3 adaptors are important for the development of mesoderm-derived embryonic structures and for regulating the cellular actin network. Molecular and cellular biology. 2003, 23 (13): 4586-4597. 10.1128/MCB.23.13.4586-4597.2003.CrossRefPubMedPubMedCentral Bladt F, Aippersbach E, Gelkop S, Strasser GA, Nash P, Tafuri A, Gertler FB, Pawson T: The murine Nck SH2/SH3 adaptors are important for the development of mesoderm-derived embryonic structures and for regulating the cellular actin network. Molecular and cellular biology. 2003, 23 (13): 4586-4597. 10.1128/MCB.23.13.4586-4597.2003.CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Buday L: Membrane-targeting of signalling molecules by SH2/SH3 domain-containing adaptor proteins. Biochem Biophys Acta. 1999, 1422: 187-204.PubMed Buday L: Membrane-targeting of signalling molecules by SH2/SH3 domain-containing adaptor proteins. Biochem Biophys Acta. 1999, 1422: 187-204.PubMed
10.
Zurück zum Zitat Buday L, Wunderlich L, Tamas P: The Nck family of adapter proteins. Regulators of actin cytoskeleton. Cellular signalling. 2002, 14 (9): 723-731. 10.1016/S0898-6568(02)00027-X.CrossRefPubMed Buday L, Wunderlich L, Tamas P: The Nck family of adapter proteins. Regulators of actin cytoskeleton. Cellular signalling. 2002, 14 (9): 723-731. 10.1016/S0898-6568(02)00027-X.CrossRefPubMed
11.
Zurück zum Zitat Cowan CA, Henkemeyer M: The SH2/SH3 adaptor Grb4 transduces B-ephrin reverse signals. Nature. 2001, 413 (6852): 174-179. 10.1038/35093123.CrossRefPubMed Cowan CA, Henkemeyer M: The SH2/SH3 adaptor Grb4 transduces B-ephrin reverse signals. Nature. 2001, 413 (6852): 174-179. 10.1038/35093123.CrossRefPubMed
12.
Zurück zum Zitat Lawe DC, Hahn C, Wong AJ: The Nck SH2/SH3 adaptor protein is present in the nucleus and associates with the nuclear protein SAM68. Oncogene. 1997, 14: 223-231. 10.1038/sj.onc.1200821.CrossRefPubMed Lawe DC, Hahn C, Wong AJ: The Nck SH2/SH3 adaptor protein is present in the nucleus and associates with the nuclear protein SAM68. Oncogene. 1997, 14: 223-231. 10.1038/sj.onc.1200821.CrossRefPubMed
13.
Zurück zum Zitat Oser M, Dovas A, Cox D, Condeelis J: Nck1 and Grb2 localization patterns can distinguish invadopodia from podosomes. Eur J Cell Biol. 2010 Oser M, Dovas A, Cox D, Condeelis J: Nck1 and Grb2 localization patterns can distinguish invadopodia from podosomes. Eur J Cell Biol. 2010
14.
Zurück zum Zitat Li W, She H: The SH2 and SH3 adapter Nck: a two-gene family and a linker between tyrosine kinases and multiple signaling networks. Histol Histopathol. 2000, 15: 947-955.PubMed Li W, She H: The SH2 and SH3 adapter Nck: a two-gene family and a linker between tyrosine kinases and multiple signaling networks. Histol Histopathol. 2000, 15: 947-955.PubMed
15.
Zurück zum Zitat Latreille M, Laberge MK, Bourret G, Yamani L, Larose L: Deletion of Nck1 attenuates hepatic ER stress signaling, improves glucose tolerance and insulin signaling in liver of obese mice. American journal of physiology. 2011, E423-E434. Latreille M, Laberge MK, Bourret G, Yamani L, Larose L: Deletion of Nck1 attenuates hepatic ER stress signaling, improves glucose tolerance and insulin signaling in liver of obese mice. American journal of physiology. 2011, E423-E434.
16.
Zurück zum Zitat Chou MM, Fajardo JE, Hanafusa H: The SH2- and SH3-Containing Nck Protein Transforms Mammalian Fibroblasts in the Absence of Elevated Phosphotyrosine Levels. Mol Cell Biol. 1992, 12 (12): 5834-5842.CrossRefPubMedPubMedCentral Chou MM, Fajardo JE, Hanafusa H: The SH2- and SH3-Containing Nck Protein Transforms Mammalian Fibroblasts in the Absence of Elevated Phosphotyrosine Levels. Mol Cell Biol. 1992, 12 (12): 5834-5842.CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Li W, Hu P, Skolnik EY, Ullrich A, Schlessinger J: The SH2 and SH3 Domain-Containing Nck Protein Oncogenic and a Common Target for Phosphorylation by Different Surface Receptors. Mol Cell Biol. 1992, 12 (12): 5824-5833.CrossRefPubMedPubMedCentral Li W, Hu P, Skolnik EY, Ullrich A, Schlessinger J: The SH2 and SH3 Domain-Containing Nck Protein Oncogenic and a Common Target for Phosphorylation by Different Surface Receptors. Mol Cell Biol. 1992, 12 (12): 5824-5833.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat de Wit NJ, Rijntjes J, Diepstra JH, van Kuppevelt TH, Weidle UH, Ruiter DJ, van Muijen GN: Analysis of differential gene expression in human melanocytic tumour lesions by custom made oligonucleotide arrays. British journal of cancer. 2005, 92 (12): 2249-2261. 10.1038/sj.bjc.6602612.CrossRefPubMedPubMedCentral de Wit NJ, Rijntjes J, Diepstra JH, van Kuppevelt TH, Weidle UH, Ruiter DJ, van Muijen GN: Analysis of differential gene expression in human melanocytic tumour lesions by custom made oligonucleotide arrays. British journal of cancer. 2005, 92 (12): 2249-2261. 10.1038/sj.bjc.6602612.CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Smalley KS, Contractor R, Haass NK, Lee JT, Nathanson KL, Medina CA, Flaherty KT, Herlyn M: Ki67 expression levels are a better marker of reduced melanoma growth following MEK inhibitor treatment than phospho-ERK levels. British journal of cancer. 2007, 96 (3): 445-449. 10.1038/sj.bjc.6603596.CrossRefPubMedPubMedCentral Smalley KS, Contractor R, Haass NK, Lee JT, Nathanson KL, Medina CA, Flaherty KT, Herlyn M: Ki67 expression levels are a better marker of reduced melanoma growth following MEK inhibitor treatment than phospho-ERK levels. British journal of cancer. 2007, 96 (3): 445-449. 10.1038/sj.bjc.6603596.CrossRefPubMedPubMedCentral
21.
Zurück zum Zitat Lussier G, Larose L: A Casein Kinase I activity is constitutively associated with Nck. J Biol Chem. 1997, 272: 2688-2694. 10.1074/jbc.272.5.2688.CrossRefPubMed Lussier G, Larose L: A Casein Kinase I activity is constitutively associated with Nck. J Biol Chem. 1997, 272: 2688-2694. 10.1074/jbc.272.5.2688.CrossRefPubMed
22.
Zurück zum Zitat Kelm JM, Timmins NE, Brown CJ, Fussenegger M, Nielsen LK: Method for generation of homogeneous multicellular tumor spheroids applicable to a wide variety of cell types. Biotechnol Bioeng. 2003, 83 (2): 173-180. 10.1002/bit.10655.CrossRefPubMed Kelm JM, Timmins NE, Brown CJ, Fussenegger M, Nielsen LK: Method for generation of homogeneous multicellular tumor spheroids applicable to a wide variety of cell types. Biotechnol Bioeng. 2003, 83 (2): 173-180. 10.1002/bit.10655.CrossRefPubMed
23.
Zurück zum Zitat Graeven U, Rodeck U, Karpinski S, Jost M, Philippou S, Schmiegel W: Modulation of Angiogenesis and Tumorigenicity of Human Melanocytic Cells by Vascular Endothelial Growth Factor and Basic Fibroblast Growth Factor. Cancer research. 2001, 61 (19): 7282-7290.PubMed Graeven U, Rodeck U, Karpinski S, Jost M, Philippou S, Schmiegel W: Modulation of Angiogenesis and Tumorigenicity of Human Melanocytic Cells by Vascular Endothelial Growth Factor and Basic Fibroblast Growth Factor. Cancer research. 2001, 61 (19): 7282-7290.PubMed
24.
Zurück zum Zitat Iliopoulos D, Ernst C, Steplewski Z, Jambrosic JA, Rodeck U, Herlyn M, Clark WH, Koprowski H, Herlyn D: Inhibition of Metastases of a Human Melanoma Xenograft by Monoclonal Antibody to the GD2/GD3 Gangliosides. J Natl Cancer Inst. 1989, 81 (6): 440-444. 10.1093/jnci/81.6.440.CrossRefPubMed Iliopoulos D, Ernst C, Steplewski Z, Jambrosic JA, Rodeck U, Herlyn M, Clark WH, Koprowski H, Herlyn D: Inhibition of Metastases of a Human Melanoma Xenograft by Monoclonal Antibody to the GD2/GD3 Gangliosides. J Natl Cancer Inst. 1989, 81 (6): 440-444. 10.1093/jnci/81.6.440.CrossRefPubMed
25.
Zurück zum Zitat Mayer BJ, Hamaguchi M, Hanafusa H: Characterization of p47gag-crk, a novel oncogene product with sequence similarity to a putative modulatory domain of protein-tyrosine kinases and phospholipase C. Cold Spring Harb Symp Quant Biol. 1988, 53 (Pt 2): 907-914.CrossRefPubMed Mayer BJ, Hamaguchi M, Hanafusa H: Characterization of p47gag-crk, a novel oncogene product with sequence similarity to a putative modulatory domain of protein-tyrosine kinases and phospholipase C. Cold Spring Harb Symp Quant Biol. 1988, 53 (Pt 2): 907-914.CrossRefPubMed
26.
Zurück zum Zitat Watanabe T, Tsuda M, Tanaka S, Ohba Y, Kawaguchi H, Majima T, Sawa H, Minami A: Adaptor Protein Crk Induces Src-Dependent Activation of p38 MAPK in Regulation of Synovial Sarcoma Cell Proliferation. Molecular Cancer Research. 2009, 7 (9): 1582-1592. 10.1158/1541-7786.MCR-09-0064.CrossRefPubMed Watanabe T, Tsuda M, Tanaka S, Ohba Y, Kawaguchi H, Majima T, Sawa H, Minami A: Adaptor Protein Crk Induces Src-Dependent Activation of p38 MAPK in Regulation of Synovial Sarcoma Cell Proliferation. Molecular Cancer Research. 2009, 7 (9): 1582-1592. 10.1158/1541-7786.MCR-09-0064.CrossRefPubMed
27.
Zurück zum Zitat Crowley E, Horwitz AF: Tyrosine phosphorylation and cytoskeletal tension regulate the release of fibroblast adhesions. The Journal of cell biology. 1995, 131 (2): 525-537. 10.1083/jcb.131.2.525.CrossRefPubMed Crowley E, Horwitz AF: Tyrosine phosphorylation and cytoskeletal tension regulate the release of fibroblast adhesions. The Journal of cell biology. 1995, 131 (2): 525-537. 10.1083/jcb.131.2.525.CrossRefPubMed
28.
Zurück zum Zitat Zamir E, Katz BZ, Aota S, Yamada KM, Geiger B, Kam Z: Molecular diversity of cell-matrix adhesions. Journal of cell science. 1999, 112 (Pt 11): 1655-1669.PubMed Zamir E, Katz BZ, Aota S, Yamada KM, Geiger B, Kam Z: Molecular diversity of cell-matrix adhesions. Journal of cell science. 1999, 112 (Pt 11): 1655-1669.PubMed
29.
Zurück zum Zitat Chan KT, Cortesio CL, Huttenlocher A: FAK alters invadopodia and focal adhesion composition and dynamics to regulate breast cancer invasion. The Journal of cell biology. 2009, 185 (2): 357-370. 10.1083/jcb.200809110.CrossRefPubMedPubMedCentral Chan KT, Cortesio CL, Huttenlocher A: FAK alters invadopodia and focal adhesion composition and dynamics to regulate breast cancer invasion. The Journal of cell biology. 2009, 185 (2): 357-370. 10.1083/jcb.200809110.CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat Mader CC, Oser M, Magalhaes MAO, Bravo-Cordero JJ, Condeelis J, Koleske AJ, Gil-Henn H: An EGFR-Src-Arg-Cortactin Pathway Mediates Functional Maturation of Invadopodia and Breast Cancer Cell Invasion. Cancer research. 2011, 71 (5): 1730-1741. 10.1158/0008-5472.CAN-10-1432.CrossRefPubMedPubMedCentral Mader CC, Oser M, Magalhaes MAO, Bravo-Cordero JJ, Condeelis J, Koleske AJ, Gil-Henn H: An EGFR-Src-Arg-Cortactin Pathway Mediates Functional Maturation of Invadopodia and Breast Cancer Cell Invasion. Cancer research. 2011, 71 (5): 1730-1741. 10.1158/0008-5472.CAN-10-1432.CrossRefPubMedPubMedCentral
31.
Zurück zum Zitat Smith-Pearson PS, Greuber EK, Yogalingam G, Pendergast AM: Abl Kinases Are Required for Invadopodia Formation and Chemokine-induced Invasion. Journal of Biological Chemistry. 2010, 285 (51): 40201-40211. 10.1074/jbc.M110.147330.CrossRefPubMedPubMedCentral Smith-Pearson PS, Greuber EK, Yogalingam G, Pendergast AM: Abl Kinases Are Required for Invadopodia Formation and Chemokine-induced Invasion. Journal of Biological Chemistry. 2010, 285 (51): 40201-40211. 10.1074/jbc.M110.147330.CrossRefPubMedPubMedCentral
32.
Zurück zum Zitat Balzer EM, Whipple RA, Thompson K, Boggs AE, Slovic J, Cho EH, Matrone MA, Yoneda T, Mueller SC, Martin SS: c-Src differentially regulates the functions of microtentacles and invadopodia. Oncogene. 2010, 29 (48): 6402-6408. 10.1038/onc.2010.360.CrossRefPubMedPubMedCentral Balzer EM, Whipple RA, Thompson K, Boggs AE, Slovic J, Cho EH, Matrone MA, Yoneda T, Mueller SC, Martin SS: c-Src differentially regulates the functions of microtentacles and invadopodia. Oncogene. 2010, 29 (48): 6402-6408. 10.1038/onc.2010.360.CrossRefPubMedPubMedCentral
33.
Zurück zum Zitat Kelley LC, Ammer AG, Hayes KE, Martin KH, Machida K, Jia L, Mayer BJ, Weed SA: Oncogenic Src requires a wild-type counterpart to regulate invadopodia maturation. Journal of cell science. 2010, 123 (22): 3923-3932. 10.1242/jcs.075200.CrossRefPubMedPubMedCentral Kelley LC, Ammer AG, Hayes KE, Martin KH, Machida K, Jia L, Mayer BJ, Weed SA: Oncogenic Src requires a wild-type counterpart to regulate invadopodia maturation. Journal of cell science. 2010, 123 (22): 3923-3932. 10.1242/jcs.075200.CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat Fantus IG, Kadota S, Deragon G, Foster B, Posner BI: Pervanadate [peroxide(s) of vanadate] mimics insulin action in rat adipocytes via activation of the insulin receptor tyrosine kinase. Biochemistry. 1989, 28 (22): 8864-8871. 10.1021/bi00448a027.CrossRefPubMed Fantus IG, Kadota S, Deragon G, Foster B, Posner BI: Pervanadate [peroxide(s) of vanadate] mimics insulin action in rat adipocytes via activation of the insulin receptor tyrosine kinase. Biochemistry. 1989, 28 (22): 8864-8871. 10.1021/bi00448a027.CrossRefPubMed
35.
Zurück zum Zitat Kadota S, Fantus IG, Deragon G, Guyda HJ, Hersh B, Posner BI: Peroxide(s) of vanadium: a novel and potent insulin-mimetic agent which activates the insulin receptor kinase. Biochemical and biophysical research communications. 1987, 147 (1): 259-266. 10.1016/S0006-291X(87)80115-8.CrossRefPubMed Kadota S, Fantus IG, Deragon G, Guyda HJ, Hersh B, Posner BI: Peroxide(s) of vanadium: a novel and potent insulin-mimetic agent which activates the insulin receptor kinase. Biochemical and biophysical research communications. 1987, 147 (1): 259-266. 10.1016/S0006-291X(87)80115-8.CrossRefPubMed
36.
Zurück zum Zitat Posner BI, Faure R, Burgess JW, Bevan AP, Lachance D, Zhang-Sun G, Fantus IG, Ng JB, Hall DA, Lum BS: Peroxovanadium compounds. A new class of potent phosphotyrosine phosphatase inhibitors which are insulin mimetics. Journal of Biological Chemistry. 1994, 269 (6): 4596-4604.PubMed Posner BI, Faure R, Burgess JW, Bevan AP, Lachance D, Zhang-Sun G, Fantus IG, Ng JB, Hall DA, Lum BS: Peroxovanadium compounds. A new class of potent phosphotyrosine phosphatase inhibitors which are insulin mimetics. Journal of Biological Chemistry. 1994, 269 (6): 4596-4604.PubMed
37.
Zurück zum Zitat Brattain MG, Strobel-Stevens J, Fine D, Webb M, Sarrif AM: Establishment of mouse colonic carcinoma cell lines with different metastatic properties. Cancer research. 1980, 40 (7): 2142-2146.PubMed Brattain MG, Strobel-Stevens J, Fine D, Webb M, Sarrif AM: Establishment of mouse colonic carcinoma cell lines with different metastatic properties. Cancer research. 1980, 40 (7): 2142-2146.PubMed
38.
Zurück zum Zitat Lacroix M, Leclercq G: Relevance of breast cancer cell lines as models for breast tumours: an update. Breast cancer research and treatment. 2004, 83 (3): 249-289. 10.1023/B:BREA.0000014042.54925.cc.CrossRefPubMed Lacroix M, Leclercq G: Relevance of breast cancer cell lines as models for breast tumours: an update. Breast cancer research and treatment. 2004, 83 (3): 249-289. 10.1023/B:BREA.0000014042.54925.cc.CrossRefPubMed
39.
Zurück zum Zitat Neve RM, Chin K, Fridlyand J, Yeh J, Baehner FL, Fevr T, Clark L, Bayani N, Coppe JP, Tong F, et al: A collection of breast cancer cell lines for the study of functionally distinct cancer subtypes. Cancer cell. 2006, 10 (6): 515-527. 10.1016/j.ccr.2006.10.008.CrossRefPubMedPubMedCentral Neve RM, Chin K, Fridlyand J, Yeh J, Baehner FL, Fevr T, Clark L, Bayani N, Coppe JP, Tong F, et al: A collection of breast cancer cell lines for the study of functionally distinct cancer subtypes. Cancer cell. 2006, 10 (6): 515-527. 10.1016/j.ccr.2006.10.008.CrossRefPubMedPubMedCentral
40.
Zurück zum Zitat Takeichi M: Morphogenetic roles of classic cadherins. Curr Opin Cell Biol. 1995, 7 (5): 619-627. 10.1016/0955-0674(95)80102-2.CrossRefPubMed Takeichi M: Morphogenetic roles of classic cadherins. Curr Opin Cell Biol. 1995, 7 (5): 619-627. 10.1016/0955-0674(95)80102-2.CrossRefPubMed
41.
Zurück zum Zitat Perl AK, Wilgenbus P, Dahl U, Semb H, Christofori G: A causal role for E-cadherin in the transition from adenoma to carcinoma. Nature. 1998, 392 (6672): 190-193. 10.1038/32433.CrossRefPubMed Perl AK, Wilgenbus P, Dahl U, Semb H, Christofori G: A causal role for E-cadherin in the transition from adenoma to carcinoma. Nature. 1998, 392 (6672): 190-193. 10.1038/32433.CrossRefPubMed
42.
Zurück zum Zitat Frixen UH, Behrens J, Sachs M, Eberle G, Voss B, Warda A, Lochner D, Birchmeier W: E-cadherin-mediated cell-cell adhesion prevents invasiveness of human carcinoma cells. The Journal of cell biology. 1991, 113 (1): 173-185. 10.1083/jcb.113.1.173.CrossRefPubMed Frixen UH, Behrens J, Sachs M, Eberle G, Voss B, Warda A, Lochner D, Birchmeier W: E-cadherin-mediated cell-cell adhesion prevents invasiveness of human carcinoma cells. The Journal of cell biology. 1991, 113 (1): 173-185. 10.1083/jcb.113.1.173.CrossRefPubMed
43.
Zurück zum Zitat Mbalaviele G, Dunstan CR, Sasaki A, Williams PJ, Mundy GR, Yoneda T: E-cadherin expression in human breast cancer cells suppresses the development of osteolytic bone metastases in an experimental metastasis model. Cancer research. 1996, 56 (17): 4063-4070.PubMed Mbalaviele G, Dunstan CR, Sasaki A, Williams PJ, Mundy GR, Yoneda T: E-cadherin expression in human breast cancer cells suppresses the development of osteolytic bone metastases in an experimental metastasis model. Cancer research. 1996, 56 (17): 4063-4070.PubMed
44.
Zurück zum Zitat Hynes RO: Integrins: versatility, modulation, and signaling in cell adhesion. Cell. 1992, 69 (1): 11-25. 10.1016/0092-8674(92)90115-S.CrossRefPubMed Hynes RO: Integrins: versatility, modulation, and signaling in cell adhesion. Cell. 1992, 69 (1): 11-25. 10.1016/0092-8674(92)90115-S.CrossRefPubMed
45.
Zurück zum Zitat Giancotti FG, Mainiero F: Integrin-mediated adhesion and signaling in tumorigenesis. Biochim Biophys Acta. 1994, 1198 (1): 47-64.PubMed Giancotti FG, Mainiero F: Integrin-mediated adhesion and signaling in tumorigenesis. Biochim Biophys Acta. 1994, 1198 (1): 47-64.PubMed
46.
Zurück zum Zitat Juliano RL, Varner JA: Adhesion molecules in cancer: the role of integrins. Curr Opin Cell Biol. 1993, 5 (5): 812-818. 10.1016/0955-0674(93)90030-T.CrossRefPubMed Juliano RL, Varner JA: Adhesion molecules in cancer: the role of integrins. Curr Opin Cell Biol. 1993, 5 (5): 812-818. 10.1016/0955-0674(93)90030-T.CrossRefPubMed
47.
Zurück zum Zitat Giancotti FG, Tarone G: Positional control of cell fate through joint integrin/receptor protein kinase signaling. Annu Rev Cell Dev Biol. 2003, 19: 173-206. 10.1146/annurev.cellbio.19.031103.133334.CrossRefPubMed Giancotti FG, Tarone G: Positional control of cell fate through joint integrin/receptor protein kinase signaling. Annu Rev Cell Dev Biol. 2003, 19: 173-206. 10.1146/annurev.cellbio.19.031103.133334.CrossRefPubMed
48.
Zurück zum Zitat Goicoechea SM, Tu Y, Hua Y, Chen K, Shen TL, Guan JL, Wu C: Nck-2 interacts with focal adhesion kinase and modulates cell motility. The international journal of biochemistry & cell biology. 2002, 34 (7): 791-805. 10.1016/S1357-2725(02)00002-X.CrossRef Goicoechea SM, Tu Y, Hua Y, Chen K, Shen TL, Guan JL, Wu C: Nck-2 interacts with focal adhesion kinase and modulates cell motility. The international journal of biochemistry & cell biology. 2002, 34 (7): 791-805. 10.1016/S1357-2725(02)00002-X.CrossRef
49.
Zurück zum Zitat Guo W, Giancotti FG: Integrin signalling during tumour progression. Nat Rev Mol Cell Biol. 2004, 5 (10): 816-826. 10.1038/nrm1490.CrossRefPubMed Guo W, Giancotti FG: Integrin signalling during tumour progression. Nat Rev Mol Cell Biol. 2004, 5 (10): 816-826. 10.1038/nrm1490.CrossRefPubMed
50.
Zurück zum Zitat Holland SJ, Gale NV, Gish GD, Roth RA, Songyang Z, Cantley LC, Henkemeyer M, Yancopoulos GD, Pawson T: Juxtamembrane tyrosine residues couple the Eph family receptor EphB2/Nuk to specific SH2 domain proteins in neuronal cells. EMBO J. 1997, 16 (13): 3877-3888. 10.1093/emboj/16.13.3877.CrossRefPubMedPubMedCentral Holland SJ, Gale NV, Gish GD, Roth RA, Songyang Z, Cantley LC, Henkemeyer M, Yancopoulos GD, Pawson T: Juxtamembrane tyrosine residues couple the Eph family receptor EphB2/Nuk to specific SH2 domain proteins in neuronal cells. EMBO J. 1997, 16 (13): 3877-3888. 10.1093/emboj/16.13.3877.CrossRefPubMedPubMedCentral
51.
Zurück zum Zitat Lee C-H, Li W, Zhou M, Batzer AG, Myers MG, White MF, Schlessinger J, Skolnik EY: Nck associates with the SH2 domain-docking protein IRS-1 in insulin-stimulated cells. Proc Natl Acad Sci USA. 1993, 90: 11713-11717. 10.1073/pnas.90.24.11713.CrossRefPubMedPubMedCentral Lee C-H, Li W, Zhou M, Batzer AG, Myers MG, White MF, Schlessinger J, Skolnik EY: Nck associates with the SH2 domain-docking protein IRS-1 in insulin-stimulated cells. Proc Natl Acad Sci USA. 1993, 90: 11713-11717. 10.1073/pnas.90.24.11713.CrossRefPubMedPubMedCentral
52.
Zurück zum Zitat Meisenhelder J, Hunter T: The SH2/SH3 Domain-Containing Protein Nck Is Recognized by Certain Anti-Phospholipase C-γ1 Monoclonal Antibodies, And Its Phosphorylation on Tyrosine Is Stimulated by Platelet-Derived Growth Factor and Epidermal Growth Factor Treatment. Mol Cell Biol. 1992, 12 (12): 5843-5856.CrossRefPubMedPubMedCentral Meisenhelder J, Hunter T: The SH2/SH3 Domain-Containing Protein Nck Is Recognized by Certain Anti-Phospholipase C-γ1 Monoclonal Antibodies, And Its Phosphorylation on Tyrosine Is Stimulated by Platelet-Derived Growth Factor and Epidermal Growth Factor Treatment. Mol Cell Biol. 1992, 12 (12): 5843-5856.CrossRefPubMedPubMedCentral
53.
Zurück zum Zitat Park D, Rhee SG: Phosphorylation of Nck in Response to a Variety of Receptors, Phorbol Myristate Acetate, and Cyclic AMP. Mol Cell Biol. 1992, 12 (12): 5816-5823.CrossRefPubMedPubMedCentral Park D, Rhee SG: Phosphorylation of Nck in Response to a Variety of Receptors, Phorbol Myristate Acetate, and Cyclic AMP. Mol Cell Biol. 1992, 12 (12): 5816-5823.CrossRefPubMedPubMedCentral
54.
Zurück zum Zitat Stein E, Huynh-Do U, Lane AA, Ceretti DP, Daniel TO: Nck Recruitment to Eph Receptor, EphB1/ELK, Couples Ligand Activation to c-Jun Kinase. J Biol Chem. 1998, 273 (3): 1303-1308. 10.1074/jbc.273.3.1303.CrossRefPubMed Stein E, Huynh-Do U, Lane AA, Ceretti DP, Daniel TO: Nck Recruitment to Eph Receptor, EphB1/ELK, Couples Ligand Activation to c-Jun Kinase. J Biol Chem. 1998, 273 (3): 1303-1308. 10.1074/jbc.273.3.1303.CrossRefPubMed
55.
Zurück zum Zitat Tu Y, Li F, Wu C: Nck-2, a Novel Src Homology 2/3-containing Adaptor Protein That Interacts with the LIM-only Protein PINCH and Components of Growth Factor Receptor Kinase-signaling Pathways. Mol Biol Cell. 1998, 9: 3367-3382.CrossRefPubMedPubMedCentral Tu Y, Li F, Wu C: Nck-2, a Novel Src Homology 2/3-containing Adaptor Protein That Interacts with the LIM-only Protein PINCH and Components of Growth Factor Receptor Kinase-signaling Pathways. Mol Biol Cell. 1998, 9: 3367-3382.CrossRefPubMedPubMedCentral
56.
Zurück zum Zitat Tu Y, Li F, Goicoechea S, Wu C: The LIM-only protein PINCH directly interacts with the integrin-linked kinase and is recruited to integrin-rich sites in spreading cells. M Cell Biol. 1999, 19: 2425-2434.CrossRef Tu Y, Li F, Goicoechea S, Wu C: The LIM-only protein PINCH directly interacts with the integrin-linked kinase and is recruited to integrin-rich sites in spreading cells. M Cell Biol. 1999, 19: 2425-2434.CrossRef
57.
Zurück zum Zitat Wu C, Dedhar S: Integrin-linked kinase (ILK) and its interactors: a new paradigm for the coupling of extracellular matrix to actin cytoskeleton and signaling complexes. The Journal of cell biology. 2001, 155 (4): 505-510. 10.1083/jcb.200108077.CrossRefPubMedPubMedCentral Wu C, Dedhar S: Integrin-linked kinase (ILK) and its interactors: a new paradigm for the coupling of extracellular matrix to actin cytoskeleton and signaling complexes. The Journal of cell biology. 2001, 155 (4): 505-510. 10.1083/jcb.200108077.CrossRefPubMedPubMedCentral
58.
Zurück zum Zitat Zhang Y, Guo L, Chen K, Wu C: A critical role of the PINCH-integrin-linked kinase interaction in the regulation of cell shape change and migration. The Journal of biological chemistry. 2002, 277 (1): 318-326.CrossRefPubMed Zhang Y, Guo L, Chen K, Wu C: A critical role of the PINCH-integrin-linked kinase interaction in the regulation of cell shape change and migration. The Journal of biological chemistry. 2002, 277 (1): 318-326.CrossRefPubMed
59.
Zurück zum Zitat Blasutig IM, New LA, Thanabalasuriar A, Dayarathna TK, Goudreault M, Quaggin SE, Li SS, Gruenheid S, Jones N, Pawson T: Phosphorylated YDXV motifs and Nck SH2/SH3 adaptors act cooperatively to induce actin reorganization. Molecular and cellular biology. 2008, 28 (6): 2035-2046. 10.1128/MCB.01770-07.CrossRefPubMedPubMedCentral Blasutig IM, New LA, Thanabalasuriar A, Dayarathna TK, Goudreault M, Quaggin SE, Li SS, Gruenheid S, Jones N, Pawson T: Phosphorylated YDXV motifs and Nck SH2/SH3 adaptors act cooperatively to induce actin reorganization. Molecular and cellular biology. 2008, 28 (6): 2035-2046. 10.1128/MCB.01770-07.CrossRefPubMedPubMedCentral
60.
Zurück zum Zitat Li X, Meriane M, Triki I, Shekarabi M, Kennedy TE, Larose L, Lamarche-Vane N: The adaptor protein Nck-1 couples the netrin-1 receptor DCC (deleted in colorectal cancer) to the activation of the small GTPase Rac1 through an atypical mechanism. The Journal of biological chemistry. 2002, 277 (40): 37788-37797. 10.1074/jbc.M205428200.CrossRefPubMed Li X, Meriane M, Triki I, Shekarabi M, Kennedy TE, Larose L, Lamarche-Vane N: The adaptor protein Nck-1 couples the netrin-1 receptor DCC (deleted in colorectal cancer) to the activation of the small GTPase Rac1 through an atypical mechanism. The Journal of biological chemistry. 2002, 277 (40): 37788-37797. 10.1074/jbc.M205428200.CrossRefPubMed
61.
Zurück zum Zitat Antoku S, Saksela K, Rivera GM, Mayer BJ: A crucial role in cell spreading for the interaction of Abl PxxP motifs with Crk and Nck adaptors. Journal of cell science. 2008, 121 (Pt 18): 3071-3082.CrossRefPubMedPubMedCentral Antoku S, Saksela K, Rivera GM, Mayer BJ: A crucial role in cell spreading for the interaction of Abl PxxP motifs with Crk and Nck adaptors. Journal of cell science. 2008, 121 (Pt 18): 3071-3082.CrossRefPubMedPubMedCentral
62.
Zurück zum Zitat Smith JM, Katz S, Mayer BJ: Activation of the Abl tyrosine kinase in vivo by Src homology 3 domains from the Src homology2/Src homology 3 adaptor Nck. J Biol Chem. 1999, 274 (39): 27956-27962. 10.1074/jbc.274.39.27956.CrossRefPubMed Smith JM, Katz S, Mayer BJ: Activation of the Abl tyrosine kinase in vivo by Src homology 3 domains from the Src homology2/Src homology 3 adaptor Nck. J Biol Chem. 1999, 274 (39): 27956-27962. 10.1074/jbc.274.39.27956.CrossRefPubMed
63.
Zurück zum Zitat Preisinger C, Kolch W: The Bcr-Abl kinase regulates the actin cytoskeleton via a GADS/Slp-76/Nck1 adaptor protein pathway. Cellular signalling. 2010, 22 (5): 848-856. 10.1016/j.cellsig.2009.12.012.CrossRefPubMed Preisinger C, Kolch W: The Bcr-Abl kinase regulates the actin cytoskeleton via a GADS/Slp-76/Nck1 adaptor protein pathway. Cellular signalling. 2010, 22 (5): 848-856. 10.1016/j.cellsig.2009.12.012.CrossRefPubMed
64.
Zurück zum Zitat Lee JH, Pyon J-K, Kim DW, Lee SH, Nam HS, Kim CH, Kang SG, Lee YJ, Park MY, Jeong DJ, et al: Elevated c-Src and c-Yes expression in malignant skin cancers. Journal of Experimental & Clinical Cancer Research. 2010, 29 (1): 116-10.1186/1756-9966-29-116.CrossRef Lee JH, Pyon J-K, Kim DW, Lee SH, Nam HS, Kim CH, Kang SG, Lee YJ, Park MY, Jeong DJ, et al: Elevated c-Src and c-Yes expression in malignant skin cancers. Journal of Experimental & Clinical Cancer Research. 2010, 29 (1): 116-10.1186/1756-9966-29-116.CrossRef
65.
Zurück zum Zitat Stylli SS, Stacey TT, Verhagen AM, Xu SS, Pass I, Courtneidge SA, Lock P: Nck adaptor proteins link Tks5 to invadopodia actin regulation and ECM degradation. Journal of cell science. 2009, 122 (Pt 15): 2727-2740.CrossRefPubMedPubMedCentral Stylli SS, Stacey TT, Verhagen AM, Xu SS, Pass I, Courtneidge SA, Lock P: Nck adaptor proteins link Tks5 to invadopodia actin regulation and ECM degradation. Journal of cell science. 2009, 122 (Pt 15): 2727-2740.CrossRefPubMedPubMedCentral
66.
Zurück zum Zitat Yamaguchi H, Lorenz M, Kempiak S, Sarmiento C, Coniglio S, Symons M, Segall J, Eddy R, Miki H, Takenawa T, et al: Molecular mechanisms of invadopodium formation: the role of the N-WASP-Arp2/3 complex pathway and cofilin. The Journal of cell biology. 2005, 168 (3): 441-452. 10.1083/jcb.200407076.CrossRefPubMedPubMedCentral Yamaguchi H, Lorenz M, Kempiak S, Sarmiento C, Coniglio S, Symons M, Segall J, Eddy R, Miki H, Takenawa T, et al: Molecular mechanisms of invadopodium formation: the role of the N-WASP-Arp2/3 complex pathway and cofilin. The Journal of cell biology. 2005, 168 (3): 441-452. 10.1083/jcb.200407076.CrossRefPubMedPubMedCentral
67.
Zurück zum Zitat Ayala I, Baldassarre M, Giacchetti G, Caldieri G, Tete S, Luini A, Buccione R: Multiple regulatory inputs converge on cortactin to control invadopodia biogenesis and extracellular matrix degradation. Journal of cell science. 2008, 121 (Pt 3): 369-378.CrossRefPubMed Ayala I, Baldassarre M, Giacchetti G, Caldieri G, Tete S, Luini A, Buccione R: Multiple regulatory inputs converge on cortactin to control invadopodia biogenesis and extracellular matrix degradation. Journal of cell science. 2008, 121 (Pt 3): 369-378.CrossRefPubMed
68.
Zurück zum Zitat Chen WT: Proteolytic activity of specialized surface protrusions formed at rosette contact sites of transformed cells. J Exp Zool. 1989, 251 (2): 167-185. 10.1002/jez.1402510206.CrossRefPubMed Chen WT: Proteolytic activity of specialized surface protrusions formed at rosette contact sites of transformed cells. J Exp Zool. 1989, 251 (2): 167-185. 10.1002/jez.1402510206.CrossRefPubMed
69.
Zurück zum Zitat Condeelis J, Segall JE: Intravital imaging of cell movement in tumours. Nature reviews. 2003, 3 (12): 921-930.PubMed Condeelis J, Segall JE: Intravital imaging of cell movement in tumours. Nature reviews. 2003, 3 (12): 921-930.PubMed
70.
Zurück zum Zitat Buccione R, Orth JD, McNiven MA: Foot and mouth: podosomes, invadopodia and circular dorsal ruffles. Nat Rev Mol Cell Biol. 2004, 5 (8): 647-657. 10.1038/nrm1436.CrossRefPubMed Buccione R, Orth JD, McNiven MA: Foot and mouth: podosomes, invadopodia and circular dorsal ruffles. Nat Rev Mol Cell Biol. 2004, 5 (8): 647-657. 10.1038/nrm1436.CrossRefPubMed
71.
Zurück zum Zitat McNiven MA, Baldassarre M, Buccione R: The role of dynamin in the assembly and function of podosomes and invadopodia. Front Biosci. 2004, 9: 1944-1953. 10.2741/1348.CrossRefPubMed McNiven MA, Baldassarre M, Buccione R: The role of dynamin in the assembly and function of podosomes and invadopodia. Front Biosci. 2004, 9: 1944-1953. 10.2741/1348.CrossRefPubMed
Metadaten
Titel
Nck2 promotes human melanoma cell proliferation, migration and invasion in vitro and primary melanoma-derived tumor growth in vivo
verfasst von
Mélissa Labelle-Côté
Julie Dusseault
Salma Ismaïl
Aude Picard-Cloutier
Peter M Siegel
Louise Larose
Publikationsdatum
01.12.2011
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2011
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/1471-2407-11-443

Weitere Artikel der Ausgabe 1/2011

BMC Cancer 1/2011 Zur Ausgabe

Umsetzung der POMGAT-Leitlinie läuft

03.05.2024 DCK 2024 Kongressbericht

Seit November 2023 gibt es evidenzbasierte Empfehlungen zum perioperativen Management bei gastrointestinalen Tumoren (POMGAT) auf S3-Niveau. Vieles wird schon entsprechend der Empfehlungen durchgeführt. Wo es im Alltag noch hapert, zeigt eine Umfrage in einem Klinikverbund.

CUP-Syndrom: Künstliche Intelligenz kann Primärtumor finden

30.04.2024 Künstliche Intelligenz Nachrichten

Krebserkrankungen unbekannten Ursprungs (CUP) sind eine diagnostische Herausforderung. KI-Systeme können Pathologen dabei unterstützen, zytologische Bilder zu interpretieren, um den Primärtumor zu lokalisieren.

Sind Frauen die fähigeren Ärzte?

30.04.2024 Gendermedizin Nachrichten

Patienten, die von Ärztinnen behandelt werden, dürfen offenbar auf bessere Therapieergebnisse hoffen als Patienten von Ärzten. Besonders gilt das offenbar für weibliche Kranke, wie eine Studie zeigt.

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.