In this study, we identified differential expression of immunoreactive matrix metalloproteinase 2 (MMP2)/gelatinase A, membrane-anchored MT1-MMP/MMP14, and human relaxin-2 (RLN2) in human benign and malignant thyroid tissues. MMP2 and MT1-MMP were detected in the majority of thyroid cancer tissues and colocalized with RLN2-positive cells. MMP2 was mostly absent in goiter tissues and, similar to RLN2, may serve as a marker for thyroid cancer. MMP2 and MT1-MMP were identified as novel RLN2 targets. RLN2 caused a significant downregulation of tissue inhibitor of MMP (TIMP) 3 protein levels but did not change the expression levels of MMP13, and TIMP1, TIMP2, and TIMP4 in human thyroid carcinoma cells. RLN2 failed to affect the expression of MMP1, 3, 8, and 9 in the thyroid carcinoma cells investigated. Stable RLN2 transfectants secreted enhanced levels of bioactive MMP2 which contributed to the increased collagenolytic activity and in vitro invasiveness into collagen matrix by human thyroid cancer cells. Three-dimensional reconstitution of confocal fluorescent microscopy images revealed larger-sized invadopodia, with intense MT1-MMP accumulation at the leading migrating edge in RLN2 transfectants when compared with enhanced green fluorescent protein clones. In RLN2 transfectants actin stress fibers contributed to pseudopodia formation. In conclusion, enhanced tumor cell invasion by RLN2 involves the formation of MT1-MMP–enriched invadopodia that lead to increased collagenolytic cell invasion by human thyroid cancer cells. Mol Cancer Res; 9(6); 673–87. ©2011 AACR.

The multifunctional heterodimeric peptide hormone relaxin (RLN), a member of the insulin-like superfamily, is an established endocrine factor in the reproductive tract, cardiovascular/neural system, and in cancer biology (1–6). Upregulated in various human cancer tissues, RLN2 contributes to tumor cell proliferation, tissue invasion, and tumor angiogenesis (7–9). Understanding the molecular mechanisms by which RLN2 enhances the tissue invasiveness of tumor cells in patients is of prognostic and therapeutic importance. High serum levels of RLN2 were shown to be associated with increased levels of metastasis in breast cancer patients (10).

The penetration of the basal lamina and remodeling of the extracellular matrix (ECM) are crucial early steps in the process of tumor cell invasion. RLN is known to contribute to ECM remodeling processes in various biological systems by: (i) inhibiting fibroblast proliferation and differentiation (11, 12); (ii) inhibiting collagen synthesis/secretion and tissue deposition in vivo (13–15); (iii) altering the composition and structural integrity of the ECM (16–19); and by (iv) increasing the production/secretion and activity of enzymes that are powerful remodeling factors of the basal lamina and ECM (20, 21). In the human thyroid gland, RLN2 is exclusively detected in thyroid cancer tissues and enhances the production and secretion of the lysosomal acid hydrolases of the cathepsin family which promote penetration of elastin matrices by human thyroid cancer cells (7). Earlier reports supported a role for RLN in ECM remodeling by upregulating cathepsin B and dipeptidyl peptidase I in the mouse pubic symphysis and urokinase and tissue type plasminogen activator in the uterus and cervix of prepubertal gilts (22–24). Members of the family of matrix metalloproteinases (MMP) and their tissue inhibitors of MMP (TIMP1–4) are another group of proteinases responsive to RLN.

The MMP/TIMP system is known to modulate normal (e.g., development, growth, and repair) and pathologic tissue compositions (e.g., fibrosis, tumor spread). Functional imbalance between MMPs and TIMPs can facilitate tumor cell invasion of basal lamina and ECM and enhances tumor angiogenesis (25). The members of the human MMP family of zinc-dependent endopeptidases segregate into 5 major subclasses (collagenases, stromelysins, gelatinases, membrane-type MMPs, and other MMPs; ref. 26). All of these MMPs combined have the ability to degrade essentially any ECM component (27). Apart from the basic structural design of secreted MMPs, which includes an N-terminal signal peptide, a propeptide, and a catalytic domain with a conserved zinc-binding site linked to a hemopexin-like domain, the membrane-type (MT)-MMPs 1–4 possess an additional anchoring transmembrane and cytoplasmic domain C-terminal to the hemopexin-like domain. MMP family members MMP2 and MT1-MMP, for example, not only degrade ECM components but can also cleave and activate growth factors, thereby promoting tumor cell migration/invasion (28, 29) and modulating angiogenesis (30).

MMP2, MT1-MMP, and TIMP2 play a significant role in thyroid cancer. These MMP and TIMP members are frequently upregulated in differentiated thyroid carcinoma and increased MMP2 expression is common in papillary and undifferentiated/anaplastic thyroid cancer, but not in follicular adenoma (31–33). Enhanced production and activation of proMMP2, MT1-MMP, and TIMP2 is positively associated with large thyroid tumor size, increased invasiveness, including lymph node metastasis, and advanced disease stage (31, 34, 35). Human normal thyrocytes and thyroid carcinoma cell lines were shown to express MMP1, 2, and 9, MT1-MMP, and TIMP1–3 (36, 37).

RLN affects the expression and/or secretion of collagenase 1 (MMP1) and 3 (MMP13), gelatinase A (MMP2) and B (MMP9), stromelysin1 (MMP3), and MT1-MMP in the periodontal apparatus and joints (38–40), lung (13), reproductive tissues (24, 41–43), human renal fibroblasts (44), and human breast cancer cells (20). We previously showed that human thyroid cancer tissue is a source of RLN2 and that RLN2 enhances elastin matrix degradation by thyroid cancer cells (7).

In this study, we have determined the tissue distribution of MMP2/gelatinase A and MT-MMP1/MMP14 in human thyroid carcinoma tissues and identified MMP2 to be predominantly present in thyroid tumor tissues. We show that both MMPs are RLN2 target molecules in thyroid carcinoma cells. Of all members of the TIMP family, RLN2 caused no changes in TIMP2 but a significant downregulation of TIMP3 in thyroid cancer cells. The RLN2-mediated upregulation of in vitro invasiveness through collagen matrices coincided with the increased presence of MT1-MMP and MMP2 at the leading migrating edge of invadopodia in human thyroid carcinoma cells. These findings implicate the MMP2–MT1-MMP–TIMP2 system and TIMP3 as potent mediators of RLN2 in promoting tissue invasion and angiogenesis in human thyroid cancer.

Reverse transcriptase PCR

One microgram of total RNA was used as a template for first strand cDNA synthesis as described previously (7). PCR amplification conditions had been optimized for each primer pair listed in Table 1.

Table 1.

List of primers used for the RT-PCR detection of MMPs and TIMPs

Primer namePrimer sequenceSize (bp)Annealing temp. Ta (°C)
F-MMP1 5′-TGGGAGCAAACACATCTGAC-3′ 560 64 
R-MMP1 5′-ATCACTTCTCCCCGAATCGT-3′   
F-MMP2 5′-GCAGATGCCTGGAATGCCAT-3′ 504 59 
R-MMP2 5′-AGGGTTCTGTGAGCCACAGA-3′   
F-MMP3 5′-GCAGTTTGCTCAGCCTATCC-3′ 214 62 
R-MMP3 5′-GAGTGTCGGAGTCCAGCTTC-3′   
F-MMP8 5′-TCTGCAAGGTTATCCCAAGG-3′ 251 58 
R-MMP8 5′-TTGGTCCACTGAAGACATGG-3′   
F-MMP9 5′-GCTATGGTTACACTCGGGG-3′ 530 64 
R-MMP9 5′-GCCATCTGCGTTTCCAAACC-3′   
F-MMP10 5′-CAGAAGTTCCTTGGGTTGGA-3′ 598 62 
R-MMP10 5′-GGGGAGGTCCGTAGAGAGAC-3′   
F-MMP13 5′-AGGAGATGCCCATTTTGATG-3′ 405 58 
R-MMP13 5′-GGAAGTTCTGGCCAAAATGA-3′   
F-MT1-MMP 5′-ACATTGGAGGAGACACCCAC-3′ 504 62 
R-MT1-MMP 5′-GGCAGTGTTGATGGACGC-3′   
F-TIMP1 5′-ATTCCGACCTCGTCATCAG-3′ 404 56 
R-TIMP1 5′-CGTCCACAAGCAATGAGTG-3′   
F-TIMP2 5′-GACGTTGGAGGAAAGAAGGA-3′ 371 59 
R-TIMP2 5′-CGTTGGAGGCCTGCTTATGG-3′   
F-TIMP3 5′-GGGTGTGTGGCATTGATGAT-3′ 428 64 
R-TIMP3 5′-ATGTACCGAGGCTTCACCAA-3′   
F-TIMP4 5′-CCACTCGGCACTTGTGATTCG-3′ 489 62 
R-TIMP4 5′-CAGGTGCCGTCAACATGCTTC-3′   
F-18S 5′-GTTGTTGGAGCGATTTGTCTGG-3′ 344 61 
R-18S 5′-AGGGCAGGGACTTAATCAACGC-3′   
Primer namePrimer sequenceSize (bp)Annealing temp. Ta (°C)
F-MMP1 5′-TGGGAGCAAACACATCTGAC-3′ 560 64 
R-MMP1 5′-ATCACTTCTCCCCGAATCGT-3′   
F-MMP2 5′-GCAGATGCCTGGAATGCCAT-3′ 504 59 
R-MMP2 5′-AGGGTTCTGTGAGCCACAGA-3′   
F-MMP3 5′-GCAGTTTGCTCAGCCTATCC-3′ 214 62 
R-MMP3 5′-GAGTGTCGGAGTCCAGCTTC-3′   
F-MMP8 5′-TCTGCAAGGTTATCCCAAGG-3′ 251 58 
R-MMP8 5′-TTGGTCCACTGAAGACATGG-3′   
F-MMP9 5′-GCTATGGTTACACTCGGGG-3′ 530 64 
R-MMP9 5′-GCCATCTGCGTTTCCAAACC-3′   
F-MMP10 5′-CAGAAGTTCCTTGGGTTGGA-3′ 598 62 
R-MMP10 5′-GGGGAGGTCCGTAGAGAGAC-3′   
F-MMP13 5′-AGGAGATGCCCATTTTGATG-3′ 405 58 
R-MMP13 5′-GGAAGTTCTGGCCAAAATGA-3′   
F-MT1-MMP 5′-ACATTGGAGGAGACACCCAC-3′ 504 62 
R-MT1-MMP 5′-GGCAGTGTTGATGGACGC-3′   
F-TIMP1 5′-ATTCCGACCTCGTCATCAG-3′ 404 56 
R-TIMP1 5′-CGTCCACAAGCAATGAGTG-3′   
F-TIMP2 5′-GACGTTGGAGGAAAGAAGGA-3′ 371 59 
R-TIMP2 5′-CGTTGGAGGCCTGCTTATGG-3′   
F-TIMP3 5′-GGGTGTGTGGCATTGATGAT-3′ 428 64 
R-TIMP3 5′-ATGTACCGAGGCTTCACCAA-3′   
F-TIMP4 5′-CCACTCGGCACTTGTGATTCG-3′ 489 62 
R-TIMP4 5′-CAGGTGCCGTCAACATGCTTC-3′   
F-18S 5′-GTTGTTGGAGCGATTTGTCTGG-3′ 344 61 
R-18S 5′-AGGGCAGGGACTTAATCAACGC-3′   

Abbreviations: F, forward primer; R, reverse primer; bp, base pairs.

Tissues

A total of 106 human thyroid tissues [25 goiter, 30 papillary (PTC), 27 follicular (FTC), and 24 undifferentiated thyroid cancer (UTC)] were obtained at the Department of General, Visceral and Vascular Surgery, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany (Table 2). Tumor size, local tissue invasion, colonization of local lymph nodes, and presence/absence of metastases were determined according to the pTNM tumor staging classification (T = tumor, N = lymph node, and M = metastasis). Tissue specimens were cryopreserved in liquid nitrogen. Human placental tissue sections were used as positive control for the immunodetection of MT1-MMP. The collection of all human tissues studied was approved by the ethical committee at the Martin Luther University, Faculty of Medicine, and all patients gave written consent.

Table 2.

List of human thyroid tissues used in this study

TissueAgeSexpTNM stageIHCMMP (no. of cases)
Goiter 51 F  ø MMP2(n = 15) 
 42  ø  
 57   
 35  ø  
 42  ø  
 61  ø  
 51  ø  
 73  ø  
 65  ø  
 41  ø  
 47  ø  
 43  ø  
 60  ø  
 74  ø  
 35  ø  
 62  (+) MT1-MMP (n = 10) 
 44  ø  
 71  ø  
 65  ø  
 74  ø  
 39  ø  
 71  ø  
 46   
 36  ø  
 60  (+)  
PTC 55 T1N0M0 MMP2 (n = 19) 
 17 T2N0MX  
 47 T2N0MX  
 36 T2N1MX ø  
 36 T2N1MX ø  
 61 T4N0MX ø  
 68 T4N0MX  
 41 T4N0MX  
 74 T4N1MX  
 32 T2N1bM0  
 72 T4N1aM1  
 10 T4N1bM0  
 26 T4N1bM1  
 28 T4N1bMX  
 66 T4NXMX ø  
 75 T4NXMX  
 47 T2N0M1  
 57 T4N0M0  
 72 T3N0M1  
 56 T1aNxMx (+) MT1-MMP (n = 8) 
 17 T2N0M0  
 57 T2NxMx ø  
 62 T3N1Mx  
 72 T4N1M1  
 64 T4NxMx  
 55 T1aN0M0  
 26 T4aN1bM1  
 74 T4NXMX ø/ø MMP2/MT1-MMP (n = 3) 
 84 T4aN1bM1 +/+  
 68 T4aN1bMx +/+  
FTC 41 T2N0MX ø MMP2 (n = 16) 
 68 T2N1MX  
 35 T3NXM1  
 68 T3NXMX  
 53 T4N0M0  
 36 T4N0M0  
 68 T4N0MX ø  
 43 T4N1aMX  
 52 T4N1bMX ø  
 61 T4N1bM1  
 28 T3N0M1 ø  
 86 T3N1aMX  
 57 T3N1bMX  
 67 T4N0M0  
 66 T4N0MX  
 77 T4N1bMX  
 55 T2NXMX MT1-MMP (n = 5) 
 13 T3NXMX  
 53 T4NxMx  
 69 T2N1MX  
 60 T4N0Mx  
 52 T3N1bcM0 +/ø MMP2/MT1-MMP (n = 6) 
 68 T2N0MX +/ø  
 50 T4N0M0 +/+  
 66 T4N0MX ø/ø  
 36 T4bN0M0 +/+  
 65 T4bN1bMX ø/ø  
UTC 52 T3N1bM0 ø MMP2 (n = 18) 
 63 T3N1bM1  
 54 T3N1bMX ø  
 76 T4N1aM1  
 59 T4N1aM1  
 74 T4N1aMX  
 71 T4N1aMX ø  
 69 T4N1bM1  
 65 T4N1bM1  
 72 T4N1bM1 ø  
 76 T4N1bM1 ø  
 87 T4N1bM1 ø  
 42 T4N1bMX ø  
 79 T4N1bMX  
 53 T4N1aMX  
 68 T4N1aMX  
 78 T4N0M0  
 69 T3NXMX  
 69 T4NxMx MT1-MMP (n = 1) 
 72 T4N2Mx ø/ø MMP2/MT1-MMP (n = 5) 
 87 T4NxMx +/+  
 28 T3N0M1 +/+  
 71 T4N1aMx +/ø  
 63 T3N1bM1 ø/+  
TissueAgeSexpTNM stageIHCMMP (no. of cases)
Goiter 51 F  ø MMP2(n = 15) 
 42  ø  
 57   
 35  ø  
 42  ø  
 61  ø  
 51  ø  
 73  ø  
 65  ø  
 41  ø  
 47  ø  
 43  ø  
 60  ø  
 74  ø  
 35  ø  
 62  (+) MT1-MMP (n = 10) 
 44  ø  
 71  ø  
 65  ø  
 74  ø  
 39  ø  
 71  ø  
 46   
 36  ø  
 60  (+)  
PTC 55 T1N0M0 MMP2 (n = 19) 
 17 T2N0MX  
 47 T2N0MX  
 36 T2N1MX ø  
 36 T2N1MX ø  
 61 T4N0MX ø  
 68 T4N0MX  
 41 T4N0MX  
 74 T4N1MX  
 32 T2N1bM0  
 72 T4N1aM1  
 10 T4N1bM0  
 26 T4N1bM1  
 28 T4N1bMX  
 66 T4NXMX ø  
 75 T4NXMX  
 47 T2N0M1  
 57 T4N0M0  
 72 T3N0M1  
 56 T1aNxMx (+) MT1-MMP (n = 8) 
 17 T2N0M0  
 57 T2NxMx ø  
 62 T3N1Mx  
 72 T4N1M1  
 64 T4NxMx  
 55 T1aN0M0  
 26 T4aN1bM1  
 74 T4NXMX ø/ø MMP2/MT1-MMP (n = 3) 
 84 T4aN1bM1 +/+  
 68 T4aN1bMx +/+  
FTC 41 T2N0MX ø MMP2 (n = 16) 
 68 T2N1MX  
 35 T3NXM1  
 68 T3NXMX  
 53 T4N0M0  
 36 T4N0M0  
 68 T4N0MX ø  
 43 T4N1aMX  
 52 T4N1bMX ø  
 61 T4N1bM1  
 28 T3N0M1 ø  
 86 T3N1aMX  
 57 T3N1bMX  
 67 T4N0M0  
 66 T4N0MX  
 77 T4N1bMX  
 55 T2NXMX MT1-MMP (n = 5) 
 13 T3NXMX  
 53 T4NxMx  
 69 T2N1MX  
 60 T4N0Mx  
 52 T3N1bcM0 +/ø MMP2/MT1-MMP (n = 6) 
 68 T2N0MX +/ø  
 50 T4N0M0 +/+  
 66 T4N0MX ø/ø  
 36 T4bN0M0 +/+  
 65 T4bN1bMX ø/ø  
UTC 52 T3N1bM0 ø MMP2 (n = 18) 
 63 T3N1bM1  
 54 T3N1bMX ø  
 76 T4N1aM1  
 59 T4N1aM1  
 74 T4N1aMX  
 71 T4N1aMX ø  
 69 T4N1bM1  
 65 T4N1bM1  
 72 T4N1bM1 ø  
 76 T4N1bM1 ø  
 87 T4N1bM1 ø  
 42 T4N1bMX ø  
 79 T4N1bMX  
 53 T4N1aMX  
 68 T4N1aMX  
 78 T4N0M0  
 69 T3NXMX  
 69 T4NxMx MT1-MMP (n = 1) 
 72 T4N2Mx ø/ø MMP2/MT1-MMP (n = 5) 
 87 T4NxMx +/+  
 28 T3N0M1 +/+  
 71 T4N1aMx +/ø  
 63 T3N1bM1 ø/+  

Cell culture

The human follicular thyroid carcinoma cell line FTC-133 and previously described FTC-133 stable transfectants expressing human RLN2 (FTC-133–RLN2) and enhanced green fluorescent protein (EGFP)-expressing FTC-133 negative controls (FTC-133–EGFP; refs. 7, 45) were cultured in Dulbecco's modified Eagle's medium/HAM-F12 medium plus 10% fetal calf serum (FCS; Gibco). Culture medium was changed every 2 to 3 days and transfectants were passaged every 5 to 6 days. Prior to the experiments, cells were either seeded in 25 cm2 flasks or in 6-, 24-, or 96-well plates (Techno Plastic Products AG) at 80% confluency. Untransfected FTC-133 at the same concentration were incubated with 100 ng/mL RLN2 (RLN2 was generously provided by Corthera Inc.). All experiments were conducted under serum-free conditions.

Immunohistochemistry

For immunohistochemistry, sections of goiter and thyroid carcinoma tissues were investigated for the presence of immunoreactive MMP2, MT1-MMP, and RLN2. Immunohistochemical analysis was done by using the DAKO LSAB Kit (Dako). Tissue sections were fixed with 3% H2O2 in cold methanol for 25 minutes, washed in PBS plus 0.1% Tween-20 (PBST), and nonspecific-binding sites were blocked with 10% goat serum in PBST (blocking buffer) for 1 hour at room temperature (RT). Sections were incubated with the primary monoclonal antibodies for MMP2 and MT1-MMP (both Calbiochem) diluted 1:100, or a polyclonal antiserum against RLN2 (Immunodiagnostik) at 1:300 with antibody diluent (Dako) at 4°C overnight (7). Sections were incubated with horseradish peroxidase (HRP)–conjugated secondary antibodies (Dako) for colorimetric detection of target MMPs and RLN2 within the thyroid tissues investigated. Immunostained sections were counterstained with hematoxylin/eosin.

Immunofluorescence and confocal laser scanning microscopy

The day before the immunofluorescence experiments, cells were washed twice with 1× PBS and grown in serum-free medium overnight. For exclusive F-actin staining, cells were incubated with phalloidin–AlexaFluor 546 conjugate (1:200; Invitrogen). Immunofluorescent confocal microscopy was done in 12-well plates with insert filters containing polyester membranes with 3.0-μm pore size (Costar). Cells were seeded on the filters and incubated in a 5% CO2 humidified incubator at 37°C for 3 hours to allow pseudopodia to reach the underside of the filter. Cells on both sides of the filters were rinsed with PBS, fixed with 3.7% paraformaldehyde in PBS for 25 minutes, permeabilized, and incubated with a rabbit polyclonal anti–MT1-MMP (1:50; ab51074; Abcam) and mouse anti-human beta 1 integrin antibody (clone JB1A; 1:1,000; ref. 46) or phalloidin-AlexaFluor 488 conjugate (1:200; Invitrogen). Goat anti-rabbit IgG AlexaFluor 633 and goat anti-mouse AlexaFluor 546 (both Invitrogen) were used for secondary antibodies. Laser-scanning confocal images were acquired by using the Zeiss LSM 710 multispectral confocal microscope and Zen 2009 acquisition software (Carl Zeiss MicroImaging; LLC). Axial plane images were generated from Z-axis fluorescence image stacks acquired at intervals and with pinhole diameters optimal for a plan-apochromatic 63×/1.4 objective (equivalent to 1 AU). Laser lines and spectral windows specific for the selected fluorophores were used as recommended by the manufacturer.

Protein extraction and Western blot analysis

FTC-133–RLN2/FTC-133–EGFP transfectants and untransfected FTC-133 were seeded in 6-well plates at 8 × 104 cells/well and cultured for 24 hours in serum-free medium. Supernatants were collected 72 hours later and centrifuged at 4,000 × g for 30 minutes at 4°C to pellet remaining cells. Cellular proteins were isolated with 2× extraction buffer (125 mmol/L Tris-HCl pH 6, 8; 4% SDS; 20% glycerol; 10% (v/v) mercaptoethanol (ME); 2% (v/v) bromophenol blue) with the addition of a protease inhibitor cocktail (all Sigma-Aldrich). Proteins (30 μg) were run on 12% SDS-PAGE gels and blotted onto Hybond-ECL nitrocellulose membranes (Amersham). Membranes were blocked for 1 hour in PBS blocking buffer containing 0.1% Tween 20, 3% nonfat dry milk, and 2% bovine serum albumin (BSA) for the detection of MMP2 and TBS containing 0.1% Tween 20 and 5% nonfat dry milk for MMP14 and TIMP2 immunodetection. Membranes were incubated for 1 hour in blocking buffer at RT and overnight at 4°C with specific primary antibodies to human TIMP2 (Santa Cruz Biotechnology), MMP2, and MT1-MMP (both Calbiochem). Membranes were washed 3 times in buffered saline followed by a 1 hour incubation at RT with the secondary antibodies, either goat anti-mouse IgG conjugated HRP (for MMP2; Santa Cruz Biotechnology) or goat anti-rabbit conjugated HRP (for TIMP2 and MT1-MMP; Dianova). For lamin A/C detection, MMP14 blots were stripped in 0.2 NaOH for 15 minutes and washed 3 times with TBST prior to blocking with 5% milk in TBST, pH 7.6, for 1 hour at room temperature. Membranes were probed overnight at 4°C with an anti-lamin A/C antibody (1:200; SC-6215; Santa Cruz, CA). Membranes were washed 3 times in blocking buffer followed by 1 hour incubation with a rabbit anti-goat conjugated HRP (1:10,000; SC-2922; Santa Cruz). Blots were visualized by using an ECL kit (Thermo Fisher). Beta-actin detection was used as a marker for equal protein loading per lane. Densitometry data were obtained by using Kodak Digital Science 1D software with the Kodak Digital Science Electrophoresis Documentation and Analysis System 120. The ratio in the levels of each of the probed protein versus β-actin in FTC-133–EGFP cells served as 100% reference for the determination of the relative contents of these proteins in the transfectants.

Pseudopodia assay

Pseudopodia assays were conducted with slight modifications as described previously (47). The day before the assay, cells were washed twice with 1× PBS and grown in serum-free medium overnight. Purification of pseudopodia was done in 6-well plates, with insert filters containing 1.0 μm porous track-etched polyethylene terephthalate membranes (BD Biosciences). Each filter was seeded with 7.5 × 105 cells and incubated in a 5% CO2 humidified incubator at 37°C for 1 hour to allow the tips of pseudopodia to reach the underside of the filter. Pseudopodia were isolated by dipping the underside of each filter in 100 μL of lysis buffer (50 mmol/L Tris-HCl, pH 7.4, 3 mmol/L NaCl, 2 mmol/L MgCl2, 0.1% SDS, 0.1% NP-40 (Igepal), protease inhibitor cocktail; all Sigma) for 10 seconds. The remaining cell body was collected by adding 100 μL of lysis buffer onto the top of each filter. Pseudopodia and cell body lysates from 3 filters were pooled separately and subjected to 3 freeze/thaw cycles (dry ice for 3 minutes and thawed at 37°C for 1 minute). Lysates were centrifuged at 13,000 × g for 20 minutes at 4°C and supernatants of pseudopodia lysates were dried in a SpeedVac, resuspended in double distilled water to make a 10× concentrated pseudopodia supernatant and protein concentration was measured by using the Micro BCA Protein Assay Kit (Pierce). Proteins from the cell body and the pseudopodia were separated on a 12% gradient gel (Invitrogen) and transferred to nitrocellulose membranes (Thermo Fisher) for the detection of MMP2 and MT1-MMP.

MMP2/MMP9 activity assay

The enzymatic activity of secreted MMP2 derived from stable FTC-133 transfectants and untransfected FTC-133 was determined by using the MMP2/MMP9 activity test (Calbiochem) according to the manufacturers' instructions. Cells (5 × 103) were seeded per 96-well and grown overnight before cells were cultured for 24 hours in serum-free medium. Supernatants collected from all transfectants and from untransfected FTC-133 treated with recombinant RLN2 (100 ng/mL) were incubated at 37°C with 20 μmol/L of the substrate. The enzymatic activity was measured after 30 minutes at 280 nm.

Migration assay

Migration assays were conducted in 24-well Transwell chambers (Costar). The upper and lower culture chambers were separated by a polycarbonate membrane with 8-μm pore size. Membranes were coated with 1 mg/mL gelatine, collagen A (collagen type I), or fibronectin (all Biochrom). FTC-133, FTC-133–RLN2, and FTC-133–EGFP (each at 1 × 104) in serum-free medium were seeded onto the filter and incubated for 24 hours at 37°C in a CO2 incubator, in the presence and absence of RLN2 at 100 ng/mL. We used the general MMP inhibitor GM-6001 (Ilomastat; Chemicon/Millipore) at a concentration that inhibits MMP2 (0.5 nmol/L). FTC-133 cells did not express MMP9 (data not shown). Cells that had migrated to the underside of the filter were washed with PBS, fixed for 10 minutes in ice-cold methanol: PBS [1:1 (v/v); Merck], followed by a 20 minutes wash in ice-cold methanol and staining in 0.1% toluidine blue solution (Merck) in sodium carbonate (Roth). Stained cells were counted by light microscopy (Zeiss) in 5 separate fields (magnification ×100) per filter.

Zymography assays

The 24-well plates were coated with 1 mg/mL gelatine (Biochrom) and dried overnight in sterile conditions. FTC-133–RLN2 and FTC-133–EGFP transfectants (2 × 104) were seeded onto the coated plates and cultured for 48 hours at 37°C in a CO2 incubator. Proteins were stained with Scarlet Bibrich solution (Wako). Stained plates were visualized by light microscopy (Zeiss). For collagen zymography, collagen-I from bovine Achilles tendon (1 g; Sigma) was dissolved in 0.2 mol/L acetic acid and stirring at 4°C overnight. The collagen solution was neutralized and adjusted to pH 7.2 with concentrated NaOH and adjusted with 50 mmol/L Tris-HCl, pH 7.4. Ten microgram of total protein extracts of FTC-133–RLN2 and FTC-133–EGFP transfectants were mixed with 4× extraction buffer (for composition see Protein extraction and Western blot analysis; without mercaptoethanol) and subjected to electrophoresis on a 10% SDS-PAGE copolymerized with 0.035% v/v of solubilized collagen. The gel was washed for 1 hour at RT with 2.5% Triton X-100 in 50 mmol/L Tris pH 7.4, 5 mmol/L CaCl2, 1 μmol/L ZnCl2, rinsed in distilled water, and incubated at 37°C for 16 hours in 0.01 mol/L sodium phosphate buffer, pH 7.4, containing 5 mmol/L CaCl2 followed by staining with 0.5% Coomassie brilliant blue R250 in 30% ethanol, 10% acetic acid for 30 minutes and then destained with 30% ethanol and 10% acetic acid. Collagenolytic activities were detected as unstained bands against the background of Coomassie brilliant blue–stained collagen.

Statistical analysis

Statistical analysis was carried out with SPSS 12.0 and Excel software and all experimental parameters were calculated for statistical significance by using Student's t test. The values of P < 0.05 were considered statistically significant. Densitometric analysis was carried out by using the Kodak Digital Science 1D software.

Immunoreactive MT1-MMP and MMP2 are increased and colocalized with RLN2 in human thyroid carcinoma

We analyzed human goiter tissues and PTC, FTC, and UTC thyroid cancer tissues for the presence of immunoreactive MMP2, MT1-MMP, and RLN2. Goiter tissues showed very weak to absent immunostaining for MMP2 (Fig. 1A), MT1-MMP (Fig. 1B), and RLN2 (Fig. 1C). Only 1 of 15 (7%) and 3 of 10 (30%) of goiter cases showed weak staining for MMP2 and MT1-MMP, respectively. None were positive for RLN2 as described previously (7). All thyroid carcinoma tissues investigated (PTC, FTC, and UTC) displayed increased presence of immunoreactive MMP2 (Fig. 1D, G, and J), MT1-MMP (Fig. 1E, H, and K), and RLN2 (Fig. 1F, I, and L). When the specific antiserum was replaced with a nonimmune serum of the same species, no specific immunostaining was obtained in any of the tissues (Fig. 1a–l). RLN2 colocalized with MMP2- and MT1-MMP–producing cells. We were unable to find a correlation for immunoreactive MMP2 or MT1-MMP thyroid cancer tissues with either gender, age, or tumor size, lymphatic/systemic metastasis (pTNM) status of the patients (Table 2). No correlation with these clinicopathologic criteria was found for the presence of both MMP2 and MT1-MMP in the few thyroid cancer tissue cases (3 PTC, 6 FTC, and 5 UTC) which we were able to test for both MMPs (Table 2). The immunohistochemical staining results suggested that MMP2 may be a suitable marker for thyroid cancer because only a minority of goiter tissue (<10%) but a high percentage (61%–77%) of thyroid cancer tissues expressed immunoreactive MMP2. MT1-MMP was present in both goiter (30%, weak staining) and thyroid cancer tissues (67%–82%; Table 3). As shown previously, MT1-MMP and MMP2 were present in fetal trophoblast cells of human placenta used as positive control (data not shown; ref. 48).

Figure 1.

Tissues of human goiter (n = 25), PTC (n = 30), FTC (n = 27), and UTC (n = 24) were investigated for the presence of immunoreactive MMP2, MT1-MMP, and RLN2. Representative immunohistochemical images of human thyroid tissues are shown immunostained for MMP2, MT1-MMP, and RLN2. Goiter tissue (A–C, a–c) showed no staining for MMP2 (A) and RLN2 (C) or inconsistent and very weak staining for MT1-MMP (B). The majority of PTC (D–F), FTC (G–I), and UTC (J–L) investigated displayed strong immunostaining for MMP2 (D, G, and J), MT1-MMP (E, H, and K), and RLN2 (F, I, and L), respectively. The insets in each image indicate the absence of staining when specific primary antibodies were replaced with a species-specific nonimmune serum (a–l).

Figure 1.

Tissues of human goiter (n = 25), PTC (n = 30), FTC (n = 27), and UTC (n = 24) were investigated for the presence of immunoreactive MMP2, MT1-MMP, and RLN2. Representative immunohistochemical images of human thyroid tissues are shown immunostained for MMP2, MT1-MMP, and RLN2. Goiter tissue (A–C, a–c) showed no staining for MMP2 (A) and RLN2 (C) or inconsistent and very weak staining for MT1-MMP (B). The majority of PTC (D–F), FTC (G–I), and UTC (J–L) investigated displayed strong immunostaining for MMP2 (D, G, and J), MT1-MMP (E, H, and K), and RLN2 (F, I, and L), respectively. The insets in each image indicate the absence of staining when specific primary antibodies were replaced with a species-specific nonimmune serum (a–l).

Close modal
Table 3.

Distribution profile of immunoreactive MMP2 and MT1-MMP in 106 human thyroid tissues investigated

TissueProtein expression (%)
MMP2MT1-MMP
Goiter 30 
PTC 77 82 
FTC 73 64 
UTC 61 67 
TissueProtein expression (%)
MMP2MT1-MMP
Goiter 30 
PTC 77 82 
FTC 73 64 
UTC 61 67 

RLN2 enhances the expression of MMP2 and MT1-MMP

We show that RLN2 increased the transcriptional activity, protein production, and secretion of gelatinase A/MMP2 in previously described RLN2-expressing stable transfectants of the human follicular thyroid carcinoma FTC-133. MT1-MMP protein in either the 64 kDa proform (Fig. 2A) and 54 kDa active form (Fig. 2B) were upregulated in 2 out of 3 or all 3 RLN2 transfectants, respectively, when compared with the EGFP control. All RLN2 clones, but not the EGFP mock, displayed strong immunoreactive bands at 72 kDa corresponding to MMP2 (Fig. 2C). TIMP2 is part of an MMP2 activation complex, which also includes proMMP2 and MT1-MMP (49). The expression of TIMP2 detected as a single immunoreactive protein at 21 kDa did not change significantly in all EGFP and RLN2 clones tested (Fig. 3D). The expression levels determined for MT1-MMP, MMP2, and TIMP2 correlated well with the densitometric measurements derived from the Western blot analysis for MT1-MMP proform (64 kDa) and active form (54 kDa), MMP2, and TIMP2 (data not shown). The upregulation in MT1-MMP and MMP2 correlated with RLN2 production as determined by RLN2 ELISA in the FTC-133–RLN2 stable transfectants (70–95 pg/mL) and negligible levels of RLN2 in the EGFP mock cells (data not shown).

Figure 2.

Results of the quantitative Western blot analysis and representative blots (n = 3) are shown for MT1-MMP (A, B), MMP2 (C), TIMP2 (D), and TIMP3 (E, F). We observed an increase in protein levels for MT1-MMP 64 kDa proform (A), the MT1-MMP 54 kDa active form (B), and MMP2 (C) in RLN2 transfectants when compared with mock cells. The upregulation of RLN2 did not significantly alter TIMP2 protein levels (D). Western blot analysis revealed a consistent downregulation of human TIMP3 dimer (48 kDa; E) and monomer (27 kDa; F) in all RLN2 transfectants as compared with mock cells. Results from 3 independent experiments with SDs are shown. Statistical analysis was done by employing SPSS software and paired Student's t test. The value of P < 0.05 was considered statistically significant as indicated with a star (*). Abbreviation: cl, clone.

Figure 2.

Results of the quantitative Western blot analysis and representative blots (n = 3) are shown for MT1-MMP (A, B), MMP2 (C), TIMP2 (D), and TIMP3 (E, F). We observed an increase in protein levels for MT1-MMP 64 kDa proform (A), the MT1-MMP 54 kDa active form (B), and MMP2 (C) in RLN2 transfectants when compared with mock cells. The upregulation of RLN2 did not significantly alter TIMP2 protein levels (D). Western blot analysis revealed a consistent downregulation of human TIMP3 dimer (48 kDa; E) and monomer (27 kDa; F) in all RLN2 transfectants as compared with mock cells. Results from 3 independent experiments with SDs are shown. Statistical analysis was done by employing SPSS software and paired Student's t test. The value of P < 0.05 was considered statistically significant as indicated with a star (*). Abbreviation: cl, clone.

Close modal
Figure 3.

Supernatants of RLN2 transfectants (A) and FTC-133 treated with human recombinant RLN2 (100 ng/mL; B) showed increased secretion of bioactive MMP2 as determined with a specific fluorescently labeled substrate. The enhanced enzymatic cleavage of this substrate observed in RLN2 clones was inhibited in the presence of the MMP inhibitor GM6001 (C). Treatment of transfectants with ethanol (EtOH) used as solvent for GM6001 had no effect (C). Collagen-I in-gel zymography revealed an upregulation of collagenolytic activity at around 72 kDa resembling proMMP2 specifically in RLN2 transfectants (D). For in situ zymography, EGFP mock and RLN2 clone 4 transfectants were embedded in gelatine matrix. A pronounced pericellular halo effect reflecting the pericellular matrix degradation was observed with cells expressing RLN2, but not EGFP mock cells (E). When the RLN2 expressing cells were cultured in the presence of the MMP inhibitor GM6001 at a concentration (0.5 nmol/L) sufficient for selective inhibition of MMP2, the pericellular degradation was abolished (E). All experiments were repeated at least 3 times and a representative result with SDs is shown. Statistics were done with SPSS software and paired Student's t test. The value of P < 0.05 was considered statistically significant as indicated with a star (*). Abbreviation: cl, clone; MW, molecular weight.

Figure 3.

Supernatants of RLN2 transfectants (A) and FTC-133 treated with human recombinant RLN2 (100 ng/mL; B) showed increased secretion of bioactive MMP2 as determined with a specific fluorescently labeled substrate. The enhanced enzymatic cleavage of this substrate observed in RLN2 clones was inhibited in the presence of the MMP inhibitor GM6001 (C). Treatment of transfectants with ethanol (EtOH) used as solvent for GM6001 had no effect (C). Collagen-I in-gel zymography revealed an upregulation of collagenolytic activity at around 72 kDa resembling proMMP2 specifically in RLN2 transfectants (D). For in situ zymography, EGFP mock and RLN2 clone 4 transfectants were embedded in gelatine matrix. A pronounced pericellular halo effect reflecting the pericellular matrix degradation was observed with cells expressing RLN2, but not EGFP mock cells (E). When the RLN2 expressing cells were cultured in the presence of the MMP inhibitor GM6001 at a concentration (0.5 nmol/L) sufficient for selective inhibition of MMP2, the pericellular degradation was abolished (E). All experiments were repeated at least 3 times and a representative result with SDs is shown. Statistics were done with SPSS software and paired Student's t test. The value of P < 0.05 was considered statistically significant as indicated with a star (*). Abbreviation: cl, clone; MW, molecular weight.

Close modal

Of the 4 TIMP protein members identified by Western blot in the FTC-133 transfectants, RLN2 caused an exclusive and marked reduction in TIMP3 isoforms at 48 kDa (Fig. 2E) and 27 kDa (Fig. 2F), without affecting the levels of TIMP1 or TIMP4 protein (Table 4). Expression of other members of the MMP family known to be associated with cancer, including MMP1 (collagenase 1), MMP3 (stromelysin 1), MMP8 (collagenase 2), MMP9 (gelatinase B), MMP10 (stromelysin 2), and MMP13 (collagenase 3) was tested in the FTC-133–EGFP and FTC-133–RLN2 transfectants by using reverse transcriptase PCR (RT-PCR). Transcripts were exclusively observed for MMP13 and remained unaltered in the presence of RLN2 (Table 4). FTC-133 cells were devoid of MMP1, 3, 8, 9, and 10 transcripts (Table 4).

Table 4.

Presence of selective MMPs and TIMP1–4 transcripts in the FTC-133–EGFP and RLN2 stable transfectants

MMP
MMP1 Negative 
MMP2 Upregulated in RLN2 clones 
MMP3 Negative 
MMP8 Negative 
MMP9 Negative 
MMP10 Negative 
MMP13 Present but unchanged 
MMP14 Upregulated in RLN2 clones 
 TIMP  
TIMP1 Present but unchanged 
TIMP2 Present but unchanged 
TIMP3 Downregulated in RLN2 clones 
TIMP4 Present but unchanged 
MMP
MMP1 Negative 
MMP2 Upregulated in RLN2 clones 
MMP3 Negative 
MMP8 Negative 
MMP9 Negative 
MMP10 Negative 
MMP13 Present but unchanged 
MMP14 Upregulated in RLN2 clones 
 TIMP  
TIMP1 Present but unchanged 
TIMP2 Present but unchanged 
TIMP3 Downregulated in RLN2 clones 
TIMP4 Present but unchanged 

NOTE: EGFP and RLN2 transfectants showed similar mRNA profiles, whereas quantitative differences were obtained for MMP2, MT1-MMP, and TIMP3, with the 2 MMPs being upregulated and TIMP3 downregulated in RLN2 clones. MMP13 (collagenase-3) transcripts were not altered between RLN2 and EGFP clones.

RLN2 enhances secretion of active MMP2 and collagenolytic activity by human thyroid cancer cells

Supernatants of RLN2 transfectants contained significantly more bioactive MMP2 when compared with the mock control as determined by MMP2/MMP9 bioactivity assay (Fig. 3A). The enzymatic processing of the substrate was likely caused by the increased MMP2 activity because RLN2 transfectants did not express MMP9 (Table 4). The addition of the general MMP inhibitor GM6001 abolished the enhanced proteolytic cleavage of the fluorescently labeled substrate by the RLN2 transfectants, but not mock transfectants (Fig. 3C). Untransfected FTC-133 thyroid cancer cells exposed to recombinant RLN2 also showed enhanced secretion of active MMP2 (Fig. 3B). Furthermore, protein extracts of RLN2 transfectants (clone 10) subjected to in-gel collagen-I zymography revealed a specific band of digested collagen-I at 72 kDa likely resembling MMP2 (Fig. 3D). In situ zymography assays also showed that the transfectants cultured in gelatine matrices differed in their ability to degrade this matrix as evidenced by the larger bright pericellular halos around RLN2 transfectants, which could be inhibited with the general MMP inhibitor GM6001 at a concentration known to inhibit MMP2 (Fig. 3E).

RLN2 increases migration through collagen matrix and recruitment of MT1-MMP to the leading edge of migration in human follicular thyroid cancer cells

We conducted migration assays to determine whether the observed increase in MMP2 and MT1-MMP production and MMP2 activity resulted in enhanced penetration of the RLN2 transfectants through collagen type I matrices. All 3 RLN2 transfectants tested (Fig. 4A) and FTC-133 cells treated with human recombinant RLN2 (Fig. 4B) were able to penetrate collagen-I matrix and migrate to the underside of the filter in significantly higher numbers than mock and untreated controls, respectively. In addition to collagen I, MMP2 and MT1-MMP are capable of digesting other extracellular matrix compounds, including gelatine and fibronectin (50). We observed a positive trend, but no significant increase, in migration through gelatine matrices and no change in the ability of clones to invade fibronectin matrix (data not shown).

Figure 4.

Migration assays were conducted by using filters with a pore size of 8 μm covered with a matrix composed of collagen type I. Cells were seeded on top of the matrix and allowed to penetrate the matrix to reach the underside of the filter where they were counted (see Materials and Methods for details). All RLN2 transfectants displayed superior penetration of the collagen-I matrix as compared with mock cells (A). FTC-133 treated with human recombinant RLN2 (100 ng/mL) also showed a statistically significant increase in migration through the collagen matrices (B). In pseudopodia assays, we used membranes with a pore size of 1 μm to analyze protein extracts obtained exclusively from cellular protrusions at the leading migrating edge of cells (EGFP and RLN2 clone 10). Quantitative Western blot analysis (C, D) of protein extracts obtained from invadopodia (bottom of the filter) and the rest of the cell body (top of the filter) showed a significant increase in MT1-MMP in the cell body and an increased concentration (borderline significant) of MT1-MMP in the pseudopodia of RLN2 clones (C, D). The representative Western blot shows that nuclear protein lamin A/C was exclusively detected in protein extracts derived from the top of the filter (C). The absence of lamin A/C in pseudopodia protein extracts excluded contamination of the invadopodia protein fraction with cell nuclei/bodies from the filter top (C). All experiments were repeated at least 3 times and a representative result is shown. Statistics were done with SPSS software and paired Student's t test. The value of P < 0.05 was considered significant (*). Abbreviation: cl, clone.

Figure 4.

Migration assays were conducted by using filters with a pore size of 8 μm covered with a matrix composed of collagen type I. Cells were seeded on top of the matrix and allowed to penetrate the matrix to reach the underside of the filter where they were counted (see Materials and Methods for details). All RLN2 transfectants displayed superior penetration of the collagen-I matrix as compared with mock cells (A). FTC-133 treated with human recombinant RLN2 (100 ng/mL) also showed a statistically significant increase in migration through the collagen matrices (B). In pseudopodia assays, we used membranes with a pore size of 1 μm to analyze protein extracts obtained exclusively from cellular protrusions at the leading migrating edge of cells (EGFP and RLN2 clone 10). Quantitative Western blot analysis (C, D) of protein extracts obtained from invadopodia (bottom of the filter) and the rest of the cell body (top of the filter) showed a significant increase in MT1-MMP in the cell body and an increased concentration (borderline significant) of MT1-MMP in the pseudopodia of RLN2 clones (C, D). The representative Western blot shows that nuclear protein lamin A/C was exclusively detected in protein extracts derived from the top of the filter (C). The absence of lamin A/C in pseudopodia protein extracts excluded contamination of the invadopodia protein fraction with cell nuclei/bodies from the filter top (C). All experiments were repeated at least 3 times and a representative result is shown. Statistics were done with SPSS software and paired Student's t test. The value of P < 0.05 was considered significant (*). Abbreviation: cl, clone.

Close modal

To further elucidate the potential mechanism by which RLN2 may facilitate the enhanced migration of thyroid cancer cells through collagen matrices, we isolated pseudopodial membrane protrusions extending into 1-μm wide filter pores after a 1 hour migration time. The narrow filter pores only permitted the collection of invadopodia rather than the whole cell body which stayed on top of the filter as shown by the exclusive presence of the nuclear protein lamin A/C in protein preparations from the top, but not the bottom, of the filter (Fig. 4C). This pseudopodia assay allowed us to determine proteins present at the leading edge of migration (bottom of filter) and the rest of the cell body (top of filter). Western blot analysis of protein extracts from the invadopodia (bottom of filter) and rest of the cell body (top of filter) confirmed the increased overall production of MT1-MMP in RLN2 clones as compared with EGFP clones (data not shown; Fig. 2A and B). Protein levels of MT1-MMP were increased in the invadopodia fraction (just below significance) and significantly in the cell body on top of the filter as shown by quantitative Western blot analysis (Fig. 4C and D). Next, we used multicolor fluorescent confocal microscopy on RLN2 and EGFP transfectants to visualize the presence of MT1-MMP in invadopodia invading the 3-μm wide filter pores. A monoclonal antibody against beta 1 integrin was used as a membrane marker. We observed pronounced immunofluorescent staining for both MT1-MMP and beta 1 integrin in the invadopodia of RLN2 clones when compared with EGFP clones at the same exposure time (Fig. 5A). Thus, one way of RLN2 to ensure increased collagen invasion was to increase the concentration of MT1-MMP within the cells and the invadopodia as compared with EGFP clones. In the invadopodia, though, MT1-MMP was distributed over a much smaller surface area (Fig. 5A) and, thus, was more concentrated as shown in the confocal fluorescent images of the membrane pore-penetrating invadopodia (Fig. 5A). RLN2 may alter the actin cytoskeleton and this may contribute to the increased invasion of ECM. Staining of F-actin by the fungal toxin phalloidin revealed distinct differences in the actin cytoskeletal organization in EGFP and RLN2 transfectants, with the latter displaying stress fibers (Fig. 5B). Increased stress fiber formation was also detected in the 3-dimensional (3D) confocal images of invadopodia derived from the RLN2 transfectants (Fig. 5C), suggesting changes to the cytoskeletal actin architecture and increased presence of MT1-MMP in pseudopodia to contribute to invadopodia formation and ECM-penetrating activity.

Figure 5.

Fluorescent microscopy was used to visualize the cellular distribution of MT1-MMP and beta 1 integrin (A) as well as MT1-MMP and F-actin (B, C) in RLN2 (clone 10) and EGFP thyroid cancer transfectants. 3D reconstitution of z-stagged confocal fluorescent images were obtained for invadopodia from transfectants that had been plated onto filter membranes with 3 μm pore size. Invadopodia are viewed from below the filter (top image) and from the side (bottom image), with the main cell body placed on top of the membrane and invadopodia protruding through the membrane pore onto the underside of the filter. Under identical experimental conditions, RLN2 transfectants displayed larger-sized pseudopodia with high-intensity fluorescence for MT1-MMP, likely aiding enhanced local collagenolytic invasiveness of the RLN2 clones as compared with EGFP clones (red channel: MT1-MMP, green channel: beta 1 integrin, yellow channel: merged images; A). Fluorescent staining of filamentous actin with phalloidin-AlexaFluor 546 conjugate revealed extensive stress fiber formation in the presence of RLN2, whereas EGFP clones showed a subcortical network of F-actin (B). 3D confocal fluorescent images confirmed the presence of green phalloidin 488 nm stained actin stress fibers located in the neck of pseudopodia of the RLN2 clones as seen in cross-sections (top image) and viewed from the side (bottom image; C). Bleb-like structures were detected on phalloidin staining of the actin cytoskeleton in invadopodia derived from EGFP clones (C). Red channel: MT1-MMP; green channel: F-actin; yellow channel: merged images; all measurement bars of confocal images: 10 μm. Abbreviation: cl, clone.

Figure 5.

Fluorescent microscopy was used to visualize the cellular distribution of MT1-MMP and beta 1 integrin (A) as well as MT1-MMP and F-actin (B, C) in RLN2 (clone 10) and EGFP thyroid cancer transfectants. 3D reconstitution of z-stagged confocal fluorescent images were obtained for invadopodia from transfectants that had been plated onto filter membranes with 3 μm pore size. Invadopodia are viewed from below the filter (top image) and from the side (bottom image), with the main cell body placed on top of the membrane and invadopodia protruding through the membrane pore onto the underside of the filter. Under identical experimental conditions, RLN2 transfectants displayed larger-sized pseudopodia with high-intensity fluorescence for MT1-MMP, likely aiding enhanced local collagenolytic invasiveness of the RLN2 clones as compared with EGFP clones (red channel: MT1-MMP, green channel: beta 1 integrin, yellow channel: merged images; A). Fluorescent staining of filamentous actin with phalloidin-AlexaFluor 546 conjugate revealed extensive stress fiber formation in the presence of RLN2, whereas EGFP clones showed a subcortical network of F-actin (B). 3D confocal fluorescent images confirmed the presence of green phalloidin 488 nm stained actin stress fibers located in the neck of pseudopodia of the RLN2 clones as seen in cross-sections (top image) and viewed from the side (bottom image; C). Bleb-like structures were detected on phalloidin staining of the actin cytoskeleton in invadopodia derived from EGFP clones (C). Red channel: MT1-MMP; green channel: F-actin; yellow channel: merged images; all measurement bars of confocal images: 10 μm. Abbreviation: cl, clone.

Close modal

In this study, we have shown the presence of MMP2 (gelatinase A) and MT1-MMP (MMP14) in human benign and malignant thyroid tissues. Of all the MMP members (MMP1, 2, 8, 9, 10, 13, and 14) and TIMPs (TIMP1–4) investigated here, RLN2 specifically induced the expression of MMP2 and MT1-MMP in human follicular thyroid carcinoma cells. Production of proMMP2 and MT1-MMP has previously been shown in human PTC and MT1-MMP expression levels correlated with the proMMP2 activation ratio, thyroid cancer cell invasion, and PTC lymph node metastasis (31, 35). Similarly, a positive correlation was reported for the presence of immunoreactive MMP2, but not TIMP2, and bad prognosis of patients with medullary thyroid cancer, a tumor derived from C-cells in the thyroid gland (51). Expression of gelatinase A (MMP2), gelatinase B (MMP9), and MT-MMP1 has also been reported in breast, colon, lung, skin, ovary, and prostate cancer and is associated with enhanced invasion and decreased overall survival (52–54). MT1-MMP is important for cell-surface activation of proMMP2 (49) and the MT1-MMP C-terminal domain is essential for promoting cell migration (55, 56). Both MMP2 and MT1-MMP are major players in tumor angiogenesis and linked to lymphangiogenesis, lymphatic invasion, and lymph node metastasis (53, 57). The targeted deletion of MMP2 impairs lymphatic vasculature and the inhibition of MMP2 and MT1-MMP attenuates the formation of both new blood and lymphatic vessels and reduces lymph node metastasis (58). The angiogenic phenotype of MT1-MMP–producing cells is associated with the upregulation of VEGF (59). Mice deficient in MT1-MMP develop skeletal deformations as a result of delayed vascular invasion of the cartilage (60) and overexpression of MT1-MMP induces tumorigenic mammary gland abnormalities in transgenic mice (61). Our finding of an upregulation of MT1-MMP/MMP2 coincided with the RLN2-induced downregulation of TIMP3. TIMP3 is known to inhibit VEGF-mediated angiogenic signalling and lack of TIMP3 has been shown to cause an increase in abnormal vessel formation (62). Our findings may in part explain the increased angiogenesis and growth we had reported previously in xenografts derived from the FTC-133 transfectants expressing RLN2 used in this study (7).

Currently, few factors have been identified which can affect the expression of MMP2 and MT1-MMP. Epidermal growth factor (EGF) was shown to mediate the invasion of differentiated thyroid cancer cells by an EGF receptor–mediated increase in the expression and activation of MMP2 (35). The membrane-bound glycoprotein and MMP1 receptor EMMPRIN (extracellular matrix metalloproteinase inducer, CD147) was reported to induce MMP1, 2, 3, and MT1-MMP, but not MMP9 expression (63). Here, we have identified RLN2 as a novel potent inducer of MMP2 and MT1-MMP in human follicular thyroid cancer cells. RLN2 is strongly and specifically expressed in thyroid cancer tissues and both RLN receptors, RXFP1 and RXFP2, are expressed in the thyroid gland (7, 64, 65). Exposure to RLN2 caused a significant RXFP1-mediated increase in the ability of human thyroid cancer cells to invade elastin matrices as a result of the enhanced production and secretion of cathepsin-L, a potent elastinolytic lysosomal hydrolase (7). Our discovery of MMP2 and MT1-MMP as novel RLN2 targets in human thyroid cancer cells identifies RLN2 as an important multifunctional regulator of a range of potent proteases with overlapping ECM-degrading qualities known to facilitate various stages of (thyroid) cancer cell invasiveness (35, 66–68).

RLN exerts autocrine/paracrine actions on tumor cell migration/invasion and angiogenesis in different human cancer models (1, 5). Follicular thyroid cancer cells and PC-3 prostate carcinoma cells expressing RLN2 displayed enhanced xenograft growth and angiogenesis (7, 69, 70). The importance of the RLN/RXFP1 ligand receptor system for tumor growth is also highlighted by 2 sets of in vivo experiments. We and others have recently shown that the intratumoral injection of a RLN antagonist peptide (71) or RXFP1 antisense coated nanoparticles (72) results in the specific in vivo silencing of RXFP1 signaling pathways in xenografts of human prostate cancer cells and causes a marked reduction in tumor size. High RLN2 expression has been associated with advanced endometrial cancer (EC) tumor stages, myometrial EC invasion, and a significantly shorter overall survival of women with EC (8). The stimulation of the human EC cell lines HEC-1B and KLE with RLN2 resulted in the RXFP1-mediated release of increased levels of MMP2 in KLE cells and MMP9 in HEC-1B cells (8). RLN2 affects breast cancer growth and invasiveness in a cell type-specific manner (73, 74). RLN2 is upregulated in neoplastic breast lesions (75) and systemic RLN2 levels were reported to be higher in women with metastatic breast cancer (10). The RLN2-induced increase in the secretion of MMP2, MMP7, and MMP9 and the upregulation of MMP2, MMP9, MMP13, and MT1-MMP transcripts resulted in enhanced in vitro invasiveness of the human breast cancer cell lines SK-BR3 and MCF7 (20). A recent study failed to show a correlation between plasma RLN levels and the presence of RLN, RXFP1, or MMP2 in the local tumor environment (76). Instead, these authors reported statistically significant correlations for the presence of RLN and RXFP1 and for RXFP1 and MMP2 in canine mammary tumor tissues and emphasized the importance of a locally active RLN/RXFP1 system for ECM remodeling in tumor tissues (76).

Currently, little information is available on the local effects of RLN2 in tumor cells and tissues. The MMP2/MT1-MMP/TIMP2 complex and αVβ3 integrin were reported to localize to specific caveolin microdomains, invadopodia, at the leading edge of migration (77, 78). This prompted us to elucidate the effects of RLN2 on the local distribution of MT1-MMP at the leading edge of migration in human follicular thyroid carcinoma cells. Pseudopodia assays resemble a modified motility assay with the ability to isolate cell components exclusively derived from the invading membrane tip of motile cells (47). We combined pseudopodia assays with Western blot analysis and confocal fluorescent microscopy to gain quantitative and visual information on the distribution of MT1-MMP at the leading migrating edge. The higher level of immunoreactive MT1-MMP in the RLN2 transfectants caused more MT1-MMP to be concentrated in the small volume of the invadopodia, likely contributing to high local levels of collagenolytic activity with resulting more effective collagen matrix degradation by RLN2 transfectants as compared with the EGFP clones. This “extrinsic” effect of RLN2, which affected the proteolytic activity at the leading migrating edge of tumor cells and resulted in ECM remodeling in the pericellular space, was not the only mechanism used by this hormone to promote tumor cell invasion. Exclusive to the RLN2 transfectants and not observed in EGFP clones, RLN2 also facilitated invasion of thyroid cancer cells by an “intrinsic” mechanism that promoted the formation of actin stress fibers by an as yet unidentified mechanism. Confocal images revealed that these actin stress fibers participated in the formation of the invadopodia in RLN2 transfectants. The remodeling of the actin cytoskeletal architecture and formation of bundles of actomyosin stress fibers are important steps in invadopodia formation and facilitate cell migration and ECM degradation (72, 79).

In summary, the specific upregulation and colocalization of RLN2, MT1-MMP, and MMP2 in human thyroid cancer tissues as well as the contribution of RLN2-induced actin stress fibers to the formation of MT1-MMP–rich invadopodia that cause local collagenolytic remodeling of the surrounding ECM microenvironment identifies RLN2 as a novel and versatile pathogenic factor in thyroid cancer invasiveness.

No potential conflicts of interest were disclosed.

C. H-Vu and T. Klonisch thank the Deutsche Forschungsgemeinschaft (DFG) for their financial support (KL1249/5-1; 5-2). T. Klonisch and S. H-Klonisch thank the Natural Sciences and Engineering Council of Canada (NSERC), the Surgery Research Fund, and the Manitoba Institute for Child Health (MICH) for their generous support. The authors thank Corthera Inc. for their generous gift of recombinant human relaxin.

This work was financially supported by Deutsche Forschungsgemeinschaft (DFG; KL1249/5-1; 5-2), Wilhelm Roux Program, Medical Faculty, Martin Luther University Halle-Wittenberg, Natural Sciences and Engineering Research Council of Canada, and the Manitoba Health Research Council, Canada.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Silvertown
JD
,
Summerlee
AJ
,
Klonisch
T
. 
Relaxin-like peptides in cancer
.
Int J Cancer
2003
;
107
:
513
9
.
2.
Parry
LJ
,
Vodstrcil
LA
. 
Relaxin physiology in the female reproductive tract during pregnancy
.
Adv Exp Med Biol
2007
;
612
:
34
48
.
3.
Ma
S
,
Gundlach
AL
. 
Relaxin-family peptide and receptor systems in brain: insights from recent anatomical and functional studies
.
Adv Exp Med Biol
2007
;
612
:
119
37
.
4.
Conrad
KP
,
Novak
J
. 
Emerging role of relaxin in renal and cardiovascular function
.
Am J Physiol Regul Integr Comp Physiol
2004
;
287
:
R250
61
.
5.
Klonisch
T
,
Bialek
J
,
Radestock
Y
,
Hoang-Vu
C
,
Hombach-Klonisch
S
. 
Relaxin-like ligand-receptor systems are autocrine/paracrine effectors in tumor cells and modulate cancer progression and tissue invasiveness
.
Adv Exp Med Biol
2007
;
612
:
104
18
.
6.
Nistri
S
,
Bani
D
. 
Relaxin in vascular physiology and pathophysiology: possible implications in ischemic brain disease
.
Curr Neurovasc Res
2005
;
2
:
225
3
.
7.
Hombach-Klonisch
S
,
Bialek
J
,
Trojanowicz
B
,
Weber
E
,
Holzhausen
HJ
,
Silvertown
JD
, et al
Relaxin enhances the oncogenic potential of human thyroid carcinoma cells
.
Am J Pathol
2006
;
169
:
617
32
.
8.
Kamat
AA
,
Feng
S
,
Agoulnik
IU
,
Kheradmand
F
,
Bogatcheva
NV
,
Coffey
D
, et al
The role of relaxin in endometrial cancer
.
Cancer Biol Ther
2006
;
5
:
71
7
.
9.
Thompson
VC
,
Hurtado-Coll
A
,
Turbin
D
,
Fazli
L
,
Lehman
ML
,
Gleave
ME
, et al
Relaxin drives Wnt signaling through upregulation of PCDHY in prostate cancer
.
Prostate
2010
;
70
:
1134
45
.
10.
Binder
C
,
Simon
A
,
Binder
L
,
Hagemann
T
,
Schulz
M
,
Emons
G
, et al
Elevated concentrations of serum relaxin are associated with metastatic disease in breast cancer patients
.
Breast Cancer Res Treat
2004
;
87
:
157
66
.
11.
Masterson
R
,
Hewitson
TD
,
Kelynack
K
,
Martic
M
,
Parry
L
,
Bathgate
R
, et al
Relaxin down-regulates renal fibroblast function and promotes matrix remodelling in vitro
.
Nephrol Dial Transplant
2004
;
19
:
544
52
.
12.
Samuel
CS
,
Unemori
EN
,
Mookerjee
I
,
Bathgate
RA
,
Layfield
SL
,
Mak
J
, et al
Relaxin modulates cardiac fibroblast proliferation, differentiation, and collagen production and reverses cardiac fibrosis in vivo
.
Endocrinology
2004
;
145
:
4125
33
.
13.
Unemori
EN
,
Pickford
LB
,
Salles
AL
,
Piercy
CE
,
Grove
BH
,
Erikson
ME
, et al
Relaxin induces an extracellular matrix-degrading phenotype in human lung fibroblasts in vitro and inhibits lung fibrosis in a murine model in vivo
.
J Clin Invest
1996
;
98
:
2739
45
.
14.
Williams
EJ
,
Benyon
RC
,
Trim
N
,
Hadwin
R
,
Grove
BH
,
Arthur
MJ
, et al
Relaxin inhibits effective collagen deposition by cultured hepatic stellate cells and decreases rat liver fibrosis in vivo
.
Gut
2001
;
49
:
577
83
.
15.
Unemori
EN
,
Beck
LS
,
Lee
WP
,
Xu
Y
,
Siegel
M
,
Keller
G
, et al
Human relaxin decreases collagen accumulation in vivo in two rodent models of fibrosis
.
J Invest Dermatol
1993
;
101
:
280
5
.
16.
McDonald
GA
,
Sarkar
P
,
Rennke
H
,
Unemori
E
,
Kalluri
R
,
Sukhatme
VP
. 
Relaxin increases ubiquitin-dependent degradation of fibronectin in vitro and ameliorates renal fibrosis in vivo
.
Am J Physiol Renal Physiol
2003
;
285
:
F59
67
.
17.
Samuel
CS
,
Sakai
LY
,
Amento
EP
. 
Relaxin regulates fibrillin 2, but not fibrillin 1, mRNA and protein expression by human dermal fibroblasts and murine fetal skin
.
Arch Biochem Biophys
2003
;
411
:
47
55
.
18.
Zhao
L
,
Samuel
CS
,
Tregear
GW
,
Beck
F
,
Wintour
EM
. 
Collagen studies in late pregnant relaxin null mice
.
Biol Reprod
2000
;
63
:
697
703
.
19.
Xu
Q
,
Chakravorty
A
,
Bathgate
RA
,
Dart
AM
,
Du
XJ
. 
Relaxin therapy reverses large artery remodeling and improves arterial compliance in senescent spontaneously hypertensive rats
.
Hypertension
2010
;
55
:
1260
6
.
20.
Binder
C
,
Hagemann
T
,
Husen
B
,
Schulz
M
,
Einspanier
A
. 
Relaxin enhances in vitro invasiveness of breast cancer cell lines by up-regulation of matrix metalloproteases
.
Mol Hum Reprod
2002
;
8
:
789
96
.
21.
Mookerjee
I
,
Unemori
EN
,
Du
XJ
,
Tregear
GW
,
Samuel
CS
. 
Relaxin modulates fibroblast function, collagen production, and matrix metalloproteinase-2 expression by cardiac fibroblasts
.
Ann N Y Acad Sci
2005
;
1041
:
190
3
.
22.
McDonald
JK
,
Schwabe
C
. 
Relaxin-induced elevations of cathepsin B and dipeptidyl peptidase I in the mouse pubic symphysis, with localization by fluorescence enzyme histochemistry
.
Ann N Y Acad Sci
1982
;
380
:
178
86
.
23.
Wang-Lee
JL
,
Lenhart
JA
,
Ohleth
KM
,
Ryan
PL
,
Bagnell
CA
. 
Regulation of urokinase- and tissue-type plasminogen activator by relaxin in the uterus and cervix of the prepubertal gilt
.
J Reprod Fertil
1998
;
114
:
119
25
.
24.
Qin
X
,
Garibay-Tupas
J
,
Chua
PK
,
Cachola
L
,
Bryant-Greenwood
GD
. 
An autocrine/paracrine role of human decidual relaxin I. Interstitial collagenase (matrix metalloproteinase-1) and tissue plasminogen activator
.
Biol Reprod
1997
;
56
:
800
11
.
25.
Westermarck
J
,
Kahari
VM
. 
Regulation of matrix metalloproteinase expression in tumor invasion
.
FASEB J
1999
;
13
:
781
92
.
26.
Kessenbrock
K
,
Plaks
V
,
Werb
Z
. 
Matrix metalloproteinases: regulators of the tumor microenvironment
.
Cell
2010
;
141
:
52
67
.
27.
Herouy
Y
. 
Matrix metalloproteinases in skin pathology (Review)
.
Int J Mol Med
2001
;
7
:
3
12
.
28.
Koshikawa
N
,
Mizushima
H
,
Minegishi
T
,
Eguchi
F
,
Yotsumoto
F
,
Nabeshima
K
, et al
Proteolytic activation of heparin-binding EGF-like growth factor by membrane-type matrix metalloproteinase-1 in ovarian carcinoma cells
.
Cancer Sci
2011
;
102
:
111
6
.
29.
Kajita
M
,
Itoh
Y
,
Chiba
T
,
Mori
H
,
Okada
A
,
Kinoh
H
, et al
Membrane-type 1 matrix metalloproteinase cleaves CD44 and promotes cell migration
.
J Cell Biol
2001
;
153
:
893
904
.
30.
Basile
JR
,
Holmbeck
K
,
Bugge
TH
,
Gutkind
JS
. 
MT1-MMP controls tumor-induced angiogenesis through the release of semaphorin 4D
.
J Biol Chem
2007
;
282
:
6899
905
.
31.
Nakamura
H
,
Ueno
H
,
Yamashita
K
,
Shimada
T
,
Yamamoto
E
,
Noguchi
M
, et al
Enhanced production and activation of progelatinase A mediated by membrane-type 1 matrix metalloproteinase in human papillary thyroid carcinomas
.
Cancer Res
1999
;
59
:
467
73
.
32.
Zedenius
J
,
Stahle-Backdahl
M
,
Enberg
U
,
Grimelius
L
,
Larsson
C
,
Wallin
G
, et al
Stromal fibroblasts adjacent to invasive thyroid tumors: expression of gelatinase A but not stromelysin 3 mRNA
.
World J Surg
1996
;
20
:
101
6
.
33.
Wasenius
VM
,
Hemmer
S
,
Kettunen
E
,
Knuutila
S
,
Franssila
K
,
Joensuu
H
. 
Hepatocyte growth factor receptor, matrix metalloproteinase-11, tissue inhibitor of metalloproteinase-1, and fibronectin are up-regulated in papillary thyroid carcinoma: a cDNA and tissue microarray study
.
Clin Cancer Res
2003
;
9
:
68
75
.
34.
Maeta
H
,
Ohgi
S
,
Terada
T
. 
Protein expression of matrix metalloproteinases 2 and 9 and tissue inhibitors of metalloproteinase 1 and 2 in papillary thyroid carcinomas
.
Virchows Arch
2001
;
438
:
121
8
.
35.
Yeh
MW
,
Rougier
JP
,
Park
JW
,
Duh
QY
,
Wong
M
,
Werb
Z
, et al
Differentiated thyroid cancer cell invasion is regulated through epidermal growth factor receptor-dependent activation of matrix metalloproteinase (MMP)-2/gelatinase A
.
Endocr Relat Cancer
2006
;
13
:
1173
83
.
36.
Aust
G
,
Hofmann
A
,
Laue
S
,
Rost
A
,
Kohler
T
,
Scherbaum
WA
. 
Human thyroid carcinoma cell lines and normal thyrocytes: expression and regulation of matrix metalloproteinase-1 and tissue matrix metalloproteinase inhibitor-1 messenger-RNA and protein
.
Thyroid
1997
;
7
:
713
24
.
37.
Hofmann
A
,
Laue
S
,
Rost
AK
,
Scherbaum
WA
,
Aust
G
. 
mRNA levels of membrane-type 1 matrix metalloproteinase (MT1-MMP), MMP-2, and MMP-9 and of their inhibitors TIMP-2 and TIMP-3 in normal thyrocytes and thyroid carcinoma cell lines
.
Thyroid
1998
;
8
:
203
14
.
38.
Henneman
S
,
Bildt
MM
,
Degroot
J
,
Kuijpers-Jagtman
AM
,
Von den Hoff
JW
. 
Relaxin stimulates MMP-2 and alpha-smooth muscle actin expression by human periodontal ligament cells
.
Arch Oral Biol
2008
;
53
:
161
7
.
39.
Kapila
S
,
Xie
Y
,
Wang
W
. 
Induction of MMP-1 (collagenase-1) by relaxin in fibrocartilaginous cells requires both the AP-1 and PEA-3 promoter sites
.
Orthod Craniofac Res
2009
;
12
:
178
86
.
40.
Kapila
S
,
Xie
Y
. 
Targeted induction of collagenase and stromelysin by relaxin in unprimed and beta-estradiol-primed diarthrodial joint fibrocartilaginous cells but not in synoviocytes
.
Lab Invest
1998
;
78
:
925
38
.
41.
Qin
X
,
Chua
PK
,
Ohira
RH
,
Bryant-Greenwood
GD
. 
An autocrine/paracrine role of human decidual relaxin. II. Stromelysin-1 (MMP-3) and tissue inhibitor of matrix metalloproteinase-1 (TIMP-1)
.
Biol Reprod
1997
;
56
:
812
20
.
42.
Lenhart
JA
,
Ryan
PL
,
Ohleth
KM
,
Palmer
SS
,
Bagnell
CA
. 
Relaxin increases secretion of matrix metalloproteinase-2 and matrix metalloproteinase-9 during uterine and cervical growth and remodeling in the pig
.
Endocrinology
2001
;
142
:
3941
9
.
43.
Lenhart
JA
,
Ryan
PL
,
Ohleth
KM
,
Palmer
SS
,
Bagnell
CA
. 
Relaxin increases secretion of tissue inhibitor of matrix metalloproteinase-1 and -2 during uterine and cervical growth and remodeling in the pig
.
Endocrinology
2002
;
143
:
91
8
.
44.
Heeg
MH
,
Koziolek
MJ
,
Vasko
R
,
Schaefer
L
,
Sharma
K
,
Muller
GA
, et al
The antifibrotic effects of relaxin in human renal fibroblasts are mediated in part by inhibition of the Smad2 pathway
.
Kidney Int
2005
;
68
:
96
109
.
45.
Bialek
J
,
Hombach-Klonisch
S
,
Fiebig
B
,
Weber
E
,
Hoang-Vu
C
,
Klonisch
T
. 
Lysosomal acid hydrolases of the cathepsin family are novel targets of INSL3 in human thyroid carcinoma cells
.
Ann N Y Acad Sci
2009
;
1160
:
361
6
.
46.
Ni
H
,
Wilkins
JA
. 
Localisation of a novel adhesion blocking epitope on the human beta 1 integrin chain
.
Cell Adhes Commun
1998
;
5
:
257
71
.
47.
Cho
SY
,
Klemke
RL
. 
Purification of pseudopodia from polarized cells reveals redistribution and activation of Rac through assembly of a CAS/Crk scaffold
.
J Cell Biol
2002
;
156
:
725
36
.
48.
Szabova
L
,
Son
MY
,
Shi
J
,
Sramko
M
,
Yamada
SS
,
Swaim
WD
, et al
Membrane-type MMPs are indispensable for placental labyrinth formation and development
.
Blood
2010
;
116
:
5752
61
.
49.
Strongin
AY
,
Collier
I
,
Bannikov
G
,
Marmer
BL
,
Grant
GA
,
Goldberg
GI
. 
Mechanism of cell surface activation of 72-kDa type IV collagenase. Isolation of the activated form of the membrane metalloprotease
.
J Biol Chem
1995
;
270
:
5331
8
.
50.
Sternlicht
MD
,
Werb
Z
. 
How matrix metalloproteinases regulate cell behavior
.
Annu Rev Cell Dev Biol
2001
;
17
:
463
516
.
51.
Cavalheiro
BG
,
Junqueira
CR
,
Brandao
LG
. 
Expression of matrix metalloproteinase 2 (MMP-2) and tissue inhibitor of metalloproteinase 2 (TIMP-2) in medullary thyroid carcinoma: prognostic implications
.
Thyroid
2008
;
18
:
865
71
.
52.
Egeblad
M
,
Werb
Z
. 
New functions for the matrix metalloproteinases in cancer progression
.
Nat Rev Cancer
2002
;
2
:
161
74
.
53.
Devy
L
,
Huang
L
,
Naa
L
,
Yanamandra
N
,
Pieters
H
,
Frans
N
, et al
Selective inhibition of matrix metalloproteinase-14 blocks tumor growth, invasion, and angiogenesis
.
Cancer Res
2009
;
69
:
1517
26
.
54.
Hotary
K
,
Allen
E
,
Punturieri
A
,
Yana
I
,
Weiss
SJ
. 
Regulation of cell invasion and morphogenesis in a three-dimensional type I collagen matrix by membrane-type matrix metalloproteinases 1, 2, and 3
.
J Cell Biol
2000
;
149
:
1309
23
.
55.
Cao
J
,
Kozarekar
P
,
Pavlaki
M
,
Chiarelli
C
,
Bahou
WF
,
Zucker
S
. 
Distinct roles for the catalytic and hemopexin domains of membrane type 1-matrix metalloproteinase in substrate degradation and cell migration
.
J Biol Chem
2004
;
279
:
14129
39
.
56.
Li
XY
,
Ota
I
,
Yana
I
,
Sabeh
F
,
Weiss
SJ
. 
Molecular dissection of the structural machinery underlying the tissue-invasive activity of membrane type-1 matrix metalloproteinase
.
Mol Biol Cell
2008
;
19
:
3221
33
.
57.
Littlepage
LE
,
Sternlicht
MD
,
Rougier
N
,
Phillips
J
,
Gallo
E
,
Yu
Y
, et al
Matrix metalloproteinases contribute distinct roles in neuroendocrine prostate carcinogenesis, metastasis, and angiogenesis progression
.
Cancer Res
2010
;
70
:
2224
34
.
58.
Nakamura
ES
,
Koizumi
K
,
Kobayashi
M
,
Saiki
I
. 
Inhibition of lymphangiogenesis-related properties of murine lymphatic endothelial cells and lymph node metastasis of lung cancer by the matrix metalloproteinase inhibitor MMI270
.
Cancer Sci
2004
;
95
:
25
31
.
59.
Sounni
NE
,
Devy
L
,
Hajitou
A
,
Frankenne
F
,
Munaut
C
,
Gilles
C
, et al
MT1-MMP expression promotes tumor growth and angiogenesis through an up-regulation of vascular endothelial growth factor expression
.
FASEB J
2002
;
16
:
555
64
.
60.
Holmbeck
K
,
Bianco
P
,
Caterina
J
,
Yamada
S
,
Kromer
M
,
Kuznetsov
SA
, et al
MT1-MMP-deficient mice develop dwarfism, osteopenia, arthritis, and connective tissue disease due to inadequate collagen turnover
.
Cell
1999
;
99
:
81
92
.
61.
Ha
HY
,
Moon
HB
,
Nam
MS
,
Lee
JW
,
Ryoo
ZY
,
Lee
TH
, et al
Overexpression of membrane-type matrix metalloproteinase-1 gene induces mammary gland abnormalities and adenocarcinoma in transgenic mice
.
Cancer Res
2001
;
61
:
984
90
.
62.
Qi
JH
,
Ebrahem
Q
,
Moore
N
,
Murphy
G
,
Claesson-Welsh
L
,
Bond
M
, et al
A novel function for tissue inhibitor of metalloproteinases-3 (TIMP3): inhibition of angiogenesis by blockage of VEGF binding to VEGF receptor-2
.
Nat Med
2003
;
9
:
407
15
.
63.
Guo
H
,
Li
R
,
Zucker
S
,
Toole
BP
. 
EMMPRIN (CD147), an inducer of matrix metalloproteinase synthesis, also binds interstitial collagenase to the tumor cell surface
.
Cancer Res
2000
;
60
:
888
91
.
64.
Hombach-Klonisch
S
,
Bialek
J
,
Radestock
Y
,
Truong
A
,
Agoulnik
AI
,
Fiebig
B
, et al
INSL3 has tumor-promoting activity in thyroid cancer
.
Int J Cancer
2010
;
127
:
521
31
.
65.
Klonisch
T
,
Mustafa
T
,
Bialek
J
,
Radestock
Y
,
Holzhausen
HJ
,
Dralle
H
, et al
Human medullary thyroid carcinoma: a source and potential target for relaxin-like hormones
.
Ann N Y Acad Sci
2005
;
1041
:
449
61
.
66.
Plehn
A
,
Gunther
D
,
Aurich
H
,
Hoang-Vu
C
,
Weber
E
. 
Influence of proliferation, differentiation and dedifferentiation factors on the expression of the lysosomal cysteine proteinase cathepsin L (CL) in thyroid cancer cell lines
.
Adv Exp Med Biol
2000
;
477
:
487
95
.
67.
She
M
,
Jim
Yeung SC
. 
Combining a matrix metalloproteinase inhibitor, a farnesyltransferase inhibitor, and a taxane improves survival in an anaplastic thyroid cancer model
.
Cancer Lett
2006
;
238
:
197
201
.
68.
Shi
Y
,
Zou
M
. 
Matrix metalloproteinases in thyroid cancer
.
Cancer Treat Res
2004
;
122
:
179
90
.
69.
Radestock
Y
,
Willing
C
,
Kehlen
A
,
Hoang-Vu
C
,
Hombach-Klonisch
S
. 
Relaxin enhances S100A4 and promotes growth of human thyroid carcinoma cell xenografts
.
Mol Cancer Res
2010
;
8
:
494
506
.
70.
Silvertown
JD
,
Ng
J
,
Sato
T
,
Summerlee
AJ
,
Medin
JA
. 
H2 relaxin overexpression increases in vivo prostate xenograft tumor growth and angiogenesis
.
Int J Cancer
2006
;
118
:
62
73
.
71.
Silvertown
JD
,
Symes
JC
,
Neschadim
A
,
Nonaka
T
,
Kao
JC
,
Summerlee
AJ
, et al
Analog of H2 relaxin exhibits antagonistic properties and impairs prostate tumor growth
.
FASEB J
2007
;
21
:
754
65
.
72.
Feng
S
,
Agoulnik
IU
,
Truong
A
,
Li
Z
,
Creighton
CJ
,
Kaftanovskaya
EM
, et al
Suppression of relaxin receptor RXFP1 decreases prostate cancer growth and metastasis
.
Endocr Relat Cancer
2010
;
17
:
1021
33
.
73.
Silvertown
JD
,
Geddes
BJ
,
Summerlee
AJ
. 
Adenovirus-mediated expression of human prorelaxin promotes the invasive potential of canine mammary cancer cells
.
Endocrinology
2003
;
144
:
3683
91
.
74.
Radestock
Y
,
Hoang-Vu
C
,
Hombach-Klonisch
S
. 
Relaxin reduces xenograft tumour growth of human MDA-MB-231 breast cancer cells
.
Breast Cancer Res
2008
;
10
:
R71
.
75.
Tashima
LS
,
Mazoujian
G
,
Bryant-Greenwood
GD
. 
Human relaxins in normal, benign and neoplastic breast tissue
.
J Mol Endocrinol
1994
;
12
:
351
64
.
76.
Lamp
O
,
Honscha
KU
,
Jakob
J
,
Lamp
J
,
Schweizer
S
,
Reischauer
A
, et al
Investigation of the local expression of the relaxin system in canine mammary tumours
.
Reprod Domest Anim
2009
;
44
Suppl 2
:
224
9
.
77.
Puyraimond
A
,
Fridman
R
,
Lemesle
M
,
Arbeille
B
,
Menashi
S
. 
MMP-2 colocalizes with caveolae on the surface of endothelial cells
.
Exp Cell Res
2001
;
262
:
28
36
.
78.
Nabeshima
K
,
Inoue
T
,
Shimao
Y
,
Okada
Y
,
Itoh
Y
,
Seiki
M
, et al
Front-cell-specific expression of membrane-type 1 matrix metalloproteinase and gelatinase A during cohort migration of colon carcinoma cells induced by hepatocyte growth factor/scatter factor
.
Cancer Res
2000
;
60
:
3364
9
.
79.
Albiges-Rizo
C
,
Destaing
O
,
Fourcade
B
,
Planus
E
,
Block
MR
. 
Actin machinery and mechanosensitivity in invadopodia, podosomes and focal adhesions
.
J Cell Sci
2009
;
122
:
3037
49
.