An official website of the United States government
Here's how you know
Official websites use .gov
A
.gov website belongs to an official
government organization in the United States.
Secure .gov websites use HTTPS
A lock (
) or https:// means you've safely
connected to the .gov website. Share sensitive
information only on official, secure websites.
As a library, NLM provides access to scientific literature. Inclusion in an NLM database does not imply endorsement of, or agreement with,
the contents by NLM or the National Institutes of Health.
Learn more:
PMC Disclaimer
|
PMC Copyright Notice
. Author manuscript; available in PMC: 2019 Feb 22.
Published in final edited form as: Nature. 2018 Aug 22;560(7720):655–660. doi: 10.1038/s41586-018-0444-0
1Department of Pharmacology and Moores Cancer Center
8State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
13Department of Cellular and Molecular Medicine, Institute of Genomic Medicine, University of California San Diego School of Medicine, 9500 Gilman Drive, La Jolla, CA 92093, USA
1Department of Pharmacology and Moores Cancer Center
2Ludwig Institute for Cancer Research,
3Bioinformatics and Systems Biology Graduate Program
4Department of Bioengineering
5Institute of Engineering in Medicine, University of California San Diego, La Jolla, California 92093, USA
6Department of Molecular Enzymology
7Center for Metabolic Regulation of Healthy Aging, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan
8State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
9Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University, Seoul 03722, Korea
10Robert J. Tomisch Pathology & Laboratory Medicine Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
11Children’s Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
12Division of Oral Biology and Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
13Department of Cellular and Molecular Medicine, Institute of Genomic Medicine, University of California San Diego School of Medicine, 9500 Gilman Drive, La Jolla, CA 92093, USA
Author contribution
Z.M. and K.L.G. conceived the project and wrote the manuscript. Z.M., K.C.L, C.F., S.L., M.P., T.M., M.L. performed in vitro cell assays, CRISPR knockout, quantitative real-time PCR, immunofluorescence, and xenograft studies. A.K., J.K.P., K.C.W., A.W.H., S.C., A.J.E. assisted in manufacturing hydrogels and immunofluorescence. S.W.P., H.W.P., provided knockout cell lines. Y.Q., Y.D., Z.Y. B.R. performed the next generation sequencing and bioinformatics analyses. X.W. performed the pathological analyses. F.X.Y., C-Y.W., B.R., A.J.E. provided the technical support. </author_notes>
†
Correspondence to Kun-Liang Guan, kuguan@ucsd.edu.
The publisher's version of this article is available at Nature
Abstract
Mammalian cells are surrounded by neighboring cells and extracellular matrix (ECM), which provide cells with structural support and mechanical cues that influence diverse biological processes1. The Hippo pathway effectors YAP and TAZ are regulated by mechanical cues and mediate cellular responses to ECM stiffness2,3. Here we identified Ras-related GTPase RAP2 as a critical intracellular signal transducer that relays ECM rigidity signals to control mechano-sensitive cellular activities through YAP/TAZ. RAP2 is activated by low ECM stiffness, and RAP2 deletion blocks YAP/TAZ regulation by stiffness signals and promotes aberrant cell growth. Mechanistically, matrix stiffness acts through phospholipase Cγ1 (PLCγ1) to influence levels of phosphatidylinositol 4,5-bisphosphate (PIP2) and its product phosphatidic acid (PA), which activates RAP2 through PDZGEF1/2. At low stiffness, active RAP2 binds to and stimulates mitogen-activated protein kinase kinase kinase kinase 4/6/7 (MAP4K4/6/7) and Rho GTPase activating protein 29 (ARHGAP29), resulting in LATS1/2 activation and YAP/TAZ inhibition. RAP2 and YAP/TAZ play pivotal roles in mechano-regulated transcription, as YAP/TAZ deletion abolishes the ECM stiffness-responsive transcriptome. Our findings reveal RAP2 as a molecular switch in mechanotransduction, thereby defining a mechanosignaling pathway from ECM stiffness to the nucleus.
YAP/TAZ function as essential effectors of mechanotransduction to regulate cell proliferation and differentiation3–7. When cells are shifted from stiff to soft matrices, YAP/TAZ translocate from the nucleus to the cytoplasm, and are thus inactivated. However, the signaling mechanism from ECM stiffness to the Hippo pathway is unclear. Because small GTPases function as molecular switches in many biological processes8, we screened for small GTPases that affect YAP/TAZ localization in cells seeded on soft (1 kPa) or stiff (40 kPa) matrices (Supplemental information). RAP2A was identified since its overexpression induced cytoplasmic translocation of YAP/TAZ even on a stiff matrix (Fig. 1a). No other GTPases, including the closely related RAP1 and RAS, showed similar activity (Extended Data Fig. 1a).
At high stiffness, both wild-type (WT) and RAP2A/B/C-triple knockout (RAP2-KO) MCF10A cells showed nuclear localization of YAP/TAZ (Fig. 1b,c). At low stiffness, WT cells exhibited mainly cytoplasmic YAP/TAZ, whereas RAP2-KO MCF10A cells retained YAP/TAZ in the nucleus (Fig. 1c). RAP2 deletion in HEK293A cells also suppressed low stiffness-induced YAP/TAZ cytoplasmic translocation (Fig. 1d,e, Extended Data Fig. 1b). YAP/TAZ target genes CTGF, CYR61, and ANKRD1 were repressed by low stiffness in WT cells, but not in the RAP2-KO cells (Fig. 1f). Similar results were observed in human mesenchymal stem cells (Extended Data Fig. 1c-e), in which RAP2 deletion suppressed their differentiation into adipocytes (Extended Data Fig. 1f,g). In the luminal breast cancer MCF7 cells, ECM stiffness modulated YAP/TAZ localization in a RAP2-dependent manner, whereas the basal type MDA-MB-468 showed constitutively cytoplasmic YAP/TAZ localization regardless of stiffness (Extended Fig. 1h-l). TWIST and β-catenin were reported to show nuclear-cytoplasmic shuttling in response to physical cues9,10. TWIST, but not β-catenin, displayed nuclear-cytoplasmic translocation in response to ECM stiffness (Extended Data Fig. 2a). However, RAP2 deletion had no obvious impact on TWIST localization.
Activity of small GTPases is switched on and off by GTP- and GDP-binding, respectively. A RalGDS-RBD pulldown assay showed that low stiffness promotes RAP2 GTP-binding (Extended Data Fig. 2b, Fig. 2a). Unlike WT RAP2A, the GTP-binding-deficient mutant RAP2A-S17N did not induce cytoplasmic translocation of YAP/TAZ (Extended Data Fig. 2b,c). RAP2 interaction with its activators PDZGEF1/211-13 was enhanced by low stiffness (Extended Data Fig. 2d). We generated PDZGEF1/2-dKO cells (Extended Data Fig. 2e,f) and discovered that they were defective in YAP/TAZ cytoplasmic translocation (Fig. 2b,c) and target gene repression (Extended Data Fig. 2g) in response to low stiffness. PDZGEF1/2 deletion blunted RAP2 activation by low stiffness (Fig. 2d), while PDZGEF1 overexpression induced YAP/TAZ cytoplasmic translocation in WT but not RAP2-KO cells (Extended Data Fig. 2h,i).
PIP2 activates RAP2 through PDZGEF1/2 at the plasma membrane after PIP2 is converted to PA by phospholipase D1/2 (PLD1/2)13. Using a GFP-tagged PIP2 reporter, we observed that PIP2 was enriched at the plasma membrane at low stiffness (Fig. 2e, Extended Data Fig. 2j). Focal adhesions decrease PIP2 through activating PLC𝛾114,15. Inhibition of PLC𝛾1 by U73122 induced PIP2 accumulation (Extended Data Fig. 2k). We hypothesized that ECM stiffness regulates RAP2 and YAP/TAZ through modulating focal adhesion and local PIP2 abundance at plasma membrane. Focal adhesion kinase (FAK) inhibitor PF573228 or PLC𝛾1 inhibitor U73122 increased RAP2 GTP-binding and YAP/TAZ cytoplasmic translocation (Extended Data Fig. 3a-c). In contrast, PLD1/2 inhibitor BML279 reduced RAP2 GTP-binding and accumulated YAP/TAZ in the nucleus (Extended Data Fig. 3d-f). The effects of PIP2 on YAP/TAZ localization were confirmed by PLC𝛾1- and PLD1/2-knockdown experiments (Extended Data Fig. 3g-l).
The nuclear-cytoplasmic shuttling of YAP/TAZ is generally controlled by LATS1/2-dependent phosphorylation2. However, the role of Hippo kinase cascade in YAP/TAZ regulation by mechanotransduction is not well-defined4–7. We found that low ECM stiffness induced phosphorylation of LATS1/2 and YAP/TAZ in WT cells, and this was significantly blunted in RAP2-KO cells (Fig. 3a). Furthermore, RAP2 induced YAP phosphorylation in a GTP-binding-dependent manner (Extended Data Fig. 3m). We proposed that RAP2 controls YAP/TAZ localization via the Hippo pathway. Consistent with this notion, deletion of LATS1/2 or combined deletion of MST1/2 and MAP4Ks abolished the YAP/TAZ regulation by ECM rigidity (Extended Data Fig. 3n,o). Hippo pathway core components were similarly required for RAP2 to induce YAP/TAZ phosphorylation and cytoplasmic translocation (Extended Data Fig. 4a,b). The role of LATS in this regulation was further confirmed in LATS1/2-dKO mouse embryonic fibroblasts, as well as NF2-KO or MOB1A/1B-dKO HEK293A cells (Extended Data Fig. 4c,d)16,17.
MAP4K4, TNIK (MAP4K7), and ARHGAP29 are RAP2 effectors18–20. Notably, ARHGAP29 is one of the RhoGAPs that are transcriptionally activated by YAP21,22. MAP4K4 kinase activity was stimulated by low stiffness in WT but not RAP2-KO cells (Extended Data Fig. 4e,f). Moreover, low stiffness induced MAP4K4 phosphorylation, as indicated by reduced mobility, in a RAP2-dependent manner (Extended Data Fig. 4g,h). Deletion of the RAP2-interacting citron domain19 in MAP4K4 abolished its regulation by RAP2 (Extended Fig. 4i,j), and the mutant also failed to rescue the YAP/TAZ translocation defect in MST1/2-MAP4Ks-8KO cells (Fig. 3b). Interestingly, a recent study showed that the Drosophila MAP4K4/6/7 homolog Msn regulates Yki in response to tension23. In addition, RAP2A overexpression led to RhoA inactivation (Extended Data Fig. 5a), a potent activator for YAP/TAZ7,16 (Extended Data Fig. 5b-d). This action of RAP2A on RhoA was mediated by ARHGAP29, because YAP phosphorylation induced by ARHGAP29 required its Rho-GAP domain and the Hippo kinase cascade (Fig. 3c, Extended Data Fig. 5e). ARHGAP29 deletion compromised RhoA inactivation caused by low stiffness (Extended Data Fig. 5f,g). Therefore, RAP2 acts through MAP4K4/6/7 and ARHGAP29 to inhibit YAP/TAZ. Consistent with this finding, MAP4K4/6/7-ARHGAP29–4KO cells were resistant to RAP2-induced cytoplasmic translocation and phosphorylation of YAP/TAZ (Fig. 3d, Extended Data Fig.5h), and displayed impaired responses to ECM stiffness (Fig. 3e, Extended Data Fig. 5i). Collectively, our data reveal a signaling axis linking matrix stiffness to YAP/TAZ regulation via focal adhesion→PLC𝛾1→PIP2→PA→PDZGEF1/2→RAP2→ARHGAP29/MAP4K→LATS1/2 (Fig. 3f), which works in parallel to the cell spreading/RhoA/cytoskeleton tension-mediated YAP/TAZ translocation mechanism proposed previously4,7.
RAP2 is activated by cell-cell contact12, which also presents mechanical cue to cells and inhibits YAP/TAZ2,17. RAP2 deletion moderately increased nuclear YAP/TAZ at high confluence (Extended Data Fig. 6a,b). Combined deletion of RAP2 and MST1/2 resulted in stronger YAP/TAZ nuclear accumulation and gene transactivation (Extended Data Fig. 6a-e). Deletion of both RAP2 and MST1/2 is required to blunt LATS phosphorylation and YAP/TAZ inactivation, suggesting that confluency signaling is rather complex and additional routes, such as cellular junctions, contribute to LATS1/2 activation.
RAP2 deletion selectively enhanced cell growth only at low stiffness (Extended Data Fig. 7a). To assess RAP2’s role in tumorigenesis, we performed three assays: acinus formation, anchorage-independent growth, and xenotransplantation. First, we applied a 3D-culture system with low stiffness hydrogels to assay MCF10A acinus formation9 (Extended Data Fig. 7b), the aberrancy of which represents irregular cell growth and malignant transformation9,24. WT cells formed normal acini while RAP2-KO cells generated multi-acinar structures (Fig. 4a,b, Extended Data Fig. 7c), and YAP/TAZ knockdown significantly reduced aberrant acini. Since MST1/2 mediate some physical signals independent of RAP2 (Extended Data Fig. 6), we generated RAP2A/B/C-MST1/2–5KO (RAP2-MST1/2-KO) MCF10A cells (Extended Data Fig. 7d). While MST1/2-dKO cells formed relatively normal acini, the RAP2-MST1/2-KO cells formed large acini with invasive behaviors (Extended Data Fig. 7e,f) even at 150 Pa, a stiffness mimicking normal breast tissue (Extended Data Fig. 7g,h). Second, colony formation assay in soft agar showed that RAP2-MST1/2-KO cells conferred anchorage-independent growth (Extended Data Fig. 8a). Third, RAP2-MST1/2-KO cells showed significant xenograft growth in immune-deficient mice while MST1/2-dKO cells failed to sustain growth (Fig. 4c, Extended Data Fig. 8b,c). RAP2-MST1/2-KO xenografts contained abundant MCF10A cells recapitulating acinus and duct formation of breast tissue, while MST1/2-dKO xenografts consisted of mainly host cells with a small number of MCF10A cells (Extended Data Fig. 8d,e). Moreover, the RAP2-MST1/2-KO cells displayed architectural and cytological atypia with signs of malignancy (Extended Data Fig. 8e). Consistently, YAP/TAZ knockdown suppressed xenograft growth (Fig. 4d, Extended Data Fig. 8f-h). The RAP2’s function in stiffness-regulated growth was confirmed in a xenograft model using H-RAS-V12 expressing MCF10A cells25,26 (Fig. 4e,f, Extended Data Fig. 8i,j). We used LOX-overexpressing fibroblasts or semi-synthetic hyaluronan-derived hydrogels (soft: 0.40 ± 0.03 kPa; stiff: 8.98 ± 0.33 kPa)27,28 to assess the effect of stiffness on cell growth in xenograft models (Extended Data Fig. 9). Under low stiffness or with control fibroblasts, RAP2-KO cells grew significantly larger xenografts than WT cells whereas the growth advantage of RAP2-KO cells diminished at high stiffness or in presence of LOX-overexpressing fibroblasts (Fig. 4g,h, Extended Data Fig. 9).
YAP/TAZ are transcriptional co-activators that generate functional output through gene transcription2. RNA-seq was performed with RAP2-KO, LATS1/2-dKO, and YAP/TAZ-dKO HEK293A cells to investigate their transcriptional responses to ECM stiffness (Fig. 4i, Extended Data Fig. 10a). 814 genes were down-regulated and 513 genes were up-regulated by low stiffness in WT cells. These genes are involved in metabolic processes, such as RNA and macromolecule biosynthesis, and morphogenesis (Supplemental information). YAP/TAZ are ‘gate-keepers’ that are responsible for virtually all the stiffness-responsive genes, as deletion of YAP/TAZ or LATS1/2 abolished most of the up- or down-regulations of these genes (Fig. 4i), such as AMOTL2 and LGR5 (Extended Data Fig. 10b). Consistent with its role in mechano-signaling, RAP2 deletion completely abolished the changes in expression of 40–50% (not including those partially blunted) of stiffness-responsive genes (Fig. 4i).
To assess the interplay of RAP2 and Hippo pathway components in stiffness-dependent gene regulation, we enriched YAP/TAZ-activating genes that were down-regulated by YAP/TAZ knockout at high stiffness and upregulated by LATS1/2 knockout at low stiffness, and YAP/TAZ-repressing genes that were conversely regulated (Extended Data Fig.10c,d). These genes defined a ECM-Hippo transcriptome that comprised nearly a third of the genes affected by ECM stiffness (Extended Data Fig. 10e,f). RAP2 deletion completely abolished the responses to ECM in about 50% of ECM-Hippo transcriptome genes (Extended Data Fig. 10g,h), not to mention that RAP2 deletion also partially compromised many genes. The RAP2-regulated ECM-Hippo transcriptome revealed that RAP2 controls genes involved in cell growth and adhesion (e.g. CTGF, CYR61), as well as morphogenesis and development (e.g. KRT8, KRT18, GDF6), through the Hippo pathway to respond to ECM stiffness (Fig. 4j, Supplemental information).
This study has uncovered RAP2 as an intracellular mechanotransducer, which relays extracellular mechanical signals to transcriptional regulation through the Hippo pathway. ECM stiffness acts through RAP2 and its downstream Hippo kinase cascade to modulate a YAP/TAZ-mediated mechano-responsive transcriptome. The identification of this new signaling axis provides mechanistic insights into how cellular machinery is driven by mechanical stimuli.
Methods
Cell culture
HEK293A were maintained in DMEM containing 10% fetal bovine serum. MCF10A cells were maintained as previously described24. The pre-malignant derivative of MCF10A cells, MCF10A-T, were generated by infecting MCF10A cells with a lentiviral vector that expressing a constitutively active H-Ras mutant (G12V) as previously described25,26. Adipocyte-derived mesenchymal stem cells (MSCs) were cultured in HMSC growth medium from Cell Applications Inc, and were differentiated into adipocytes and stained according to a modified protocol29. MCF7 and MDA-MB-468 were maintained in DMEM/F12 with 10% FBS. Insulin (0.01 mg/ml) was used for maintaining MCF7 cells.
Plasmids
Flag-pLJM1-RAP2A and pRK5-HA-GST-RAP2A plasmids were kindly provided by Dr. David M. Sabatini (Addgene #19311 & 14952). PDZGEF1 and ARHGAP29 coding sequences were subcloned from cDNA clones BC117321 and BC093741 (Transomics Technology), respectively.
GFP-C1-PLCdelta-PH was a gift from Dr. Tobias Meyer (Addgene #21179).
CRISPR/Cas9 system was used to delete genes in HEK293A, MCF10A, MSC, MCF7 and MDA-MB-468 cells. The plasmids px459 v2 and lentiCRISPR v2 were provided by Dr. Feng Zhang (Addgene #62988 & #52961).
The sgRNA sequences targeting individual genes were as following:
GST-RalGDS-RBD-expressing BL21 strain was kindly provided by Dr. Rick Firtel, and GST-Rhotekin-RBD-expressing BL21 strain was a gift from Dr. Joan Heller Brown at the University of California, San Diego. The recombinant proteins were purified and stored as bound to glutathione-agarose beads. The binding of RAP2-GTP and RHOA-GTP from cell lysates to RalGDS-RBD-agarose and Rhotekin-RBD-agarose beads, respectively, was performed in a buffer containing 50 mM Tris-HCl pH 7.5, 150 mM NaCl, 25 mM MgCl2, 10% glycerol, 0.5% NP-40 substitute, 1 mM DTT.
Cell culture with two-dimensional or three-dimensional polyacrylamide-based hydrogels
Two-dimensional culture on hydrogels of high (40.40 ± 2.39 kPa) or low (1.00 ± 0.31 kPa) stiffness was described elsewhere30. 10 μg/ml human placenta fibronectin or 25–50 ug/ml rat tail Collagen I were used to coat the Sulfo-Sanpah-activated hydrogels according to the preferences of the cell lines. Three-dimensional culture followed a protocol with the modification that the Matrigel base was replaced with fibronectin-coated hydrogels9,24.
Staining and microscopy
For immunofluorescence, cells were fixed in 4% formaldehyde/PBS for 10 min and then were treated with 0.1% Triton X-100 or Saponin (only for staining PIP2 GFP reporter) for 15 min. After blocking, the cells were stained with corresponding antibodies. Most images were captured with a Nikon Eclipse Ti confocal microscope and then were exported from NIS elements imaging software. Images in Fig. 2d, Ext Data Fig. 4c,d were taken with Olympus FV1000 confocal microscope, and Image J was used to merge the signals from channels. For immunohistochemistry, xenografts were subject to heat-induced antigen retrieval using 10 mM sodium citrate buffer followed by 3% H2O2 for 30 min to quench endogenous peroxidase activity. Sections were incubated overnight at 4°C with YAP/TAZ antibody and detected using Vectastain elite ABC kit and DAB Peroxidase Substrate kit (Vector Laboratories) according to the manufacturer’s protocol.
Preparation of semi-synthetic hyaluronan-derived hydrogels
Under aseptic conditions, Glycosil (ESI Bio, cat # GS222), Gelin-S, (ESI Bio, cat # GS231), and Extralink (ESI Bio, cat # GS3006) were dissolved in degassed water (ESI Bio, cat # GS240) according to the manufacturer’s directions. To make soft hydrogels, stock concentrations of 10 mg/mL Glycosil, 10 mg/mL Gelin-S, and 5 mg/mL Extralink were made per manufacturer’s directions. To make stiff hydrogels, concentrated stocks of Glycosil and Extralink were prepared by solubilization in reduced volumes to make 2x Glycosil and 5x Extralink. Prior to use, aliquots were taken from each vial to make solutions at 1:5 ratios of Extralink: (Glycosil + Gelin-S) and 5x Extralink: (2x Glycosil + Gelin-S) for soft and stiff hydrogels respectively. For all conditions, the amount of Gelin-S was kept constant to ensure the same number of gelatin-based cell binding sites.
In order to perform atomic force microscopy, the Glycosil and Gelin-S components were mixed thoroughly and then the Extralink was added to initiate gelation. Fifty μL of the mixture was added drop-wise to DCMS-treated glass slides and a methacrylated coverslip was placed on top. Each sample was prepared in triplicate. The slides were then incubated at 37°C until complete gelation. For the samples tested, this occurred within 30–40 minutes of incubation at 37°C. Upon gelation, hydrogel stiffness was measured by AFM. Hydrogels were then placed in PBS containing 1% antibiotic/antimycotic at 37°C and the stiffness was measured at 1, 24, and 48 hours after mixing.
AFM measurement procedure
AFM was performed to measure hydrogel stiffness as previously described31. Briefly, indentations were performed using a pyrex-nitride probe with a pyramid tip (spring constant ~0.04 N/m, 35º half-angle opening, NanoAndMore USA Corporation, cat # PNP-TR) connected to a MFP-3D Bio Atomic Force Microscope (Oxford Instruments) mounted on a Ti-U fluorescent inverted microscope (Nikon Instruments). Probes were calibrated using the Igor 6.34A software (WaveMetrics). Samples were then loaded on the AFM, submersed in PBS, and indented at a velocity of 2 μm/s with a trigger force of 2 nN. ~20 force measurements were performed over a 90 μm x 90 μm region per gel. Measurements were made each day for three separate gels per condition. Elastic modulus was calculated based on a Hertz-based fit using a built-in code written in the Igor 6.34A software.
Animal studies.
8–9 weeks old female NOD/SCID mice were purchased from Jackson Lab, and 8–9 weeks old female nude mice were provided by the animal care program at University of California, San Diego. The mice were hosted in a special pathogen-free room under standard 12:12-h light/dark cycle, fed with standard rodent chow and water ad libitum, randomized before experiments. The sample size choice was not pre-determined for each experiment. When comparing RAP2-MST1/2-KO MCF10A cell and MST1/2-dKO MCF10A cells, a total of 5 × 106 MCF10A cells in 50% high concentration Matrigel (BD Bioscience) dissolved in PBS were subcutaneously inoculated in a NOD/SCID mouse. When comparing RAP2-KO MCF10A and MCF10A-T cells with WT MCF10A and MCF10A-T cells, 5 × 106 cells in 50% Matrigel were injected into nude mice subcutaneously. When comparing RAP2-KO with WT MCF7 cells, 2 × 106 MCF7 cells and 4 × 105 LOX-expressing or control NIH3T3 cells in 50% Matrigel/PBS were co-injected subcutaneously into nude mice. For the semi-synthetic hyaluronan-derived hydrogels, 2 × 106 MCF7 cells in 20 μl PBS suspension were embedded into 200 μl of soft and stiff formulations as described in the preparation of hydrogels. After brief gelation (5 min) at room temperature, the cell-laden hydrogels were subcutaneously injected into nude mice. The investigators are blinded to group allocations during data collection and analyses. All the procedures followed the NIH guidelines for the care and use of laboratory animals and the IACUC at the University of California, San Diego approved the experiments. For subcutaneous tumor growth, the maximum single tumor cannot exceed 2 cm in diameter in mice according to the guidelines provided by the animal care program at University of California, San Diego, and no experiments in this study generated tumor burden over this limit.
RNA interference
Lentiviral vectors pLKO.1-hygromycin (Addgene #24150) and pLKO.1-Blasticidin (Addgene #26655) were used to clone the following sense sequences to knock down human YAP or TAZ:
Negative control: CCTAAGGTTAAGTCGCCCTCG (cloned into pLKO.1-hygromycin and pLKO.1-blasticidin); YAP#1: GCCACCAAGCTAGATAAAGAA (pLKO.1-hygromycin); YAP#2: GACATCTTCTGGTCAGAGATA (pLKO.1-hygromycin); TAZ#1: GCGTTCTTGTGACAGATTATA (pLKO.1-blasticidin); TAZ#2: GCTCATGAGTATGCCCAATGC (pLKO.1-blasticidin). The resulting plasmids were used to package lentiviruses and the target cells were infected with an MOI of 0.5.
Duplex siRNAs targeting PLC𝛾1 and PLD1/2 were purchased from Integrated DNA Technologies, Inc. and transfected into cells with RNAiMAX (ThermoFisher Scientific). The sequences are as following
PLC𝛾1 #1 sense strand 5’-rGrArCrCrUrCrArUrCrArGrCrUrArCrUrArUrGrArGrArAAC-3’; anti-sense strand 5’-rGrUrUrUrCrUrCrArUrArGrUrArGrCrUrGrArUrGrArGrGrUrCrArA-3’.
PLC𝛾1 #2 sense strand 5’-rGrGrCrArArGrArArGrUrUrCrCrUrUrCrArGrUrArCrArATC-3’; anti-sense strand 5’-rGrArUrUrGrUrArCrUrGrArArGrGrArArCrUrUrCrUrUrGrCrCrUrU-3’.
PLD1 #1 sense strand 5’-rGrUrGrGrArUrArArArUrUrArCrArUrArUrCrArUrUrCrUGT-3’; antisense strand 5’- rArCrArGrArArUrGrArUrArUrGrUrArArUrUrUrArUrCrCrArCrUrG-3’.
PLD1 #2 sense strand 5’-rArCrUrGrGrArArGrArUrUrArCrUrUrGrArCrArArArGrATA-3’; anti-sense strand 5’-rUrArUrCrUrUrUrGrUrCrArArGrUrArArUrCrUrUrCrCrArGrUrUrG-3’.
PLD2 #1 sense strand 5’-rArArCrCrArArGrArArGrArArArUrArCrCrGrUrCrArUrUTT-3’; anti-sense strand 5’-rArArArArUrGrArCrGrGrUrArUrUrUrCrUrUrCrUrUrGrGrUrUrGrU-3’.
PLD2 #2 sense strand 5’-rCrUrCrUrArCrArUrUrGrArGrArArUrCrArGrUrUrCrUrUCA-3’; anti-sense strand 5’-rUrGrArArGrArArCrUrGrArUrUrCrUrCrArArUrGrUrArGrArGrGrA-3’.
Quantitative real-time PCR
Total RNAs were extracted with a kit from Qiagen. Reverse transcription was performed with iScript from Bio-Rad. The real-time PCR was performed with the Applied Biosystems 7300 with primers targeting CTGF, CYR61, and ANKRD1: CTGF-Forward: 5’-CCAATGACAACGCCTCCTG-3’, CTGF-Reverse: 5’-TGGTGCAGCCAGAAAGCTC-3’; CYR61-Forward: 5’-AGCCTCGCATCCTATACAACC-3’, CYR61-Reverse: 5’-TTCTTTCACAAGGCGGCACTC-3’; ANKRD1-Forward: 5’-GTGTAGCACCAGATCCATCG-3’, ANKRD1-Reverse: 5’-CGGTGAGACTGAACCGCTAT-3’. The gene expression was normalized to GAPDH: Forward: 5’-TGCACCACCAACTGCTTAGC-3’; Reverse: 5’- GGCATGGACTGTGGTCATGAG-3’.
RNA sequencing and bioinformatics analysis
Total RNAs were extracted by TRIzol (Thermo Fisher Scientific) from HEK293A cells seeded on high and low stiffness fibronectin-coated hydrogels. Three replicates for each sample were generated and analyzed. The resulting RNA was then used to prepare libraries using Illumina TruSeq Stranded mRNA Library Prep Kit Set A (Illumina, RS- 122–2101) or Set B (Illumina, RS-122–2102). The libraries were sequenced using Illumina HiSeq 4000 (single end 50bp). Reads were aligned to the hg19 reference genome using STAR32. Only uniquely mapped reads were kept for further analysis. Number of reads for each gene were counted using htseq-count33 according to Gencode human annotation release 24. DeSeq234 was used to identify differential expressed genes with default parameters. Genes with adjusted p-value < 0.1 were considered as differentially expressed. GO and KEGG enrichment analysis of differential expressed genes was performed using DAVID35.
Statistical analysis
Microsoft Excel was used for t-tests, and Graphpad Prism v6 was used for two-way ANOVA tests. When p value is smaller than 0.0001, Graphpad Prism v6 does not provide a precise p value and instead only shows a range of p < 0.0001.
Data availability
The source data for the graph representations can be found in the online version of the paper. For uncropped images of Western blot data, see Supplementary Figure 1. The RNA sequencing data is available in GEO DataSets with the accession number GSE98547. All other data that support the findings of this study are available upon request from the corresponding author.
K.C.L., A.W.H., and S.W.P. are supported by the T32 GM007752 training grant, A.K. is by T32AR060712, and J.K.P by F32HL126406. K.L.G. is supported by grants from NIH (CA196878, CA217642, GM51586, DEO15964) as is A.J.E. (R21CA217735, R01CA206880).
A. J. E is also supported by NSF grant 1463689, and A.K. is supported by NSF graduate research fellowship program and ARCS/Roche Foundation Scholar Award in Life Science.
Footnotes
Competing interests
Dr. Kun-Liang Guan is a co-founder of Vivace Therapeutics.
Reference:
1.Humphrey JD, Dufresne ER & Schwartz MA
Mechanotransduction and extracellular matrix homeostasis. Nat Rev Mol Cell Biol
15, 802–812, doi: 10.1038/nrm3896 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
2.Meng Z, Moroishi T & Guan KL
Mechanisms of Hippo pathway regulation. Genes Dev
30, 1–17, doi: 10.1101/gad.274027.115 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
3.Halder G, Dupont S & Piccolo S
Transduction of mechanical and cytoskeletal cues by YAP and TAZ. Nat Rev Mol Cell Biol
13, 591–600, doi: 10.1038/nrm3416 (2012). [DOI] [PubMed] [Google Scholar]
4.Aragona M
et al.
A mechanical checkpoint controls multicellular growth through YAP/TAZ regulation by actin-processing factors. Cell
154, 1047–1059, doi: 10.1016/j.cell.2013.07.042 (2013). [DOI] [PubMed] [Google Scholar]
5.Codelia VA, Sun G & Irvine KD
Regulation of YAP by mechanical strain through Jnk and Hippo signaling. Curr Biol
24, 2012–2017, doi: 10.1016/j.cub.2014.07.034 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
6.Wada K, Itoga K, Okano T, Yonemura S & Sasaki H
Hippo pathway regulation by cell morphology and stress fibers. Development
138, 3907–3914, doi: 10.1242/dev.070987 (2011). [DOI] [PubMed] [Google Scholar]
7.Dupont S
et al.
Role of YAP/TAZ in mechanotransduction. Nature
474, 179–183, doi: 10.1038/nature10137 (2011). [DOI] [PubMed] [Google Scholar]
8.Cherfils J & Zeghouf M
Regulation of small GTPases by GEFs, GAPs, and GDIs. Physiol Rev
93, 269–309, doi: 10.1152/physrev.00003.2012 (2013). [DOI] [PubMed] [Google Scholar]
9.Wei SC
et al.
Matrix stiffness drives epithelial-mesenchymal transition and tumour metastasis through a TWIST1-G3BP2 mechanotransduction pathway. Nat Cell Biol
17, 678–688, doi: 10.1038/ncb3157 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
11.de Rooij J
et al.
PDZ-GEF1, a guanine nucleotide exchange factor specific for Rap1 and Rap2. J Biol Chem
274, 38125–38130 (1999). [DOI] [PubMed] [Google Scholar]
12.Monteiro AC
et al.
Trans-dimerization of JAM-A regulates Rap2 and is mediated by a domain that is distinct from the cis-dimerization interface. Mol Biol Cell
25, 1574–1585, doi: 10.1091/mbc.E14-01-0018 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
13.Gloerich M
et al.
Rap2A links intestinal cell polarity to brush border formation. Nat Cell Biol
14, 793–801, doi: 10.1038/ncb2537 (2012). [DOI] [PubMed] [Google Scholar]
14.Carloni V, Romanelli RG, Pinzani M, Laffi G & Gentilini P
Focal adhesion kinase and phospholipase C gamma involvement in adhesion and migration of human hepatic stellate cells. Gastroenterology
112, 522–531 (1997). [DOI] [PubMed] [Google Scholar]
15.Zhang X
et al.
Focal adhesion kinase promotes phospholipase C-gamma1 activity. Proc Natl Acad Sci U S A
96, 9021–9026 (1999). [DOI] [PMC free article] [PubMed] [Google Scholar]
16.Plouffe SW
et al.
Characterization of Hippo Pathway Components by Gene Inactivation. Mol Cell
64, 993–1008, doi: 10.1016/j.molcel.2016.10.034 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
17.Meng Z
et al.
MAP4K family kinases act in parallel to MST1/2 to activate LATS1/2 in the Hippo pathway. Nat Commun
6, 8357, doi: 10.1038/ncomms9357 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
18.Myagmar BE
et al.
PARG1, a protein-tyrosine phosphatase-associated RhoGAP, as a putative Rap2 effector. Biochem Biophys Res Commun
329, 1046–1052, doi: 10.1016/j.bbrc.2005.02.069 (2005). [DOI] [PubMed] [Google Scholar]
19.Machida N
et al.
Mitogen-activated protein kinase kinase kinase kinase 4 as a putative effector of Rap2 to activate the c-Jun N-terminal kinase. J Biol Chem
279, 15711–15714, doi: 10.1074/jbc.C300542200 (2004). [DOI] [PubMed] [Google Scholar]
20.Taira K
et al.
The Traf2- and Nck-interacting kinase as a putative effector of Rap2 to regulate actin cytoskeleton. J Biol Chem
279, 49488–49496, doi: 10.1074/jbc.M406370200 (2004). [DOI] [PubMed] [Google Scholar]
21.Qiao Y
et al.
YAP Regulates Actin Dynamics through ARHGAP29 and Promotes Metastasis. Cell Rep
19, 1495–1502, doi: 10.1016/j.celrep.2017.04.075 (2017). [DOI] [PubMed] [Google Scholar]
22.Porazinski S
et al.
YAP is essential for tissue tension to ensure vertebrate 3D body shape. Nature
521, 217–221, doi: 10.1038/nature14215 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
23.Li Q
et al.
Ingestion of Food Particles Regulates the Mechanosensing Misshapen-Yorkie Pathway in Drosophila Intestinal Growth. Dev Cell
45, 433–449 e436, doi: 10.1016/j.devcel.2018.04.014 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
24.Debnath J, Muthuswamy SK & Brugge JS
Morphogenesis and oncogenesis of MCF-10A mammary epithelial acini grown in three-dimensional basement membrane cultures. Methods
30, 256–268 (2003). [DOI] [PubMed] [Google Scholar]
25.Yoh KE
et al.
Repression of p63 and induction of EMT by mutant Ras in mammary epithelial cells. Proc Natl Acad Sci U S A
113, E6107–E6116, doi: 10.1073/pnas.1613417113 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
26.Dawson PJ, Wolman SR, Tait L, Heppner GH & Miller FR
MCF10AT: a model for the evolution of cancer from proliferative breast disease. Am J Pathol
148, 313–319 (1996). [PMC free article] [PubMed] [Google Scholar]
27.Serban MA, Scott A & Prestwich GD
Use of hyaluronan-derived hydrogels for three-dimensional cell culture and tumor xenografts. Curr Protoc Cell Biol
Chapter 10, Unit 10 14, doi: 10.1002/0471143030.cb1014s40 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
29.Wen JH
et al.
Interplay of matrix stiffness and protein tethering in stem cell differentiation. Nat Mater
13, 979–987, doi: 10.1038/nmat4051 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
30.Tse JR & Engler AJ
Preparation of hydrogel substrates with tunable mechanical properties. Curr Protoc Cell Biol
Chapter 10, Unit 10 16, doi: 10.1002/0471143030.cb1016s47 (2010). [DOI] [PubMed] [Google Scholar]
31.Kaushik G, Fuhrmann A, Cammarato A & Engler AJ
In situ mechanical analysis of myofibrillar perturbation and aging on soft, bilayered Drosophila myocardium. Biophys J
101, 2629–2637, doi: 10.1016/j.bpj.2011.10.042 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
33.Anders S, Pyl PT & Huber W
HTSeq--a Python framework to work with high-throughput sequencing data. Bioinformatics
31, 166–169, doi: 10.1093/bioinformatics/btu638 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
34.Love MI, Huber W & Anders S
Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol
15, 550, doi: 10.1186/s13059-014-0550-8 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
35.Dennis G Jr.
et al.
DAVID: Database for Annotation, Visualization, and Integrated Discovery. Genome Biol
4, P3 (2003). [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
The source data for the graph representations can be found in the online version of the paper. For uncropped images of Western blot data, see Supplementary Figure 1. The RNA sequencing data is available in GEO DataSets with the accession number GSE98547. All other data that support the findings of this study are available upon request from the corresponding author.