Tumor cell invasion is promoted by interstitial flow-induced matrix priming by stromal fibroblasts.

Interstitial flow emanates from tumors into the microenvironment where it promotes tumor cell invasion. Fibroblasts are key constituents of the tumor stroma that modulate the mechanical environment by matrix remodeling and contraction. Here, we explore how interstitial fluid flow affects fibroblast-tumor cell interactions. Using a 3-dimensional invasion assay and MDA-MB-435S cells cocultured with dermal fibroblasts in a collagen matrix, we showed a synergistic enhancement of tumor cell invasion by fibroblasts in the presence of interstitial flow. Interstitial flow also drove transforming growth factor (TGF)-β1 and collagenase-dependent fibroblast migration, consistent with previously described mechanisms in which flow promotes invasion through autologous chemotaxis and increased motility. Concurrently, migrating fibroblasts enhanced tumor cell invasion by matrix priming via Rho-mediated contraction. We propose a model in which interstitial flow promotes fibroblast migration through increased TGF-β1 activation and collagen degradation, positioning fibroblasts to locally reorganize collagen fibers via Rho-dependent contractility, in turn enhancing tumor cell invasion via mechanotactic cues. This represents a novel mechanism in which interstitial flow causes fibroblast-mediated stromal remodeling that facilitates tumor invasion.

[1]  E. Sahai,et al.  Fibroblast-led collective invasion of carcinoma cells with differing roles for RhoGTPases in leading and following cells , 2007, Nature Cell Biology.

[2]  Mikala Egeblad,et al.  Matrix Crosslinking Forces Tumor Progression by Enhancing Integrin Signaling , 2009, Cell.

[3]  Kevin W. Eliceiri,et al.  Matrix density-induced mechanoregulation of breast cell phenotype, signaling, and gene expression through a FAK-ERK linkage , 2009, Oncogene.

[4]  B. Hinz,et al.  Myofibroblasts and mechano-regulation of connective tissue remodelling , 2002, Nature Reviews Molecular Cell Biology.

[5]  Paolo P. Provenzano,et al.  Collagen reorganization at the tumor-stromal interface facilitates local invasion , 2006, BMC medicine.

[6]  Raghu Kalluri,et al.  Fibroblasts in cancer , 2006, Nature Reviews Cancer.

[7]  John A. Pedersen,et al.  Effects of extracellular fiber architecture on cell membrane shear stress in a 3D fibrous matrix. , 2007, Journal of biomechanics.

[8]  W Matthew Petroll,et al.  Quantitative assessment of local collagen matrix remodeling in 3-D culture: the role of Rho kinase. , 2006, Experimental cell research.

[9]  R. Wells Faculty Opinions recommendation of Myofibroblast contraction activates latent TGF-beta1 from the extracellular matrix. , 2008 .

[10]  L. Coussens,et al.  Tumor stroma and regulation of cancer development. , 2006, Annual review of pathology.

[11]  M. Stack,et al.  Multi-step pericellular proteolysis controls the transition from individual to collective cancer cell invasion , 2007, Nature Cell Biology.

[12]  P. Friedl,et al.  Migration of highly aggressive MV3 melanoma cells in 3-dimensional collagen lattices results in local matrix reorganization and shedding of alpha2 and beta1 integrins and CD44. , 1997, Cancer research.

[13]  B. Hinz,et al.  Regulation of myofibroblast activities: calcium pulls some strings behind the scene. , 2010, Experimental cell research.

[14]  P. Khaw,et al.  TGF-β1, -β2, and -β3 In Vitro: Biphasic Effects on Tenon’s Fibroblast Contraction, Proliferation, and Migration , 2000 .

[15]  V. Niggli,et al.  Differential roles of Rho-kinase and myosin light chain kinase in regulating shape, adhesion, and migration of HT1080 fibrosarcoma cells. , 2006, Biochemical and Biophysical Research Communications - BBRC.

[16]  Z. Werb,et al.  HSulf-2, an extracellular endoglucosamine-6-sulfatase, selectively mobilizes heparin-bound growth factors and chemokines: effects on VEGF, FGF-1, and SDF-1 , 2006, BMC Biochemistry.

[17]  M. Goumans,et al.  Balancing the activation state of the endothelium via two distinct TGF‐β type I receptors , 2002, The EMBO journal.

[18]  John A. Pedersen,et al.  Cells in 3D matrices under interstitial flow: effects of extracellular matrix alignment on cell shear stress and drag forces. , 2010, Journal of biomechanics.

[19]  M. Neeman,et al.  Overexpression of vascular endothelial growth factor 165 drives peritumor interstitial convection and induces lymphatic drain: magnetic resonance imaging, confocal microscopy, and histological tracking of triple-labeled albumin. , 2002, Cancer research.

[20]  Cynthia A. Reinhart-King,et al.  Tensional homeostasis and the malignant phenotype. , 2005, Cancer cell.

[21]  Kenneth M. Yamada,et al.  Myosin IIA regulates cell motility and actomyosin–microtubule crosstalk , 2007, Nature Cell Biology.

[22]  Z. Werb,et al.  Matrix Metalloproteinases: Regulators of the Tumor Microenvironment , 2010, Cell.

[23]  Frederick Grinnell,et al.  Fibroblast biology in three-dimensional collagen matrices. , 2003, Trends in cell biology.

[24]  P. Khaw,et al.  TGF-beta1, -beta2, and -beta3 in vitro: biphasic effects on Tenon's fibroblast contraction, proliferation, and migration. , 2000, Investigative ophthalmology & visual science.

[25]  Sanjay Kumar,et al.  The mechanical rigidity of the extracellular matrix regulates the structure, motility, and proliferation of glioma cells. , 2009, Cancer research.

[26]  Thomas Hawighorst,et al.  Induction of tumor lymphangiogenesis by VEGF-C promotes breast cancer metastasis , 2001, Nature Medicine.

[27]  A. Ruddell,et al.  Tumor-induced sentinel lymph node lymphangiogenesis and increased lymph flow precede melanoma metastasis. , 2007, The American journal of pathology.

[28]  D. Rifkin,et al.  An assay for transforming growth factor-beta using cells transfected with a plasminogen activator inhibitor-1 promoter-luciferase construct. , 1994, Analytical biochemistry.

[29]  Brian Bierie,et al.  Tumour microenvironment: TGFβ: the molecular Jekyll and Hyde of cancer , 2006, Nature Reviews Cancer.

[30]  H. Moses,et al.  Stimulation of the chemotactic migration of human fibroblasts by transforming growth factor beta , 1987, The Journal of experimental medicine.

[31]  Melody A Swartz,et al.  Autologous chemotaxis as a mechanism of tumor cell homing to lymphatics via interstitial flow and autocrine CCR7 signaling. , 2007, Cancer cell.

[32]  J. Pollard,et al.  Microenvironmental regulation of metastasis , 2009, Nature Reviews Cancer.

[33]  Kevin W Eliceiri,et al.  Contact guidance mediated three-dimensional cell migration is regulated by Rho/ROCK-dependent matrix reorganization. , 2008, Biophysical journal.

[34]  G. Jenkins The role of proteases in transforming growth factor-beta activation. , 2008, The international journal of biochemistry & cell biology.

[35]  Amit Jain,et al.  Probing cellular mechanobiology in three-dimensional culture with collagen-agarose matrices. , 2010, Biomaterials.

[36]  C. Hofmann,et al.  Transforming growth factor-beta 1 induces intestinal myofibroblast differentiation and modulates their migration. , 2009, World journal of gastroenterology.

[37]  M. Swartz,et al.  Fibroblast alignment under interstitial fluid flow using a novel 3-D tissue culture model. , 2003, American journal of physiology. Heart and circulatory physiology.

[38]  Melody A Swartz,et al.  Autologous morphogen gradients by subtle interstitial flow and matrix interactions. , 2006, Biophysical journal.

[39]  Melody A. Swartz,et al.  Interstitial fluid flow induces myofibroblast differentiation and collagen alignment in vitro , 2005, Journal of Cell Science.

[40]  Sanjay Kumar,et al.  Mechanics, malignancy, and metastasis: The force journey of a tumor cell , 2009, Cancer and Metastasis Reviews.

[41]  Hiroshi Hosoya,et al.  Traction forces of fibroblasts are regulated by the Rho-dependent kinase but not by the myosin light chain kinase. , 2006, Archives of biochemistry and biophysics.

[42]  Fumio Matsumura,et al.  Distinct roles of MLCK and ROCK in the regulation of membrane protrusions and focal adhesion dynamics during cell migration of fibroblasts , 2004, The Journal of cell biology.

[43]  B. Hinz,et al.  Myofibroblast contraction activates latent TGF-β1 from the extracellular matrix , 2007, The Journal of cell biology.

[44]  Dai Fukumura,et al.  Tumor microenvironment abnormalities: Causes, consequences, and strategies to normalize , 2007, Journal of cellular biochemistry.

[45]  John M Tarbell,et al.  Interstitial flow promotes vascular fibroblast, myofibroblast, and smooth muscle cell motility in 3-D collagen I via upregulation of MMP-1. , 2009, American journal of physiology. Heart and circulatory physiology.

[46]  B. Hinz,et al.  Integrins and the activation of latent transforming growth factor beta1 - an intimate relationship. , 2008, European journal of cell biology.

[47]  D. Rifkin,et al.  In vitro and in vivo evidence for shear-induced activation of latent transforming growth factor-beta1. , 2008, Blood.

[48]  Y. Inoue,et al.  Functional diversity between Rho-kinase- and MLCK-mediated cytoskeletal actions in a myofibroblast-like hepatic stellate cell line. , 2003, Biochemical and biophysical research communications.

[49]  Paul A. Janmey,et al.  Non-Linear Elasticity of Extracellular Matrices Enables Contractile Cells to Communicate Local Position and Orientation , 2009, PloS one.

[50]  E. Kubista,et al.  MMP-2 and MMP-9 Expression in Breast Cancer-Derived Human Fibroblasts is Differentially Regulated by Stromal-Epithelial Interactions , 2002, Breast Cancer Research and Treatment.

[51]  Federica Boschetti,et al.  Synergy between interstitial flow and VEGF directs capillary morphogenesis in vitro through a gradient amplification mechanism. , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[52]  C. Rueden,et al.  Bmc Medicine Collagen Density Promotes Mammary Tumor Initiation and Progression , 2022 .