Abstract
A defining hallmark of cancer and cancer development is upregulated angiogenesis. The vasculature formed in tumors is structurally abnormal, not organized in the conventional hierarchical arrangement, and more permeable than normal vasculature. These features contribute to leaky, tortuous, and dilated blood vessels, which act to create heterogeneous blood flow, compression of vessels, and elevated interstitial fluid pressure. As such, abnormalities in the tumor vasculature not only affect the delivery of nutrients and oxygen to the tumor, but also contribute to creating an abnormal tumor microenvironment that further promotes tumorigenesis. The role of chemical signaling events in mediating tumor angiogenesis have been well researched; however, the relative contribution of physical cues and mechanical regulation of tumor angiogenesis is less understood. Growing research indicates that the physical tumor microenvironment plays a significant role in tumor progression and promoting abnormal tumor vasculature. Here, we review how mechanical cues found in the tumor microenvironment promote aberrant tumor angiogenesis. Specifically, we discuss the influence of matrix stiffness and mechanical stresses in tumor tissue on tumor vasculature, as well as the mechanosensory pathways utilized by endothelial cells to respond to the physical cues found in the tumor microenvironment. We also discuss the impact of the resulting aberrant tumor vasculature on tumor progression and therapeutic treatment.
Keywords: VE-cadherin, VEGF, Matrix stiffness, MMP, Contractility, Fluid shear stress, Interstitial pressure, Mechanotransduction, Mechanosensitivity, Barrier function
6.1. Introduction
Like normal tissue, tumor tissue requires an adequate supply of nutrients and oxygen provided by blood vessels to meet metabolic needs, remove waste products, and survive. To meet these needs during tumor growth, blood vessels are developed through angiogenesis, the sprouting of new blood vessels from existing blood vessels [1, 2]. In normal tissue, the initiation of angiogenesis, known as the angiogenic switch, is tightly regulated; however, during tumor progression the appropriate balance of pro- and anti-angiogenic cues is lost, and the angiogenic switch is almost always activated [1]. Notably, while nutrient requirements can differ between tumor types and during tumor progression, the generation of a tumor blood supply is a rate-limiting step in solid tumor growth [3]. Consequently, solid tumors develop vasculature with many abnormal features [2, 4]. Solid tumor vasculature is exceptionally variable in size, shape, as well as architecture and is not organized in the conventional hierarchical arrangement found in normal tissue [5, 6]. This is due to the abnormal properties acquired by tumor endothelial cells [7, 8]. In the blood vessels of mouse mammary carcinomas, tumor endothelial cells have been shown to be poorly connected, grow on top of one another, and project into the lumen of the vessels [9]. Additionally, in many different types of solid tumors, the tumor vessel walls contain many openings, widened cell-cell junctions, and irregular or deficient basement membrane coverage [9–11]. Together, these abnormal features contribute to create hyperpermeable, tortuous, and dilated blood vessels, which generate heterogeneous blood flow and limited perfusion throughout the tumor.
A principal determinant of phenotypic differences found in tumors is the surrounding microenvironment [7]. Endothelial cells of recently formed blood vessels in the tumor are subjected to distinct extracellular signals including hypoxia, low pH, a deregulated and disorganized extracellular matrix (ECM), mechanical stresses, and soluble mediators released by surrounding tumor and stromal cells. Angiogenesis is tightly controlled by numerous chemical and mechanical signaling events, and these differences in extracellular cues have a profound effect on the formation of new capillaries. As such, the abnormal features of the tumor vasculature are believed to result from the disproportionate balance of pro- and anti-angiogenic cues found in the tumor microenvironment. Overexpression of vascular endothelial growth factor (VEGF) and other pro-angiogenic growth factors within the tumor microenvironment have been extensively investigated as major contributing factors in the formation of abnormal tumor vasculature. However, recent work have indicated that mechanical cues and forces within the tumor microenvironment play an important role in promoting a tumor vasculature phenotype [12].
Understanding the components of the tumor stroma such as the vasculature, has become key to understanding tumor growth and progression [3]. The tumor vasculature has been demonstrated to not only influence tumor growth but also be instrumental in facilitating metastasis and creating an irregular tumor microenvironment that assists in tumor progression [6, 7, 13]. This chapter will provide an overview of the mechanical cues and forces found in the tumor microenvironment and discuss their respective impact on tumor angiogenesis and promoting abnormal tumor vasculature. The mechanosensory pathways that are employed by endothelial cells to respond to mechanical stimuli, specifically aberrant mechanosensory pathways found in tumor endothelial cells, will be reviewed. Finally, this chapter will briefly discuss the clinical impact of abnormal tumor vasculature and its influence on cancer treatment.
6.2. Mechanical Cues in the Tumor Microenvironment
In the past few decades, there has been an increasing interest on how physical and mechanical cues in the tumor microenvironment influence cancer cells and cancer progression. As tumors stimulate neovascularization and angiogenesis to meet growth needs, the tumor vasculature is exposed to a mechanically abnormal and highly heterogeneous microenvironment (Fig. 6.1a). A critical component of the tumor microenvironment is the ECM, which is a complex three-dimensional assembly of macromolecules and interconnected cell-scale fibers with distinct physical and biomechanical properties [14–17]. The ECM determines the mechanical properties of a tissue as well as provides a dynamic and bioactive structure that fundamentally controls cell behavior through chemical and mechanical signals [17]. Tight regulation of the ECM is essential to maintaining tissue homeostasis, and abnormal ECM dynamics contribute to many pathological conditions, including cancer [18–20].
Fig. 6.1.
Mechanical cues in the tumor microenvironment influence tumor angiogenesis. (a) To meet nutrient needs, tumors upregulate angiogenesis and produce a vasculature network. The resulting tumor vasculature has many abnormal characteristics and is highly disordered. (b) In the tumor microenvironment, stiffening of the ECM modulates cell-cell junctions and localization of VE-cadherin, which results in disrupted barrier function and increased permeability. (c) Growth-induced solid stress from ECM deposition and proliferating stromal and cancer cells causes tumor vessel compression. (d) Elevated interstitial fluid pressure (IFP) in the tumor often exceeds that of the microvascular pressure (MVP), causing limited perfusion and disrupting flow patterns
6.2.1. Increased Matrix Stiffness During Tumor Progression
During solid tumor progression, the ECM commonly becomes deregulated and disorganized, creating solid tumor tissue with heterogeneous three-dimensional matrix features, organization, rigidity, and composition [14, 21–23]. Such changes to the ECM can significantly alter biochemical properties, alter cell response to growth factors, and disrupt cell behaviors [14–16, 24, 25]. Notably, increased ECM stiffness and density, caused primarily from increased collagen deposition and increased crosslinking within the stroma during the progression of many solid tumors, have been demonstrated to be cell-instructive and involved in promoting a malignant phenotype [14, 26–28]. Compared to normal tissue, many solid tumors are markedly stiffer (Table 6.1).
Table 6.1.
Mechanical properties of normal and tumor tissue
Tissue | State | Stiffness | Interstitial fluid pressure | Solid stress | References |
---|---|---|---|---|---|
Breast | Normal | 0.4–2.0 kPa | 0.0–3.0 mmHg | [26, 29–35] | |
Breast carcinoma | 4.0–12.0 kPa | 4.0–53.0 mmHg | 75.1–142.5 mmHg | ||
Lung | Normal | 10.0 kPa | −7.0 mmHg | [31, 35–38] | |
Lung carcinoma | 25.0–35.0 kPa | 1.0–27.0 mmHg | – | ||
Brain | Normal | 0.26–0.49 kPa | 0.0 mmHg | [35, 39–45] | |
Glioblastoma | 7.0–26.0 kPa | −0.5–15.0 mmHg | 1.56 mmHg | ||
Liver | Normal | 0.3–0.6 kPa | −2.2 mmHg | [46–48] | |
Hepatoma | 1.6–20.0 kPa | 0.0–30.0 mmHg | – | ||
Colorectal | Normal | 0.9–4.0 kPa | 14.0 mmHg | [32, 35, 44, 49–53] | |
Colorectal carcinoma | 7.5–30.0 kPa | 16.0–45.0 mmHg | 7.5 mmHg | ||
Kidney | Normal | 2.0 kPa | 6.0 mmHg | [32, 45, 54, 55] | |
Renal cell carcinoma | 13.0 kPa | 38.0 mmHg | – | ||
Skin | Normal | 35.0–300.0 kPa | −2.0–0.4 mmHg | [31, 32, 35, 45, 56, 57] | |
Metastatic melanoma | 400.0 kPa | 0.0–60.0 mmHg | – | ||
Pancreatic | Normal | 1.0 kPa | 8.0 mmHg | [44, 58, 59] | |
Pancreatic ductal adenocarcinoma | 2.0–4.0 kPa | 75.0–130.0 mmHg | 52.5 mmHg | ||
Bone | Normal | 2.0–14.0 GPa | 2.9 mmHg | [30, 34, 60–62] | |
Osteosarcoma | >689 MPa | 35.5 mmHg | 35.3–48.3 mmHg |
In vascular biology, the ECM drives capillary morphogenesis by providing necessary organization cues to endothelial cells [63]. Endothelial cell capillary-like network formation is influenced by ECM concentration [64–66], ECM composition [67, 68], as well as matrix stiffness [69–72]. Collectively, these and other studies clearly demonstrate the important role of the ECM in directing endothelial cell network formation. Compared to normal endothelial cells, tumor endothelial cells are exposed to a highly mechanically heterogeneous and abnormal microenvironment [14, 21, 73]. These abnormal physical cues in the tumor microenvironment continuously alter cell-ECM force balances that can influence tumor endothelial gene expression and cell behavior [74–77]. Indeed, tumor endothelial cells are notably phenotypically different from normal endothelial cells, and the tumor endothelium displays distinct gene expression profiles from the normal epithelium [78]. Tumor endothelial cells also demonstrate constant expression of endothelial activation, enhanced pro-adhesion and angiogenic properties, upregulated cell survival pathways, as well as altered mechanosensitivity [79, 80]. After isolation from tumors, tumor endothelial cell maintained these properties in cell culture, indicating a persistent alteration in phenotype. As such, tumor endothelial cells may be phenotypically adapted to stiffer ECM conditions in the tumor microenvironment by undergoing reprogramming of signaling pathways, possibly causing some of their aberrant functions [6].
Recent work has identified that altering matrix mechanics alone can induce a tumor vasculature phenotype. Increasing three-dimensional collagen stiffness without altering matrix architecture via non-enzymatic glycation increased angiogenic outgrowth and vascular branching density of in vitro endothelial cell spheroids, creating a morphology reminiscent of tumor vasculature [12]. Other methods of increasing collagen matrix stiffness in vitro have demonstrated comparable increases in angiogenic response in stiffer matrices (Table 6.2) [81–85]. Similar modulation of angiogenic outgrowth and branching by ECM stiffness was observed in vivo. In a MMTV-PyMT mouse tumor model1 [86], β-aminopropionitrile (BAPN), a lysyl oxidase inhibitor, was used to modulate the stiffness of mammary tumors from approximately 4.5 kPa in control mice to 3 kPa in BAPN-treated mice. It was shown that decreasing matrix stiffness via BAPN treatment significantly reduced the extent of angiogenesis and vascular branching density within tumors. Changes in matrix stiffness were also revealed to modulate endothelial cell-cell junctional properties and endothelial cell permeability both in vitro and in vivo [12]. Notably, the changes observed in vascular phenotype were due solely to mechanical alterations to the ECM. For endothelial cells cultured on compliant (0.2 kPa) or stiff (10 kPa) polyacrylamide substrates, stiffer matrices impaired barrier function and localization of vascular endothelial cadherin (VE-cadherin), contributing to increased vessel permeability (Fig. 6.1b). Endothelial cells on stiffer matrices demonstrated punctate VE-cadherin and β-catenin positive endothelial cell-cell junctions, as well as stress-mediated localization of tight junction protein zona occuldens 1 (ZO-1) that matched VE-cadherin. In vivo staining of VE-cadherin, β-catenin, and ZO-1 also demonstrated changes in junctional architecture in stiffer tumors. Interestingly, the altered vascular phenotype and increased angiogenic response required upregulation of matrix metalloproteinase (MMP) activity, specifically membrane-type 1 MMP (MT1-MMP). This finding suggests MMPs play an important role in promoting angiogenesis [12]. MMP activity has been shown to be important for ECM degradation and basement membrane remodeling during angiogenesis. MT-MMPs in particular are able to provide addition control over degradation events by providing spatial control of matrix degradation at the cell membrane surface [87]. Previous work has identified MT1-MMP activation is dependent on cell contractility and matrix stiffness [88]. Together, these findings demonstrate the importance of changing ECM cues during tumor progression, chiefly increased matrix stiffness, in promoting aberrant tumor vasculature.
Table 6.2.
The effects of matrix stiffening on angiogenesis in three-dimensional in vitro models
Matrix | Method of altering matrix stiffness | Stiffness (kPa) | Angiogenic response | References |
---|---|---|---|---|
Collagen | Non-enzymatic glycation with ribose | ~0.18–0.50 | Increasing matrix stiffness resulted in increased angiogenic outgrowth and branching density | [12] |
Collagen | Non-enzymatic glycation with glucose-6-phosphate | – | Decreased sprouting, but increased branching and tortuosity in crosslinked gels | [81] |
Collagen | Transglutaminase | 0.45–0.89 | Increasing matrix stiffness resulted in increased angiogenic sprouting, invasion, and remodeling | [82] |
Collagen | Varying oligomer: monomer ratio | ~0.06–0.26 | Increasing stiffness increased network length, branching, and vascularized area | [83] |
Collagen | EDC/NHS | – | Increased crosslinking resulted in increased capillary number and spoke-like vessel structure | [84] |
Collagen | Varying pH of polymerization solution | ~5–20 | Thicker, deeper capillary networks on more rigid three-dimensional collagen gels. Formation of large lumen on rigid gels compared to flexible gels | [85] |
6.2.2. Physical Forces in the Tumor Microenvironment
In addition to changes to the stromal ECM during tumorigenesis, solid tumors are also exposed to physical forces during tumor progression. As physical forces grow during solid tumor growth, increased tension in the tissue impacts not only tumor growth, but it also deforms the tumor vasculature [89]. These mechanical forces found in the tumor microenvironment can be categorized as solid or fluid stresses.
Solid stress is defined as the combined mechanical forces from the non-fluid, structural components of the tumor, predominantly cancer cells, various host cells, and the ECM [89]. Within solid tumors, solid stress is significantly elevated due to elevated cell and matrix densities (Table 6.1). Solid stresses accumulate as the tumor tissue becomes stiffer than the normal surrounding tissue and the constrained production of mechanical forces by tumor components dislocates the surrounding normal tissue [89]. Furthermore, as cancer and stromal cells proliferate and migrate through the ECM, growth-induced solid stresses are generated and transmitted through the ECM [34]. Interestingly, the total solid stresses in the tumor are compressive in the interior of the tumor, but forces are compressive in the radial direction and tensile in the circumferential direction at the tumor-host interface [34, 90]. The ECM components of the tumor stroma, notably collagen, can also help to transmit these forces across the tumor and to surrounding host tissue. Tumor-associated collagen signatures including dense collagen, tense collagen fibers, and aligned collagen fibers have been identified in tumors and are associated with tumor progression [21]. Collagen fibers are extraordinarily stiff in tension and offer tensile strength to tissue and can also supply solid stress when highly contractile cancer cells apply forces to them [89]. Long-range stress transmission (250–1000 μm) between cells in fibrous matrices is well appreciated [91–93]. Tension-driven fiber alignment, fiber stiffness, as well as fiber strain-hardening all permit and facilitate long-range mechanical interactions [94]. Notably, the range of these mechanical interactions increases with increasing cellular polarization and contractility [94]. Tumor stromal cells such as fibroblasts have been shown to be highly contractile and generate tensional forces by contraction of the surrounding matrix. Tissue tension, such as that generated by activated fibroblasts, has been demonstrated to influence vascular growth. Ingrowth and expansion of vascular tissue are associated with and directed by tissue contraction, where endothelial cells outgrow along the direction of tensional forces [95, 96]. Such translocation of functional vascular formations into tissue has previously been described for tumor-induced neovascularization of mouse cornea [97] and in human dermal wound healing models [98]. These data help to establish the concept of biomechanical regulation of tissue vascularization.
Fluid stress in the tumor microenvironment is the combined forces exerted by the fluid components of the tumor, namely the microvascular fluid pressure, interstitial fluid pressure, and shear stress, applied by the blood flow and interstitial flow [89]. Within tumors, elevated interstitial fluid pressure from leaking blood vessels and ineffective intratumor lymphatics leads to abnormal tumor vasculature due to the resulting transmural pressure (Table 6.1) [10, 90, 99]. In both experimental and human solid tumors, interstitial fluid pressure has been reported to commonly range from 4 to 60 mmHg in neoplastic regions [32, 35, 42, 100] and has been reported as high as 130 mmHg in mouse pancreatic ductal adenocarcinomas [59]. The subsequent abnormal structure of the tumor microvasculature increases geometric and viscous resistances to blood flow, further contributing to aberrant flow and limited perfusion in tumor tissue [89]. Aberrant flow in the tumor vasculature is significant and can influence endothelial cell function. Distinct flow patterns in the different regions of normal vessels are important in regulating molecular and morphological differences needed for endothelial cell specialization [101]. Flow and shear stresses have a well-established effect on endothelial cells. Fluid shear stress enacts signaling cascades that influence endothelial morphology as well as trigger remodeling of vascular networks [102]. Precisely, fluid shear stresses affect vascular endothelial growth factor receptor (VEGFR) conformational changes [103], tubule formation [104], and barrier function [105] and ultimately direct endothelial morphogenesis and sprout formation [106, 107]. Basal-to-apical transendothelial flow has also been demonstrated to induce an invasive phenotype through focal adhesion kinase (FAK)-mediated signaling and extensive endothelial cell-cell junction remodeling [108]. Endothelial cells lining tumor vessels are subjected to such transendothelial pressure and flow, and these findings are in agreement with early observations that tumor angiogenesis emerges predominately from the venous side of the circulation [109]. Together, these data demonstrate that fluid stresses not only influence tumor vessel perfusion but also contributes to abnormal vessel structure and function.
Collectively, solid and fluid stresses in the tumor microenvironment act to compress tumor vessels and significantly alter blood flow through the tumor. Growth-induced solid stress in solid tumors has been reported to commonly range from 10 to 142 mmHg [89, 90], while interstitial fluid pressure within tumor tissue has been reported to commonly range from about 4 to 60 mmHg (Table 6.1) [32, 35, 42, 100, 110]. Together, these forces act to compress blood vessels in the tumor, causing limited perfusion and hypoxia throughout the tumor tissue (Fig. 6.1c, d). Notably, solid stress in the tumor, rather than increased interstitial fluid pressure, has been identified to be the predominant cause of vessel compression [90, 110]. Removal of the mechanical forces in solid tumor tissue can recover some of the aberrant features of the tumor vasculature. Depletion of the structural components that contribute to solid stress in the tumor microenvironment – cancer cells, fibroblasts, or collagen – significantly reduces solid stress and improves perfusion through the tumor tissue in breast, pancreatic, and melanoma tumor models [34]. Together, the physical forces that accumulate during tumor growth considerably impact vessel architecture, permeability, and perfusion. Better understanding of these physical forces, and their influence on tumorigenesis, will be important for improving therapeutic treatment.
6.3. Mechanosensory Pathways in Tumor Angiogenesis
Conventionally, biochemical signals have been believed to serve as the principal means that signaling pathways are activated in endothelial cells; however, mechanical forces have more recently also been demonstrated to regulate endothelial cell phenotype and function. Recent work has shown that mechanical forces control endothelial cell proliferation, survival, migration, and ECM remodeling, all of which play prominent roles in angiogenesis [111, 112]. Dynamic cellular response to mechanical forces is essential to vascular biology. For instance, fluid shear stress from blood flow plays a critical role in regulating vessel morphogenesis, sprouting, and barrier function [113, 114]. To convert mechanical forces and biophysical cues into intracellular biochemical signaling cascades, endothelial cells employ an interconnected system of mechanosensors to sense and respond to mechanical cues. These mechanosensors include the actin cytoskeleton, integrins, cell-cell adhesion receptors, receptor tyrosine kinases, and other membrane proteins including ion channels and G-protein-coupled receptors (Table 6.3). Often in cancer, and in tumor endothelial cells specifically, many of these mechanosensory pathways become deregulated and/or malfunction leading to abnormal tumor endothelial cell function.
Table 6.3.
Prominent mechanosensory pathways in tumor angiogenesis
Mechanosensor | Location | Mechanical activation | Relevant function | Role in tumor angiogenesis | References |
---|---|---|---|---|---|
PECAM1 | Adherens junctions, apicolateral membrane | Fluid shear stress, circumferential strain | Phosphorylated in response to mechanical forces, transactivates VEGFR | Important in changes to cytoskeletal architecture. Activates VEGFR and downstream signaling events | [114, 146, 147, 149, 151] |
VE-cadherin | Adherens junctions | Fluid shear stress, circumferential strain | Transmembrane scaffolding of PECAM1 and VEGFR2/3. Important in maintaining barrier function | Disrupted VE-cadherin endothelial cell-cell junctions are observed in stiff environments and tumor vasculature | [12, 113, 114, 152, 156] |
VEGFR2 | Adherens junctions, apical membrane | Fluid shear stress, circumferential strain | Shear stress causes ligand-independent phosphorylation, activates MAPK/PI3K/Akt | Elevated expression in tumor blood vessels. Involved in tumor EC barrier integrity. Major signal transducer for angiogenesis | [147, 149, 157–159] |
VEGFR3 | Adherens junctions, apical membrane | Fluid shear stress, circumferential strain | Shear stress causes ligand-independent phosphorylation, activates MAPK/PI3K/Akt | Involved in maintaining tumor EC barrier integrity. Inhibition reduces vascular density | [149, 160, 161] |
Integrins | Basal adhesion complexes | Fluid shear stress, cell-ECM stress | Shear stress causes downstream activation by PI3K to regulate cell orientation. Important in sensing and applying cell-ECM stresses | Inhibition of α1β1 and α2β1, α5β1, as well as αvβ5 and αvβ3 suppress tumor angiogenesis. αvβ3 and αvβ5 integrin expression linked to grade of neuroblastoma | [117, 126, 130–133, 141] |
Actin cytoskeleton | Cortical plasma membrane, cytoplasmic, perinuclear | Fluid shear stress, circumferential strain, cell-ECM stress | Fluid shear stress causes filament deformations. Inhibition blocks many responses to mechanical cues | Tumor endothelial cells demonstrate increased cellular contractility and aberrant mechanosensitivity | [80, 113, 116–118] |
TRPV4 | Apical membrane | Fluid shear stress, circumferential strain | Regulates mechanosensitivity and Rho/ROCK activity | Tumor endothelial cells have reduced TRPV4 expression, leading to aberrant Rho/ROCK mechanosensitivity | [122, 174–176] |
EP2 | Apical/basal membrane | Fluid shear stress, cell-ECM stress | Induces VEGF expression via ERK2/JNK1 activation | Released from cancer cells to elicit a pro-angiogenic response | [178, 179, 181] |
S1PR | Apical/basal membrane | Fluid shear stress, cell-ECM stress | Activation leads to Rac-Cdc42 signaling and correlates with ERK1 and ERK2 activation | Important role in regulating endothelial cell cytoskeletal structure, migration, capillary-like network formation, and vascular maturation | [178, 179, 182] |
PAR1 | Apical/basal membrane | Fluid shear stress, cell-ECM stress | Modulates Rho GTPase activity | Influences endothelial cell permeability. PAR1 expression increased in cancer | [178, 179, 183, 184] |
6.3.1. The Actin Cytoskeleton and Integrins
The actin cytoskeleton and integrins act as principal mechanotransducers in cells. Early experiments identified molecular connections between integrins, cytoskeletal filaments, and nuclear scaffolds, where exogenous force on integrins caused cytoskeletal filament reorientation, nuclei distortion, and nucleoli redistribution [115]. The cytoskeleton serves as the load-bearing architecture of the cell as well as a mechanical coupler to the ECM. As such, the cytoskeleton is vital to cellular response to environmental cues [116–118]. Adhesion proteins, known as integrins, serve as the main receptors that mediate the connection of the cytoskeleton to the surrounding ECM. ECM components bind to integrins that are linked intracellularly to the actin cytoskeleton. Mechanical stresses distributed throughout the ECM then converge on integrins [117]. The short cytoplasmic tail of integrins enable intracellular signaling cascades in response to mechanical cues, which can regulate various cell functions including cell survival, proliferation, and migration [119–121]. In endothelial cells, 130 pN force exerted on integrins has been demonstrated to elicit Rho-mediated cytoskeletal tension [122], which precedes both stress fiber and focal adhesion formation [123]. Recent work has implicated changes in cell mechanics in the pathogenesis of many diseases, including cancer. Cancer cells exhibit significantly distinct mechanical properties compared to their non-tumorigenic counterparts. As such, disruption of cytoskeletal regulation has been linked to cancer progression. Alterations to cytoskeletal organization as well as upregulation of cytoskeletal scaffolding proteins and signaling circuits contribute to an altered mechanical state and have been tied to tumorigenesis [124]. Cancer cells are associated with increased contractility, where cellular traction stresses increase with increasing metastatic potential in breast, prostate, and lung cancer models [125]. Similarly, many integrin signaling pathways are exploited in cancer to support tumor progression. Together, these alterations manipulate cell function in order to better manipulate the host microenvironment and provide abundant vasculature to the tumor to support tumor growth [126].
Changes to the ECM during tumor progression, such as ECM stiffening, are sensed through the cytoskeleton and integrin receptors. ECM stiffening causes enhanced integrin-mediated Rho/Rho-associated protein kinase (ROCK) activity and contraction in tumor epithelial cells [26, 127] as well as tumor endothelial cells [80]. Abnormal Rho-mediated sensing of mechanical forces has been suggested to contribute toward the aberrant behaviors observed in tumor endothelial cells that produce structural abnormalities [80]. Tumor endothelial cells have abnormal mechanosensitivity to uniaxial cyclic strain transmitted through the ECM [80], which has been shown to be mediated by dynamic regulation of Rho activity and cytoskeletal tension [128]. Interestingly, tumor endothelial cells also displayed thicker stress fibers, stronger adhesion strength, enhanced cytoskeletal tension, and constitutively high baseline activity of Rho and ROCK. However, normal and tumor endothelial cells express comparable levels of active β1 and β3 integrins, indicating these observations are a result of higher intrinsic Rho- and ROCK-dependent cytoskeletal tension [80]. These differences in response to mechanical cues between normal and tumor endothelial cells suggests that the abnormal mechanical and structural components of the tumor microenvironment may cause tumor endothelial cells to gradually obtain an altered phenotype. Such alteration in mechanosensitivity may additionally enable tumor endothelial cells to spread and form capillary networks over a wider range of matrix stiffness compared to normal endothelial cells [80].
Specific integrins have been demonstrated to contribute to not only angiogenesis but also tumor angiogenesis and tumor progression [129–131]. Expression of α1β1 and α2β1 integrins is upregulated by VEGF in endothelial cells [132], and combined antagonism of α1β1 and α2β1 reduces tumor growth and tumor angiogenesis of human squamous cell carcinoma xenographs [133]. The α5β1 integrin is selectively expressed in angiogenic vasculature and is necessary for proper angiogenesis [131, 134]. Endothelial cells undergoing angiogenesis upregulate αvβ3 and αvβ5 integrins in order to facilitate growth and survival of newly forming vessels [126, 135]. Cytokine-dependent pathways of angiogenesis have been shown to have a necessity for αv integrins. Integrin αvβ3 is necessary for angiogenic pathways activated by basic fibroblast growth factor (bFGF) or tumor necrosis factor α (TNF-α), and integrin αvβ5 is necessary for angiogenic pathways activated by VEGF or transforming growth factor α (TGF-α) [136]. Specifically, the αvβ5 integrin pathway downstream of VEGF causes activation of FAK and Src kinase [137]. Many of these pro-angiogenic factors have been implicated in promoting tumor angiogenesis [3]. The αvβ3 integrin has also been demonstrated to be required for angiogenesis [138], as well as associate with VEGFR2 and be involved with VEGFR2 recycling events [126]. Consequently, binding of αvβ3 to its corresponding ECM ligands has been shown to increase VEGF signaling [139, 140]. Moreover, αvβ3 and αvβ5 integrins are selectively expressed in tumor vasculature [130]. Integrin αvβ3 is highly expressed on angiogenic vessels of malignant breast carcinoma [141], and the level of expression of αvβ3 and αvβ5 integrins in tumor endothelial cells has been tied to the grade of malignancy in neuroblastoma [142]. Inhibition of αvβ3 suppressed angiogenesis and reduced tumor growth of breast carcinoma in a severe combined immunodeficient (SCID) mouse/human chimeric model [141] as well as resulted in tumor reduction in human clinical trials [143]. Combined inhibition of αvβ3 and αvβ5 integrins also significantly reduced growth of human melanoma xenografts in SCID mice [144]. Integrin α6β4 signaling has likewise been demonstrated to be involved in cancer cell invasion and selectively expressed in tumor vasculature. Specifically, integrin α6β4 is involved in the promotion and onset of the invasive phase of pathological angiogenesis. The β4 substrate domain promotes bFGF- and VEGF-mediated angiogenesis and regulates angiogenic sprouting by promoting nuclear translocation of activated ERK and NF-κB as endothelial cells migrate [129]. Furthermore, melanoma, lung, lymphoma, and fibrosarcoma tumors in mice carrying targeted deletion of the signaling portion of the integrin β4 subunit had significant reduction in size and microvascular density compared to wild-type mice, indicating the β4 substrate domain promotes tumor angiogenesis [129]. Together, these data demonstrate the role of cytoskeletal- and integrin-mediated mechanosensory pathways in facilitating tumor angiogenesis.
6.3.2. Cell-Cell Adhesion Receptors
Endothelial cells form mechanical connections to neighboring cells through a multiprotein cell-cell adhesion structure known as adherens junctions. Adherens junctions are important in endothelial monolayer integrity, contact inhibition of growth, and apoptosis [145, 146]. Within adherens junctions is a mechanosensory complex comprised of platelet endothelial cell adhesion molecule 1 (PECAM1), VE-cadherin, and VEGFR2/3. Within this complex, PECAM1 directly transmits mechanical force, VE-cadherin acts as an adaptor, and VEGFR2 activates biochemical signaling (Fig. 6.2) [113, 147]. Notably, small GTPase activity is required for the functioning of this mechanosensory complex [148].
Fig. 6.2.
The PECAM1, VE-cadherin, VEGFR mechanosensory complex utilized by endothelial cells in response to mechanical forces. (a) In response to force, tension is applied to PECAM1, followed by VE-cadherin assisted association of PECAM1 and VEGFR. (b) PECAM1 triggers Src-mediated activation of the Src family kinase Fyn, which phosphorylates and activates VEGFR2/3. VEGFR2/3 activates RhoA, PI3K, MAPK, and Akt signaling cascades that influence endothelial cell function and promote angiogenesis
PECAM1 is a transmembrane immunoglobulin family protein that participates in homophilic adhesion at cell-cell junctions. In response to mechanical stimuli, PECAM1 triggers Src-mediated activation of a Src family kinase, possibly the Src family tyrosine kinase Fyn, which phosphorylates and activates VEGFR2 [147, 149]. PECAM1 is vital to proper vascular development, and PECAM polymorphisms have been linked to pathological vessels [150]. PECAM1 and VE-cadherin based adhesions are essential for flow-induced integrin activation, and PECAM1–VE-cadherin mechanosensory response has been thought to be dependent on direct force exerted on PECAM1 [146]. Focal adhesion growth and adaptive cellular stiffening in endothelial cells occur due to integrin-dependent RhoA activation from force transduction via PECAM1. Furthermore, local mechanical stimulation of PECAM1 has been demonstrated to elicit a global cellular response, specifically force-dependent activation of PI3K and RhoA activity [151]. Together, this mechanochemical signaling response enables changes to cytoskeletal architecture and adaptive cytoskeletal stiffening.
VE-cadherin assists the association of PECAM1 and VEGFR2 through its transmembrane domain to stimulate downstream activity of VEGFR in response to mechanical activation of PECAM1 [113]. VE-cadherin also plays an important role in sensing and responding to changes in matrix stiffness. Specifically, VE-cadherin modulates cytoskeletal mechanics in response to changes in matrix stiffness through small Rho GTPases [152]. Comparable to integrin-mediated changes in contractility, cadherin-mediated increases in contractility are actin-dependent. As such, endothelial cell-cell junction integrity is maintained by VE-cadherin. In quiescent endothelial cell networks, VE-cadherin is localized linearly beside cell-cell borders to form continuous, stable adherens junctions, while VE-cadherin is organized in short linear structures perpendicular to cell-cell borders in endothelial cells with reduced network integrity [153]. In response to increased matrix stiffness, disruption of VE-cadherin-mediated cell-cell junctions results in disrupted barrier integrity and increased endothelial cell monolayer permeability in both in vitro and ex vivo models [154]. Such disruptions are also observed in tumor neovasculature. Aberrant tumor vessels demonstrate decreased levels of junctional VE-cadherin, which contributes to lowered barrier tightness and increased vascular permeability [155, 156]. However, cell contractility increases with matrix stiffness, and inhibition of Rho-mediated cell contractility has been demonstrated to decrease VE-cadherin cell-cell separation distance and restored monolayer integrity [154] as well as normalize tumor endothelial cell behavior [80]. These data demonstrate the importance of mechanical cues on VE-cadherin function and cell-cell and cell-matrix connectivity.
6.3.3. VEGFRs and VEGF Signaling
VEGFR signaling is critical for normal endothelial cell migration, proliferation, and angiogenesis. VEGFRs are transmembrane receptor tyrosine kinases (RTKs) that mediate most of the angiogenic effects of VEGF. VEGF-induced activation of RhoA is necessary for endothelial cell cytoskeleton reorganization and migration, and these changes are also accompanied by the formation of small cell-cell openings that contribute to increased permeability [157]. In response to shear stress, VEGFR2 undergoes rapid induction and nuclear translocation, followed by ligand-independent phosphorylation that causes activation of MAPK, PI3K, and Akt signaling pathways that are involved in promoting angiogenesis (Fig. 6.2) [149, 158, 159]. VEGFR2 phosphorylation is additionally accompanied by VEGFR2 membrane clustering and downstream signaling [158]. Cyclic strain prompts dissociation of VEGFR2 from VE-cadherin, which can increase vascular permeability [113]. Similarly, VEGFR3 has recently been recognized as a member of this mechanosensory complex [149], and has been suggested to be involved in maintaining endothelial barrier integrity during tumor angiogenesis [160]. Antibody inference of VEGFR3 function significantly reduced tumor growth of lung, pancreatic, renal, colon, and prostate tumor xenografts in immunocompromised mice. Notably, the blood vessel density was decreased and the amount of hypoxic and necrotic tissue was increased in these anti-VEGFR3 treated tumors [161]. Depletion of VEGFR2 and/or VEGFR3 leads to significantly diminished endothelial cell response to mechanical cues. More specifically, depletion of either VEGFR significantly lessened shear-induced integrin activation and cell alignment as well as weakly reduced PI3K and AKT signaling; however, all effectors were strongly inhibited through depletion of both VEGFRs [149].
ECM stiffness influences VEGFR expression and vascular development in vitro and in vivo. GATA2 and VEGFR2 expression is increased with increasing substrate stiffness, where GATA2 mediates p190RhoGAP-dependent control of VEGFR2 expression [162]. Matrix stiffness has also been demonstrated to alter cell response to growth factors. Substrate stiffness has recently been shown to modify the coordinated actions of VEGF-matrix binding that is critical for VEGF internalization [163]. In endothelial cells, VEGF induced changes in stress fiber organization and contractile response are mediated by VEGFR2 and ROCK signaling [157, 164]. Elevated expression of VEGFRs has also been linked to many cancers. For example, VEGFR2, the predominant receptor tyrosine kinase that mediates VEGF signaling and VEGF-mediated angiogenesis, has been identified in bladder, brain, breast, cervical, colon, endometrial, gastric, head and neck, hepatocellular, lung, melanoma, mesothelioma, multiple myeloma, myeloid leukemia, esophageal, ovarian, pancreatic, prostate, renal cell carcinoma, squamous, and thyroid human cancers [165]. In many of these tumors, VEGFR expression has been correlated with either poor survival, disease progression, and/or recurrence [165]. This increased VEGFR expression has been seen on both tumor cells and endothelial cells. Notably, compared to normal blood vessels, the expression of VEGFR1 (FLT1) as well as VEGFR2 (KDR) is enhanced in tumor blood vessels [166–168]. These data suggest that cell response to growth factor signaling is closely linked to matrix stiffness, and altered sensitivity may play an important role in tumor angiogenesis.
6.3.4. Membrane Proteins
The cell membrane offers a large target for external mechanical forces to act upon, and as such mechanosensitive ion channels present in the membrane serve as one of the earliest responses to mechanical force and changes to the microenvironment. As key operators of cell signaling, ion channels have been implicated in tumorigenesis and have altered expression in tumor cells as well as stromal and endothelial cells [169]. Recent work has demonstrated that the transient receptor potential (TRP) ion channel superfamily is linked with an array of cancers [170], and abnormal TRP ion channel function can cause sustained proliferation, evasion of growth suppressors, and resistance to cell death [171, 172].
TRP channels have also been identified to be critical to endothelial cell function, and TRP ion channel malfunction and/or dysregulation is associated with endothelial cell dysfunction including disruption of angiogenic competence and barrier maintenance [173]. Specifically, transient receptor potential vanilloid 4 (TRPV4) has been shown to regulate tumor angiogenesis and tumor endothelial cell function by modulating cellular mechanosensitivity. Tumor endothelial cells demonstrate reduced TRPV4 expression correlated with aberrant mechanosensitivity toward ECM stiffness. Together, these changes in TRPV4 expression lead to increased migration and abnormal angiogenesis [174]. Loss of TRPV4 in TPRV4 knockout endothelial cells leads to significantly increased proliferation, migration, and basal Rho activation reminiscent of tumor-derived endothelial cells [175]. Further, the absence of TRPV4 in TRPV4 knockout mice was found to result in increased vascular density, increased vessel diameter, and reduced pericyte coverage within lung carcinoma tumors compared to wild-type mice – all principle characteristics of abnormal tumor angiogenesis [174]. Either overexpression or pharmacological activation of TRPV4 or pharmacological inhibition of the downstream Rho/ROCK pathway was able to normalize tumor vasculature, reduce tumor growth, and improve cancer therapy of lung tumors in a mouse model [174, 175]. These findings provide further support that aberrant Rho/ROCK mechanosensitivity is a significant contributor to abnormal tumor endothelial cell function. Interestingly, some data also suggests that integrins and mechanosensitive ion channels are well connected [122]. Cyclic strain to endothelial cells causes activation of TRPV4, which then activates supplementary integrins and triggers downstream cytoskeletal reorganization [176]. While TRPV4 has been the most studied TRP channel in tumor angiogenesis, other TRP superfamily channels have been implicated as contributors of abnormal tumor angiogenesis as well [169]. These data further demonstrate the role of abnormal mechanosensory pathways in tumor endothelial cell function and tumor angiogenesis.
The large family of cell-surface G-protein-coupled receptors (GPCRs) have additionally been identified as contributors of tumor angiogenesis and aberrant tumor endothelial cell function. Normally, GPCRs are activated when an extracellular ligand binds to or induces an active conformation. However, fluid shear stress and increased membrane tension have also been reported to induce conformational transitions and activation of GPCRs in endothelial cells, suggesting GPCRs are involved in mediating mechanochemical signaling in endothelial cells [177]. Many GPCRs are overexpressed in various cancers. During tumor progression, cancer cells frequently take over the natural physiological functions of GPCRs to proliferate, evade immune detection, invade surrounding tissue and metastasize, as well as increase angiogenesis [178]. The GPCRs prostaglandin E2 (PGE2) receptor EP2, sphingosine-1 phosphate receptors (S1PRs), and protease-activated receptor 1 (PAR1) have all been strongly implicated in eliciting a pro-angiogenic response in breast, head and neck, colon, non-small-cell lung, and prostate cancers [178–180]. The release of PGE2 from tumor cells, due to unregulated expression of COX2, stimulates expression of EP2 receptors on endothelial cells and induces VEGF expression via ERK2/JNK1 activation [181]. S1PR1 activation has been linked to endothelial cell survival, chemotactic motility, and capillary-like network formation as well as release of pro-angiogenic cytokines from tumor cells [182]. PAR1 activation has been shown to modulate Rho GTPase activity and play an important role in endothelial adherens junction disassembly and vascular permeability [178, 183]. Notably, PAR1 expression is directly correlated with invasiveness of breast cancer, where highly metastatic human breast cell lines and breast carcinoma biopsy specimens express high levels of PAR1 [184]. Taken together, these GPCRs provoke a pro-angiogenic response in tumors via activation of a network of small GTPases, Akt, and MAPK signaling that stimulates endothelial cell migration, survival, and growth.
6.4. Clinical Impact of Abnormal Tumor Vasculature
6.4.1. Impaired Barrier Function and Delivery of Chemotherapeutics
Together, the mechanical forces found in tumors work to produce a functionally abnormal tumor vasculature with impaired barrier function. Solid tumor vasculature is often leaky with a defective endothelium. Indeed, the tumor vasculature is characterized by its defective endothelial monolayer, large intercellular openings and holes, and abnormal sprouts that all work to impair barrier function [6]. Normal endothelial cells form uniform monolayers; however, tumor endothelial cells are irregular in shape and size, have cytoplasmic projections into the vessel lumen, and form an incomplete endothelium. Tumor blood vessels have large intracellular gaps between tumor endothelial cells, highlighted by transcellular holes, fenestra, and channels [6]. Additionally, high tumor endothelial cell motility and turnover may hinder the formation of intercellular junctions, further promoting larger intercellular openings [6]. Endothelial junctions are also highly dynamic and sensitive to extracellular stimuli. As such, VE-cadherin based junctions are susceptible to continuous reorganization due to the dynamically changing tumor ECM and the aberrant mechanosensitivity of tumor endothelial cells [80]. Consequently, tumor blood vessel hyperpermeability and impaired barrier function arises due to the combined effects of tumor vessels lacking or having abnormal function of endothelial cells, pericytes, and/or basement membrane [185].
Leakiness of the tumor vasculature not only impacts tumor growth and metastasis but also has a profound impact on drug delivery to the tumor. Traditionally, vessel leakiness is believed to be due to overexpression of pro-angiogenic growth factors; however, emerging work has demonstrated that the physical environment plays an important role in impairing endothelial cell barrier integrity. Elevated ECM stiffness increases endothelial cell-cell junctional properties and endothelial permeability in vitro and in vivo [12]. Vessel compression due to mechanical forces in the tumor microenvironment causes large areas of the tumor to have limited perfusion and limited systemic administration of therapeutic agents [186–188]. Vessel compression along with the highly tortuous and disorganized arrangement of tumor blood vessels creates sluggish and heterogeneous blood flow, which can affect microvascular pressure [89, 189]. While accurate measurements of microvascular pressure are challenging to obtain, it has been reported that increased tumor interstitial fluid pressure is also accompanied by increased microvascular pressure [190]. For example, microvascular pressure in normal tissue is approximately 15 to 25 mmHg, while the microvascular pressure in tumor tissue has been reported to range from 5.5 to 34 mmHg in MCaIV mouse mammary carcinoma tumors [35]. Importantly, the elevated interstitial fluid pressure found in tumor tissue is often nearly as high as or can exceed microvascular pressure, eliminating pressure gradients across tumor vessels and inhibiting convective transport of drugs [89]. Combined, these factors severely limit the efficacy of traditional cancer treatments.
Efficient and uniform systemic delivery of cancer therapeutics is a critical challenge in cancer treatment. To increase the delivery and efficacy of therapeutics, an emerging cancer treatment strategy seeks to normalize the tumor vasculature [191]. The anti-angiogenic drug bevacizumab, an antibody targeted against VEGF, has been used in combination with chemotherapy and has produced a five month increase in survive in colorectal cancer patients [192]. Other preclinical studies have demonstrated that anti-angiogenic agents can increase perfusion and drug uptake in tumors [42, 193]. As such, this has led to many pro- and anti-angiogenic therapies that seek to restore normal vessel densities [194]; however, angiogenic signaling is robust and redundant, and inhibition of individual signaling molecules can be overcome by escape mechanisms [194, 195]. For example, initial response to anti-angiogenic therapies targeting the VEGF pathway is followed by a restoration of tumor progression. In both clinical and preclinical settings, emerging data describe that tumors develop either evasive resistance or intrinsic resistance to these treatments. [196]. For these reasons, it is essential to pursue novel methods for tumor vasculature normalization, and targeting mechanical forces in the tumor and/or mechanosensory pathways may be one possible strategy.
6.4.2. Promotion of an Aggressive Tumor Phenotype
As the vasculature not only provides oxygen and nutrients but is also a conduit for the removal of waste products, abnormalities in the vasculature are a major contributor to other abnormalities that exist in the tumor microenvironment [6]. Vascular abnormalities lead to a hypoxic and acidic tumor microenvironment [197]. It is well established that tumor blood vessels are heterogeneous in organization and structure, and tumor blood vessels are often more abundant at the tumor-host interface compared to more central regions of the tumor. Furthermore, vascular density has been reported to decrease during tumor progression [198]. As previously discussed, these heterogeneities and abnormal organization arises from changes to the ECM and accumulation of stresses during tumor progression. Consequently, the spatial disorganization and abnormal architecture of the tumor vasculature create diffusion-limited hypoxia throughout the tumor tissue as intercapillary distances often exceed 100–200 μm, the maximum nutrient and oxygen diffusion limits [2].
Such a harsh microenvironment was originally thought to starve the tumor and decrease cancer cell survival; however, it has been established that hypoxia helps to promote a more aggressive and difficult-to-treat tumor phenotype (Fig. 6.3). Specifically, the abnormal tumor microenvironment employs selective pressures that cause cancer cell populations to dynamically adapt [13]. Not only do cancer cells prosper in this harsh environment, but such selection pressures contribute to the propagation of cancer cells [6]. Hypoxia provokes proteome, induce pro-survival changes in gene expression, control the anabolic switch in central metabolism, as well as help to drive malignant progression through genomic changes in neoplastic cells [199, 200]. Additionally, a hypoxic and acidic microenvironment affects host immune response. Hypoxia and acidosis reprogram local macrophages into an immunosuppressive phenotype that helps cancer cells evade immune detection as well as diminishes the killing potential of immune effectors cells within the tumor microenvironment [13].
Fig. 6.3.
Abnormal tumor vasculature that develops during tumor progression helps to promote an abnormal tumor microenvironment that promotes a more aggressive tumor phenotype. (a) In vivo tumors from MMTV-PyMT mice treated with BAPN to soften the tumor tissue or vehicle controls showing 2 MDa FITC-labeled vasculature (green) and extravasating Evans Blue (red) (scale bar = 150 μm). Control tumors demonstrate more abnormal vascular architecture and increased vascular permeability compared to softened tumors. (b) During tumor progression, increased mechanical cues in the tumor microenvironment contribute to creating abnormal tumor vasculature that is highly permeability and inefficient in delivering oxygen and nutrients. Limited diffusion in the tumor creates a hypoxic and acidic environment that not only promote angiogenesis but also promotes genomic instability, an anabolic switch in metabolism, resistance to apoptosis, malignant progression, induction of a cancer stem cell phenotype, as well as resistance to many cancer therapies
Hypoxia also influences cancer cell response to radiation and many chemotherapeutics. This can occur through a variety of mechanisms [199]. The most widely occurring mechanisms of hypoxia-mediated resistance to cytotoxic therapy include extracellular acidification causing decreased drug uptake, resistance to apoptosis, and genomic instability that causes further mutagenesis of cancer cells. For many bio-reductive prodrugs that are intended to be metabolized, inadequate extravascular penetration of the drug significantly contributes to chemoresistance [199, 201]. Together, these findings indicate that abnormalities in the tumor vasculature help to make cancer treatments exceedingly challenging due to a rapidly altering cancer cell phenotype and resistance to many traditional therapies.
6.5. Conclusions
Mechanical forces in the tumor microenvironment play an important role in directing tumor growth and promoting abnormal tumor vasculature. Stiffening of the tumor ECM promotes abnormal branching patterns, vascular density, as well as increased endothelial cell-cell junctions and permeability, whereas mechanical stresses in the tumor compress tumor blood vessels and limit perfusion. Growing evidence indicates that such mechanical alterations in the tumor microenvironment help to alter tumor endothelial cell phenotype and mechanosensitivity. This abnormal mechanosensitivity is now being tied to deregulated or malfunctioning mechanosensors in tumor endothelial cells. While it is clear that the mechanical microenvironment mediates tumor angiogenesis, much work still remains to fully understand specific mechanosensory pathways utilized by endothelial cells to respond to aberrant mechanical cues. Identifying these pathways will better our understanding of mechanical regulation in tumor angiogenesis and provide new methods to tame the physical forces in tumors. Such findings will provide important understanding to how changes in the tumor microenvironment facilitate tumor progression and may present new therapeutic targets to normalize the tumor vasculature.
6.6. Acknowledgements
The authors gratefully acknowledge support from the National Heart, Lung and Blood Institute (HL127499) to CAR-K and a National Science Foundation Graduate Research Fellowship under Grant No. DGE-1650441 to MRZ.
Footnotes
The MMTV-PyMT transgenic mouse model is widely used to study mammary tumor progression and metastasis. In the MMTV-PyMT model, mammary gland-specific expression of the polyoma middle T antigen (PyMT) oncogene driven by the upstream mouse mammary tumor virus (MMTV) long terminal repeat promoter results in mammary epithelium transformation and rapid development of multifocal mammary adenocarcinomas and metastatic lesions.
References
- 1.Hanahan D, Folkman J (1996) Patterns and emerging mechanisms of the angiogenic switch during tumorigenesis. Cell 86:353–364 [DOI] [PubMed] [Google Scholar]
- 2.Carmeliet P, Jain RK (2000) Angiogenesis in cancer and other diseases. Nature 407:249–257 [DOI] [PubMed] [Google Scholar]
- 3.Bergers G, Benjamin LE (2003) Tumorigenesis and the angiogenic switch. Nat Rev Cancer 3(6):401–410 [DOI] [PubMed] [Google Scholar]
- 4.Less JR et al. (1991) Microvascular architecture in a mammary carcinoma: branching patterns and vessel dimensions. Cancer Res 51(265):265–273 [PubMed] [Google Scholar]
- 5.Baluk P, Hashizume H, McDonald DM (2005) Cellular abnormalities of blood vessels as targets in cancer. Curr Opin Genet Dev 15(1):102–11 [DOI] [PubMed] [Google Scholar]
- 6.Dudley AC (2012) Tumor endothelial cells. Cold Spring Harb Perspect Med 2(3):1–18 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Aird WC (2012) Endothelial cell heterogeneity. Cold Spring Harb Perspect Med 2(1):a006429–a006429 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Aird WC (2009) Molecular heterogeneity of tumor endothelium. Cell Tissue Res 335(1):271–281 [DOI] [PubMed] [Google Scholar]
- 9.Hashizume H et al. (2000) Openings between defective endothelial cells explain tumor vessel leakiness. Am J Pathol 156(4):1363–1380 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Hobbs SK et al. (1998) Regulation of transport pathways in tumor vessels: role of tumor type and microenvironment. Proc Natl Acad Sci 95(8):4607–4612 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Dvorak HF et al. (1999) Vascular permeability fac- tor/vascular endothelial growth factor and the significance of microvascular hyperpermeability in angiogenesis. Curr Top Microbiol Immunol 237:97–132 [DOI] [PubMed] [Google Scholar]
- 12.Bordeleau F et al. (2017) Matrix stiffening promotes a tumor vasculature phenotype. Proc Natl Acad Sci 114(3):492–497 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Jain RK (2014) Antiangiogenesis strategies revisited: from starving tumors to alleviating hypoxia. Cancer Cell 26(5):605–622 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Lu P, Weaver VM, Werb Z (2012) The extracellular matrix: a dynamic niche in cancer progression. J Cell Biol 196(4):395–406 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Daley WP, Peters SB, Larsen M (2008) Extracellular matrix dynamics in development and regenerative medicine. J Cell Sci 121(3):255–264 [DOI] [PubMed] [Google Scholar]
- 16.Kim DH et al. (2012) Matrix nanotopography as a regulator of cell function. J Cell Biol 197(3):351–360 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Hynes RO (2009) The extracellular matrix: not just pretty fibrils. Science 326(5957):1216–1219 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Mongiat M et al. (2016) Extracellular matrix, a hard player in angiogenesis. Int J Mol Sci 17(11): 1822. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Bonnans C, Chou J, Werb Z (2014) Remodelling the extracellular matrix in development and disease. Nat Rev Mol Cell Biol 15(12):786–801 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Mammoto T, Ingber DE (2010) Mechanical control of tissue and organ development. Development 137(9):1407–1420 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Provenzano PP et al. (2006) Collagen reorganization at the tumor-stromal interface facilitates local inva- sion. BMC Med 4(1):38–38 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Wiseman BS (2002) Stromal effects on mammary gland development and breast cancer. Science 296(5570):1046–1049 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Butcher DT, Alliston T, Weaver VM (2009) A tense situation: forcing tumour progression. Nat Rev Cancer 9(2):108–122 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Grassian AR, Coloff JL, Brugge JS (2011) Extra- cellular matrix regulation of metabolism and implications for tumorigenesis. Cold Spring Harb Symp Quant Biol 76:313–324 [DOI] [PubMed] [Google Scholar]
- 25.Morris BA et al. (2016) Collagen matrix density drives the metabolic shift in breast cancer cells. EBioMedicine 13:146–156 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Paszek MJ et al. (2005) Tensional homeostasis and the malignant phenotype. Cancer Cell 8(3):241–254 [DOI] [PubMed] [Google Scholar]
- 27.Wozniak MA et al. (2003) ROCK-generated contractility regulates breast epithelial cell differentiation in response to the physical properties of a three-dimensional collagen matrix. J Cell Biol 163(3):583–595 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Provenzano PP et al. (2009) Matrix density-induced mechanoregulation of breast cell phenotype, signaling and gene expression through a FAK-ERK linkage. Oncogene 28(49):4326–4343 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Samani A et al. (2003) Measuring the elastic modulus of ex vivo small tissue samples. Phys Med Biol 48(14):2183–2198 [DOI] [PubMed] [Google Scholar]
- 30.Gefen A, Dilmoney B (2007) Mechanics of the normal woman’s breast. Technol Health Care 15(4):259–271 [PubMed] [Google Scholar]
- 31.Jain RK (1994) Barriers to drug delivery in solid tumors. Sci Am 271(1):58–65 [DOI] [PubMed] [Google Scholar]
- 32.Less JR et al. (1992) Interstitial hypertension in human breast and colorectal tumors. Cancer Res 52(22):6371–6374 [PubMed] [Google Scholar]
- 33.Nathanson SD, Nelson L (1994) Interstitial fluid pressure in breast cancer, benign breast conditions, and breast parenchyma. Ann Surg Oncol 1(4):333–338 [DOI] [PubMed] [Google Scholar]
- 34.Stylianopoulos T et al. (2012) Causes, consequences, and remedies for growth-induced solid stress in murine and human tumors. Proc Natl Acad Sci 109(38):15101–15108 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Jain RK, Tong RT, Munn LL (2007) Effect of vascular normalization by antiangiogenic therapy on interstitial hypertension, peritumor edema, and lymphatic metastasis: insights from a mathematical model. Cancer Res 67(6):2729–2735 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Ebihara T et al. (2000) Changes in extracellular matrix and tissue viscoelasticity in bleomycin-induced lung fibrosis. Temporal aspects. Am J Respir Crit Care Med 162(4):1569–1576 [DOI] [PubMed] [Google Scholar]
- 37.Aukland K, Reed RK (1993) Interstitial-lymphatic mechanisms in the control of extracellular fluid volume. Physiol Rev 73(1):1–78 [DOI] [PubMed] [Google Scholar]
- 38.Mori T et al. (2015) Interstitial fluid pressure correlates clinicopathological factors of lung cancer. Ann Thorac Cardiovasc Surg 21(3):201–208 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Kumar S, Weaver VM (2009) Mechanics, malignancy, and metastasis: the force journey of a tumor cell. Cancer Metastasis Rev 28(1–2):113–127 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Boucher Y et al. (1997) Interstitial fluid pressure in intracranial tumours in patients and in rodents. Br J Cancer 75(6):829–836 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Arbit E, Lee J, Diresta G (1994) Interstitial hypertension in human brain tumors: possible role in peritumoral edema formation In: Nagai H, Kamlya K (eds) Intracranial pressure, 9th edn Springer, Tokyo [Google Scholar]
- 42.Goel S et al. (2011) Normalization of the vasculature for treatment of cancer and other diseases. Physiol Rev 91(3):1071–1121 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Navalitloha Y et al. (2006) Therapeutic implications of tumor interstitial fluid pressure in subcutaneous RG-2 tumors. Neuro-Oncology 8(3):227–233 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Nia HT et al. (2016) Solid stress and elastic energy as measures of tumour mechanopathology. Nat Biomed Eng 1 10.1038/s41551-016-0004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Guyton AC, Hall JE (2006) Textbook of medical physiology, Guyton physiology series. Elsevier Saunders, Amsterdam [Google Scholar]
- 46.Wells RG (2008) The role of matrix stiffness in regulating cell behavior. Hepatology 47(4):1394–1400 [DOI] [PubMed] [Google Scholar]
- 47.Yeh WC et al. (2002) Elastic modulus measurements of human liver and correlation with pathology. Ultrasound Med Biol 28(4):467–474 [DOI] [PubMed] [Google Scholar]
- 48.Hori K et al. (1986) Increased tumor tissue pressure in association with the growth of rat tumors. Jpn J Cancer Res 77(1):65–73 [PubMed] [Google Scholar]
- 49.Kawano S et al. (2015) Assessment of elasticity of colorectal cancer tissue, clinical utility, pathological and phenotypical relevance. Cancer Sci 106(9):1232–1239 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Johnson LA et al. (2013) Matrix stiffness corresponding to strictured bowel induces a fibrogenic response in human colonic fibroblasts. Inflamm Bowel Dis 19(5):891–903 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Nebuloni M et al. (2016) Insight on colorectal carcinoma infiltration by studying perilesional extracellular matrix. Sci Rep 6:22522. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Netti PA et al. (2000) Role of extracellular matrix assembly in interstitial transport in solid tumors role of extracellular matrix assembly in interstitial transport in solid tumors. Cancer Res 60:2497–2503 [PubMed] [Google Scholar]
- 53.Stanczyk M et al. (2010) Lack of functioning lymphatics and accumulation of tissue fluid/lymph in interstitial “lakes” in colon cancer tissue. Lymphology 43(4):158–167 [PubMed] [Google Scholar]
- 54.Levental I, Georges PC, Janmey PA (2007) Soft biological materials and their impact on cell function. Soft Matter 3:2990306. [DOI] [PubMed] [Google Scholar]
- 55.Lee JW et al. (2011) Palpation device for the identification of kidney and bladder cancer: a pilot study. Yonsei Med J 52(5):768–772 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Wortman T, Hsu F, Slocum A (2016) A novel phantom tissue model for skin elasticity quantification. ASME J Med Devices 10(2):020961 [Google Scholar]
- 57.Boucher Y et al. (1991) Interstitial hypertension in superficial metastatic melanomas in humans. Cancer Res 51(24):6691–6694 [PubMed] [Google Scholar]
- 58.Rice AJ et al. (2017) Matrix stiffness induces epithelial-mesenchymal transition and promotes chemoresistance in pancreatic cancer cells. Oncogene 6(7):e352. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Provenzano PP et al. (2012) Enzymatic targeting of the stroma ablates physical barriers to treatment of pancreatic ductal adenocarcinoma. Cancer Cell 21(3):418–429 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Zysset PK et al. (1999) Elastic modulus and hardness of cortical and trabecular bone lamellae measured by nanoindentation in the human femur. J Biomech 32(10):1005–1012 [DOI] [PubMed] [Google Scholar]
- 61.Odgaard A, Linde F (1991) The underestimation of Young’s modulus in compressive testing of cancellous bone specimens. J Biomech 24(8): 691–698 [DOI] [PubMed] [Google Scholar]
- 62.Nathan SS et al. (2008) Tumor interstitial fluid pressure may regulate angiogenic factors in osteosarcoma. J Orthop Res 26(11):1520–1525 [DOI] [PubMed] [Google Scholar]
- 63.Davis GE, Senger DR (2005) Endothelial extracellular matrix: biosynthesis, remodeling, and functions during vascular morphogenesis and neovessel stabilization. Circ Res 97(11):1093–1107 [DOI] [PubMed] [Google Scholar]
- 64.Ingber DE, Folkman J (1989) Mechanochemical switching between growth and differentiation during fibroblast growth factor-stimulated angiogenesis in vitro: role of extracellular matrix. J Cell Biol 109(1):317–330 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Vailheé B et al. (1997) In vitro angiogenesis is modulated by the mechanical properties of fibrin gels and is related to alpha(v)beta3 integrin localization. In Vitro Cell Dev Biol Anim 33(10): 763–773 [DOI] [PubMed] [Google Scholar]
- 66.Ghajar CM et al. (2008) The effect of matrix density on the regulation of 3-D capillary morphogenesis. Biophys J 94(5):1930–1941 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Rao RR et al. (2012) Matrix composition regulates three-dimensional network formation by endothelial cells and mesenchymal stem cells in collagen fibrin materials. Angiogenesis 15(2):253–264 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Kniazeva E, Putnam AJ (2009) Endothelial cell traction and ECM density influence both capillary morphogenesis and maintenance in 3-D. Am J Physiol Cell Physiol 297(1):C179–C187 [DOI] [PubMed] [Google Scholar]
- 69.Mason BN et al. (2013) Tuning three-dimensional collagen matrix stiffness independently of collagen concentration modulates endothelial cell behavior. Acta Biomater 9(1):4635–4644 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Sieminski AL, Hebbel RP, Gooch KJ (2004) The relative magnitudes of endothelial force generation and matrix stiffness modulate capillary morphogenesis in vitro. Exp Cell Res 297(2):574–584 [DOI] [PubMed] [Google Scholar]
- 71.LaValley DJ, Reinhart-King CA (2014) Matrix stiffening in the formation of blood vessel. Adv Regen Biol 1:1–18 [Google Scholar]
- 72.Wu Y, Al-Ameen MA, Ghosh G (2014) Integrated effects of matrix mechanics and vascular endothelial growth factor (VEGF) on capillary sprouting. Ann Biomed Eng 42(5):1024–1036 [DOI] [PubMed] [Google Scholar]
- 73.Cox TR, Erler JT (2011) Remodeling and homeostasis of the extracellular matrix: implications for fibrotic diseases and cancer. Dis Model Mech 4(2):165–178 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Bershadsky AD, Balaban NQ, Geiger B (2003) Adhesion-dependent cell mechanosensitivity. Annu Rev Cell Dev Biol 19(1):677–695 [DOI] [PubMed] [Google Scholar]
- 75.Ingber DE (1991) Integrins as mechanochemical transducers. Curr Opin Cell Biol 3(5):841–848 [DOI] [PubMed] [Google Scholar]
- 76.Polet F, Feron O (2013) Endothelial cell metabolism and tumour angiogenesis: glucose and glutamine as essential fuels and lactate as the driving force. J Intern Med 273(2):156–165 [DOI] [PubMed] [Google Scholar]
- 77.Liu Z et al. (2010) Mechanical tugging force regulates the size of cell-cell junctions. Proc Natl Acad Sci 107(22):9944–9949 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Croix BS et al. (2000) Genes expressed in human tumor endothelium. Science 289(5482):1197–1202 [DOI] [PubMed] [Google Scholar]
- 79.Bussolati B et al. (2003) Altered angiogenesis and survival in human tumor-derived endothelial cells. FASEB J 17(9):1159–1161 [DOI] [PubMed] [Google Scholar]
- 80.Ghosh K et al. (2008) Tumor-derived endothelial cells exhibit aberrant Rho-mediated mechanosensing and abnormal angiogenesis in vitro. Proc Natl Acad Sci 105(32):11305–11310 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Francis-Sedlak ME et al. (2010) Collagen glycation alters neovascularization in vitro and in vivo. Microvasc Res 80(1):3–9 [DOI] [PubMed] [Google Scholar]
- 82.Lee PF et al. (2013) Angiogenic responses are enhanced in mechanically and microscopically characterized, microbial transglutaminase crosslinked collagen matrices with increased stiffness. Acta Biomater 9(7):7178–7190 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Whittington CF, Yoder MC, Voytik-Harbin SL (2013) Collagen-polymer guidance of vessel net- work formation and stabilization by endothelial colony forming cells in vitro. Macromol Biosci 13(9):1135–1149 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Yao C et al. (2008) The effect of cross-linking of collagen matrices on their angiogenic capability. Biomaterials 29(1):66–74 [DOI] [PubMed] [Google Scholar]
- 85.Yamamura N et al. (2007) Effects of the mechanical properties of collagen gel on the in vitro formation of microvessel networks by endothelial cells. Tissue Eng 13(7):1443–1453 [DOI] [PubMed] [Google Scholar]
- 86.Fantozzi A, Christofori G (2006) Mouse models of breast cancer metastasis. Breast Cancer Res 8(4):212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Sternlicht MD, Werb Z (2001) How matrix metalloproteinases regulate cell behavior. Annu Rev Cell Dev Biol 17(1):463–516 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Haage A, Schneider IC (2014) Cellular contractility and extracellular matrix stiffness regulate matrix metalloproteinase activity in pancreatic cancer cells. FASEB J 28(8):3589–3599 [DOI] [PubMed] [Google Scholar]
- 89.Jain RK, Martin JD, Stylianopoulos T (2014) The role of mechanical forces in tumor growth and therapy. Annu Rev Biomed Eng 16(1):321–346 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Stylianopoulos T et al. (2013) Coevolution of solid stress and interstitial fluid pressure in tumors during progression: implications for vascular collapse. Cancer Res 73(13):3833–3841 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Harris AK, Stopak D, Wild P (1981) Fibroblast traction as a mechanism for collagen morphogenesis. Nature 290(5803):249–251 [DOI] [PubMed] [Google Scholar]
- 92.Miron-Mendoza M, Seemann J, Grinnell F (2008) Collagen fibril flow and tissue translocation coupled to fibroblast migration in 3D collagen matrices. Mol Biol Cell 19(5):2051–2058 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Shi Q et al. (2014) Rapid disorganization of mechanically interacting systems of mammary acini. Proc Natl Acad Sci 111(2):658–663 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Wang H et al. (2014) Long-range force transmission in fibrous matrices enabled by tension-driven align- ment of fibers. Biophys J 107(11):2592–2603 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Kilarski WW et al. (2009) Biomechanical regulation of blood vessel growth during tissue vascularization. Nat Med 15(6):657–664 [DOI] [PubMed] [Google Scholar]
- 96.Korff T, Augustin HG (1999) Tensional forces in fibrillar extracellular matrices control directional capillary sprouting. J Cell Sci 112(19):3249–3258 [DOI] [PubMed] [Google Scholar]
- 97.Kenyon BM et al. (1996) A model of angiogenesis in the mouse cornea. Invest Ophthalmol Vis Sci 37(8):1625–1632 [PubMed] [Google Scholar]
- 98.Lockhart AC et al. (2003) A clinical model of dermal wound angiogenesis. Wound Repair Regen 11(4):306–313 [DOI] [PubMed] [Google Scholar]
- 99.Padera TP et al. (2004) Pathology: cancer cells compress intratumour vessels. Nature 427(6976):695. [DOI] [PubMed] [Google Scholar]
- 100.Boucher Y, Jain RK (1992) Microvascular pressure is the principal driving force for interstitial hyper- tension in solid tumors: implications for vascular collapse. Cancer Res 52(18):5110–5114 [PubMed] [Google Scholar]
- 101.Potente M, Maäkinen T (2017) Vascular heterogeneity and specialization in development and disease. Nat Rev Mol Cell Biol 18(8):477. [DOI] [PubMed] [Google Scholar]
- 102.Helmlinger G et al. (1991) Effects of pulsatile flow on cultured vascular endothelial cell morphology. J Biomech Eng 113(2):123–131 [DOI] [PubMed] [Google Scholar]
- 103.Wang Y et al. (2007) Selective adapter recruitment and differential signaling networks by VEGF vs. shear stress. Proc Natl Acad Sci 104(21):8875–8879 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Kappas NC et al. (2008) The VEGF receptor Flt-1 spatially modulates Flk-1 signaling and blood vessel branching. J Cell Biol 181(5):847–858 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Price GM et al. (2010) Effect of mechanical fac- tors on the function of engineered human blood microvessels in microfluidic collagen gels. Biomaterials 31(24):6182–6189 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Song JW, Munn LL (2011) Fluid forces control endothelial sprouting. Proc Natl Acad Sci 108(37):15342–15347 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Pa Galie et al. (2014) Fluid shear stress threshold regulates angiogenic sprouting. Proc Natl Acad Sci 111(22):7968–7973 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Vickerman V, Kamm RD (2012) Mechanism of a flow-gated angiogenesis switch: early signaling events at cell–matrix and cell–cell junctions. Integr Biol 4(8):863–874 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Ausprunk DH, Folkman J (1977) Migration and proliferation of endothelial cells in preformed and newly formed blood vessels during tumor angiogenesis. Microvasc Res 14(1):53–65 [DOI] [PubMed] [Google Scholar]
- 110.Chauhan VP et al. (2014) Compression of pancreatic tumor blood vessels by hyaluronan is caused by solid stress and not interstitial fluid pressure. Cancer Cell 26(1):14–15 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Li S, Huang NF, Hsu S (2005) Mechanotransduction in endothelial cell migration. J Cell Biochem 96(6):1110–1126 [DOI] [PubMed] [Google Scholar]
- 112.Li YSJ, Haga JH, Chien S (2005) Molecular basis of the effects of shear stress on vascular endothelial cells. J Biomech 38(10):1949–1971 [DOI] [PubMed] [Google Scholar]
- 113.Kutys ML, Chen CS (2016) Forces and mechanotransduction in 3D vascular biology. Curr Opin Cell Biol 42:73–79 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Conway DE et al. (2013) Fluid shear stress on endothelial cells modulates mechanical tension across VE-cadherin and PECAM-1. Curr Biol 23(11):1024–1030 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Maniotis AJ, Chen CS, Ingber DE (1997) Demon- stration of mechanical connections between inte- grins, cytoskeletal filaments, and nucleoplasm that stabilize nuclear structure. Proc Natl Acad Sci 94(3):849–854 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Stevenson RP, Veltman D, Machesky LM (2012) Actin-bundling proteins in cancer progression at a glance. J Cell Sci 125:1073–1079 [DOI] [PubMed] [Google Scholar]
- 117.Alenghat FJ, Ingber DE (2002) Mechanotransduction: all signals point to cytoskeleton, matrix, and integrins. Sci Signal 2002(119):pe6. [DOI] [PubMed] [Google Scholar]
- 118.Schwarz US, Gardel ML (2012) United we stand –integrating the actin cytoskeleton and cell–matrix adhesions in cellular mechanotransduction. J Cell Sci 125(13):3051–3060 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Katsumi A et al. (2004) Integrins in mechanotransduction. J Biol Chem 279(13):12001–12004 [DOI] [PubMed] [Google Scholar]
- 120.Giancotti FG, Ruoslahti E (1999) Integrin signaling. Science 285(5430):1028–1033 [DOI] [PubMed] [Google Scholar]
- 121.Miranti CK, Brugge JS (2002) Sensing the environment: a historical perspective on integrin signal transduction. Nat Cell Biol 4(4):E83–E90 [DOI] [PubMed] [Google Scholar]
- 122.Matthews BD et al. (2006) Cellular adaptation to mechanical stress: role of integrins, Rho, cytoskeletal tension and mechanosensitive ion channels. J Cell Sci 119(3):508–518 [DOI] [PubMed] [Google Scholar]
- 123.Reinhart-King CA, Dembo M, Hammer DA (2005) The dynamics and mechanics of endothelial cell spreading. Biophys J 89(1):676–689 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Zanotelli MR, Bordeleau F, Reinhart-King CA (2017) Subcellular regulation of cancer cell mechanics. Curr Opin Biomed Eng 1:8–14 [Google Scholar]
- 125.Kraning-Rush CM, Califano JP, Reinhart-King CA (2012) Cellular traction stresses increase with increasing metastatic potential. PLoS One 7(2):e32572–e32572 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Weis SM, Cheresh DA (2011) αV integrins in angiogenesis and cancer. Cold Spring Harb Perspect Med 1(1):1–14 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Paszek MJ, Weaver VM (2004) The tension mounts: mechanics meets morphogenesis and malignancy. J Mammary Gland Biol Neoplasia 9(4):325–342 [DOI] [PubMed] [Google Scholar]
- 128.Tzima E et al. (2001) Activation of integrins in endothelial cells by fluid shear stress mediates Rho-dependent cytoskeletal alignment. EMBO J 20(17):4639–4647 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Nikolopoulos SN et al. (2004) Integrin β4 signaling promotes tumor angiogenesis. Cancer Cell 6(5):471–483 [DOI] [PubMed] [Google Scholar]
- 130.Ruoslahti E (2000) Targeting tumor vasculature with homing peptides from phage display. Semin Cancer Biol 10(6):435–442 [DOI] [PubMed] [Google Scholar]
- 131.Ruoslahti E (2002) Specialization of tumour vasculature. Nat Rev Cancer 2(2):83–90 [DOI] [PubMed] [Google Scholar]
- 132.Senger DR et al. (1997) Angiogenesis promoted by vascular endothelial growth factor: regulation through α1β1 and α2β1 integrins. Proc Natl Acad Sci 94(25):13612–13617 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Senger D et al. (2002) The α1β1 and α2β1 integrins provide critical support for vascular endothelial growth factor signaling, endothelial cell migration, and tumor angiogenesis. Am J Pathol 160(1):195–204 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Kim S et al. (2000) Regulation of angiogenesis in vivo by ligation of integrin α5β1 with the central cell-binding domain of fibronectin. Am J Pathol 156(4):1345–1362 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Brooks PC et al. (1994) Integrin αvβ3 antagonists promote tumor regression by inducing apoptosis of angiogenic blood vessels. Cell 79(7):1157–1164 [DOI] [PubMed] [Google Scholar]
- 136.Friedlander M et al. (1995) Definition of two angiogenic pathways by distinct αv integrins. Science 270(5241):1500–1502 [DOI] [PubMed] [Google Scholar]
- 137.Hood JD et al. (2003) Differential alphav integrin- mediated Ras-ERK signaling during two pathways of angiogenesis. J Cell Biol 162(5):933–943 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Brooks PC, Clark RAF, Cheresh DA (1994) Requirement of vascular integrin αvβ3 for angiogenesis. Science 264(5158):569–571 [DOI] [PubMed] [Google Scholar]
- 139.Soldi R et al. (1999) Role of αvβ3 integrin in the activation of vascular endothelial growth factor receptor-2. EMBO J 18(4):882–892 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Borges E, Jan Y, Ruoslahti E (2000) Platelet-derived growth factor receptor beta and vascular endothelial growth factor receptor 2 bind to the beta 3 integrin through its extracellular domain. J Biol Chem 275(51):39867–39873 [DOI] [PubMed] [Google Scholar]
- 141.Brooks PC et al. (1995) Antiintegrin αvβ3 blocks human breast cancer growth and angiogenesis in human skin. J Clin Investig 96(4):1815–1822 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Erdreich-Epstein A et al. (2000) Integrins alpha(v)beta3 and alpha(v)beta5 are expressed by endothelium of high-risk neuroblastoma and their inhibition is associated with increased endogenous ceramide. Cancer Res 60(3):712–721 [PubMed] [Google Scholar]
- 143.Eliceiri BP, Cheresh Da (1999) The role of αv integrins during angiogenesis: insights into potential mechanisms of action and clinical development. J Clin Dev 103(9):1227–12330 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Kumar CC et al. (2001) Inhibition of angiogenesis and tumor growth by SCH221153, a dual alpha(v)beta3 and alpha(v)beta5 integrin receptor antagonist. Cancer Res 61(5):2232–2238 [PubMed] [Google Scholar]
- 145.Bazzoni G, Dejana E (2004) Endothelial cell-to- cell junctions: molecular organization and role in vascular homeostasis. Physiol Rev 84(3):869–901 [DOI] [PubMed] [Google Scholar]
- 146.Dorland YL, Huveneers S (2017) Cell–cell junctional mechanotransduction in endothelial remodeling. Cell Mol Life Sci 74(2):279–292 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Hahn C, Schwartz MA (2009) Mechanotransduction in vascular physiology and atherogenesis. Nat Rev Mol Cell Biol 10(1):53–62 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Lakshmikanthan S et al. (2015) Rap1 promotes endothelial mechanosensing complex formation, NO release and normal endothelial function. EMBO Rep 16(5):628–637 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Coon BG et al. (2015) Intramembrane binding of VE-cadherin to VEGFR2 and VEGFR3 assembles the endothelial mechanosensory complex. J Cell Biol 208(7):975–986 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Conway D, Schwartz MA (2012) Lessons from the endothelial junctional mechanosensory com- plex. F1000 Biol Rep 4(1):2–7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Collins C et al. (2012) Localized tensional forces on PECAM-1 elicit a global mechanotransduction response via the integrin-RhoA pathway. Curr Biol 22(22):2087–2094 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Murakami M, Simons M (2009) Regulation of vascular integrity. J Mol Med 87(6):571–582 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Fraccaroli A et al. (2015) Endothelial alpha-parvin controls integrity of developing vasculature and is required for maintenance of cell–cell junctions. Circ Res 117(1):19–40 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Huynh J et al. (2011) Age-related intimal stiffening enhances endothelial permeability and leukocyte transmigration. Sci Transl Med 3(112):112ra122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Mazzone M et al. (2009) Heterozygous deficiency of PHD2 restores tumor oxygenation and inhibits metastasis via endothelial normalization. Cell 136(5):839–851 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Giannotta M, Trani M, Dejana E (2013) VE- cadherin and endothelial adherens junctions: active guardians of vascular integrity. Dev Cell 26(5):441–454 [DOI] [PubMed] [Google Scholar]
- 157.van Nieuw Amerongen GP et al. (2003) Involvement of RhoA/Rho kinase signaling in VEGF-induced endothelial cell migration and angiogenesis in vitro. Arterioscler Thromb Vasc Biol 23(2):211–217 [DOI] [PubMed] [Google Scholar]
- 158.Shay-salit A et al. (2002) VEGF receptor 2 and the adherens junction as a mechanical transducer in vascular endothelial cells. Proc Natl Acad Sci 99(14):9462–9467 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Koch S, Claesson-Welsh L (2012) Signal transduc- tion by vascular endothelial growth factor receptors. Cold Spring Harb Perspect Med 2(7):a006502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Kubo H et al. (2000) Involvement of vascular endothelial growth factor receptor-3 in maintenance of integrity of endothelial cell lining during tumor angiogenesis. Blood 96(2):546–553 [PubMed] [Google Scholar]
- 161.Laakkonen P et al. (2007) Vascular endothelial growth factor receptor 3 is involved in tumor angio- genesis and growth. Cancer Res 67(2):593–599 [DOI] [PubMed] [Google Scholar]
- 162.Mammoto A et al. (2009) A mechanosensitive transcriptional mechanism that controls angiogenesis. Nature 457(7233):1103–1108 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Sack KD, Teran M, Nugent MA (2016) Extra-cellular matrix stiffness controls vegf signaling and processing in endothelial cells. J Cell Physiol 231(9):2026–2039 [DOI] [PubMed] [Google Scholar]
- 164.Yang MT, Reich DH, Chen CS (2011) Measurement and analysis of traction force dynamics in response to vasoactive agonists. Integr Biol 3(6):663–674 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Goel HL, Mercurio AM (2013) VEGF targets the tumour cell. Nat Rev Cancer 13(12):871–882 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Ogawa K et al. (2000) The ephrin-A1 ligand and its receptor, EphA2, are expressed during tumor neovascularization. Oncogene 19(52):6043–6052 [DOI] [PubMed] [Google Scholar]
- 167.Shin D et al. (2001) Expression of EphrinB2 identifies a stable genetic difference between arterial and venous vascular smooth muscle as well as endothelial cells, and marks subsets of microvessels at sites of adult neovascularization. Dev Biol 230(2):139–150 [DOI] [PubMed] [Google Scholar]
- 168.Gale NW et al. (2001) Ephrin-B2 selectively marks arterial vessels and neovascularization sites in the adult, with expression in both endothelial and smooth-muscle cells. Dev Biol 230(2):151–160 [DOI] [PubMed] [Google Scholar]
- 169.Martial S (2016) Involvement of ion channels and transporters in carcinoma angiogenesis and metastasis. Am J Physiol Cell Physiol 310(9):C710–C727 [DOI] [PubMed] [Google Scholar]
- 170.Liberati S et al. (2013) Oncogenic and anti- oncogenic effects of transient receptor potential channels. Curr Top Med Chem 13(3):344–366 [DOI] [PubMed] [Google Scholar]
- 171.Lehen’kyi V, Prevarskaya N (2011) Oncogenic TRP channels. Adv Exp Med Biol 704:929–945 [DOI] [PubMed] [Google Scholar]
- 172.Lehen’kyi V, Prevarskaya N (2011) Study of TRP channels in cancer cells In: Zhu MX (ed) TRP channels. CRC Press/Taylor & Francis. Llc, Boca Raton (FL) [PubMed] [Google Scholar]
- 173.Kwan H-Y, Huang Y, Yao X (2007) TRP channels in endothelial function and dysfunction. Biochim Biophys Acta 1772(8):907–914 [DOI] [PubMed] [Google Scholar]
- 174.Adapala RK et al. (2016) Activation of mechanosensitive ion channel TRPV4 normalizes tumor vasculature and improves cancer therapy. Oncogene 35(3):314–322 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Thoppil RJ et al. (2016) TRPV4 channels regulate tumor angiogenesis via modulation of Rho/Rho kinase pathway. Oncotarget 7(18): 25849–25861 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Thodeti CK et al. (2009) TRPV4 channels mediate cyclic strain-induced endothelial cell reorienta- tion through integrin-to-integrin signaling. Circ Res 104(9):1123–1130 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Chachisvilis M, Zhang YL, Frangos JA (2006) G protein-coupled receptors sense fluid shear stress in endothelial cells. Proc Natl Acad Sci 103(42):15463–15468 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Dorsam RT, Gutkind JS (2007) G-protein-coupled receptors and cancer. Nat Rev Cancer 7(2):79–94 [DOI] [PubMed] [Google Scholar]
- 179.Richard DE, Vouret-Craviari V, Pouysseégur J (2001) Angiogenesis and G-protein-coupled receptors: signals that bridge the gap. Oncogene 20(1):1556–1562 [DOI] [PubMed] [Google Scholar]
- 180.O’Hayre M, Degese MS, Gutkind JS (2014) Novel insights into G protein and G protein-coupled receptor signaling in cancer. Curr Opin Cell Biol 27:126–135 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Pai R et al. (2001) PGE2 stimulates VEGF ex- pression in endothelial cells via ERK2/JNK1 signaling pathways. Biochem Biophys Res Commun 286(5):923–928 [DOI] [PubMed] [Google Scholar]
- 182.Visentin B et al. (2006) Validation of an anti- sphingosine-1-phosphate antibody as a potential therapeutic in reducing growth, invasion, and angiogenesis in multiple tumor lineages. Cancer Cell 9(3):225–238 [DOI] [PubMed] [Google Scholar]
- 183.Vouret-Craviari V, Grall D, Van Obberghen-Schilling E (2003) Modulation of Rho GTPase activity in endothelial cells by selective proteinase-activated receptor (PAR) agonists. J Thromb Haemost 1(5):1103–1111 [DOI] [PubMed] [Google Scholar]
- 184.Evan-Ram S et al. (1998) Thrombin receptor over-expression in malignant and physiological invasion processes. Nature 4(8):909–914 [DOI] [PubMed] [Google Scholar]
- 185.McDonald DM, Baluk P (2002) Significance of blood vessel leakiness in cancer. Cancer Res 62(18):5381–5385 [PubMed] [Google Scholar]
- 186.Pries AR et al. (2010) The shunt problem: control of functional shunting in normal and tumour vasculature. Nat Rev Cancer 10(8):587–593 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Kamoun WS et al. (2010) Simultaneous measurement of RBC velocity, flux, hematocrit and shear rate in vascular networks. Nat Methods 7(8):655–660 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Baish JW et al. (2011) Scaling rules for diffusive drug delivery in tumor and normal tissues. Proc Natl Acad Sci 108(5):1799–1803 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Chauhan VP et al. (2011) Delivery of molecular and nanoscale medicine to tumors: transport barriers and strategies. Annu Rev Chem Biomol Eng 2(1):281–298 [DOI] [PubMed] [Google Scholar]
- 190.Heldin C-H et al. (2004) High interstitial fluid pressure — an obstacle in cancer therapy. Nat Rev Cancer 4(10):806–813 [DOI] [PubMed] [Google Scholar]
- 191.Jain RK (2013) Normalizing tumor microenvironment to treat cancer: bench to bedside to biomarkers. J Clin Oncol 31(17):2205–2218 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Hurwitz H (2004) Integrating the anti-VEGF-A humanized monoclonal antibody bevacizumab with chemotherapy in advanced colorectal cancer. Clin Colorectal Cancer 4(Suppl 2):S62–S68 [DOI] [PubMed] [Google Scholar]
- 193.Maes H et al. (2014) Tumor vessel normalization by chloroquine independent of autophagy. Cancer Cell 26(2):190–206 [DOI] [PubMed] [Google Scholar]
- 194.Potente M, Gerhardt H, Carmeliet P (2011) Ba- sic and therapeutic aspects of angiogenesis. Cell 146(6):873–887 [DOI] [PubMed] [Google Scholar]
- 195.Welti J et al. (2013) Recent molecular discoveries in angiogenesis and antiangiogenic therapies in cancer. J Clin Investig 123(8):3190–3200 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Bergers G, Hanahan D (2008) Modes of resistance to anti-angiogenic therapy. Nat Rev Cancer 8(8):592–603 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Jain RK (2005) Normalization of tumor vasculature: an emerging concept in antiangiogenic therapy. Science 307(5706):58–62 [DOI] [PubMed] [Google Scholar]
- 198.Nagy JA et al. (2009) Why are tumour blood vessels abnormal and why is it important to know? Br J Cancer 100(6):865–869 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Wilson WR, Hay MP (2011) Targeting hypoxia in cancer therapy. Nat Rev Cancer 11(6):393–410 [DOI] [PubMed] [Google Scholar]
- 200.Hockel M, Vaupel P (2001) Tumor hypoxia: definitions and current clinical, biologic, and molecular aspects. J Natl Cancer Inst 93(4):266–276 [DOI] [PubMed] [Google Scholar]
- 201.Minchinton AI, Tannock IF (2006) Drug penetration in solid tumours. Nat Rev Cancer 6(8):583–592 [DOI] [PubMed] [Google Scholar]