Skip to main content
Erschienen in: Angiogenesis 3/2021

Open Access 17.03.2021 | Brief Communication

Phosphorylation of pericyte FAK-Y861 affects tumour cell apoptosis and tumour blood vessel regression

verfasst von: Delphine M. Lees, Louise E. Reynolds, Ana Rita Pedrosa, Marina Roy-Luzarraga, Kairbaan M. Hodivala-Dilke

Erschienen in: Angiogenesis | Ausgabe 3/2021

Abstract

Focal adhesion kinase (FAK) is a non-receptor tyrosine kinase that is overexpressed in many cancer types and in vivo studies have shown that vascular endothelial cell FAK expression and FAK-phosphorylation at tyrosine (Y) 397, and subsequently FAK-Y861, are important in tumour angiogenesis. Pericytes also play a vital role in regulating tumour blood vessel stabilisation, but the specific involvement of pericyte FAK-Y397 and FAK-Y861 phosphorylation in tumour blood vessels is unknown. Using PdgfrβCre + ;FAKWT/WT, PdgfrβCre + ;FAKY397F/Y397F and PdgfrβCre + ;FAKY861F/Y861F mice, our data demonstrate that Lewis lung carcinoma tumour growth, tumour blood vessel density, blood vessel perfusion and pericyte coverage were affected only in late stage tumours in PdgfrβCre + ;FAKY861F/Y861F but not PdgfrβCre + ;FAKY397F/Y397F mice. Further examination indicates a dual role for pericyte FAK-Y861 phosphorylation in the regulation of tumour vessel regression and also in the control of pericyte derived signals that influence apoptosis in cancer cells. Overall this study identifies the role of pericyte FAK-Y861 in the regulation of tumour vessel regression and tumour growth control and that non-phosphorylatable FAK-Y861F in pericytes reduces tumour growth and blood vessel density.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1007/​s10456-021-09776-8.
DelphineM. Lees and LouiseE. Reynolds are Joint authorship.
The original online version of this article was revised: Figure 1 is updated.
A correction to this article is available online at https://​doi.​org/​10.​1007/​s10456-021-09802-9.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

Angiogenesis is a complex biological process that involves multiple cell types, crosstalk between these cells and responses to different stimuli [13]. Interactions between pericytes and endothelial cells play important roles in angiogenesis. Pericyte recruitment to newly forming blood vessels is thought to promote neovessel stabilisation and is an essential step in neovascularisation [4, 5]. Failure to recruit pericytes to blood vessels can affect vascular remodelling, vessel regression and vessel leakage [1, 2, 6]. This has led to pericyte-targeted therapies being developed and a better understanding of the role of pericytes in tumour angiogenesis [7, 8].
It is well documented that focal adhesion kinase (FAK) plays a central role in different aspects of tumour growth and is overexpressed in many types of cancer [911]. One of the many roles of endothelial cell FAK, in the promotion of tumour growth, is in the initiation of angiogenesis [12]. FAK regulates growth, survival, migration and invasion through its dual role as a kinase and as a scaffolding protein. FAK kinase activity results in tyrosine (Y) 397-phosphorylation, which, in turn, allows proteins containing the Src-homology (SH2) domain to bind to FAK, e.g., Src and PI3K. FAK-Src complexing is thought to potentiate further FAK phosphorylation at other FAK domains including FAK-Y861 [13, 14]. Most studies have pointed towards essential roles for phosphorylation of FAK-Y397 but much less in known about the requirement of FAK-Y861. We and others have shown previously that endothelial cell (EC) FAK is required for tumour growth since EC FAK loss leads to a reduction in tumour growth, accompanied by a reduction in tumour vascular density [12]. Endothelial specific loss of FAK inhibits brain tumour formation and leads to normalisation of the vasculature [15]. Global constitutive deletion of exon 5, which encodes FAK-Y397, also leads to an embryonic lethal phenotype with vascular permeability defects [16]. More recently, we have shown that non-phosphorylatable mutations of tyrosines 397 and 861 (Y397 and Y861) in endothelial cells have differential effects on tumour angiogenesis [17] and that endothelial cell FAK regulates angiocrine signalling in the control of doxorubicin sensitivity in malignant cells [18]. Whilst FAK has been studied extensively in endothelial cells, the function of FAK in pericytes during tumour growth and angiogenesis is starting to emerge, with loss of pericyte FAK enhancing tumour angiogenesis [19]. Given that pericytes are an important cell type in the regulation of tumour growth and paracrine signalling [2022], here we have examined the role of FAK point mutations in pericytes during pathological angiogenesis. Specifically, we have examined the effect of non-phosphorylatable mutations of tyrosines 397 and 861 (Y397F and Y861F) – FAK residues using pericyte-specific FAKY397F/Y397F and FAKY861F/Y861F mice.
In this study, we show that in a subcutaneous Lewis Lung Carcinoma (LLC) tumour model, both angiogenesis and tumour growth are reduced only in mice which have the pericyte-specific FAK-Y861F mutation. This correlates with a significant increase in vessel regression. Furthermore, examination of the secretome and protein expression in FAK-Y861F pericytes highlight an altered pericyte signature involving cytokines and protein secretion that are involved in cancer cell apoptosis. Thus, pericyte FAK-Y861 plays a role in the control of tumour growth.

Results

Generation of pericyte-specific FAK mutant mice

We generated a new mouse model that enables us to study the effect of endogenous deletion of Y397 and Y861 of FAK in pericytes, during pathological angiogenesis. In this knock-out/knockin mouse model we initially generated myc-tagged chicken-WT-FAK, or non-phosphorylatable -Y397F or -Y861F mutant FAK constructs (preceded by a STOP sequence flanked by loxP sites) targeted to the Rosa26 (R26) locus. These mice were bred with PC-specific PdgfrβCre;FAKfl/fl mice to generate mutant FAK-knockin and endogenous FAK-knockout in pericytes under Cre control (Supplementary Fig. 1a, b). These mice show no defects in Mendelian ratios, gender distribution or weights and had no obvious adverse phenotype (Fig. 1a-c). Pericytes isolated from PdgfrβCre + ;FAKWT/WT, PdgfrβCre + ;FAKY861F/Y861F and PdgfrβCre + ;FAKY397F/Y397F mice confirmed the presence of the myc-tag indicating chicken FAK-knockin and normal levels of total FAK (Supplementary Fig. 1c). We also confirmed reduced pY397 in FAK-Y397F pericytes and reduced pY861 in FAK-Y861F pericytes (Fig. 1d). Similarly to work published in endothelial cells [17], Y397F mutation in pericytes correlates with reduced levels of pY861F (Fig. 1e). Phosphorylated Src was reduced in Y397F pericytes, but not in Y861F pericytes (Fig. 1f).

Tumour growth and blood vessel density are reduced in PdgfrβCre + ;FAKY861F/Y861F but not PdgfrβCre + ;FAKY397F/Y397F mice

To examine the effects of pericyte FAK-Y397F and Y861F mutations on tumour growth and angiogenesis, PdgfrβCre + ;FAKWT/WT control mice, PdgfrβCre + ;FAKY861F/Y861F and PdgfrβCre + ;FAKY397F/Y397F mice were injected subcutaneously with Lewis Lung Carcinoma cells (LLC). In vivo tumour growth was reduced in PdgfrβCre + ;FAKY861F/Y861F, but not PdgfrβCre + ;FAKY397F/Y397F mice. Furthermore, the PdgfrβCre + ;FAKY861F/Y861F but not PdgfrβCre + ;FAKY397F/Y397F mice had significantly reduced blood vessel density (as determined by the number of endomucin‐positive vessels per mm2 of age‐matched, size‐matched tumours) compared with PdgfrβCre + ;FAKWT/WT control mice (Fig. 1g, h).
In addition to the above, blood vessel perfusion (determined by the percentage of endomucin-positive blood vessels that were also positive for PE-PECAM antibody after ante-mortem perfusion) and pericyte coverage (determined by the percentage of vessels with NG2-positive mural cell association) were both reduced in PdgfrβCre + ;FAKY861F/Y861F but not PdgfrβCre + ;FAKY397F/Y397F mice (Fig. 2 a, b). Loss of blood vessels can result from partial or persistent regression of vascular endothelial cells. Blood vessel regression is associated with endothelial cell loss and pericyte dropout leaving behind empty collagen IV basement membrane sleeves [23, 24]. Thus, vessel regression can be determined by the presence of endothelial cell-negative, collagen IV-positive basement membrane blood vessel sleeves. Collagen IV / endomucin double-immunostaining of day 14–21 tumours (when the rate of tumour growth in PdgfrβCre + ;FAKY861F/Y861F mice slows down) from PdgfrβCre + ;FAKWT/WT, PdgfrβCre + ;FAKY397F/Y397F and PdgfrβCre + ;FAKY861F/Y861F mice showed a significant increase in endomucin-negative / collagen IV-positive blood vessel sleeves in tumours from PdgfrβCre + ;FAKY861F/Y861F compared with tumours from PdgfrβCre + ;FAKWT/WT, PdgfrβCre + ;FAKY397F/Y397F mice (Fig. 2c). These results correlate with the reduction in tumour growth and suggest that tumour blood vessel regression may be involved in the tumour growth phenotype in PdgfrβCre + ;FAKY861F/Y861F mice.

Increased tumour necrosis in early stage tumours of PdgfrβCre + ;FAKY861F/Y861F mice

Examination of early stage tumours was undertaken to determine at what stage, during growth of the tumour, angiogenesis was being affected. In early stage tumours (day 12 post tumour cell inoculation) LLC tumour size, blood vessel density and blood vessel perfusion were similar between PdgfrβCre + ;FAKWT/WT and PdgfrβCre + ;FAKY861F/Y861F mice (Fig. 3a-c), suggesting that loss of blood vessels occur at a later stage of tumour growth. Indeed to examine whether FAK-Y861F PCs could directly affect the initial stages of microvessel sprouting, aortic rings from PdgfrβCre + ;FAKY861F/Y861F and PdgfrβCre + ;FAKWT/WT mice were embedded in collagen and stimulated, or not, with VEGF (30 ng/ml). VEGF treatment significantly increased angiogenic sprouting in both genotypes to the same extent suggesting that mutation of FAK-Y861F in pericytes is not sufficient to directly affect angiogenic sprouting in a tumour-free environment (Supplementary Fig. 2).
Examination of early stage tumours, using H&E stained sections, revealed that tumours from PdgfrβCre + ;FAKY861F/Y861F mice had a significant increase in tumour necrosis compared with PdgfrβCre + ;FAKWT/WT control mice (Fig. 3d). These results suggest that tumour necrosis precedes the onset of vessel regression and the reduced late stage tumour blood vessel density and tumour growth in PdgfrβCre + ;FAKY861F/Y861F mice.

FAK-Y861F pericyte secretome enhances LLC tumour cell apoptosis

Paracrine or juxtacrine signals from endothelial cells, angiocrine signals, have been implicated in the control of tumour growth where endothelial cell derived signals directly affect tumour cells by altering secreted cytokine profiles [18, 25, 26]. The role of pericytes in this type of signalling is also emerging [27, 28]. Thus, we turned our attention to the possible effects of FAK-Y861F pericytes on tumour cell apoptosis. Using R&D protein profiler arrays, lysates from FAK-Y861F pericytes showed a decrease in levels of thrombospondin, MCP-1, proliferin, TIMP-1 and sICAM/CD54 together with an increase in IGFBP-2, endostatin, TNF-alpha, SDF1 and ADAMTS-1 compared with lysates from control WT pericytes. Similar effects on these cytokines were not observed in FAK-Y397F pericytes compared with WT pericytes and SDF-1/CXCL12 was apparently reduced (Fig. 4a). We focused on the role of MCP-1 in the apoptotic phenotype in tumours from PdgfrβCre + ;FAKY861F/Y861F mice since, in vitro, MCP-1 promotes mural cell recruitment [29] and in vivo pharmacological inhibition of MCP-1 reduces tumour growth and macrophage recruitment resulting in increased tumour necrosis [30], whilst loss of MCP-1 delays mammary tumourigenesis [31]. Therefore, we treated LLC with MCP-1, after exposure to Y861F conditioned medium (CM), and asked if MCP-1 could be involved in controlling tumour cell apoptosis. CM from FAK-WT and FAK-Y861F pericytes were incubated with cultured LLC cells and apoptosis quantified by TUNEL staining. Y861F CM caused a significant increase in LLC apoptosis compared with LLCs incubated with WT CM. Indeed the pro-apoptotic phenotype of Y861F CM was reduced by the addition of exogenous recombinant MCP-1 to the pericyte CM (Fig. 4b), suggesting that the reduction of MCP-1 in Y861F pericytes is at least partially involved in the pro-apoptotic phenotype in tumours grown in PdgfrβCre + ;FAKY861F/Y861F (Y861F) mice. This pro-apoptotic tumour cell phenotype was confirmed, in vivo, where tumours from PdgfrβCre + ;FAKY861F/Y861F mice displayed significantly increased levels of apoptosis compared with tumours from either PdgfrβCre + ;FAKWT/WT or PdgfrβCre + ;FAKY397F/Y397F mice (Fig. 4c).
FAK kinase activity leads to phosphorylation of Y397-FAK which, in turn, allows protein containing the Src-homology (SH2) domain to bind to FAK, including Src. FAK-Src complexing are thought to potentiate further FAK phosphorylation at other FAK domains including Y861. Since phosphorylated Src was not reduced in Y861F pericytes, we wanted to determine if the phenotype observed in Y861F pericytes was dependent on Src. To address this, Src levels were depleted in FAK-WT, FAK-Y397F and FAK-Y861F pericytes followed by examination of the effect of CM from these cells on LLC cell death. CM from Src-depleted FAK-WT and FAK-Y397F pericytes significantly increased LLC apoptosis. Conversely, CM from FAK-Y861 Src-depleted pericytes (Supplementary Fig. 3) had no additional effect on LLC apoptosis above LLC exposed to non-treated CM (Fig. 4d).
These results imply a direct regulation of tumour cells through pericyte paracrine signalling—a direct effect on vascular disruption and ultimately tumour growth that is regulated by FAK-Y861 phosphorylation, and independently of Src.

Discussion

Our data demonstrate that FAK-Y861F pericytes decrease tumour burden, at least in part, by directly affecting tumour cell apoptosis, and call for further consideration of the role of tumour pericytes in the direct control of tumour growth, in addition to their effects on vessel stabilisation. Indeed, we have recently shown that pericytes play a central role in the control of primary tumour growth, through cross-talk with multiple cell types in the tumour microenvironment [19, 20].
FAK is upregulated in many cancer types and is currently being targeted as a potential anti-cancer agent because of its essential roles in tumour growth and angiogenesis [12, 3234]. Our previous work identified distinct roles for FAK-Y397 and FAK-Y861 phosphorylation in tumour endothelial cells where endothelial cell FAK-Y397 but not FAK-Y861 reduced tumour growth and angiogenesis, and linking these tyrosine domains to specific signaling pathways downstream of FAK and regulation of an inside out signal affecting endothelial cells surface receptor expression [17, 35]. Pericytes are thought to be essential in stabilising the tumour vasculature and thus attention has been given to the potential of targeting pericytes to induce vascular disruption in cancer control [36]. However, recent conflicting studies have emerged challenging this idea [37] and suggest possible additional roles for pericytes in regulating tumour growth. In contrast to the effect of FAK-Y397F mutation in endothelial cells [17], our mouse models show that phosphorylation of FAK-Y397 in pericytes is not a prerequisite for the control of LLC tumour growth or angiogenesis. The reason for this apparent discrepancy likely reflect a different requirement for FAK-Y397-phosphorylation in these two cells types in tumour blood vessels. Additionally, complete loss of pericyte FAK is compensated by an increase in pericyte p-Pyk2, which is sufficient to drive a downstream signaling cascade to promote tumour growth, through effects on both malignant cells and angiogenesis [19]. Importantly, one cannot directly compare pericyte FAK-KO with pericyte FAK- Y397F, since phosphorylation of Src is not affected in pericyte FAK-KO mice, whereas in FAK-Y397F mice, the kinase domain is not affected (as it acts upstream of Y397 in the signaling cascade of FAK activation).
FAK-Y861F mutation in pericytes is associated with reduced tumour growth and angiogenesis that is preceded by an increased tendency for tumour cell necrosis and subsequent blood vessel regression. Since tumour necrosis has been demonstrated to have vascular disrupting effects [38, 39] it is tempting to speculate that the early stage enhanced tumour necrosis may initiate vascular disruption. Additionally, given that the Y861F mutation reduces pericyte association with tumour blood vessels, it is likely that vessel stability and maturation is compromised and this reduced pericyte coverage is also likely to be responsible for the decrease in the numbers of functional blood vessels and reduced tumour growth.
Studies have shown that vascular endothelial cells can control tumour growth via angiocrine signals, including cytokines and chemokines [18, 40]. In our study, the protein signature of FAK-Y861F pericytes is a combination of reduced levels of TSP-2, MCP-1, proliferin, TIMP-1 and sICAM/CD54 together with an increase in IGFBP-2, endostatin, TNF-alpha, SDF1 and ADAMTS-1. This molecular signature associates with pro-apoptotic effects on tumour cells but apparently has little effect on VEGF-stimulated angiogenic sprouting in aortic ring assays in a tumour-free context. The lack of an effect on VEGF-induced aortic ring sprouting suggests that the effect of FAK-Y861F pericyte enhancement on tumour cell apoptosis may be a contributing factor to subsequent vessel regression in a tumour environment. The anti-angiogenic effect of TSP-2 is blocked by VEGF thus providing a possible explanation of why VEGF-stimulated angiogenesis is not affected in aortic ring assays from PdgfrβCre + ;FAKY861F/Y861F mice where the decrease in TSP-2 might otherwise have increased angiogenesis [41]. The pro-tumoural effects of MCP-1, proliferin, TIMP-1 and soluble ICAM-1 [4245] whilst the anti-cancer effects of IGFBP-2, endostatin and ADAMTS-1 [4648] have been published and suggest that this secretome signature may well provide a combination of effects on cancer growth. Indeed MCP-1 addition to conditioned medium from FAK-Y861F pericytes was able to rescue the pro-apoptotic effect of this cocktail on tumour cells.
Furthermore, we have shown here that loss of pericyte FAK-Y861 phosphorylation intrinsically regulates a paracrine signal that controls apoptosis of tumour cells, independent of Src and does not affect phosphorylation of Y416-Src. We and others have previously shown that loss of FAK, or a non-phosphorylatable Y397F mutation in endothelial cells, disrupts active Src-FAK interactions, driving Src degradation, possibly activating other FAK-Src independent signalling pathways [17, 4951].
The molecular mechanism by which FAK-Y861 affects this secretome signature is the subject of the future study. Our data support the notion that pericytes are not simply vessel supporting cells, but that via FAK-Y861, can regulate tumour growth via pericyte derived signals directly affecting tumour cell apoptosis.

Methods

Mice

To examine the effect of pericyte-specific FAK mutations in vivo, we developed PdgfrβCre;FAKW/WT mice (FAK-WT), PdgfrβCre;FAKY397F/Y397F (Y397F) mice and PdgfrβCre;FAKY861F/Y861F (Y861F) mice [52]. All experiments were approved by United Kingdom Home Office Regulations. For animals bred in-house, health screens (quarterly) were conducted in accordance with FELASA guidelines for health monitoring of rodent colonies, to confirm their free status of known pathogens in accordance with FELASA screens. No clinical signs were detected. Animals were housed in groups of 4–6 mice per individually ventilated cage in a 12 h light dark cycle (06:30–18:30 light; 18:30–06:30 dark), with controlled room temperature (21 ± 1 °C) and relative humidity (40–60%). The cages contained 1–1.5 cm layer of animal bedding, and with environmental enrichment including cardboard Box-tunnel and crinkled paper nesting material. Animals had access to food and water ad libitum.

Tumour growth

Male and female mice aged 12–16 weeks were injected subcutaneously with 0.5 × 106 Lewis lung carcinoma cells (LLC, ATCC) into the flanks. Tumour dimensions were measured over time and tumour growth was determined using the formula: length x width2 × 0.52. When tumours reached the maximum legal size allowed, mice were killed, tumour sizes measured and tumour samples were either snap-frozen, fixed in 4% paraformaldehyde (PFA) or fixed in 4% PFA/sucrose for histological analysis. For early tumour growth studies, tumours were harvested at day 12 post tumour cell injection.

Blood vessel density

Five µm frozen tumour sections were air-dried for 10 min, washed once in PBS, fixed in acetone for 10 min at -20 °C, washed in PBS three times and then blocked with 5% normal goat serum for 30 min at room temperature. After blocking, sections were incubated with primary antibodies overnight at 4 °C. Primary antibodies used were directed against endomucin (clone V.7C7; Santa Cruz; sc-65495, 1:100). Sections were then washed with PBS and incubated with Alexa-Fluor®-conjugated secondary antibody (1:100, Invitrogen) for 45 min at room temperature before mounting the slides with Prolong® Gold anti-fade reagent (Invitrogen, P36934). Tumour blood vessels were counted across entire midline sections, and the numbers were expressed as vessels/mm2. For examination of blood vessel regression, tumours were fixed with 4% PFA/sucrose then frozen in OCT.

Blood vessel perfusion

For analysis of the % of functional tumour vessels, 100 μl PE-PECAM antibody (clone 390, neat; Biolegend, 102,407) was injected via the tail vein 10 min prior to culling mice. Tumours were dissected immediately, snap-frozen and sectioned. Frozen sections were then immunostained for endomucin, as described above. To calculate the % number of functional vessels, the number of PE-PECAM-positive blood vessels was divided by the total number of endomucin-positive blood vessels.

Pericyte coverage

Frozen tumour sections were double immunostained as described in “Blood vessel density” using the pericyte-specific antibody NG2 (AB5320; Millipore, 1:100) and endomucin. The percentage of endomucin-positive vessels with NG2-positive cells associated was calculated.

Blood vessel regression/Collagen IV staining

Frozen tumour sections were air-dried for 30 min at room temperature, permeabilised for 3 min with PBS + 0.5% TritonX-10 then blocked with 5% BSA/PBS for 1 h at room temperature. Sections were then incubated overnight at 4 °C with Collagen IV (Abcam; 1:100 dilution, ab6586) and endomucin (clone V.7C7, Santa Cruz; 1:100) antibodies. The following day, sections were washed three times with PBS and incubated for 1 h at room temperature with AlexaFluor conjugated secondary antibodies (1:100). Finally, sections were washed twice in PBS and once in distilled water then mounted (Prolong Gold with DAPI) with a glass slide and images were acquired using a confocal spinning disk microscope and sCMOS confocal camera (Nikon). Image analysis was performed using ImageJ software by making maximum intensity projections of z stacks and the numbers of vessels counted manually.

Tumour necrosis

Early stage LLC tumours were fixed in 4% formaldehyde and bisected. Tumour sections were H&E stained, scanned using a Panoramic scanner and the area of necrosis (as identified by acellular regions of tumour tissue) quantified with ImageJ software.

TUNEL staining

Tumour cell apoptosis was examined in frozen tumour sections using the In Situ Cell Death Detection Kit, TMR red (Roche, 12156792910) following the manufacturer’s instructions. Stained sections were scanned using a Panoramic scanner and the TUNEL-positive area was calculated as the percentage of TUNEL-positive area/ tumour area.

LLC and pericyte conditioned medium experiments

LLC were plated on coverslips at a density 5 × 104 cells in DMEM + 10% FCS. At the same time WT and 861F pericytes were grown to 50–60% confluency and conditioned medium (CM) removed. CM was centrifuged to remove cell debris and added to LLCs after removal of DMEM and two PBS washes. Recombinant mouse MCP-1 (Biotechne, MAB479) was added to the CM at either 10 or 50 ng/ml. Cells were incubated with CM for 24 or 48 h after which cells were stained to detect DNA fragmentation in apoptosis using the BrdU-Red DNA (TUNEL) kit (Abcam, ab66110) following the manufacturer’s instructions. Within 3 h of staining the cells were analysed for BrdU using a Zeiss microscope and Axiovision software. The percentage of TUNEL-positive cells was calculated by counting the total number of cell nuclei and the number of nuclei that were TUNEL positive. For Src siRNA experiments, WT, Y397F and Y861F pericytes were transfected for 48 hr with SMARTpool Src siRNA or non-targeted siRNA (both 10uM final concentration) as a control. After 48 hr cells were lysed for protein extraction to measure Src levels by Western blot analysis. Conditioned medium was also removed and added to LLCs as described above, prior to TUNEL staining.

Primary cell cultures

Primary mouse brain pericytes were isolated from the mice, cultured and characterised as previously described [53, 54]. Briefly, brains were removed from mice, minced, digested for 1 h in 0.1% collagenase, centrifuged in the presence of 22% BSA, and cultured in endothelial cell growth media (pMLEC) with the medium changed every 3 days. On reaching confluency, cultures were harvested with trypsin and passaged. During the first two passages, pericyte cultures were grown in pMLEC, and on the third passage they were grown in pericyte medium (PM; ScienCell Research Laboratories) containing 2% FBS and antibiotics. Tissue culture plates for all experiments were coated with a mixture of collagen (30 µg/ml), gelatin (0.1%) and fibronectin (10 µg/ml).

Aortic ring assay

Aortic rings were isolated from all mouse genotypes as previously described [55].

Angiogenesis and cytokine arrays

Pericyte angiogenesis and cytokine profiles were compared using the angiogenesis array (ARY015, R&D Biosystems) and cytokine array (ARY006, R&D Biosystems). Briefly, cell lysates were prepared as follows: sample buffer was added to the cell culture, the cells were scraped and transferred into a 1.5 ml Eppendorf tube. After sonication, samples were adjusted to the array conditions and mixed with a Detection Antibody Cocktail as indicated by the manufacturer’s instructions. Lysates were incubated overnight at 4 °C on dot-blot membranes. Membranes were washed, incubated with streptavidin-HRP for 30 min at RT, washed again and ECL was applied to the membrane to reveal the dots. Quantification of dot intensity was performed using ImageJ™ software.

Western blot analysis

Primary brain pericytes were grown to 70–80% confluency then lysed in RIPA buffer. 15–30 μg protein was run on 8% polyacrylamide gels then transferred to nitrocellulose membranes. Membranes were probed with primary antibody overnight at 4 °C. Myc-tag (Cell Signaling, clone 9B11, 2276, 1:1000), total FAK (Cell Signaling, 3258, 1:1000), phospho-397 FAK (Invitrogen, 44-624G, 1:1000), phospho-861 FAK (Invitrogen, 44-626G, 1:1000), PDGFRβ (Cell Signaling, clone 28E10, 3169, 1:1000), endomucin (V7.C7, Santa Cruz, 1:1000), Src (Cell Signaling, 2108, 1:1000), pSrc (Cell Signaling, 2101, 1:1000). The anti-HSC70 (Santa Cruz, clone B-6, sc-7298) or GAPDH (Millipore, MAB374) antibody, for loading controls, were used at 1:5000 dilution. Densitometric readings of band intensities were obtained using the ImageJ™ software.

Statistical analysis

Statistical significance was calculated using Prism 8 software and P < 0.05 was considered statistically significant, unless otherwise indicated. For tumour growth statistics, non-parametric two-sided Mann–Whitney U rank sum test was performed to compare tumour volumes each day. One-way ANOVA was performed for the in vitro TUNEL and aortic ring assay, two-way ANOVA for the blood vessel regression and in vivo TUNEL study. Wilcoxon test was performed for tumour necrosis.

Acknowledgements

We would like to thank Julie Holdsworth and Bruce Williams from Barts Cancer Institute (QMUL, UK) for their expert technical assistance and Professor Ralf Adams (Max Planck Institute for Molecular Biomedicine, Münster, Germany) for the kind gift of the PdgfrβCre mice.

Compliance with ethical standards

Conflict of interest

KHD is a scientific advisor for Ellipses. KHD is joint applicant on Patent claims N421127GB and N417173GB. The other authors declare no conflicts of interest.

Ethical approval

All procedures were approved by our local animal ethics committee, Queen Mary University of London, and were executed in accordance with United Kingdom Home Office regulations.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

Die Chirurgie

Print-Titel

Das Abo mit mehr Tiefe

Mit der Zeitschrift Die Chirurgie erhalten Sie zusätzlich Online-Zugriff auf weitere 43 chirurgische Fachzeitschriften, CME-Fortbildungen, Webinare, Vorbereitungskursen zur Facharztprüfung und die digitale Enzyklopädie e.Medpedia.

Bis 30. April 2024 bestellen und im ersten Jahr nur 199 € zahlen!

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Anhänge

Supplementary Information

Literatur
1.
Zurück zum Zitat Armulik A, Genove G, Betsholtz C (2011) Pericytes: developmental, physiological, and pathological perspectives, problems, and promises. Dev Cell 21(2):193–215CrossRefPubMed Armulik A, Genove G, Betsholtz C (2011) Pericytes: developmental, physiological, and pathological perspectives, problems, and promises. Dev Cell 21(2):193–215CrossRefPubMed
3.
Zurück zum Zitat Geevarghese A, Herman IM (2014) Pericyte-endothelial crosstalk: implications and opportunities for advanced cellular therapies. Transl Res 163(4):296–306PubMedPubMedCentralCrossRef Geevarghese A, Herman IM (2014) Pericyte-endothelial crosstalk: implications and opportunities for advanced cellular therapies. Transl Res 163(4):296–306PubMedPubMedCentralCrossRef
4.
Zurück zum Zitat von Tell D, Armulik A, Betsholtz C (2006) Pericytes and vascular stability. Exp Cell Res 312(5):623–629CrossRef von Tell D, Armulik A, Betsholtz C (2006) Pericytes and vascular stability. Exp Cell Res 312(5):623–629CrossRef
5.
Zurück zum Zitat Stratman AN, Davis GE (2012) Endothelial cell-pericyte interactions stimulate basement membrane matrix assembly: influence on vascular tube remodeling, maturation, and stabilization. Microsc Microanal 18(1):68–80PubMedCrossRef Stratman AN, Davis GE (2012) Endothelial cell-pericyte interactions stimulate basement membrane matrix assembly: influence on vascular tube remodeling, maturation, and stabilization. Microsc Microanal 18(1):68–80PubMedCrossRef
6.
Zurück zum Zitat Hellstrom M, Gerhardt H, Kalen M et al (2001) Lack of pericytes leads to endothelial hyperplasia and abnormal vascular morphogenesis. J Cell Biol 153(3):543–553PubMedPubMedCentralCrossRef Hellstrom M, Gerhardt H, Kalen M et al (2001) Lack of pericytes leads to endothelial hyperplasia and abnormal vascular morphogenesis. J Cell Biol 153(3):543–553PubMedPubMedCentralCrossRef
7.
Zurück zum Zitat Chen M, Lei X, Shi C et al (2017) Pericyte-targeting prodrug overcomes tumor resistance to vascular disrupting agents. J Clin Invest 127(10):3689–3701PubMedPubMedCentralCrossRef Chen M, Lei X, Shi C et al (2017) Pericyte-targeting prodrug overcomes tumor resistance to vascular disrupting agents. J Clin Invest 127(10):3689–3701PubMedPubMedCentralCrossRef
9.
Zurück zum Zitat Owens LV, Xu L, Craven RJ et al (1995) Overexpression of the focal adhesion kinase (p125FAK) in invasive human tumors. Cancer Res 55(13):2752–2755PubMed Owens LV, Xu L, Craven RJ et al (1995) Overexpression of the focal adhesion kinase (p125FAK) in invasive human tumors. Cancer Res 55(13):2752–2755PubMed
10.
Zurück zum Zitat Schmitz KJ, Grabellus F, Callies R et al (2005) High expression of focal adhesion kinase (p125FAK) in node-negative breast cancer is related to overexpression of HER-2/neu and activated Akt kinase but does not predict outcome. Breast Cancer Res 7(2):R194-203PubMedPubMedCentralCrossRef Schmitz KJ, Grabellus F, Callies R et al (2005) High expression of focal adhesion kinase (p125FAK) in node-negative breast cancer is related to overexpression of HER-2/neu and activated Akt kinase but does not predict outcome. Breast Cancer Res 7(2):R194-203PubMedPubMedCentralCrossRef
11.
Zurück zum Zitat Ji HF, Pang D, Fu SB et al (2013) Overexpression of focal adhesion kinase correlates with increased lymph node metastasis and poor prognosis in non-small-cell lung cancer. J Cancer Res Clin Oncol 139(3):429–435PubMedCrossRef Ji HF, Pang D, Fu SB et al (2013) Overexpression of focal adhesion kinase correlates with increased lymph node metastasis and poor prognosis in non-small-cell lung cancer. J Cancer Res Clin Oncol 139(3):429–435PubMedCrossRef
13.
14.
Zurück zum Zitat Lechertier T, Hodivala-Dilke K (2012) Focal adhesion kinase and tumour angiogenesis. J Pathol 226(2):404–412PubMedCrossRef Lechertier T, Hodivala-Dilke K (2012) Focal adhesion kinase and tumour angiogenesis. J Pathol 226(2):404–412PubMedCrossRef
15.
Zurück zum Zitat Lee J, Borboa AK, Chun HB et al (2010) Conditional deletion of the focal adhesion kinase FAK alters remodeling of the blood-brain barrier in glioma. Cancer Res 70(24):10131–10140PubMedPubMedCentralCrossRef Lee J, Borboa AK, Chun HB et al (2010) Conditional deletion of the focal adhesion kinase FAK alters remodeling of the blood-brain barrier in glioma. Cancer Res 70(24):10131–10140PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat Corsi JM, Houbron C, Billuart P et al (2009) Autophosphorylation-independent and -dependent functions of focal adhesion kinase during development. J Biol Chem 284(50):34769–34776PubMedPubMedCentralCrossRef Corsi JM, Houbron C, Billuart P et al (2009) Autophosphorylation-independent and -dependent functions of focal adhesion kinase during development. J Biol Chem 284(50):34769–34776PubMedPubMedCentralCrossRef
17.
Zurück zum Zitat Pedrosa AR, Bodrug N, Gomez-Escudero J et al (2019) Tumor angiogenesis is differentially regulated by phosphorylation of endothelial cell focal adhesion kinase tyrosines-397 and -861. Can Res 79(17):4371–4386CrossRef Pedrosa AR, Bodrug N, Gomez-Escudero J et al (2019) Tumor angiogenesis is differentially regulated by phosphorylation of endothelial cell focal adhesion kinase tyrosines-397 and -861. Can Res 79(17):4371–4386CrossRef
18.
Zurück zum Zitat Tavora B, Reynolds LE, Batista S et al (2014) Endothelial-cell FAK targeting sensitizes tumours to DNA-damaging therapy. Nature 514(7520):112–116PubMedPubMedCentralCrossRef Tavora B, Reynolds LE, Batista S et al (2014) Endothelial-cell FAK targeting sensitizes tumours to DNA-damaging therapy. Nature 514(7520):112–116PubMedPubMedCentralCrossRef
19.
Zurück zum Zitat Lechertier T, Reynolds LE, Kim H et al (2020) Pericyte FAK negatively regulates Gas6/Axl signalling to suppress tumour angiogenesis and tumour growth. Nat Commun 11(1):2810PubMedPubMedCentralCrossRef Lechertier T, Reynolds LE, Kim H et al (2020) Pericyte FAK negatively regulates Gas6/Axl signalling to suppress tumour angiogenesis and tumour growth. Nat Commun 11(1):2810PubMedPubMedCentralCrossRef
20.
Zurück zum Zitat Wong PP, Munoz-Felix JM, Hijazi M et al (2020) Cancer burden is controlled by Mural cell-beta3-integrin regulated crosstalk with tumor cells. Cell 181(6):1346–1363PubMedCrossRef Wong PP, Munoz-Felix JM, Hijazi M et al (2020) Cancer burden is controlled by Mural cell-beta3-integrin regulated crosstalk with tumor cells. Cell 181(6):1346–1363PubMedCrossRef
21.
Zurück zum Zitat Viski C, Konig C, Kijewska M et al (2016) Endosialin-expressing pericytes promote metastatic dissemination. Can Res 76(18):5313–5325CrossRef Viski C, Konig C, Kijewska M et al (2016) Endosialin-expressing pericytes promote metastatic dissemination. Can Res 76(18):5313–5325CrossRef
22.
Zurück zum Zitat Murgai M, Ju W, Eason M et al (2017) KLF4-dependent perivascular cell plasticity mediates pre-metastatic niche formation and metastasis. Nat Med 23(10):1176–1190PubMedPubMedCentralCrossRef Murgai M, Ju W, Eason M et al (2017) KLF4-dependent perivascular cell plasticity mediates pre-metastatic niche formation and metastasis. Nat Med 23(10):1176–1190PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Baluk P, Lee CG, Link H et al (2004) Regulated angiogenesis and vascular regression in mice overexpressing vascular endothelial growth factor in airways. Am J Pathol 165(4):1071–1085PubMedPubMedCentralCrossRef Baluk P, Lee CG, Link H et al (2004) Regulated angiogenesis and vascular regression in mice overexpressing vascular endothelial growth factor in airways. Am J Pathol 165(4):1071–1085PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Inai T, Mancuso M, Hashizume H et al (2004) Inhibition of vascular endothelial growth factor (VEGF) signaling in cancer causes loss of endothelial fenestrations, regression of tumor vessels, and appearance of basement membrane ghosts. Am J Pathol 165(1):35–52PubMedPubMedCentralCrossRef Inai T, Mancuso M, Hashizume H et al (2004) Inhibition of vascular endothelial growth factor (VEGF) signaling in cancer causes loss of endothelial fenestrations, regression of tumor vessels, and appearance of basement membrane ghosts. Am J Pathol 165(1):35–52PubMedPubMedCentralCrossRef
26.
Zurück zum Zitat Cao Z, Ding BS, Guo P et al (2014) Angiocrine factors deployed by tumor vascular niche induce B cell lymphoma invasiveness and chemoresistance. Cancer Cell 25(3):350–365PubMedPubMedCentralCrossRef Cao Z, Ding BS, Guo P et al (2014) Angiocrine factors deployed by tumor vascular niche induce B cell lymphoma invasiveness and chemoresistance. Cancer Cell 25(3):350–365PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Armulik A, Genove G, Mae M et al (2010) Pericytes regulate the blood-brain barrier. Nature 468(7323):557–561PubMedCrossRef Armulik A, Genove G, Mae M et al (2010) Pericytes regulate the blood-brain barrier. Nature 468(7323):557–561PubMedCrossRef
28.
Zurück zum Zitat Chen J, Luo Y, Hui H et al (2017) CD146 coordinates brain endothelial cell-pericyte communication for blood-brain barrier development. Proc Natl Acad Sci USA 114(36):E7622–E7631PubMedPubMedCentralCrossRef Chen J, Luo Y, Hui H et al (2017) CD146 coordinates brain endothelial cell-pericyte communication for blood-brain barrier development. Proc Natl Acad Sci USA 114(36):E7622–E7631PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Aplin AC, Fogel E, Nicosia RF (2010) MCP-1 promotes mural cell recruitment during angiogenesis in the aortic ring model. Angiogenesis 13(3):219–226PubMedPubMedCentralCrossRef Aplin AC, Fogel E, Nicosia RF (2010) MCP-1 promotes mural cell recruitment during angiogenesis in the aortic ring model. Angiogenesis 13(3):219–226PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Gazzaniga S, Bravo AI, Guglielmotti A et al (2007) Targeting tumor-associated macrophages and inhibition of MCP-1 reduce angiogenesis and tumor growth in a human melanoma xenograft. J Invest Dermatol 127(8):2031–2041PubMedCrossRef Gazzaniga S, Bravo AI, Guglielmotti A et al (2007) Targeting tumor-associated macrophages and inhibition of MCP-1 reduce angiogenesis and tumor growth in a human melanoma xenograft. J Invest Dermatol 127(8):2031–2041PubMedCrossRef
31.
Zurück zum Zitat Cranford TL, Velazquez KT, Enos RT et al (2017) Loss of monocyte chemoattractant protein-1 expression delays mammary tumorigenesis and reduces localized inflammation in the C3(1)/SV40Tag triple negative breast cancer model. Cancer Biol Ther 18(2):85–93PubMedPubMedCentralCrossRef Cranford TL, Velazquez KT, Enos RT et al (2017) Loss of monocyte chemoattractant protein-1 expression delays mammary tumorigenesis and reduces localized inflammation in the C3(1)/SV40Tag triple negative breast cancer model. Cancer Biol Ther 18(2):85–93PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Roberts WG, Ung E, Whalen P et al (2008) Antitumor activity and pharmacology of a selective focal adhesion kinase inhibitor, PF-562,271. Can Res 68(6):1935–1944CrossRef Roberts WG, Ung E, Whalen P et al (2008) Antitumor activity and pharmacology of a selective focal adhesion kinase inhibitor, PF-562,271. Can Res 68(6):1935–1944CrossRef
33.
Zurück zum Zitat Duxbury MS, Ito H, Zinner MJ et al (2004) Focal adhesion kinase gene silencing promotes anoikis and suppresses metastasis of human pancreatic adenocarcinoma cells. Surgery 135(5):555–562PubMedCrossRef Duxbury MS, Ito H, Zinner MJ et al (2004) Focal adhesion kinase gene silencing promotes anoikis and suppresses metastasis of human pancreatic adenocarcinoma cells. Surgery 135(5):555–562PubMedCrossRef
34.
Zurück zum Zitat Stokes JB, Adair SJ, Slack-Davis JK et al (2011) Inhibition of focal adhesion kinase by PF-562,271 inhibits the growth and metastasis of pancreatic cancer concomitant with altering the tumor microenvironment. Mol Cancer Ther 10(11):2135–2145PubMedPubMedCentralCrossRef Stokes JB, Adair SJ, Slack-Davis JK et al (2011) Inhibition of focal adhesion kinase by PF-562,271 inhibits the growth and metastasis of pancreatic cancer concomitant with altering the tumor microenvironment. Mol Cancer Ther 10(11):2135–2145PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Alexopoulou AN, Lees DM, Bodrug N et al (2017) Focal Adhesion Kinase (FAK) tyrosine 397E mutation restores the vascular leakage defect in endothelium-specific FAK-kinase dead mice. J Pathol 242(3):358–370PubMedPubMedCentralCrossRef Alexopoulou AN, Lees DM, Bodrug N et al (2017) Focal Adhesion Kinase (FAK) tyrosine 397E mutation restores the vascular leakage defect in endothelium-specific FAK-kinase dead mice. J Pathol 242(3):358–370PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Huang FJ, You WK, Bonaldo P et al (2010) Pericyte deficiencies lead to aberrant tumor vascularizaton in the brain of the NG2 null mouse. Dev Biol 344(2):1035–1046PubMedPubMedCentralCrossRef Huang FJ, You WK, Bonaldo P et al (2010) Pericyte deficiencies lead to aberrant tumor vascularizaton in the brain of the NG2 null mouse. Dev Biol 344(2):1035–1046PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Cooke VG, LeBleu VS, Keskin D et al (2012) Pericyte depletion results in hypoxia-associated epithelial-to-mesenchymal transition and metastasis mediated by met signaling pathway. Cancer Cell 21(1):66–81PubMedPubMedCentralCrossRef Cooke VG, LeBleu VS, Keskin D et al (2012) Pericyte depletion results in hypoxia-associated epithelial-to-mesenchymal transition and metastasis mediated by met signaling pathway. Cancer Cell 21(1):66–81PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Yoshida S, Kawai H, Eguchi T et al (2019) Tumor angiogenic inhibition triggered necrosis (TAITN) in oral cancer. Cells 8:7CrossRef Yoshida S, Kawai H, Eguchi T et al (2019) Tumor angiogenic inhibition triggered necrosis (TAITN) in oral cancer. Cells 8:7CrossRef
39.
Zurück zum Zitat Ramanujan S, Koenig GC, Padera TP et al (2000) Local imbalance of proangiogenic and antiangiogenic factors: a potential mechanism of focal necrosis and dormancy in tumors. Can Res 60(5):1442–1448 Ramanujan S, Koenig GC, Padera TP et al (2000) Local imbalance of proangiogenic and antiangiogenic factors: a potential mechanism of focal necrosis and dormancy in tumors. Can Res 60(5):1442–1448
40.
41.
Zurück zum Zitat Armstrong LC, Bjorkblom B, Hankenson KD et al (2002) Thrombospondin 2 inhibits microvascular endothelial cell proliferation by a caspase-independent mechanism. Mol Biol Cell 13(6):1893–1905PubMedPubMedCentralCrossRef Armstrong LC, Bjorkblom B, Hankenson KD et al (2002) Thrombospondin 2 inhibits microvascular endothelial cell proliferation by a caspase-independent mechanism. Mol Biol Cell 13(6):1893–1905PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Li M, Knight DA, As L et al (2013) A role for CCL2 in both tumor progression and immunosurveillance. Oncoimmunology. 2(7):25474CrossRef Li M, Knight DA, As L et al (2013) A role for CCL2 in both tumor progression and immunosurveillance. Oncoimmunology. 2(7):25474CrossRef
43.
Zurück zum Zitat Yang X, Qiao D, Meyer K et al (2012) Angiogenesis induced by signal transducer and activator of transcription 5A (STAT5A) is dependent on autocrine activity of proliferin. J Biol Chem 287(9):6490–6502PubMedCrossRef Yang X, Qiao D, Meyer K et al (2012) Angiogenesis induced by signal transducer and activator of transcription 5A (STAT5A) is dependent on autocrine activity of proliferin. J Biol Chem 287(9):6490–6502PubMedCrossRef
44.
Zurück zum Zitat Carpen T, Sorsa T, Jouhi L et al (2019) High levels of tissue inhibitor of metalloproteinase-1 (TIMP-1) in the serum are associated with poor prognosis in HPV-negative squamous cell oropharyngeal cancer. Cancer Immunol Immunother 68(8):1263–1272PubMedPubMedCentralCrossRef Carpen T, Sorsa T, Jouhi L et al (2019) High levels of tissue inhibitor of metalloproteinase-1 (TIMP-1) in the serum are associated with poor prognosis in HPV-negative squamous cell oropharyngeal cancer. Cancer Immunol Immunother 68(8):1263–1272PubMedPubMedCentralCrossRef
45.
Zurück zum Zitat Takahara M, Nagato T, Komabayashi Y et al (2013) Soluble ICAM-1 secretion and its functional role as an autocrine growth factor in nasal NK/T cell lymphoma cells. Exp Hematol 41(8):711–718PubMedCrossRef Takahara M, Nagato T, Komabayashi Y et al (2013) Soluble ICAM-1 secretion and its functional role as an autocrine growth factor in nasal NK/T cell lymphoma cells. Exp Hematol 41(8):711–718PubMedCrossRef
46.
Zurück zum Zitat Tang Z, Gillatt D, Rowe E et al (2019) IGFBP-2 acts as a tumour suppressor and plays a role in determining chemosensitivity in bladder cancer cells. Oncotarget 10(66):7043–7057PubMedPubMedCentralCrossRef Tang Z, Gillatt D, Rowe E et al (2019) IGFBP-2 acts as a tumour suppressor and plays a role in determining chemosensitivity in bladder cancer cells. Oncotarget 10(66):7043–7057PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Folkman J (2006) Antiangiogenesis in cancer therapy–endostatin and its mechanisms of action. Exp Cell Res 312(5):594–607PubMedCrossRef Folkman J (2006) Antiangiogenesis in cancer therapy–endostatin and its mechanisms of action. Exp Cell Res 312(5):594–607PubMedCrossRef
48.
Zurück zum Zitat Vazquez F, Hastings G, Ortega MA et al (1999) METH-1, a human ortholog of ADAMTS-1, and METH-2 are members of a new family of proteins with angio-inhibitory activity. J Biol Chem 274(33):23349–23357PubMedCrossRef Vazquez F, Hastings G, Ortega MA et al (1999) METH-1, a human ortholog of ADAMTS-1, and METH-2 are members of a new family of proteins with angio-inhibitory activity. J Biol Chem 274(33):23349–23357PubMedCrossRef
49.
Zurück zum Zitat Sandilands E, Serrels B, McEwan DG et al (2011) Autophagic targeting of Src promotes cancer cell survival following reduced FAK signalling. Nat Cell Biol 14(1):51–60PubMedCrossRef Sandilands E, Serrels B, McEwan DG et al (2011) Autophagic targeting of Src promotes cancer cell survival following reduced FAK signalling. Nat Cell Biol 14(1):51–60PubMedCrossRef
50.
Zurück zum Zitat Beausejour M, Noel D, Thibodeau S et al (2012) Integrin/Fak/Src-mediated regulation of cell survival and anoikis in human intestinal epithelial crypt cells: selective engagement and roles of PI3-K isoform complexes. Apoptosis 17(6):566–578PubMedPubMedCentralCrossRef Beausejour M, Noel D, Thibodeau S et al (2012) Integrin/Fak/Src-mediated regulation of cell survival and anoikis in human intestinal epithelial crypt cells: selective engagement and roles of PI3-K isoform complexes. Apoptosis 17(6):566–578PubMedPubMedCentralCrossRef
51.
Zurück zum Zitat Lim ST, Chen XL, Tomar A et al (2010) Knock-in mutation reveals an essential role for focal adhesion kinase activity in blood vessel morphogenesis and cell motility-polarity but not cell proliferation. J Biol Chem 285(28):21526–21536PubMedPubMedCentralCrossRef Lim ST, Chen XL, Tomar A et al (2010) Knock-in mutation reveals an essential role for focal adhesion kinase activity in blood vessel morphogenesis and cell motility-polarity but not cell proliferation. J Biol Chem 285(28):21526–21536PubMedPubMedCentralCrossRef
52.
Zurück zum Zitat Tavora B, Batista S, Alexopoulou AN et al (2014) Generation of point-mutant FAK knockin mice. Genesis 52(11):907–915PubMedCrossRef Tavora B, Batista S, Alexopoulou AN et al (2014) Generation of point-mutant FAK knockin mice. Genesis 52(11):907–915PubMedCrossRef
53.
Zurück zum Zitat Tigges U, Welser-Alves JV, Boroujerdi A et al (2012) A novel and simple method for culturing pericytes from mouse brain. Microvasc Res 84(1):74–80PubMedPubMedCentralCrossRef Tigges U, Welser-Alves JV, Boroujerdi A et al (2012) A novel and simple method for culturing pericytes from mouse brain. Microvasc Res 84(1):74–80PubMedPubMedCentralCrossRef
54.
Zurück zum Zitat Reynolds LE, D’Amico G, Lechertier T et al (2017) Dual role of pericyte alpha6beta1-integrin in tumour blood vessels. J Cell Sci 130(9):1583–1595PubMedPubMedCentral Reynolds LE, D’Amico G, Lechertier T et al (2017) Dual role of pericyte alpha6beta1-integrin in tumour blood vessels. J Cell Sci 130(9):1583–1595PubMedPubMedCentral
55.
Zurück zum Zitat Baker M, Robinson SD, Lechertier T et al (2012) Use of the mouse aortic ring assay to study angiogenesis. Nat Protoc 7(1):89–104CrossRef Baker M, Robinson SD, Lechertier T et al (2012) Use of the mouse aortic ring assay to study angiogenesis. Nat Protoc 7(1):89–104CrossRef
Metadaten
Titel
Phosphorylation of pericyte FAK-Y861 affects tumour cell apoptosis and tumour blood vessel regression
verfasst von
Delphine M. Lees
Louise E. Reynolds
Ana Rita Pedrosa
Marina Roy-Luzarraga
Kairbaan M. Hodivala-Dilke
Publikationsdatum
17.03.2021
Verlag
Springer Netherlands
Erschienen in
Angiogenesis / Ausgabe 3/2021
Print ISSN: 0969-6970
Elektronische ISSN: 1573-7209
DOI
https://doi.org/10.1007/s10456-021-09776-8

Weitere Artikel der Ausgabe 3/2021

Angiogenesis 3/2021 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Echinokokkose medikamentös behandeln oder operieren?

06.05.2024 DCK 2024 Kongressbericht

Die Therapie von Echinokokkosen sollte immer in spezialisierten Zentren erfolgen. Eine symptomlose Echinokokkose kann – egal ob von Hunde- oder Fuchsbandwurm ausgelöst – konservativ erfolgen. Wenn eine Op. nötig ist, kann es sinnvoll sein, vorher Zysten zu leeren und zu desinfizieren. 

Umsetzung der POMGAT-Leitlinie läuft

03.05.2024 DCK 2024 Kongressbericht

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

Proximale Humerusfraktur: Auch 100-Jährige operieren?

01.05.2024 DCK 2024 Kongressbericht

Mit dem demographischen Wandel versorgt auch die Chirurgie immer mehr betagte Menschen. Von Entwicklungen wie Fast-Track können auch ältere Menschen profitieren und bei proximaler Humerusfraktur können selbst manche 100-Jährige noch sicher operiert werden.

Die „Zehn Gebote“ des Endokarditis-Managements

30.04.2024 Endokarditis Leitlinie kompakt

Worauf kommt es beim Management von Personen mit infektiöser Endokarditis an? Eine Kardiologin und ein Kardiologe fassen die zehn wichtigsten Punkte der neuen ESC-Leitlinie zusammen.

Update Innere Medizin

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