scholarly journals Endothelial deletion of SHP2 suppresses tumor angiogenesis and promotes vascular normalization

2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Zhiyong Xu ◽  
Chunyi Guo ◽  
Qiaoli Ye ◽  
Yueli Shi ◽  
Yihui Sun ◽  
...  

AbstractSHP2 mediates the activities of multiple receptor tyrosine kinase signaling and its function in endothelial processes has been explored extensively. However, genetic studies on the role of SHP2 in tumor angiogenesis have not been conducted. Here, we show that SHP2 is activated in tumor endothelia. Shp2 deletion and pharmacological inhibition reduce tumor growth and microvascular density in multiple mouse tumor models. Shp2 deletion also leads to tumor vascular normalization, indicated by increased pericyte coverage and vessel perfusion. SHP2 inefficiency impairs endothelial cell proliferation, migration, and tubulogenesis through downregulating the expression of proangiogenic SRY-Box transcription factor 7 (SOX7), whose re-expression restores endothelial function in SHP2-knockdown cells and tumor growth, angiogenesis, and vascular abnormalization in Shp2-deleted mice. SHP2 stabilizes apoptosis signal-regulating kinase 1 (ASK1), which regulates SOX7 expression mediated by c-Jun. Our studies suggest SHP2 in tumor associated endothelial cells is a promising anti-angiogenic target for cancer therapy.

2007 ◽  
Vol 293 (4) ◽  
pp. C1404-C1411 ◽  
Author(s):  
Yong Deng ◽  
Jianhua Yang ◽  
Marya McCarty ◽  
Bing Su

Mitogen-activated protein kinase kinase kinase 3 (MEKK3) plays an essential role in embryonic angiogenesis, but its role in tumor growth and angiogenesis is unknown. In this study, we further investigated the role of MEKK3 in embryonic angiogenesis, tumor angiogenesis, and angiogenic factor production. We found that endothelial cells from Mekk3-deficient embryos showed defects in cell proliferation, apoptosis, and interactions with myocardium in the heart. We also found that MEKK3 is required for angiopoietin-1 (Ang1)-induced p38 and ERK5 activation. To study the role of MEKK3 in tumor growth and angiogenesis, we established both wild-type and Mekk3-deficient tumor-like embryonic stem cell lines and transplanted them subcutaneously into nude mice to assess their ability to grow and induce tumor angiogenesis. Mekk3-deficient tumors developed and grew similarly as control Mekk3 wild-type tumors and were also capable of inducing tumor angiogenesis. In addition, we found no differences in the production of VEGF in Mekk3-deficient tumors or embryos. Taken together, our results suggest that MEKK3 plays a critical role in Ang1/Tie2 signaling to control endothelial cell proliferation and survival and is required for endothelial cells to interact with the myocardium during early embryonic development. However, MEKK3 is not essential for tumor growth and angiogenesis.


2010 ◽  
Vol 48 (08) ◽  
Author(s):  
N Azoitei ◽  
GV Pusapati ◽  
A Kleger ◽  
C Brunner ◽  
F Genze ◽  
...  

2007 ◽  
Vol 18 (11) ◽  
pp. 4210-4221 ◽  
Author(s):  
Karine Raymond ◽  
Maaike Kreft ◽  
Ji-Ying Song ◽  
Hans Janssen ◽  
Arnoud Sonnenberg

An increased expression of the integrin α6β4 is correlated with a poor prognosis in patients with squamous cell carcinomas. However, little is known about the role of α6β4 in the early stages of tumor development. We have isolated cells from mouse skin (mouse tumor-initiating cells [mTICs]) that are deficient in both p53 and Smad4 and carry conditional alleles of the β4 gene (Itgb4). The mTICs display many features of multipotent epidermal stem cells and produce well-differentiated tumors after subcutaneous injection into nude mice. Deletion of Itgb4 led to enhanced tumor growth, indicating that α6β4 mediates a tumor-suppressive effect. Reconstitution experiments with β4-chimeras showed that this effect is not dependent on ligation of α6β4 to laminin-5, but on the recruitment by this integrin of the cytoskeletal linker protein plectin to the plasma membrane. Depletion of plectin, like that of β4, led to increased tumor growth. In contrast, when mTICs had been further transformed with oncogenic Ras, α6β4 stimulated tumor growth, as previously observed in human squamous neoplasms. Expression of different effector-loop mutants of RasV12 suggests that this effect depends on a strong activation of the Erk pathway. Together, these data show that depending on the mutations involved, α6β4 can either mediate an adhesion-independent tumor-suppressive effect or act as a tumor promotor.


2009 ◽  
Vol 47 (09) ◽  
Author(s):  
N Azoitei ◽  
GV Pusapati ◽  
F Genze ◽  
G Adler ◽  
T Seufferlein

Blood ◽  
2010 ◽  
Vol 115 (22) ◽  
pp. 4605-4613 ◽  
Author(s):  
Alexander Zaslavsky ◽  
Kwan-Hyuck Baek ◽  
Ryan C. Lynch ◽  
Sarah Short ◽  
Jenny Grillo ◽  
...  

Abstract The sequential events leading to tumor progression include a switch to the angiogenic phenotype, dependent on a shift in the balance between positive and negative angiogenic regulators produced by tumor and stromal cells. Although the biologic properties of many angiogenesis regulatory proteins have been studied in detail, the mechanisms of their transport and delivery in vivo during pathologic angiogenesis are not well understood. Here, we demonstrate that expression of one of the most potent angiogenesis inhibitors, thrombospondin-1, is up-regulated in the platelets of tumor-bearing mice. We establish that this up-regulation is a consequence of both increased levels of thrombospondin-1 mRNA in megakaryocytes, as well as increased numbers of megakaryocytes in the bone marrow of tumor-bearing mice. Through the use of mouse tumor models and bone marrow transplantations, we show that platelet-derived thrombospondin-1 is a critical negative regulator during the early stages of tumor angiogenesis. Collectively, our data suggest that the production and delivery of the endogenous angiogenesis inhibitor thrombospondin-1 by platelets may be a critical host response to suppress tumor growth through inhibiting tumor angiogenesis. Further, this work implicates the use of thrombospondin-1 levels in platelets as an indicator of tumor growth and regression.


Cancers ◽  
2018 ◽  
Vol 10 (8) ◽  
pp. 268 ◽  
Author(s):  
Luciano Pirola ◽  
Oskar Ciesielski ◽  
Aneta Balcerczyk

Approximately 50 years ago, Judah Folkman raised the concept of inhibiting tumor angiogenesis for treating solid tumors. The development of anti-angiogenic drugs would decrease or even arrest tumor growth by restricting the delivery of oxygen and nutrient supplies, while at the same time display minimal toxic side effects to healthy tissues. Bevacizumab (Avastin)—a humanized monoclonal anti VEGF-A antibody—is now used as anti-angiogenic drug in several forms of cancers, yet with variable results. Recent years brought significant progresses in our understanding of the role of chromatin remodeling and epigenetic mechanisms in the regulation of angiogenesis and tumorigenesis. Many inhibitors of DNA methylation as well as of histone methylation, have been successfully tested in preclinical studies and some are currently undergoing evaluation in phase I, II or III clinical trials, either as cytostatic molecules—reducing the proliferation of cancerous cells—or as tumor angiogenesis inhibitors. In this review, we will focus on the methylation status of the vascular epigenome, based on the genomic DNA methylation patterns with DNA methylation being mainly transcriptionally repressive, and lysine/arginine histone post-translational modifications which either promote or repress the chromatin transcriptional state. Finally, we discuss the potential use of “epidrugs” in efficient control of tumor growth and tumor angiogenesis.


2010 ◽  
Vol 2010 ◽  
pp. 1-6 ◽  
Author(s):  
Peter Oettgen

Angiogenesis is a critical component of tumor growth. A number of growth factors, including VEGF, FGF, and HGF, have been implicated as angiogenic growth factors that promote tumor angiogenesis in different types of cancer. Ets-1 is the prototypic member of the Ets transcription factor family. Ets-1 is known to be a downstream mediator of angiogenic growth factors. Expression of Ets-1 in a variety of different tumors is associated with increased angiogenesis. A role for other selected members of the Ets transcription factor family has also been shown to be important for the development of tumor angiogenesis. Because Ets factors also express a number of other important genes involved in cell growth, they contribute not only to tumor growth, but to disease progression. Targeting Ets factors in mouse tumor models through the use of dominant-negative Ets proteins or membrane permeable peptides directed at competitively inhibiting the DNA binding domain has now demonstrated the therapeutic potential of inhibiting selected Ets transcription factors to limit tumor growth and disease progression.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 416-416
Author(s):  
Murat O. Arcasoy ◽  
Matthew E. Hardee ◽  
Xiaohong Jiang ◽  
Mark W. Dewhirst

Abstract Erythropoietin is a pleiotropic cytokine with diverse non-hematopoietic functions. Epo and its receptor EpoR have been implicated in the physiologic angiogenesis that occurs in the mouse embryo, female genital tract and during wound healing. Epo is involved in the pathologic angiogenesis of diabetic retinopathy but the role of Epo in tumor angiogenesis, a pathologic process that is essential for tumor progression, is not established. We investigated the hypothesis that Epo may play an important role in tumor cell-induced angiogenesis and progression. Fluorescently-labeled rodent mammary carcinoma cells were implanted in dorsal skin-fold window chambers in nu/nu mice, a model that allows direct, non-invasive, serial visualization and real-time assessment of tumor cells and neovascularization simultaneously during the initial stages of tumor formation. Tumor angiogenesis (vascular length density, VLD) and growth (tumor area) were measured serially over 8 days using intravital microscopy and computerized image analysis. Co-injection of Epo with implanted tumor cells significantly increased VLD by 78% compared to vehicle-injected controls (p<0.001 by repeated measures ANOVA and Bonferroni test, n=8 mice/group). This early proangiogenic effect was associated with significant stimulation of tumor growth by 66% (p<0.001). Implantation of R3230-GFP cells engineered to stably express a constitutively active EpoR mutant (R129C) resulted in significant stimulation of neovascularization by 76% (p<0.001, n=7) and increased tumor growth by 79% compared to empty vector-transfected control cells (p<0.001). To target endogenous Epo function in window chambers, recombinant soluble EpoR (sEpoR) or a neutralizing anti-Epo monoclonal antibody (mAb) were co-injected at the time of tumor cell implantation. By day 8, marked inhibition of neovascularization was observed in response to treatment with sEpoR (44% reduction) or mAb (47% reduction) compared to vehicle-injected controls (p<0.001, n=7). This anti-angiogenic effect was associated with significant decrease in tumor size by 37% (sEpoR) and 39% (mAb), respectively (p<0.001). Stable expression of a secreted Epo antagonist (Epo-R103A) protein in tumor cells was associated with a remarkable anti-angiogenic effect with 50% reduction in VLD (p<0.001, n=7) and near complete disappearance of tumor cells by day 8 compared to controls (p<0.001). Analysis of two independent single cell clones of each transfected cell line showed similar results. All transfected cell lines exhibited similar in vitro growth characteristics and cell cycle profile. To further assess in vivo tumor growth, cells were implanted orthotopically in the mammary fat pad of female nu/nu mice (10 animals/group). EpoR-R129C expression was associated with significantly increased tumor volume (531±41 mm3, n=10) compared to vector-transfected cells (232±46 mm3, n=7). EpoR-R129C tumors exhibited significantly increased numbers of positive cells for proliferation marker Ki67 and blood vessel marker CD31 per high power field (p=0.0006 and 0.0008, respectively). Remarkably, tumor growth was completely absent after mammary fat pad implantation of R3230-GFP cells secreting the antagonist R103A-Epo protein (n=10 mice) compared to controls (183±46 mm3, n=8). Taken together, these data indicate that 1)-Epo is an important angiogenic factor that modulates tumor cell-induced angiogenesis and 2)-Suppression of tumor angiogenesis and progression by Epo blockade suggests that Epo may constitute a potential target for the therapeutic modulation of angiogenesis in cancer.


Sign in / Sign up

Export Citation Format

Share Document