Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2018 Jan 1.
Published in final edited form as: Pharmacol Res. 2016 Nov 23;115:107–123. doi: 10.1016/j.phrs.2016.11.022

Modulation of VEGF Receptor 2 signaling by Protein Phosphatases

Federico Corti 1, Michael Simons 1
PMCID: PMC5205541  NIHMSID: NIHMS832484  PMID: 27888154

Abstract

Phosphorylation of serines, threonines, and tyrosines is a central event in signal transduction cascades in eukaryotic cells. The phosphorylation state of any particular protein reflects a balance of activity between kinases and phosphatases. Kinase biology has been exhaustively studied and is reasonably well understood, however, much less is known about phosphatases. A large body of evidence now shows that protein phosphatases do not behave as indiscriminate signal terminators, but can function both as negative or positive regulators of specific signaling pathways. Genetic models have also shown that different protein phosphatases play precise biological roles in health and disease. Finally, genome sequencing has unveiled the existence of many protein phosphatases and associated regulatory subunits comparable in number to kinases. A wide variety of roles for protein phosphatase roles have been recently described in the context of cancer, diabetes, hereditary disorders and other diseases.

In particular, there have been several recent advances in our understanding of phosphatases involved in regulation of vascular endothelial growth factor receptor 2 (VEGFR2) signaling. The receptor is the principal signaling molecule mediating a wide spectrum of VEGF signal and, thus, is of paramount significance in a wide variety of diseases ranging from cancer to cardiovascular to ophthalmic. This review focuses on the current knowledge about protein phosphatases' regulation of VEGFR2 signaling and how these enzymes can modulate affect its biological effects.

Keywords: Protein phosphatases, VEGFR2 signaling, endothelial cells, blood vessels, angiogenesis

Graphical abstract

graphic file with name nihms832484u1.jpg

1. Introduction

The human genome encodes 518 protein kinases and approximately 150 protein phosphatases [1, 2]. Members of both groups are historically classified according to their amino acid preference, serine (Ser)/threonine (Thr) or tyrosine (Tyr). A comparison between these categories shows that Protein Ser/Thr Kinases (PSKs) are roughly 10 times more numerous than their counterparts Protein Ser/Thr Phosphatases (PSPs) (∼ 428 vs 40) [1, 3]. However, there is a comparable number of Protein Tyr Kinases (PTKs) and Protein Tyr Phosphatases (PTPs) (∼ 90 vs 107) [1, 4]. Recent classifications have indicated a slightly different number of PTPs [5, 6]. A classification that considers only enzymatically active phosphatases (and includes nonprotein phosphatases) limits PTPs number to 96 [5]. However, a broader PTP definition, which includes criteria of function, structure and sequence similarities, has increased the total number of PTPs to 125 (namely the extended human PTPome)[6].

Classical PTPs only recognize p-Tyr residues and are divided into receptor PTPs (PTPR), which are localized at the cell membrane, and non-receptor intracellular PTPs (PTPN). A distinct PTP subgroup (∼ 65 members) recognizes both p-Tyr and p-Ser/p-Thr residues and are referred to as Dual Specificity Phosphatases (DUSPs) [7]. The PTP catalytic domain contains the conserved sequence HC-X5-R whose Cys-residue is necessary for the phosphatase activity and is susceptible to inactivation by reactive oxygen species (ROS)-mediated oxidation. PTPRs contain an extracellular domain that may bind soluble ligands or interact with extracellular matrix components, while the catalytic domain is located in the intracellular part of the protein. Conversely, PTPNs are found in the cytosol or bound to intracellular membrane compartments and presents a variety of regulatory domains that are critical for correct spatial and temporal localization.

PSPs mainly exist as holoenzymes consisting of a catalytic subunit and one or more regulatory subunits. PSP catalytic mechanisms are less conserved compared to PTPs and are usually classified into three families: phosphoprotein phosphatases (PPPs), metal-dependent protein phosphatases (PPMs), and aspartate-based phosphatases. Although there is a limited number of PSP catalytic subunits (∼30) [3], a large number of regulatory subunits that can associate with the same catalytic subunit may provide substrate and localization specificity. The reader is referred to recent reviews on this subject for a more exhaustive description of phosphatases classification, mechanism of action and molecular structure, [2, 3, 7].

VEGFR2 is a Receptor Tyrosine Kinase (RTK) predominantly expressed in endothelial cells (ECs) and their embryonic precursors [8-10], although its expression can be detected in neuronal cells and hematopoietic stem cells [11]. Deletion of VEGFR2 [12] or its main ligand VEGFA [13, 14] leads to early embryonic lethality in mice (E8.5/9.5 and E9.5/10.5, respectively) due to severe impairment of vascular development and hematopoietic cell maturation. Furthermore, deletion of a single VEGFA allele is sufficient to induce embryonic death at E11-12 [13, 14]. VEGFA was initially named as the Vascular Permeability Factor (VPF) after its ability to induce Evans Blue extravascular leakage in guinea pig back skin [15], however, it is now evident that VEGFA plays a larger role in both embryonic development and adult life [16, 17]. The activation of VEGFR2 by VEGFA can initiate multiple signaling pathways that orchestrate a variety of complex biological effects, such as endothelial cells maturation, vessel lumen formation, vascular permeability, vasodilation, angiogenesis and arteriogenesis [18-21]. Furthermore, knock-in mice which express only selected VEGFA splicing isoforms show marked phenotype differences; this suggests some degree of function segregation between the isoforms [16, 19]

Mounting evidence during the last decade demonstrates that phosphatases do not merely “switch-off”, but can finely modulate molecular and cellular responses by regulating phosphorylation events in negative or positive nodes of a signaling pathway [22]. A number of recent reviews have addressed the role of protein phosphatases in animal disease models and human diseases such as cancer [23, 24], diabetes [25], antiplatelet therapy [26], and hereditary disorders [27]. This review will focus on phosphatases known to affect VEGFR2 signaling (and its biological effects) by means of targeting phosphorylation of the receptor itself (Fig.1) or that of its downstream signal transducers (Fig. 2). VEGFR2 signaling plays an important role in diseases such as cancer [16], cardiovascular disease [28], retinopathies (age-related macular degeneration, AMD; diabetic macular edema, DME; retinopathy of prematurity, ROP; and others) [29], cerebrovascular disorders [30] and stroke [31], therefore phosphatases that modulate its signaling (Tables 1-2) may represent novel pharmacological targets to improve therapy of these diseases.

Fig. 1. Summary of PTPs that dephosphorylate VEGFR2.

Fig. 1

Multiples PTPs can dephosphorylate VEGFR2 and modulate its biological effects. Protein phosphatases often have domains that dictate their interaction partners or specific localization to subcellular compartments. DEP-1 and VE-PTP contain fibronectin type-III repeats implicated in cell adhesion. PTP1B and TC-PTP contain a C-terminal ER-anchoring motif. Shp1 and Shp2 have two SH2 domains that can bind phosphotyrosine residues and control activation of its PTP domain. PTP-MEG2 contains an N-terminal Sec14p-homology domain that targets this PTP to secretory vesicles.

Fig. 2. Summary of phosphatases that modulate VEGFR2 downstream effectors.

Fig. 2

Multiple protein phosphatases can regulate VEGFR2 signaling and its biological effects by modulating phosphorylation of its downstream effectors. The figure shows VEGFR2 signaling nodes where dephosphorylation (by different phosphatases) results in a negative (left panel) or a positive (right panel) effect. Negative phosphatases oppose a VEGFR2-induced downstream effect, while positive phosphatase enhance/promote it. Detailed description of each phosphatase role can be found in the text.

Table 1. Role of protein phosphatases in VEGFR2 signaling.

Protein Name Gene name Type Expression (vascular system) Effect on VEGFR2 signaling Ref.
Cell Membrane Phosphatases

VE-PTP PTPRB PTPR Endothelium only Negative: dephosphorylates VEGFR2 [38, 40]

DEP-1 PTPRJ PTPR Endothelium, SMC, platelets, white blood cell lineages Negative: dephosphorylates VEGFR2 [51, 60]
Positive: dephosphorylates Src inhibitory site (Y527) [51, 62]

RPTPζ PTPRZ1 PTPR Endothelial cells (and likely other cell types) Positive: dephosphorylates Src inhibitory site (Y527) [199]

Intracellular Phosphatases

PTP1B PTPN1 PTPN All tissues Negative: dephosphorylates internalized VEGFR2 and inhibits PLCγ/ERK pathway [81, 83, 89]

Shp1 PTPN6 PTPN All tissues (high in hematopoietic lineages) Negative: dephosphorylates VEGFR2 [99]

Shp2 PTPN11 PTPN All tissues Negative: dephosphorylates VEGFR2 [113]
Positive: inhibits Csk activation which in turn leads to reduced phosphorylation of Src inhibitory site (Y527) [120]

PTEN PTEN DUSP/ lipid phosphatase All tissues Negative: dephosphorylates PIP3 thus antagonizing VEGFA-induced PI3K/AKT pathway [134]

LMW-PTP ACP1 PTPN All tissues Negative: dephosphorylates VEGFR2 [143]

PTP-MEG2 PTPN9 PTPN White blood cell lineages, platelets, endothelium Negative: dephosphorylates VEGFR2 [149]

TC-PTP PTPN2 PTPN All tissues (high in hematopoietic lineages) Negative: dephosphorylates VEGFR2 [151]

Calcineurin PPP3 Ser/Thr All tissues Positive: dephosphorylates NFATc which can translocate to nucleus and promotes VEGFR2-downstream gene expression [161, 173]

PP2A PPP2 Ser/Thr All tissues Negative: dephosphorylates RAF, eNOS and AKT [182, 184]

PRL-3 PTP4A3 DUSP Endothelial cells (high in development and tumor EC) Positive: Increases VEGFR2 phosphorylation and Src activation (Y416) [192]

MKP-1 DUSP1 Ser/Thr All tissues Positive: enhances VEGFA-induced CX3CL1 upregulation (a proangiogenic gene) [194]

Table 2. Summary of phenotypes in phosphatase mutant mice.

Name Mouse phenotype of phosphatase mutants Ref.
VE-PTP - Global deletion: death at E9.5-10 due to failure of vascular plexus remodeling [33, 35]
- Postnatal EC deletion: increased Tie2 phosphorylation and higher stability of EC junctions, decreased vascular permeability to various stimuli (e.g. thrombin, VEGFA, histamine) [42]
- Mutant mice in which VE-PTP forms a stable complex with VE-Cadherin: impaired VEGFA-induced vascular permeability [41]

DEP-1 - Global deletion: viable and fertile [67, 68]
- Global deletion: decreased cerebrovascular flow recovery after carotid occlusion; normal arteriogenesis at day 7 of hind limb ischemia (HLI) [71]
- Global deletion: decreased VEGFA-induced permeability, normal basal permeability and response to histamine; decreased arteriogenesis at day 14 of HLI [65]
- Knockin mice expressing DEP-1 with eGFP in place of the intracellular domain: death at E10.5/11.5 with sever angiogenesis defects [69]

PTP1B - Global deletion: viable and fertile with enhanced insulin sensitivity and no obesity [84, 85]
- EC-specific deletion: increased arteriogenesis [83]

Shp1 - Global deletion: death within 3 weeks of birth due to chronic systemic inflammation [103, 104]
- 60% reduced expression (mice with CCN1 postnatal EC-deletion): increased VEGFR2 activation, EC hyperproliferation and delayed vessel outgrowth in retina [106]

Shp2 - Global deletion: early embryonic lethality [123]
- Expression a constitutive active mutant in EC leads to cardiac defects similar to Noonan Syndrome (myocardial thinning, septal and valves defects) [124]

PTEN - Global deletion: death at E9.5 [135, 136]
- Single allele deletion in EC: viable mice with increased pathological angiogenesis [138]
- Homozygous deletion in EC: severe angiogenesis defects and death at E11.5 [138]
- Postnatal deletion in EC: excessive vasculature branching and larger diameter vessels in the retina [137]

LMW-PTP - Global deletion: mice are viable and protected from heart failure [144]

PTP-MEG2 - Global deletion: death during late embryonic stages due to incomplete neuronal tube development and extensive hemorrhages [147]

TC-PTP - Global deletion: death within 5 weeks of birth due to anemia, severe immunosuppression and systemic inflammation [154, 155]

Calcineurin - Knockin mice expressing an inactive CN die at E10.5/11.5 due to disorganized remodeling of the vascular plexus [176]
- EC specific deletion (Tie2 promoter) leads to defective coronary EC patterning and death at ∼E13.5 [163]

PRL-3 - Global deletion: viable but show impaired VEGFA-induced skin permeability [192]

MKP-1 - Global deletion: viable but display reduced adult arteriogenesis [194]

RPTPζ - Global deletion: viable and fertile with increased number of hematopoietic stem cells [247, 248]

2. Cell membrane phosphatases

Binding of VEGFA to VEGFR2 induces receptor dimerization and autophosphorylation at multiple tyrosine sites including Y1054/1059, Y1175, Y951, and Y1214 and others less characterized [32]. Each site is thought to promote unique downstream signaling pathways, which are linked to different cellular responses such as proliferation, migration, survival and permeability [20, 21]. Both receptor and non-receptor protein phosphatases can regulate VEGFR2 signaling and will be discussed in turn.

2.1 VE-PTP (Vascular Endothelial PTP)

VE-PTP (also called hPTP-β, R-PTPβ) is an endothelial-specific receptor PTP [33, 34]. It is found in the endothelium of all blood vessels and in the endocardium (with a high level of expression in arteries and cardiac valves) [33, 35]. VE-PTP substrates include angiopoietin-1 receptor TIE2 [34], VE-Cadherin [36], γ-catenin (Plakoglobin) [37], and VEGFR2 [38].

An association between VE-PTP and VEGR2 has been shown by a proximity ligation assay [38]. However, binding of a VE-PTP substrate-trapping mutant (a phosphatase mutant that allows stable binding with its substrate without dephosphorylation [39]) to VEGFR2 requires formation of a trimeric complex with Tie2 [40]. The VE-PTP catalytic domain dephosphorylates immunoprecipitated VEGFR2 [40] while silencing of VE-PTP in ECs increases VEGFA-induced VEGFR2 phosphorylation at multiple sites (Y951, Y1175) in immortalized microvascular EC [38]. Baseline VEGFR2 phosphorylation is not affected by VE-PTP silencing [38, 40]. VE-PTP silencing also enhances VEGFA-induced EC proliferation in vitro [38]. In addition, embryonic bodies that lack VE-PTP form excessive vessel sprouts and filopodia in response to VEGFA due to persistent VEGFR2 Y1175 phosphorylation in stalk cells and increased ECs proliferation [40].

VE-PTP activity is important in regulation of VEGFA-driven lumen formation and endothelial cell polarity. Vessel sprouts from VE-PTP null embryonic bodies show disorganized podocalyxin (a marker for vessel luminal side) distribution and impaired lumen formation [40]. This defect is rescued by lowering VEGFA concentration, suggesting that this phenotype is due to VEGFR2 hyper-activation in the absence of VE-PTP. Similarly, VE-PTP knockdown in zebrafish markedly increases the frequency of lumen-less intersomitic vessels that lack blood flow [40].

Generation of VE-PTP null mice has been achieved with two different strategies. In one case truncation of a VE-PTP sequence led to production of a secreted protein, thereby eliminating its plasma membrane expression [33]. A second strategy involved replacing the VE-PTP gene with a LacZ reporter gene [35]. In both cases VE-PTP null mice died by E9.5-E10 due to severe cardiovascular defects. These include failure of the primary vascular plexus to remodel into hierarchical branched structures, lack of functional blood vessels in the yolk sac, and multiple heart defects (e.g. lack of trabeculation and pericardial edema) [33, 35]. In contrast, VE-PTP heterozygous mice appear normal and phenotypically identical to wild type mice.

In the endothelium of adult vasculature, VE-PTP localizes in a stable junctional complex with VE-Cadherin, which contributes to maintain VE-Cadherin in an unphosphorylated state, thus enhancing its barrier function [36, 37]. This complex is disrupted following VEGFR2 activation [37]. The importance of this event is demonstrated by the loss of VEGFA-induced permeability in a knock-in mouse in which VE-PTP and VE-Cadherin are unable to dissociate [41]. Nevertheless, postnatal deletion of VE-PTP results in viable mice with no evident defects in basal permeability and with a generalized reduction in permeability response to multiple factors, including VEGFA and histamine [42]. This effect is linked to a marked increase in Tie2 phosphorylation (a VE-PTP substrate) and formation of highly stabilized endothelial junctions. Notably, Tie2-mediated enhancement of barrier integrity overrides the simultaneous loss of VE-CAD stabilization due to VE-PTP deletion. Inhibition of VE-PTP by a small molecule (AKB-9778) or a VE-PTP blocking antibody inhibits retinal and choroidal neovascularization. This may represent a novel therapeutic tool for multiple ocular diseases [43]. The mechanism of this effect has also been linked to enhanced activation of Angiopoietin-1/Tie2 signaling and improved vascular integrity. A similar mechanism has been proposed for a beneficial effect of VE-PTP inhibition on tumor vascular normalization [44].

2.2 DEP-1 (Density Enhanced Phosphatase 1)

In the vascular system, DEP-1 (also called CD148, PTPη, SCC1) is expressed in endothelial cells, smooth muscle cells, leukocytes, and platelets [26, 45-48]. DEP-1 substrates include PDGFRβ [49, 50], VEGFR2 [51], insulin receptor [52-55], p120 [56], β-catenin [57] and Src [58]. As suggested by its name, DEP-1 expression is markedly upregulated when cells reach confluency [59].

DEP-1 substrate-trapping mutant binds to phosphorylated VEGFR2 [51]. Silencing DEP-1 in HUVEC or mouse ECs enhances VEGFA-induced VEGFR2 phosphorylation at all major phosphorylation sites [51, 60]. DEP-1 is involved in contact inhibition of cell growth as shown by high VEGFA-induced BrdU incorporation in confluent endothelial cells that lack DEP-1 [60]. Finally, DEP-1 peptide activators inhibit VEGFA-induced VEGFR2 phosphorylation and proliferation in HUVEC [61]. Despite its dephosphorylation of VEGFR2, DEP-1 favors VEGFA-induced Src activation (Y416 phosphorylation) [51, 62] due to its ability to dephosphorylate Src inhibitory site (Y527). Consistent with this role, silencing of DEP-1 in HUVEC inhibits VEGFA-induced in vitro permeability [62], a process that is dependent on Src activation [63, 64]. This effect is also seen in vivo as evidenced global DEP-1 null mice display impaired VEGFA-induced skin permeability. The defect is specific to VEGFA because both histamine-induced and basal tissue permeability are normal in DEP-1 null mice [65]

Multiple genetic strategies have been used to asses DEP-1 functions in vivo. DEP-1 global null mice are viable, fertile and do not display any gross phenotype [66]. Similarly, mice expressing a modified DEP-1 that lacks a transmembrane domain (which results in a secreted extracellular DEP-1) are viable and fertile [67, 68]. Finally, an in-frame replacement of DEP-1 intracellular domain with eGFP leads to early embryonic lethality (E10.5/E11.5) due to a number of severe angiogenesis defects. Among these are a collapse of the dorsal aorta, absence of mature intersomitic vessels, disorganized and enlarged peripheral vessels, disrupted extraembryonic circulation, and endothelial cells hyperproliferation in the yolk sac [69]. The reason for these phenotype differences has not been defined, but may include GFP-driven disruption of endothelial junctional complexes and hyperactivation of VEGFR2.

Characterization of the DEP-1 role in adult vasculature has generated discordant data. A monoclonal antibody, which targets DEP-1 extracellular domain and promotes its phosphatase activity, inhibits angiogenesis in a cornea pocket assay; thus suggesting that DEP1 may act as negative regulator of pathological angiogenesis [70]. Conversely, global DEP-1-null mice display reduced VEGFA-induced angiogenesis in a Matrigel plug assay; thus indicating that DEP-1 expression can enhance angiogenesis in vivo [65].

DEP-1 global null mice are unable to increase cerebrovascular blood flow (CBF) following permanent occlusion of the common carotid artery [71]. The same study did not detect differences in blood flow recovery and collateral vessel formation after seven days in a hind limb ischemia model. However, a recent report shows that DEP-1 global null mice have reduced blood flow recovery and arteriogenesis fourteen days following the induction of hind limb ischemia [65].

In general, the role of DEP-1 in the vasculature appears complex and context-dependent. This is probably the consequence of its opposite actions upon two key molecules such as Src and VEGFR2. Furthermore, DEP-1 is expressed in multiple cell types in the vasculature, including smooth muscle cells in which it regulates PDFGRβ signaling. Future studies using conditional knockout models will help to dissect the role of DEP-1 in angiogenesis and arteriogenesis.

3. Intracellular Phosphatases

Intracellular phosphatases lack a transmembrane domain and can be found in the cytosol or bound to specific subcellular structures [7, 72]. Shortly after dimerization, VEGFR2 is internalized via a classical clathrin-dependent mechanism and moves across the cytoplasm in a series of endosomal vesicles [73, 74]. During endocytosis and the subsequent intracellular trafficking, the VEGFR2 C-terminal domain retains full or partial phosphorylation and continues to signal [75, 76]. Consequently, in addition to plasma membrane, VEGFR2 can also be exposed to intracellular phosphatases that sit in specific subcellular compartments providing a more complex spatiotemporal regulation of its signal [77].

3.1 PTP1B

PTP1B, a ubiquitously expressed PTP, anchored to the endoplasmic reticulum (ER) membrane with the catalytic domain facing the cytosol [78]. PTP1B substrates include the insulin receptor [79, 80], VEGFR2 [81], and β-catenin [82].

PTP1B dephosphorylates immunoprecipitated VEGFR2 and silencing of PTP1B in HUVEC leads to increased VEGFA-induced VEGFR2 phosphorylation (tested Y1175 only) and proliferation [81]. Similar findings show that primary mouse EC isolated from PTP1B null mice display increased VEGFA-induced VEGFR2 phosphorylation and augmented proliferation and migration in response to VEGFA [83].

Mice carrying a global homozygous deletion of PTP1B are viable, fertile, and show enhanced insulin sensitivity and resistance to obesity [84, 85]. These mice also exhibit accelerated skin wound-associated angiogenesis and a faster rate of wound closure compared to wild-type mice [86]. Endothelial-specific PTP1B deletion leads to increased angiogenesis, as demonstrated in multiple in vivo models including developmental retinal angiogenesis, in vivo Matrigel assay and wound healing [83].

Importantly, PTP1B is a negative regulator of arteriogenesis [83]. This role of the phosphatase came to light during studies of dysfunctional arteriogenesis in myosin-VI [87] and synectin null mice [88], as well as mice carrying a knock-in of a mutant Nrp1 gene with a deleted cytoplasmic domain [89]. All three mutants are characterized by abnormally slow cytoplasmic trafficking of internalized VEGFR2, which leads to a prolonged contact between the internalized receptor in early endosomes and the endoplasmic reticulum-bound PTP1B. This leads to a partial down regulation of the VEGFR2-PLCγ-MAPK cascade that plays a crucial role in arteriogenesis [90]. Defects in arteriogenesis and angiogenesis reported in endothelial and smooth muscle cell–derived neuropilin-like protein (ESDN) null mice have also been linked to PTP1B action [91].

3.2 Shp1 (Src Homology-2 domain-containing Phosphatase 1)

Shp1 (also called HCP, PTP-1C, SH-PTP1, SHP) is highly expressed in hematopoietic cells, but is also found in other tissues [92-94]. Shp1 has been implicated in negative regulation of various signaling pathways initiated by RTKs, integrins, and cytokine receptors [94-96]. This phosphatase contains two N-terminal Src homology-2 (SH2) domains and a C-terminal PTP domain. SH2 domains allow Shp1 to bind phosphotyrosine residues in receptors and adapters. This event leads to activation of its PTP domain which can then dephosphorylates downstream targets [94, 97].

A role for Shp1 in VEGFR2 signaling modulation was initially proposed after the observation that it associates with VEGFR2 after VEGFA stimulation [98]. Subsequent studies have confirmed and extended this observation. Silencing of Shp1 in HUVEC increases VEGFA-induced phosphorylation at Y1175 and Y054/59, but not Y951 [99]. Functionally, VEGFA-induced proliferation is increased after Shp1 knockdown, but migration is not affected [99]. Several ECM-associated proteins with antiangiogenic properties (e.g. TSP-1, Thrombospondin-1; TIMP-2, Tissue Inhibitor of Metalloproteinase-2; CCN1, CCN family member 1) exert their effect by regulating Shp1 activity and/or expression. In microvascular EC (MVEC), binding of TSP-1 to its receptor CD36 promotes an association between Shp1 and VEGFR2, which leads to inhibition of VEGFA-induced signaling and migration [100]. Indeed, silencing of Shp1 renders the TSP-1/CD36 axis unable to exert its inhibitory action toward VEGFR2 signaling [100]. TIMP-2 antiangiogenic activity in vitro and in vivo is also Shp1-dependent [101, 102]. Mice that lack Shp1 expression (“motheaten mice or me/me mice”) suffer from a chronic systemic inflammation and die of interstitial pneumonia within three weeks of birth [103, 104]. EC-specific Shp1 deletion has not been reported, however indirect evidences points to an important role of this phosphatase in VEGFR2 signaling and angiogenesis in vivo. Silencing of Shp1 via siRNA injection in vivo results in increased capillary density in a hind limb ischemia model [105]. Furthermore, CCN1 (a matricellular protein that can bind integrins and proteoglycans) promotes Shp1 expression, which results in negative regulation of VEGFR2 signaling [106]. Retinas from mice with a postnatal endothelial CCN1 deletion have ∼60% reduction in Shp1 expression in EC and display a general VEGFR2 signaling augmentation . This results in retinal EC hyperproliferation and formation of a highly dense and immature vasculature network with delayed vessel outgrowth [106].

3.3 Shp2 (Src Homology-2 domain-containing Phosphatase 2)

Shp2 (also called PTP-1D, SH-PTP2, SH-PTP3, Syp, PTP-2C) is a ubiquitously expressed PTP with a structure similar to that of Shp1, but with distinct molecular and functional roles [94, 97, 107]. Shp2 substrates include GAB1 [108], RasGAP [109], β-Catenin [110, 111], and RTKs such as PDGFRβ [112] and VEGFR2 [113]. Shp2 is a positive regulator of the Ras-ERK pathway and multiple limes of evidences indicate that Shp2 is upstream of RAS [114]. Indeed, removal of Shp2 impairs ERK activation downstream of various RTKs, integrins, and cytokines [97, 115, 116]. Finally, expression of Shp2 in the endothelium reduces baseline phosphorylation of junctional proteins (e.g. VE-cadherin and β-catenin), thus contributing to maintenance of endothelial barrier integrity [117, 118].

Similar to Shp1, Shp2 was initially shown to associate with VEGFR2 upon VEGFA stimulation [98]. Shp2 dephosphorylates immunoprecipitated VEGFR2 [113] while silencing of Shp2 in HUVEC enhances VEGFA-induced VEGFR2 phosphorylation [119]. Expression of a Shp2 dominant-negative mutant (C459S) leads to an increase in both VEGFA-induced VEGFR2 phosphorylation and VEGFA-induced cell migration [113]. Interestingly, VEGFA-induced ERK1/2 activation is enhanced following silencing of Shp2 in HUVEC [120], which is in contrast with observations of Shp2 as a positive regulator of RAS-ERK pathway. One potential explanation for this difference is experimental evidence, which indicate that VEGR2-induced ERK1/2 activation is RAS independent [121].

Finally, silencing of Shp2 in HUVEC leads to decreased activation of Src (Y416) and downstream AKT in response to VEGFA [120]. The positive effect on Src activation is indirect and is the consequence of Shp2-mediated dephosphorylation of PAG/Cbp, a regulator of the C-terminal SRC kinase (Csk). In the absence of Shp2, Csk becomes more active and promotes hyperphosphorylation of the C-terminal Src inhibitory site (Y527), thus resulting in a general Src inhibition [122].

Currently, there are no in vivo data regarding the role of Shp2 in VEGFR2 signaling. A global deletion of the phosphatase leads to early embryonic lethality [123], while overexpression of a constitutive active Shp2 in the endothelial cells leads to severe cardiac defects (myocardial thinning, valves and AV septum defects) and early embryonic lethality [124]. The phenotype recapitulates the major defects present in the Noonan syndrome, which is associated with a Shp2 gain-of-function mutation in 50% of the cases [125]. It remains to be defined whether this phenotype is partly due to aberrant VEGFR2 signaling remains to be defined.

2.3 PTEN (Phosphate and tensing homolog deleted on chromosome 10)

PTEN (also called MMAC, TEP1) was initially identified as a dual specificity protein phosphatase [126, 127]. However, further characterization showed that PTEN also has a lipid phosphatase activity, which allows efficient dephosphorylation of phosphatidylinositol-3,4,5-trisphosphate (PIP3) generated at the plasma cell membrane [128]. Due to its ability to inhibit PI3K/AKT signaling, PTEN is a potent tumor suppressor and it is frequently mutated in tumors [129]. PTEN is largely expressed in the cytoplasm and nuclei, but it is quickly recruited to the plasma membrane where it converts PIP3 back to PIP2 [130]. VEGFR2-mediated PI3K activation promotes PIP3 formation and AKT recruitment/activation at the plasma membrane, an event that is thought to play an important role in tumor angiogenesis [131], adult arteriogenesis [132], and developmental angiogenesis [133]. In recent years, PTEN protein phosphatase activity has also emerged as a potential regulator of biological effects such as cell migration [127]. A number of novel PTEN substrates for this function have been proposed; they include FAK, Src, and their upstream regulators [127]. The protein phosphatase activity of PTEN in EC biology needs to be defined.

Although PTEN does not directly dephosphorylate VEGFR2, it counteracts PIP3 formation at the plasma membrane level and indirectly inhibits VEGFR2-induced AKT activation [134]. PTEN overexpression in EC inhibits VEGFA-induced migration, proliferation, and pro-survival effect in vitro [134]. Furthermore, VEGFA-induced angiogenesis is decreased in the aortic ring assay after adenovirus-mediated PTEN overexpression [134].

A global deletion of PTEN leads to embryonic lethality at E9.5 [135, 136]. Embryonic bodies that lack PTEN develop larger and longer vessel sprouts upon VEGFA stimulation [137]. A constitutive deletion of endothelial PTEN leads to embryonic mortality at E11.5 due to endothelial cells hyperproliferation, enlarged capillary network, failure of vascular plexus remodeling, severe hemorrhage, and cardiac failure [138]. The loss of a single allele of PTEN in the endothelium results in viable mice that manifest increased VEGFA-induced angiogenesis in Matrigel and tumor angiogenesis assays [138]. Mice with a postnatal endothelial PTEN deletion display a phenotype characterized by excessive branching and larger vessel diameter, but have normal radial extension and number of vessel sprouts in the developing retinal vasculature [137]. The phenotype is the consequence of increased EC proliferation at the sprouting front due to loss of the Notch antiproliferative effect in stalk cells [137].

Recent findings show that PTEN is a crosstalk node between Activin-receptor Like Kinase 1 (ALK1) and VEGFR2 signaling pathways. Indeed, silencing of ALK1 or blocking its ligands, BMP9 and 10 in ECs, leads to decreased PTEN activity at the plasma membrane and hyperactivation of the VEGFR2/PI3K pathway. This mechanism is responsible for the excessive angiogenesis and arteriovenous malformations observed in deficient mice in BMP signaling. Finally, inhibition of PI3K in this model rescues vascular malformations and suggests a novel therapeutic target for Hereditary Hemorrhagic Telangiectasia 2 (HHT2)[139].

4. Additional phosphatases involved in regulation of VEGFR2 biological effects

In addition to the extensively studied protein phosphatases discussed above, a certain number of other phosphatases have also been implicated in VEGFR2 signaling. These are discussed below.

4.1 LMW-PTP (Low Molecular Weight-PTP)

LMW-PTPs (also called HCPTP, Red Cell Acid Phosphatase) are a group of five small cytosolic tyrosine phosphatases (∼18 KDa) derived from splice variants of same gene [140, 141]. LMW-PTPs are ubiquitously expressed and conserved phosphatases [4] that have attracted particular interest for the development of inhibitors to use in cancer therapy [141, 142]. The isoform HCPTP-A has been shown to interact with VEGFR2 in a yeast two-hybrid system and to dephosphorylate the recombinant VEGFR2 [143]. Overexpression of HCPTP-A in endothelial cells inhibits VEGFA-induced phosphorylation (tested anti-pTyr only), migration, and proliferation [143]. Similarly, overexpression of HCPTP-A in rat aortic rings leads to reduced number and length of VEGFA-induced sprouts [143]. LMW-PTP null mice are viable, fertile, and display protection from heart failure [144].

4.2 PTP-MEG2

PTP-MEG2 (also called MEG2) is found in the cytosol, membrane fractions, and secretory vesicles [145, 146]. It is widely expressed in most organs and is detected in lymphocytes, platelets, and endothelial cells [93, 147, 148]. PTP-MEG2 substrate-trapping mutant is able to bind phosphorylated VEGFR2 [149]. Overexpression of PTP-MEG2 reduces VEGFA-induced VEGR2 phosphorylation in HUVEC (tested Y1175 only), while silencing PTP-MEG2 leads to an increase in VEGFR2-downstream signaling [149]. The majority of PTP-MEG2 null mice (∼90%) do not survive in utero development and die during late embryonic stages due to incomplete neuronal tube development and extensive hemorrhages [147].

4.3 TC-PTP (T cell PTP)

TC-PTP is a ubiquitously expressed PTP localized in the endoplasmic reticulum, nuclei, and the cytoplasm [150]. It is highly expressed in various hematopoietic cell lineages, but is also found in blood vessels [150, 151]. Although TC-PTP shares 70% sequence identity with PTP1B and shows certain redundancy of functions, the two phosphatases have remarkably different roles and a distinct set of substrates [152]. PTP1B has a well known role in metabolism and regulation of insulin signaling [153], while TC-PTP is a critical regulator in inflammation and plays an important role in immune system development and response [150].

TC-PTP dephosphorylates immunoprecipitated VEGFR2 (affected sites Y1054/59 and Y1214; no changes in Y1175) and a TC-PTP substrate-trapping mutant binds phosphorylated VEGFR2 [151]. Silencing of TC-PTP in HUVEC increases VEGFA-induced proliferation, while TC-PTP overexpression abrogates VEGFA-induced migration [151]. A global deletion of TC-PTP in mice leads to death within five weeks of birth due to anemia, severe immunosuppression, and systemic inflammation [154, 155].

4.4 Calcineurin (CN)

CN (also called PP2B, i.e. Protein Phosphatase 2B) is a Calcium/Calmodulin (Ca2+/CaM)-dependent Ser/Thr phosphatase that consists of a catalytic subunit (CnA) and a regulatory subunit (CnB) [3, 156]. CN dephosphorylates the members of the NFATc transcription factor family (c1-c4). Unphosphorylated NFATc translocate to the nucleus where it drives expression of target genes. Classical immunosuppressants such as cyclosporine and FK-506 are potent CN inhibitors and lead to a block in NFATc dephosphorylation/nuclear translocation. The major consequence of the block is a decreased expression of T-cell immune response genes that otherwise would drive rejection of transplanted organs [157]. CN also plays an important role in the development of the cardiovascular system [158].

The CN/NFATc axis is activated by VEGFA-induced increase in intracellular Ca2+ levels (via the VEGFR2/PLCγ/IP3 cascade) [159-161]. This launches context-dependent gene programs that contribute to modulation of EC behavior during heart development [162, 163] and angiogenesis in inflammation and cancer [164-166].

CN inhibition reduces VEGFA-induced migration/proliferation in vitro [167, 168] and VEGFA-induced angiogenesis in a cornea pocket assay [168]. These effects correlate with loss of cyclooxygenase-2 (COX-2) upregulation which is driven by VEGFA-induced NFATc nuclear translocation [168]. COX-2 is a known positive regulator of angiogenesis in chronic and acute inflammation due to its ability to increase synthesis of proangiogenic molecules such as PGE2 [169, 170] and other prostanoids [171]. Intravitreal injection of CN inhibitors has been shown to decrease pathological neovascularization in a ROP model in rats [172].

CN inhibition reduces VEGFA-induced NFATc1 nuclear translocation and proliferation in human pulmonary valve endothelial cells in vitro [173]. In line with these data, global homozygous NFATc1 deletion in mice results in impaired development of pulmonary and aortic valves [174, 175] partly due to decreased proliferation of the valve endocardial lineage [162]. This mice also display early embryonic lethality (∼E14.5) [174, 175].

Knock-in mice carrying a global inactive CN (CnB*/*) die at E10.5/E11.5 due to disorganized remodeling of the vascular plexus although the initial endothelial cell differentiation and vasculogenesis appear normal [176]. Double NFATc3/c4 null mice recapitulate most features of CnB*/* mice and die at ∼E11.5 [176]. Both phenotypes appear to be due, at least in part, to a dysregulated local VEGFA expression [176]. CN inhibition (between E9.5 and E12.5) also impairs correct patterning of coronary endothelial cells [163], a coordinated process that relies on myocardial VEGFA to drive ventricular endocardial cells to form the coronary arterial network [177]. Tie2-cre driven CnB-deletion (begin ∼ E8.0) recapitulates the coronary EC patterning defects, which are observed with CN inhibitors [163].

4.5 PP2A (Protein Phosphatase 2A)

PP2A is a ubiquitously expressed, highly conserved Ser/Thr phosphatase [3]. The active enzyme is a heterotrimeric complex that contains a scaffold subunit (A) bound to catalytic (C) and regulatory (B) subunits. The existence of numerous B isoforms permits the formation of multiple holoenzyme combinations (∼80), which can have different substrate specificities or subcellular localizations [3, 178]. PP2A substrates include central transducers of VEGFR2 signaling such as AKT [179], RAF [180], and eNOS [181].

Prolonged exposure of endothelial cells to fatty acids or lipid metabolites (e.g. ceramide) increases baseline PP2A activity, which in turn suppresses major VEGFA-induced responses such as AKT, eNOS and ERK activation [182]. This effect may contribute to the onset of endothelial dysfunction and arteriogenesis defects observed in high-fat diet fed mice [182, 183]. Vasoinhibins, a family of prolactin-derived antiangiogenic peptides, promote PP2A activation in EC and inhibit VEGFA-induced eNOS activation and permeability in vitro [184]. Intravitreal injections of vasoinhibins also blocks VEGFA-induced permeability in rat retinas [184]. Finally, inhibition of VEGFA-induced eNOS activation by antiangiogenic peptide endostatin requires PP2A activity [185].

4.6 PRL-3 (Protein of Regenerating Liver-3)

PRL-3 is a dual specificity phosphatase that localizes at the plasma membrane and endosomes following prenylation at the C-terminal end [186]. PRL-3 is highly expressed in heart and blood vessels during development [187] and is upregulated in tumor endothelium [188-191]. It may act as a positive regulator of VEGFR2 signaling. Primary mouse EC isolated from PRL-3 null mice display reduced VEGFA-induced VEGFR2 phosphorylation and Src activation, although the mechanism behind this defect has not been clarified [192]. A PRL-3 inhibitor reduces VEGFA-induced migration in HUVEC [192]. Finally, VEGFA-induced permeability is impaired in PRL-3 null mice [192].

4.7 MKP-1 (Mitogen activated Kinase Phosphatases-1)

MKP-1 (also called CL100) is a dual specificity nuclear phosphatase that acts on multiple MAPKs (i.e. ERK, p38, c-Jun N-Terminal Kinase) [193]. Silencing of MKP-1 reduces VEGFA-induced EC migration and proliferation in vitro [194]. VEGFA-induced sprout formation is reduced in aortic rings isolated from MKP-1 null mice [195]. MKP-1 null mice also show delayed distal blood flow recovery in a hind-limb ischemia model due to reduced angiogenesis and arteriogenesis [194]. In the absence of MKP-1, its substrate histone H3 Serine 10 (H3S10) [196] remains phosphorylated and inhibits VEGFA-induced CX3CL1 transcription [194], a proangiogenic gene [194, 197, 198].

4.8 RPTPζ (Receptor Protein Tyrosine Phosphatase zeta)

RPTPζ (also called phosphacan, RPTPβ/ζ, PTPRZ) is a receptor tyrosine phosphatase that may play a positive role in VEGFR2-Src activation. RPTPζ dephosphorylates the Src inhibitory site (Y527) [199] and silencing of RPTPζ inhibits VEGFA-induced migration in HUVEC [200]. Pleiotrophin (PTN, HARP), a natural PTPRZ ligand that inhibits its tyrosine phosphatase activity [201], reduces VEGFA-induced proliferation in HUVEC and VEGFA-induced angiogenesis in a Matrigel plug assay in vivo [202]. However, this effect is linked to the PTN ability to bind and neutralize VEGFA rather than RPTPζ inhibition [202].

5. Therapeutic opportunities

During the last 15 years, a plethora of studies in animal models has highlighted the diversity of VEGFR2 biological effects in health and disease [16]. Furthermore, the role of VEGFR2 in human biology has been further clarified with approval of anti-VEGFA monoclonal antibodies in clinical settings [203]. While beneficial in a number of settings, the experience with these antibodies has shown that a blockade of VEGFR2 signaling can lead to a number of adverse effects such as hypertension, renal toxicity, bowel perforation, hemorrhages, wound healing delay, and others [204, 205]. Thus, it has become evident that activation of VEGFR2 signaling can have both positive and negative consequences [30, 206, 207] because various VEGFR2 downstream pathways promote different biological effects [21]. Conceptually, future therapeutic approaches should aim to modulate some, but not other, VEGFR2 signaling pathways in order to increase therapeutic efficacy and decrease side effect potential.

In this scenario, targeting phosphatases may offer a new way to fine-tune VEGFR2 signaling. Mechanisms through which different phosphatases can modulate the net effect of a signal are multiples and includes phosphosite specificity, differential subcellular localization, different level of expression and activity, modulation by ligands and regulatory subunits [3, 7, 72]. Below are some examples of disease where targeting phosphatases to modulate VEGFR2 downstream effects may prove to be therapeutically useful.

5.1 Ischemic stroke

Occlusion of a cerebral artery leads to focal ischemia and VEGFA upregulation as early as six hours after vessel occlusion [31, 208]. This results in VEGFR2 signaling activation throughout stroke progression with both deleterious and beneficial consequences [31, 209, 210]. The harmful effects include BBB disruption, vascular leakage, and brain edema [211-213]. On the positive side, VEGFR2 activation enhances tissue oxygenation by promoting vasodilation, increases arteriogenesis in the penumbra area (the less viable area around the necrotic core), and exerts certain neuroprotective and neurogenic effects [31, 211]. Preclinical studies have consistently shown that VEGFA administration immediately after a vessel occlusion can result in severe adverse effects such increased brain edema, injury, and hemorrhagic stroke transformation [31]. Conversely, VEGFA administration following the acute window (two to three days after occlusion) can lead to important beneficial effects including improved vascular function, decreased ischemic injury and enhanced neurological recovery [31]. Although promising, administration of VEGFA (especially systemic) still poses elevated risk of acute adverse effects that can overcome long-term benefits [30, 31]. In order to achieve safety and maximize benefits, novel strategies aim to dissociate VEGFA permeability effects from its neurogenic and proangiogenic ones [30].

Inhibition of VEGFR2 signaling in ischemic stroke has been also linked to therapeutic benefits such as reduced edema and infarct volume [31, 212, 214]. These beneficial effects are mainly attributed to inhibition of VEGFA-induced vascular permeability. In agreement, inhibition of Src (which drives VEGFA-induced permeability) has been shown to reduce edema and infarct volume together with an improved neurological score [215]. This led to the idea of Src inhibition as a potential treatment in cerebral ischemia and stroke [64, 216]. Small molecule inhibitors with broad target for multiple kinases, including Src family kinases, have been approved for treatment of various oncological diseases [217]. However, therapeutic applications of these new molecules in the settings of stroke still need to be investigated.

Interestingly, deletion of certain protein phosphatases leads to specific inhibition of VEGFR2- induced Src activation, resulting in impairment of VEGFA-induced permeability (i.e. DEP-1, PRL-3) [65, 192]. Thus, it is conceivable that safety of VEGFA administration following ischemic stroke could be improved by contemporaneous inhibition of these phosphatases. Alternatively, inhibition alone during the acute stroke phase may be considered in order to block permeability effects of endogenously produced VEGFA. Finally, inhibition of other VEGFR2-related phosphatases (e.g. Shp1, LMW-PTPs) has been shown to improve the VEGFA-induced proangiogenic response [105, 143] with no reported impairment in either basal or VEGFA-induced permeability [218, 219]. Post-acute inhibition of these phosphatases may be feasible to boost long-term VEGFA proangiogenic and neuroprotective activities with minimal risk of adverse effects.

Animal studies demonstrated that several other neurological disorders (e.g. Amyotrophic Lateral Sclerosis, Parkinson Disease, and others) could benefit from VEGFA therapy with a proper control of its on- vs. off-target different biological effects [30]. Thus, a fine tailoring of VEGFR2 activation by phosphatases could represent a valuable therapeutic option in multiple neurological disorders in addition to stroke.

5.2 Peripheral artery disease

Peripheral artery disease (PAD) is caused by reduction of arterial blood flow in major arteries in the limbs and in the brain. The disease is associated with an increased risk of stroke, myocardial infarction, cardiovascular death [220]. It often progresses to critical limb ischemia (CLI), a conditioned characterized by severe impairment of blood flow to peripheral tissues in the legs leading to gangrene and/or necrosis. It can further lead to complications such as skin ulceration, severe pain, claudication, and limb amputation. In these settings, proangiogenic therapy has been tested as a means to promote blood flow restoration in the affected limbs. Various trials of VEGFA therapy failed to show meaningful functional benefits, such as decreased limb amputation frequency or cardiovascular mortality [220, 221]. The reasons for such a sub-optimal response to VEGFA therapy are poorly understood, but include decreased expression and impaired activation of VEGFR2 [222, 223].

Recent advances in our understanding of VEGFR2 signaling [21] include realization of the central role played by endothelial activation of ERK signaling in arteriogenesis [90, 224], an event negatively regulated by PTP1B [83], that is upregulated in the ischemic tissue [81]. Indeed, specific endothelial deletion of PTP1B increases VEGFA-induced VEGFR2 phosphorylation at Y1175 and correlates with improved arteriogenesis and blood flow recovery following hind limb ischemia (a mouse model for PAD), even in mice with impaired arteriogenic response [83]. Alternatively, inhibition of phosphatases inactivation of ERK may also achieve the same result. Thus, inhibition of phosphatases that selectively regulate VEGFR2/ERK signaling may offer a potential new therapeutic target to improve proarteriogenic therapy in PAD.

5.3 Cancer

The idea of “starving cancer to death” idea has driven the development of antiangiogenic therapies. The advent of therapies that block VEGFA/VEGFR2 signaling, such as TKIs and monoclonal antibodies, and inhibit tumor angiogenesis represents a significant improvement in treatment of certain cancers [203]. Yet, this approach has not met the highest expectation, the complete arrest of tumor growth [206, 225, 226]. A multitude of clinical trials has shown that the benefits of antiangiogenic therapy are limited to an increase in progression-free survival and sometimes overall survival on the order of months [206, 225, 226]. Moreover, certain cancer types completely fail to respond to antiangiogenic therapy [203, 226]. The current view is that aggressive antiangiogenic therapy confers a transitory slow-down because tumors put in place alternative mechanisms to escape hypoxia. Multiple reasons have been proposed to explain acquired resistance to antiangiogenic therapy in cancer. These have been widely described in recent reviews [206, 226-228]. A plethora of phosphatases have now been associated with either oncogenic (e.g. Shp2, PTP1B) or tumor suppressor (e.g. PTEN, PP2A) functions [23, 229]; they have attracted interest as novel pharmacological targets in cancer therapy [23, 24, 230]. Similarly, phosphatases that control VEGFR2 signaling or other proangiogenic pathways may be targeted to improve the efficacy of antiangiogenic therapies.

Furthermore, experimental evidences suggests that normalization of the tumor vasculature rather than complete VEGF antagonism and vessel disruption could achieve better therapeutic effects [231]. The general idea is to develop new therapeutic strategies to balance the pruning of leaky immature vessels while stabilizing a sufficient healthy vessel network to insure uniform and enhanced chemotherapy delivery with minimal hypoxia (thus limiting the onset of resistance) [206, 232, 233]. VEGFA is naturally present in tumor microenvironment, therefore, phosphatases that control VEGFR2 signaling could be targeted to promote or modulate vascular normalization. Indeed, certain phosphatases have already been implicated in tumor angiogenesis and vascular normalization (e.g. VEPTP and Shp1) [44, 234]. Further data from mouse tumor models are required in order to understand the role of the different phosphatases in shaping the vascular tumor network.

5.4 Other diseases

In addition to the diseases described above, regulation of VEGFR2 signaling has been the focus of multiple clinical trials for cardiovascular disease [221, 235] and retinopathies [236]. Furthermore, animal studies have shown that VEGFR2 signaling is involved in heart failure [237] and atherosclerosis [238]. Mouse mutants of phosphatases that target VEGFR2 signaling are now available, thus preclinical studies using animal models of these diseases could help to unveil novel therapeutic targets and shed light into the relative contribution of VEGFR2 and related phosphatases.

6. Perspectives and challenges

The common idea that phosphatases are housekeeping genes which are unable to influence the net effect of signaling and not able to achieve complex signaling modulation has turned out to be incorrect (see Ref. [22, 239] for a complete historical perspective). Multiple experimental evidence over the years has demonstrated that: first, dephosphorylation does not necessarily lead to deactivation of the targeted molecule (e.g. Src activation requires Y527 dephosphorylation); second, inactivation of a pathway node by dephosphorylation can still result in a global positive effect (there are double inhibition circuits); third, dephosphorylation cannot be associated a priori to termination of a pathway with functional outcomes (e.g. Shp2 activating mutations promote RAS/ERK pathway and are associated to Noonan syndrome and multiple cancer types); fourth, diseases and pathological events can arise from both increased or decreased activity of specific phosphatases, thus phosphatase inhibitors as well as activators are desirable. Furthermore, the relatively high number of phosphatase genes [1, 2], the wide spectrum of phenotypes in mouse knockout models, and a degree of substrate specificity [240] suggest that phosphatases have well defined biological roles, which make them potential candidates for pharmacological targeting.

Unfortunately, the R&D process for phosphatase-targeting drugs has proved to be difficult due to the necessity of designing highly negatively charged molecules that target the catalytic domain [241]. These compounds do not move well across cell membranes, therefore, they result in low bioavailability and poor therapeutic effect. However, progress has been made and more efficient ways to target phosphatases are emerging. These include molecules that can bind to allosteric sites or regulatory subunits. Identification of both selective inhibitors and activators using these strategies has been recently reported [230, 242-244]. Novel inhibitors that can target the catalytic domain with different mechanisms are also being developed [245, 246]. An update on molecules that modulate the activity of phosphatases involved in VEGFR2 signaling regulation is given in Table 3. The list includes only compounds that have shown biological effects in vascular-related functions or that have been tested in advanced preclinical models or clinical trials.

Table 3. Status of compounds that target phosphatases described in this review.

Target Compound Status and therapeutic applications Ref / Clinical trial ID
VE-PTP AKB-9778 (small molecule inhibitor) Clinical trials: Diabetic Macular Edema, Diabetic Retinopathy (Phase 2a completed) [249]
Preclinical studies: improved vascular normalization; decreased tumor growth and metastasis [42, 44]

DEP-1 Nonapeptides (activators); monoclonal antibody (activator) Preclinical studies: inhibition of cancer cells and EC proliferation; inhibition of angiogenesis in a mouse cornea pocket assay [61, 70]

PTP1B IONIS-PTP1BRx (antisense oligomer that reduces PTP1B expression) Clinical trials: Type-2 diabetes (Phase 2 completed) [250, 251] NCT01918865
Trodusquemine/MS-1436 (small molecule inhibitor) Clinical trials: metastatic breast cancer (Phase 1 on-going) NCT02524951
(3-bromo-7-cyano-2-naphthyl)(difluoro)methyl] phosphonic acid
graphic file with name nihms832484t1.jpg
Preclinical studies: decreased tumor growth and metastasis; antidiabetic effect; increased arteriogenesis in hind-limb ischemia [87, 252, 253]

Shp1 Sodium Stibogluconate (small molecule inhibitor used for treatment of Leishmaniasis; preferential Shp1 but also inhibits Shp2 and PTP1B) Clinical Trials: advanced melanoma or other cancer in combination with classical chemotherapeutic agents (Phase 1 completed) [254] NCT00498979
TP1 (small molecule inhibitor) Preclinical studies: decreased tumor growth [255]
siRNA (decreased Shp1 expression) Preclinical studies: improved angiogenesis in hind-limb ischemia [105]

Shp2 II-B08 (small molecule inhibitor) Preclinical studies: increased mouse survival in a model of mast-cell leukemia [256]
SHP099 (small molecule inhibitor, selective and orally bioavailable) Preclinical studies: tumor growth inhibition in an orthotopic model of human-derived acute myeloid leukemia [244]

PTEN Bisperoxovanadate-based compounds (inhibitors) Preclinical studies: decreased myocardial infarct size, reduced reperfusion damage in liver surgery and transplantation [257, 258]

LMW-PTP Various lead compounds (natural and synthetic) Proposed therapeutic applications: cancer, diabetes, infectious disease, heart failure [141, 142, 144, 259]

PTP-MEG2 PTP-MEG2-Inhibitor 7 Preclinical studies: increased insulin signaling in primary hepatocytes, improved insulin sensitivity in a diet-induced obese mouse model [260]

TC-PTP Spermidine and related compounds (activators) Preclinical studies: negative regulation of EGFR and VEGFR2 signaling; reduced VEGFA-induced sprout formation in vitro [261]

PP2A FTY720 and related compounds (activators) Proposed therapeutic applications: tumor suppression, inhibition of tumor growth and tumor angiogenesis [262, 263]

PRL-3 Various lead compounds (inhibitors) Proposed therapeutic applications: cancer [24, 241, 264]

MKP-1 Various lead compounds (inhibitors and activators) Proposed therapeutic applications: inflammatory and autoimmune diseases, neurological disorders, cancer, cardiovascular disease [265-268]

RPTPζ SCB4380 (small molecule inhibitor) Preclinical studies: decreased tumor growth in a glioblastoma model [269]

VEGFR2, the main VEGFA receptor, can initiate multiple signaling pathways that orchestrate a variety of biological effects including endothelial cell maturation, sprouting, migration, vascular permeability, vasodilation, and angiogenesis [18-20, 73]. A significant amount of experimental evidence have demonstrated that phosphatases can act either as a positive or negative regulator of signaling. This notion also appears valid for VEGFR2 signaling, as described in this review. A plethora of phosphatases have now been associated with either oncogenic or tumor suppressor functions [23, 229] and have attracted interest as novel pharmacological targets in cancer therapy [23, 24, 230]. Similarly, phosphatases that control VEGFR2 signaling or other proangiogenic pathways may be targeted to improve the efficacy of antiangiogenic therapies. Conversely, certain diseases may benefit from better proangiogenic effects (e.g. peripheral artery disease [220]), thus modulation of phosphatase activity may be a useful tool to sustain local VEGFR2 activation.

Further studies will improve our knowledge of phosphatase function in physiological and pathological processes; these enzymes may soon provide a novel set of pharmacological targets in the context of multiple diseases.

Acknowledgments

The authors wish to thank Dr. Mary Jo Mulligan-Kehoe for the useful suggestions to improve this work.

Funding Sources: This work is supported by NIH Grant R01 HL053793 and HL084619

Abbreviations

AMD

age-related macular degeneration

BrdU

bromodeoxyuridine

Cys

cysteine

DR

diabetic retinopathy

DUSP

dual specificity phosphatase

E

embryonic day

EC

endothelial cells

ECM

extracellular matrix

ER

endoplasmic reticulum

HUVEC

human umbilical vein endothelial cells

p-Ser/p-Thr

phospho-serine/phospho-threonine

p-Tyr

phospho-tyrosine

PTP

protein tyrosine phosphatase

R&D

research and development

ROP

retinopathy of prematurity

ROS

reactive oxygen species

RTK

receptor tyrosine kinase

SMC

smooth muscle cells

Src

tyrosine kinase c-SRC

TKI

tyrosine kinase inhibitor(s)

VEGFR2

vascular endothelial growth factor receptor type 2

Y

tyrosine

Footnotes

Conflict of Interest: None

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Bibliography

  • 1.Manning G, Whyte DB, Martinez R, Hunter T, Sudarsanam S. The protein kinase complement of the human genome. Science. 2002;298(5600):1912–34. doi: 10.1126/science.1075762. [DOI] [PubMed] [Google Scholar]
  • 2.Sacco F, Perfetto L, Castagnoli L, Cesareni G. The human phosphatase interactome: An intricate family portrait. FEBS Lett. 2012;586(17):2732–9. doi: 10.1016/j.febslet.2012.05.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Shi Y. Serine/threonine phosphatases: mechanism through structure. Cell. 2009;139(3):468–84. doi: 10.1016/j.cell.2009.10.006. [DOI] [PubMed] [Google Scholar]
  • 4.Alonso A, Sasin J, Bottini N, Friedberg I, Friedberg I, Osterman A, Godzik A, Hunter T, Dixon J, Mustelin T. Protein tyrosine phosphatases in the human genome. Cell. 2004;117(6):699–711. doi: 10.1016/j.cell.2004.05.018. [DOI] [PubMed] [Google Scholar]
  • 5.Li X, Wilmanns M, Thornton J, Kohn M. Elucidating human phosphatase-substrate networks. Sci Signal. 2013;6(275):rs10. doi: 10.1126/scisignal.2003203. [DOI] [PubMed] [Google Scholar]
  • 6.Alonso A, Pulido R. The extended human PTPome: a growing tyrosine phosphatase family. FEBS J. 2016;283(8):1404–29. doi: 10.1111/febs.13600. [DOI] [PubMed] [Google Scholar]
  • 7.Tonks NK. Protein tyrosine phosphatases: from genes, to function, to disease. Nature reviews. 2006;7(11):833–46. doi: 10.1038/nrm2039. [DOI] [PubMed] [Google Scholar]
  • 8.Oelrichs RB, Reid HH, Bernard O, Ziemiecki A, Wilks AF. NYK/FLK-1: a putative receptor protein tyrosine kinase isolated from E10 embryonic neuroepithelium is expressed in endothelial cells of the developing embryo. Oncogene. 1993;8(1):11–8. [PubMed] [Google Scholar]
  • 9.Quinn TP, Peters KG, De Vries C, Ferrara N, Williams LT. Fetal liver kinase 1 is a receptor for vascular endothelial growth factor and is selectively expressed in vascular endothelium. Proc Natl Acad Sci U S A. 1993;90(16):7533–7. doi: 10.1073/pnas.90.16.7533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Millauer B, Wizigmann-Voos S, Schnurch H, Martinez R, Moller NP, Risau W, Ullrich A. High affinity VEGF binding and developmental expression suggest Flk-1 as a major regulator of vasculogenesis and angiogenesis. Cell. 1993;72(6):835–46. doi: 10.1016/0092-8674(93)90573-9. [DOI] [PubMed] [Google Scholar]
  • 11.Katoh O, Tauchi H, Kawaishi K, Kimura A, Satow Y. Expression of the vascular endothelial growth factor (VEGF) receptor gene, KDR, in hematopoietic cells and inhibitory effect of VEGF on apoptotic cell death caused by ionizing radiation. Cancer Res. 1995;55(23):5687–92. [PubMed] [Google Scholar]
  • 12.Shalaby F, Rossant J, Yamaguchi TP, Gertsenstein M, Wu XF, Breitman ML, Schuh AC. Failure of blood-island formation and vasculogenesis in Flk-1-deficient mice. Nature. 1995;376(6535):62–6. doi: 10.1038/376062a0. [DOI] [PubMed] [Google Scholar]
  • 13.Carmeliet P, Ferreira V, Breier G, Pollefeyt S, Kieckens L, Gertsenstein M, Fahrig M, Vandenhoeck A, Harpal K, Eberhardt C, Declercq C, Pawling J, Moons L, Collen D, Risau W, Nagy A. Abnormal blood vessel development and lethality in embryos lacking a single VEGF allele. Nature. 1996;380(6573):435–9. doi: 10.1038/380435a0. [DOI] [PubMed] [Google Scholar]
  • 14.Ferrara N, Carver-Moore K, Chen H, Dowd M, Lu L, O'Shea KS, Powell-Braxton L, Hillan KJ, Moore MW. Heterozygous embryonic lethality induced by targeted inactivation of the VEGF gene. Nature. 1996;380(6573):439–42. doi: 10.1038/380439a0. [DOI] [PubMed] [Google Scholar]
  • 15.Senger DR, Galli SJ, Dvorak AM, Perruzzi CA, Harvey VS, Dvorak HF. Tumor cells secrete a vascular permeability factor that promotes accumulation of ascites fluid. Science. 1983;219(4587):983–5. doi: 10.1126/science.6823562. [DOI] [PubMed] [Google Scholar]
  • 16.Chung AS, Ferrara N. Developmental and pathological angiogenesis. Annu Rev Cell Dev Biol. 2011;27:563–84. doi: 10.1146/annurev-cellbio-092910-154002. [DOI] [PubMed] [Google Scholar]
  • 17.Shibuya M. VEGF-VEGFR Signals in Health and Disease. Biomol Ther (Seoul) 2014;22(1):1–9. doi: 10.4062/biomolther.2013.113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Weis SM, Cheresh DA. Pathophysiological consequences of VEGF-induced vascular permeability. Nature. 2005;437(7058):497–504. doi: 10.1038/nature03987. [DOI] [PubMed] [Google Scholar]
  • 19.Olsson AK, Dimberg A, Kreuger J, Claesson-Welsh L. VEGF receptor signalling - in control of vascular function. Nature reviews. 2006;7(5):359–71. doi: 10.1038/nrm1911. [DOI] [PubMed] [Google Scholar]
  • 20.Koch S, Claesson-Welsh L. Signal transduction by vascular endothelial growth factor receptors. Cold Spring Harb Perspect Med. 2012;2(7):a006502. doi: 10.1101/cshperspect.a006502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Simons M, Gordon E, Claesson-Welsh L. Mechanisms and regulation of endothelial VEGF receptor signalling. Nature reviews. 2016 doi: 10.1038/nrm.2016.87. [DOI] [PubMed] [Google Scholar]
  • 22.Tonks NK. Protein tyrosine phosphatases--from housekeeping enzymes to master regulators of signal transduction. FEBS J. 2013;280(2):346–78. doi: 10.1111/febs.12077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Julien SG, Dube N, Hardy S, Tremblay ML. Inside the human cancer tyrosine phosphatome. Nat Rev Cancer. 2011;11(1):35–49. doi: 10.1038/nrc2980. [DOI] [PubMed] [Google Scholar]
  • 24.Fontanillo M, Köhn M. Phosphatases: Their Roles in Cancer and Their Chemical Modulators. In: Böldicke T, editor. Protein Targeting Compounds : Prediction, Selection and Activity of Specific Inhibitors. 2015. pp. 209–240. [Google Scholar]
  • 25.Gurzov EN, Stanley WJ, Brodnicki TC, Thomas HE. Protein tyrosine phosphatases: molecular switches in metabolism and diabetes. Trends in endocrinology and metabolism: TEM. 2015;26(1):30–9. doi: 10.1016/j.tem.2014.10.004. [DOI] [PubMed] [Google Scholar]
  • 26.Tautz L, Senis YA, Oury C, Rahmouni S. Perspective: Tyrosine phosphatases as novel targets for antiplatelet therapy. Bioorg Med Chem. 2015;23(12):2786–97. doi: 10.1016/j.bmc.2015.03.075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Hendriks WJ, Pulido R. Protein tyrosine phosphatase variants in human hereditary disorders and disease susceptibilities. Biochim Biophys Acta. 2013;1832(10):1673–96. doi: 10.1016/j.bbadis.2013.05.022. [DOI] [PubMed] [Google Scholar]
  • 28.Khurana R. Role of Angiogenesis in Cardiovascular Disease: A Critical Appraisal. Circulation. 2005;112(12):1813–1824. doi: 10.1161/CIRCULATIONAHA.105.535294. [DOI] [PubMed] [Google Scholar]
  • 29.Tah V, Orlans HO, Hyer J, Casswell E, Din N, Sri Shanmuganathan V, Ramskold L, Pasu S. Anti-VEGF Therapy and the Retina: An Update. J Ophthalmol. 2015;2015:627674. doi: 10.1155/2015/627674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Lange C, Storkebaum E, de Almodovar CR, Dewerchin M, Carmeliet P. Vascular endothelial growth factor: a neurovascular target in neurological diseases. Nat Rev Neurol. 2016;12(8):439–54. doi: 10.1038/nrneurol.2016.88. [DOI] [PubMed] [Google Scholar]
  • 31.Ma Y, Zechariah A, Qu Y, Hermann DM. Effects of vascular endothelial growth factor in ischemic stroke. Journal of Neuroscience Research. 2012;90(10):1873–1882. doi: 10.1002/jnr.23088. [DOI] [PubMed] [Google Scholar]
  • 32.Holmes K, Roberts OL, Thomas AM, Cross MJ. Vascular endothelial growth factor receptor-2: structure, function, intracellular signalling and therapeutic inhibition. Cellular signalling. 2007;19(10):2003–12. doi: 10.1016/j.cellsig.2007.05.013. [DOI] [PubMed] [Google Scholar]
  • 33.Bäumer S, Keller L, Holtmann A, Funke R, August B, Gamp A, Wolburg H, Wolburg-Buchholz K, Deutsch U, Vestweber D. Vascular endothelial cell-specific phosphotyrosine phosphatase (VE-PTP) activity is required for blood vessel development. Blood. 2006;107(12):4754–62. doi: 10.1182/blood-2006-01-0141. [DOI] [PubMed] [Google Scholar]
  • 34.Fachinger G, Deutsch U, Risau W. Functional interaction of vascular endothelial-protein-tyrosine phosphatase with the angiopoietin receptor Tie-2. Oncogene. 1999;18(43):5948–53. doi: 10.1038/sj.onc.1202992. [DOI] [PubMed] [Google Scholar]
  • 35.Dominguez MG, Hughes VC, Pan L, Simmons M, Daly C, Anderson K, Noguera-Troise I, Murphy AJ, Valenzuela DM, Davis S, Thurston G, Yancopoulos GD, Gale NW. Vascular endothelial tyrosine phosphatase (VE-PTP)-null mice undergo vasculogenesis but die embryonically because of defects in angiogenesis. Proc Natl Acad Sci U S A. 2007;104(9):3243–8. doi: 10.1073/pnas.0611510104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Nawroth R, Poell G, Ranft A, Kloep S, Samulowitz U, Fachinger G, Golding M, Shima DT, Deutsch U, Vestweber D. VE-PTP and VE-cadherin ectodomains interact to facilitate regulation of phosphorylation and cell contacts. The EMBO journal. 2002;21(18):4885–95. doi: 10.1093/emboj/cdf497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Nottebaum AF, Cagna G, Winderlich M, Gamp AC, Linnepe R, Polaschegg C, Filippova K, Lyck R, Engelhardt B, Kamenyeva O, Bixel MG, Butz S, Vestweber D. VE-PTP maintains the endothelial barrier via plakoglobin and becomes dissociated from VE-cadherin by leukocytes and by VEGF. The Journal of experimental medicine. 2008;205(12):2929–45. doi: 10.1084/jem.20080406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Mellberg S, Dimberg A, Bahram F, Hayashi M, Rennel E, Ameur A, Westholm JO, Larsson E, Lindahl P, Cross MJ, Claesson-Welsh L. Transcriptional profiling reveals a critical role for tyrosine phosphatase VE-PTP in regulation of VEGFR2 activity and endothelial cell morphogenesis. FASEB J. 2009;23(5):1490–502. doi: 10.1096/fj.08-123810. [DOI] [PubMed] [Google Scholar]
  • 39.Flint AJ, Tiganis T, Barford D, Tonks NK. Development of “substrate-trapping” mutants to identify physiological substrates of protein tyrosine phosphatases. Proc Natl Acad Sci U S A. 1997;94(5):1680–5. doi: 10.1073/pnas.94.5.1680. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Hayashi M, Majumdar A, Li X, Adler J, Sun Z, Vertuani S, Hellberg C, Mellberg S, Koch S, Dimberg A, Koh GY, Dejana E, Belting HG, Affolter M, Thurston G, Holmgren L, Vestweber D, Claesson-Welsh L. VE-PTP regulates VEGFR2 activity in stalk cells to establish endothelial cell polarity and lumen formation. Nat Commun. 2013;4:1672. doi: 10.1038/ncomms2683. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Broermann A, Winderlich M, Block H, Frye M, Rossaint J, Zarbock A, Cagna G, Linnepe R, Schulte D, Nottebaum AF, Vestweber D. Dissociation of VE-PTP from VE-cadherin is required for leukocyte extravasation and for VEGF-induced vascular permeability in vivo. The Journal of experimental medicine. 2011;208(12):2393–401. doi: 10.1084/jem.20110525. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Frye M, Dierkes M, Kuppers V, Vockel M, Tomm J, Zeuschner D, Rossaint J, Zarbock A, Koh GY, Peters K, Nottebaum AF, Vestweber D. Interfering with VE-PTP stabilizes endothelial junctions in vivo via Tie-2 in the absence of VE-cadherin. The Journal of experimental medicine. 2015;212(13):2267–87. doi: 10.1084/jem.20150718. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Shen J, Frye M, Lee BL, Reinardy JL, McClung JM, Ding K, Kojima M, Xia H, Seidel C, Lima e Silva R, Dong A, Hackett SF, Wang J, Howard BW, Vestweber D, Kontos CD, Peters KG, Campochiaro PA. Targeting VE-PTP activates TIE2 and stabilizes the ocular vasculature. J Clin Invest. 2014;124(10):4564–76. doi: 10.1172/JCI74527. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Goel S, Gupta N, Walcott BP, Snuderl M, Kesler CT, Kirkpatrick ND, Heishi T, Huang Y, Martin JD, Ager E, Samuel R, Wang S, Yazbek J, Vakoc BJ, Peterson RT, Padera TP, Duda DG, Fukumura D, Jain RK. Effects of vascular-endothelial protein tyrosine phosphatase inhibition on breast cancer vasculature and metastatic progression. Journal of the National Cancer Institute. 2013;105(16):1188–201. doi: 10.1093/jnci/djt164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Borges LG, Seifert RA, Grant FJ, Hart CE, Disteche CM, Edelhoff S, Solca FF, Lieberman MA, Lindner V, Fischer EH, Lok S, Bowen-Pope DF. Cloning and characterization of rat density-enhanced phosphatase-1, a protein tyrosine phosphatase expressed by vascular cells. Circ Res. 1996;79(3):570–80. doi: 10.1161/01.res.79.3.570. [DOI] [PubMed] [Google Scholar]
  • 46.Autschbach F, Palou E, Mechtersheimer G, Rohr C, Pirotto F, Gassler N, Otto HF, Schraven B, Gaya A. Expression of the membrane protein tyrosine phosphatase CD148 in human tissues. Tissue Antigens. 1999;54(5):485–98. doi: 10.1034/j.1399-0039.1999.540506.x. [DOI] [PubMed] [Google Scholar]
  • 47.Cohen S, Dadi H, Shaoul E, Sharfe N, Roifman CM. Cloning and characterization of a lymphoid-specific, inducible human protein tyrosine phosphatase, Lyp. Blood. 1999;93(6):2013–24. [PubMed] [Google Scholar]
  • 48.Kappert K, Peters KG, Bohmer FD, Ostman A. Tyrosine phosphatases in vessel wall signaling. Cardiovasc Res. 2005;65(3):587–98. doi: 10.1016/j.cardiores.2004.08.016. [DOI] [PubMed] [Google Scholar]
  • 49.Kovalenko M, Denner K, Sandstrom J, Persson C, Gross S, Jandt E, Vilella R, Bohmer F, Ostman A. Site-selective dephosphorylation of the platelet-derived growth factor beta-receptor by the receptor-like protein-tyrosine phosphatase DEP-1. The Journal of biological chemistry. 2000;275(21):16219–26. doi: 10.1074/jbc.275.21.16219. [DOI] [PubMed] [Google Scholar]
  • 50.Kappert K, Paulsson J, Sparwel J, Leppanen O, Hellberg C, Ostman A, Micke P. Dynamic changes in the expression of DEP-1 and other PDGF receptor-antagonizing PTPs during onset and termination of neointima formation. FASEB J. 2007;21(2):523–34. doi: 10.1096/fj.06-6219com. [DOI] [PubMed] [Google Scholar]
  • 51.Chabot C, Spring K, Gratton JP, Elchebly M, Royal I. New role for the protein tyrosine phosphatase DEP-1 in Akt activation and endothelial cell survival. Mol Cell Biol. 2009;29(1):241–53. doi: 10.1128/MCB.01374-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Walchli S, Curchod ML, Gobert RP, Arkinstall S, Hooft van Huijsduijnen R. Identification of tyrosine phosphatases that dephosphorylate the insulin receptor. A brute force approach based on “substrate-trapping” mutants. The Journal of biological chemistry. 2000;275(13):9792–6. doi: 10.1074/jbc.275.13.9792. [DOI] [PubMed] [Google Scholar]
  • 53.Kruger J, Trappiel M, Dagnell M, Stawowy P, Meyborg H, Bohm C, Bhanot S, Ostman A, Kintscher U, Kappert K. Targeting density-enhanced phosphatase-1 (DEP-1) with antisense oligonucleotides improves the metabolic phenotype in high-fat diet-fed mice. Cell Commun Signal. 2013;11:49. doi: 10.1186/1478-811X-11-49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Kruger J, Brachs S, Trappiel M, Kintscher U, Meyborg H, Wellnhofer E, Thone-Reineke C, Stawowy P, Ostman A, Birkenfeld AL, Bohmer FD, Kappert K. Enhanced insulin signaling in density-enhanced phosphatase-1 (DEP-1) knockout mice. Mol Metab. 2015;4(4):325–36. doi: 10.1016/j.molmet.2015.02.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Shintani T, Higashi S, Takeuchi Y, Gaudio E, Trapasso F, Fusco A, Noda M. The R3 receptor-like protein tyrosine phosphatase subfamily inhibits insulin signalling by dephosphorylating the insulin receptor at specific sites. Journal of biochemistry. 2015;158(3):235–43. doi: 10.1093/jb/mvv045. [DOI] [PubMed] [Google Scholar]
  • 56.Holsinger LJ, Ward K, Duffield B, Zachwieja J, Jallal B. The transmembrane receptor protein tyrosine phosphatase DEP1 interacts with p120(ctn) Oncogene. 2002;21(46):7067–76. doi: 10.1038/sj.onc.1205858. [DOI] [PubMed] [Google Scholar]
  • 57.Takahashi K, Matafonov A, Sumarriva K, Ito H, Lauhan C, Zemel D, Tsuboi N, Chen J, Reynolds A, Takahashi T. CD148 tyrosine phosphatase promotes cadherin cell adhesion. PLoS One. 2014;9(11):e112753. doi: 10.1371/journal.pone.0112753. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Pera IL, Iuliano R, Florio T, Susini C, Trapasso F, Santoro M, Chiariotti L, Schettini G, Viglietto G, Fusco A. The rat tyrosine phosphatase eta increases cell adhesion by activating c-Src through dephosphorylation of its inhibitory phosphotyrosine residue. Oncogene. 2005;24(19):3187–95. doi: 10.1038/sj.onc.1208510. [DOI] [PubMed] [Google Scholar]
  • 59.Ostman A, Yang Q, Tonks NK. Expression of DEP-1, a receptor-like protein-tyrosine-phosphatase, is enhanced with increasing cell density. Proc Natl Acad Sci U S A. 1994;91(21):9680–4. doi: 10.1073/pnas.91.21.9680. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Grazia Lampugnani M, Zanetti A, Corada M, Takahashi T, Balconi G, Breviario F, Orsenigo F, Cattelino A, Kemler R, Daniel TO, Dejana E. Contact inhibition of VEGF-induced proliferation requires vascular endothelial cadherin, beta-catenin, and the phosphatase DEP-1/CD148. The Journal of cell biology. 2003;161(4):793–804. doi: 10.1083/jcb.200209019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Ortuso F, Paduano F, Carotenuto A, Gomez-Monterrey I, Bilotta A, Gaudio E, Sala M, Artese A, Vernieri E, Dattilo V, Iuliano R, Brancaccio D, Bertamino A, Musella S, Alcaro S, Grieco P, Perrotti N, Croce CM, Novellino E, Fusco A, Campiglia P, Trapasso F. Discovery of PTPRJ agonist peptides that effectively inhibit in vitro cancer cell proliferation and tube formation. ACS Chem Biol. 2013;8(7):1497–506. doi: 10.1021/cb3007192. [DOI] [PubMed] [Google Scholar]
  • 62.Spring K, Chabot C, Langlois S, Lapointe L, Trinh NT, Caron C, Hebda JK, Gavard J, Elchebly M, Royal I. Tyrosine phosphorylation of DEP-1/CD148 as a mechanism controlling Src kinase activation, endothelial cell permeability, invasion, and capillary formation. Blood. 2012;120(13):2745–56. doi: 10.1182/blood-2011-12-398040. [DOI] [PubMed] [Google Scholar]
  • 63.Giannotta M, Trani M, Dejana E. VE-cadherin and endothelial adherens junctions: active guardians of vascular integrity. Developmental cell. 2013;26(5):441–54. doi: 10.1016/j.devcel.2013.08.020. [DOI] [PubMed] [Google Scholar]
  • 64.Eliceiri BP, Paul R, Schwartzberg PL, Hood JD, Leng J, Cheresh DA. Selective requirement for Src kinases during VEGF-induced angiogenesis and vascular permeability. Molecular cell. 1999;4(6):915–24. doi: 10.1016/s1097-2765(00)80221-x. [DOI] [PubMed] [Google Scholar]
  • 65.Fournier P, Dussault S, Fusco A, Rivard A, Royal I. Tyrosine Phosphatase PTPRJ/DEP-1 Is an Essential Promoter of Vascular Permeability, Angiogenesis, and Tumor Progression. Cancer research. 2016;76(17):5080–91. doi: 10.1158/0008-5472.CAN-16-1071. [DOI] [PubMed] [Google Scholar]
  • 66.Trapasso F, Drusco A, Costinean S, Alder H, Aqeilan RI, Iuliano R, Gaudio E, Raso C, Zanesi N, Croce CM, Fusco A. Genetic ablation of Ptprj, a mouse cancer susceptibility gene, results in normal growth and development and does not predispose to spontaneous tumorigenesis. DNA Cell Biol. 2006;25(6):376–82. doi: 10.1089/dna.2006.25.376. [DOI] [PubMed] [Google Scholar]
  • 67.Zhu JW, Brdicka T, Katsumoto TR, Lin J, Weiss A. Structurally distinct phosphatases CD45 and CD148 both regulate B cell and macrophage immunoreceptor signaling. Immunity. 2008;28(2):183–96. doi: 10.1016/j.immuni.2007.11.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Katsumoto TR, Kudo M, Chen C, Sundaram A, Callahan EC, Zhu JW, Lin J, Rosen CE, Manz BN, Lee JW, Matthay MA, Huang X, Sheppard D, Weiss A. The phosphatase CD148 promotes airway hyperresponsiveness through SRC family kinases. J Clin Invest. 2013;123(5):2037–48. doi: 10.1172/JCI66397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Takahashi T, Takahashi K, St John PL, Fleming PA, Tomemori T, Watanabe T, Abrahamson DR, Drake CJ, Shirasawa T, Daniel TO. A Mutant Receptor Tyrosine Phosphatase, CD148, Causes Defects in Vascular Development. Molecular and Cellular Biology. 2003;23(5):1817–1831. doi: 10.1128/MCB.23.5.1817-1831.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Takahashi T, Takahashi K, Mernaugh RL, Tsuboi N, Liu H, Daniel TO. A monoclonal antibody against CD148, a receptor-like tyrosine phosphatase, inhibits endothelial-cell growth and angiogenesis. Blood. 2006;108(4):1234–42. doi: 10.1182/blood-2005-10-4296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Hackbusch D, Dulsner A, Gatzke N, Kruger J, Hillmeister P, Nagorka S, Blaschke F, Ritter Z, Thone-Reineke C, Bohmer FD, Buschmann I, Kappert K. Knockout of Density-Enhanced Phosphatase-1 impairs cerebrovascular reserve capacity in an arteriogenesis model in mice. Biomed Res Int. 2013;2013:802149. doi: 10.1155/2013/802149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.den Hertog J, Ostman A, Bohmer FD. Protein tyrosine phosphatases: regulatory mechanisms. FEBS J. 2008;275(5):831–47. doi: 10.1111/j.1742-4658.2008.06247.x. [DOI] [PubMed] [Google Scholar]
  • 73.Simons M. An inside view: VEGF receptor trafficking and signaling. Physiology (Bethesda) 2012;27(4):213–22. doi: 10.1152/physiol.00016.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Lampugnani MG, Orsenigo F, Gagliani MC, Tacchetti C, Dejana E. Vascular endothelial cadherin controls VEGFR-2 internalization and signaling from intracellular compartments. The Journal of cell biology. 2006;174(4):593–604. doi: 10.1083/jcb.200602080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Bruns AF, Herbert SP, Odell AF, Jopling HM, Hooper NM, Zachary IC, Walker JH, Ponnambalam S. Ligand-stimulated VEGFR2 signaling is regulated by co-ordinated trafficking and proteolysis. Traffic. 2010;11(1):161–74. doi: 10.1111/j.1600-0854.2009.01001.x. [DOI] [PubMed] [Google Scholar]
  • 76.Horowitz A, Seerapu HR. Regulation of VEGF signaling by membrane traffic. Cellular signalling. 2012;24(9):1810–20. doi: 10.1016/j.cellsig.2012.05.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Clegg LW, Mac Gabhann F. Site-Specific Phosphorylation of VEGFR2 Is Mediated by Receptor Trafficking: Insights from a Computational Model. PLoS Comput Biol. 2015;11(6):e1004158. doi: 10.1371/journal.pcbi.1004158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Stuible M, Tremblay ML. In control at the ER: PTP1B and the down-regulation of RTKs by dephosphorylation and endocytosis. Trends Cell Biol. 2010;20(11):672–9. doi: 10.1016/j.tcb.2010.08.013. [DOI] [PubMed] [Google Scholar]
  • 79.Bandyopadhyay D, Kusari A, Kenner KA, Liu F, Chernoff J, Gustafson TA, Kusari J. Protein-tyrosine phosphatase 1B complexes with the insulin receptor in vivo and is tyrosine-phosphorylated in the presence of insulin. J Biol Chem. 1997;272(3):1639–45. doi: 10.1074/jbc.272.3.1639. [DOI] [PubMed] [Google Scholar]
  • 80.Seely BL, Staubs PA, Reichart DR, Berhanu P, Milarski KL, Saltiel AR, Kusari J, Olefsky JM. Protein tyrosine phosphatase 1B interacts with the activated insulin receptor. Diabetes. 1996;45(10):1379–85. doi: 10.2337/diab.45.10.1379. [DOI] [PubMed] [Google Scholar]
  • 81.Nakamura Y, Patrushev N, Inomata H, Mehta D, Urao N, Kim HW, Razvi M, Kini V, Mahadev K, Goldstein BJ, McKinney R, Fukai T, Ushio-Fukai M. Role of protein tyrosine phosphatase 1B in vascular endothelial growth factor signaling and cell-cell adhesions in endothelial cells. Circ Res. 2008;102(10):1182–91. doi: 10.1161/CIRCRESAHA.107.167080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Xu G, Craig AW, Greer P, Miller M, Anastasiadis PZ, Lilien J, Balsamo J. Continuous association of cadherin with beta-catenin requires the non-receptor tyrosine-kinase Fer. J Cell Sci. 2004;117(Pt 15):3207–19. doi: 10.1242/jcs.01174. [DOI] [PubMed] [Google Scholar]
  • 83.Lanahan AA, Lech D, Dubrac A, Zhang J, Zhuang ZW, Eichmann A, Simons M. PTP1b is a physiologic regulator of vascular endothelial growth factor signaling in endothelial cells. Circulation. 2014;130(11):902–9. doi: 10.1161/CIRCULATIONAHA.114.009683. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Elchebly M, Payette P, Michaliszyn E, Cromlish W, Collins S, Loy AL, Normandin D, Cheng A, Himms-Hagen J, Chan CC, Ramachandran C, Gresser MJ, Tremblay ML, Kennedy BP. Increased insulin sensitivity and obesity resistance in mice lacking the protein tyrosine phosphatase-1B gene. Science. 1999;283(5407):1544–8. doi: 10.1126/science.283.5407.1544. [DOI] [PubMed] [Google Scholar]
  • 85.Klaman LD, Boss O, Peroni OD, Kim JK, Martino JL, Zabolotny JM, Moghal N, Lubkin M, Kim YB, Sharpe AH, Stricker-Krongrad A, Shulman GI, Neel BG, Kahn BB. Increased energy expenditure, decreased adiposity, and tissue-specific insulin sensitivity in protein-tyrosine phosphatase 1B-deficient mice. Mol Cell Biol. 2000;20(15):5479–89. doi: 10.1128/mcb.20.15.5479-5489.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Zhang J, Li L, Li J, Liu Y, Zhang CY, Zhang Y, Zen K. Protein tyrosine phosphatase 1B impairs diabetic wound healing through vascular endothelial growth factor receptor 2 dephosphorylation. Arteriosclerosis, thrombosis, and vascular biology. 2015;35(1):163–74. doi: 10.1161/ATVBAHA.114.304705. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Lanahan AA, Hermans K, Claes F, Kerley-Hamilton JS, Zhuang ZW, Giordano FJ, Carmeliet P, Simons M. VEGF receptor 2 endocytic trafficking regulates arterial morphogenesis. Developmental cell. 2010;18(5):713–24. doi: 10.1016/j.devcel.2010.02.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Chittenden TW, Claes F, Lanahan AA, Autiero M, Palac RT, Tkachenko EV, Elfenbein A, Ruiz de Almodovar C, Dedkov E, Tomanek R, Li W, Westmore M, Singh JP, Horowitz A, Mulligan-Kehoe MJ, Moodie KL, Zhuang ZW, Carmeliet P, Simons M. Selective regulation of arterial branching morphogenesis by synectin. Developmental cell. 2006;10(6):783–95. doi: 10.1016/j.devcel.2006.03.012. [DOI] [PubMed] [Google Scholar]
  • 89.Lanahan A, Zhang X, Fantin A, Zhuang Z, Rivera-Molina F, Speichinger K, Prahst C, Zhang J, Wang Y, Davis G, Toomre D, Ruhrberg C, Simons M. The neuropilin 1 cytoplasmic domain is required for VEGF-A-dependent arteriogenesis. Developmental cell. 2013;25(2):156–68. doi: 10.1016/j.devcel.2013.03.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Ren B, Deng Y, Mukhopadhyay A, Lanahan AA, Zhuang ZW, Moodie KL, Mulligan-Kehoe MJ, Byzova TV, Peterson RT, Simons M. ERK1/2-Akt1 crosstalk regulates arteriogenesis in mice and zebrafish. J Clin Invest. 2010;120(4):1217–28. doi: 10.1172/JCI39837. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Nie L, Guo X, Esmailzadeh L, Zhang J, Asadi A, Collinge M, Li X, Kim JD, Woolls M, Jin SW, Dubrac A, Eichmann A, Simons M, Bender JR, Sadeghi MM. Transmembrane protein ESDN promotes endothelial VEGF signaling and regulates angiogenesis. J Clin Invest. 2013;123(12):5082–97. doi: 10.1172/JCI67752. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Yi TL, Cleveland JL, Ihle JN. Protein tyrosine phosphatase containing SH2 domains: characterization, preferential expression in hematopoietic cells, and localization to human chromosome 12p12-p13. Mol Cell Biol. 1992;12(2):836–46. doi: 10.1128/mcb.12.2.836. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Wright MB, Seifert RA, Bowen-Pope DF. Protein-tyrosine phosphatases in the vessel wall: differential expression after acute arterial injury, Arteriosclerosis. thrombosis, and vascular biology. 2000;20(5):1189–98. doi: 10.1161/01.atv.20.5.1189. [DOI] [PubMed] [Google Scholar]
  • 94.Neel BG, Chan G, Dhanji S. SH2 Domain-Containing Protein-Tyrosine Phosphatases. In: Bradshaw RA, Dennis EA, editors. Handbook of Cell Signaling. 2009. pp. 771–810. [Google Scholar]
  • 95.Zhang J, Somani AK, Siminovitch KA. Roles of the SHP-1 tyrosine phosphatase in the negative regulation of cell signalling. Semin Immunol. 2000;12(4):361–78. doi: 10.1006/smim.2000.0223. [DOI] [PubMed] [Google Scholar]
  • 96.Hendriks WJAJ, Böhmer FD. Non-transmembrane PTPs in Cancer. In: Neel BG, Tonks N, editors. Protein Tyrosine Phosphatases in Cancer. 2016. pp. 47–114. [Google Scholar]
  • 97.Neel BG, Gu H, Pao L. The ‘Shp'ing news: SH2 domain-containing tyrosine phosphatases in cell signaling. Trends Biochem Sci. 2003;28(6):284–293. doi: 10.1016/S0968-0004(03)00091-4. [DOI] [PubMed] [Google Scholar]
  • 98.Kroll J, Waltenberger J. The vascular endothelial growth factor receptor KDR activates multiple signal transduction pathways in porcine aortic endothelial cells. The Journal of biological chemistry. 1997;272(51):32521–7. doi: 10.1074/jbc.272.51.32521. [DOI] [PubMed] [Google Scholar]
  • 99.Bhattacharya R, Kwon J, Wang E, Mukherjee P, Mukhopadhyay D. Src homology 2 (SH2) domain containing protein tyrosine phosphatase-1 (SHP-1) dephosphorylates VEGF Receptor-2 and attenuates endothelial DNA synthesis, but not migration*. J Mol Signal. 2008;3:8. doi: 10.1186/1750-2187-3-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Chu LY, Ramakrishnan DP, Silverstein RL. Thrombospondin-1 modulates VEGF signaling via CD36 by recruiting SHP-1 to VEGFR2 complex in microvascular endothelial cells. Blood. 2013;122(10):1822–32. doi: 10.1182/blood-2013-01-482315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Seo DW, Li H, Guedez L, Wingfield PT, Diaz T, Salloum R, Wei By, Stetler-Stevenson WG. TIMP-2 Mediated Inhibition of Angiogenesis. Cell. 2003;114(2):171–180. doi: 10.1016/s0092-8674(03)00551-8. [DOI] [PubMed] [Google Scholar]
  • 102.Seo DW, Li H, Qu CK, Oh J, Kim YS, Diaz T, Wei B, Han JW, Stetler-Stevenson WG. Shp-1 mediates the antiproliferative activity of tissue inhibitor of metalloproteinase-2 in human microvascular endothelial cells. The Journal of biological chemistry. 2006;281(6):3711–21. doi: 10.1074/jbc.M509932200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Tsui HW, Siminovitch KA, de Souza L, Tsui FW. Motheaten and viable motheaten mice have mutations in the haematopoietic cell phosphatase gene. Nat Genet. 1993;4(2):124–9. doi: 10.1038/ng0693-124. [DOI] [PubMed] [Google Scholar]
  • 104.Shultz LD, Sidman CL. Genetically determined murine models of immunodeficiency. Annu Rev Immunol. 1987;5:367–403. doi: 10.1146/annurev.iy.05.040187.002055. [DOI] [PubMed] [Google Scholar]
  • 105.Sugano M, Tsuchida K, Maeda T, Makino N. SiRNA targeting SHP-1 accelerates angiogenesis in a rat model of hindlimb ischemia. Atherosclerosis. 2007;191(1):33–9. doi: 10.1016/j.atherosclerosis.2006.04.021. [DOI] [PubMed] [Google Scholar]
  • 106.Chintala H, Krupska I, Yan L, Lau L, Grant M, Chaqour B. The matricellular protein CCN1 controls retinal angiogenesis by targeting VEGF, Src homology 2 domain phosphatase-1 and Notch signaling. Development. 2015;142(13):2364–74. doi: 10.1242/dev.121913. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Lorenz U. SHP-1 and SHP-2 in T cells: two phosphatases functioning at many levels. Immunol Rev. 2009;228(1):342–59. doi: 10.1111/j.1600-065X.2008.00760.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Zhang SQ, Tsiaras WG, Araki T, Wen G, Minichiello L, Klein R, Neel BG. Receptor-specific regulation of phosphatidylinositol 3′-kinase activation by the protein tyrosine phosphatase Shp2. Mol Cell Biol. 2002;22(12):4062–72. doi: 10.1128/MCB.22.12.4062-4072.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Agazie YM, Hayman MJ. Molecular mechanism for a role of SHP2 in epidermal growth factor receptor signaling. Mol Cell Biol. 2003;23(21):7875–86. doi: 10.1128/MCB.23.21.7875-7886.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Timmerman I, Hoogenboezem M, Bennett AM, Geerts D, Hordijk PL, van Buul JD. The tyrosine phosphatase SHP2 regulates recovery of endothelial adherens junctions through control of beta-catenin phosphorylation. Mol Biol Cell. 2012;23(21):4212–25. doi: 10.1091/mbc.E12-01-0038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Ukropec JA, Hollinger MK, Salva SM, Woolkalis MJ. SHP2 association with VE-cadherin complexes in human endothelial cells is regulated by thrombin. The Journal of biological chemistry. 2000;275(8):5983–6. doi: 10.1074/jbc.275.8.5983. [DOI] [PubMed] [Google Scholar]
  • 112.Klinghoffer RA, Kazlauskas A. Identification of a putative Syp substrate, the PDGF beta receptor. The Journal of biological chemistry. 1995;270(38):22208–17. doi: 10.1074/jbc.270.38.22208. [DOI] [PubMed] [Google Scholar]
  • 113.Mitola S, Brenchio B, Piccinini M, Tertoolen L, Zammataro L, Breier G, Rinaudo MT, den Hertog J, Arese M, Bussolino F. Type I collagen limits VEGFR-2 signaling by a SHP2 protein-tyrosine phosphatase-dependent mechanism 1. Circ Res. 2006;98(1):45–54. doi: 10.1161/01.RES.0000199355.32422.7b. [DOI] [PubMed] [Google Scholar]
  • 114.Chan G, Neel BG. Role of PTPN11 (SHP2) in Cancer. In: Neel BG, Tonks N, editors. Protein Tyrosine Phosphatases in Cancer. 2016. pp. 115–143. [Google Scholar]
  • 115.Ostman A, Hellberg C, Bohmer FD. Protein-tyrosine phosphatases and cancer. Nat Rev Cancer. 2006;6(4):307–20. doi: 10.1038/nrc1837. [DOI] [PubMed] [Google Scholar]
  • 116.Lauriol J, Jaffre F, Kontaridis MI. The role of the protein tyrosine phosphatase SHP2 in cardiac development and disease. Semin Cell Dev Biol. 2015;37:73–81. doi: 10.1016/j.semcdb.2014.09.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Grinnell KL, Casserly B, Harrington EO. Role of protein tyrosine phosphatase SHP2 in barrier function of pulmonary endothelium. Am J Physiol Lung Cell Mol Physiol. 2010;298(3):L361–70. doi: 10.1152/ajplung.00374.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Hatanaka K, Lanahan AA, Murakami M, Simons M. Fibroblast growth factor signaling potentiates VE-cadherin stability at adherens junctions by regulating SHP2. PLoS One. 2012;7(5):e37600. doi: 10.1371/journal.pone.0037600. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Sinha S, Vohra PK, Bhattacharya R, Dutta S, Sinha S, Mukhopadhyay D. Dopamine regulates phosphorylation of VEGF receptor 2 by engaging Src-homology-2-domain-containing protein tyrosine phosphatase 2. Journal of cell science. 2009;122(Pt 18):3385–92. doi: 10.1242/jcs.053124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Ha CH, Bennett AM, Jin ZG. A novel role of vascular endothelial cadherin in modulating c-Src activation and downstream signaling of vascular endothelial growth factor. The Journal of biological chemistry. 2008;283(11):7261–70. doi: 10.1074/jbc.M702881200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Shibuya M. VEGFR and type-V RTK activation and signaling. Cold Spring Harb Perspect Biol. 2013;5(10):a009092. doi: 10.1101/cshperspect.a009092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Zhang SQ, Yang W, Kontaridis MI, Bivona TG, Wen G, Araki T, Luo J, Thompson JA, Schraven BL, Philips MR, Neel BG. Shp2 Regulates Src Family Kinase Activity and Ras/Erk Activation by Controlling Csk Recruitment. Molecular cell. 2004;13(3):341–355. doi: 10.1016/s1097-2765(04)00050-4. [DOI] [PubMed] [Google Scholar]
  • 123.Saxton TM, Henkemeyer M, Gasca S, Shen R, Rossi DJ, Shalaby F, Feng GS, Pawson T. Abnormal mesoderm patterning in mouse embryos mutant for the SH2 tyrosine phosphatase Shp-2. EMBO J. 1997;16(9):2352–64. doi: 10.1093/emboj/16.9.2352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Araki T, Chan G, Newbigging S, Morikawa L, Bronson RT, Neel BG. Noonan syndrome cardiac defects are caused by PTPN11 acting in endocardium to enhance endocardial-mesenchymal transformation. Proc Natl Acad Sci U S A. 2009;106(12):4736–41. doi: 10.1073/pnas.0810053106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Tartaglia M, Gelb BD. Noonan syndrome and related disorders: genetics and pathogenesis. Annu Rev Genomics Hum Genet. 2005;6:45–68. doi: 10.1146/annurev.genom.6.080604.162305. [DOI] [PubMed] [Google Scholar]
  • 126.Li J, Yen C, Liaw D, Podsypanina K, Bose S, Wang SI, Puc J, Miliaresis C, Rodgers L, Mc Combie R, Bigner SH, Giovanella BC, Ittmann M, Tycko B, Hibshoosh H, Wigler MH, Parsons R. PTEN, a putative protein tyrosine phosphatase gene mutated in human brain, breast, and prostate cancer. Science. 1997;275(5308):1943–7. doi: 10.1126/science.275.5308.1943. [DOI] [PubMed] [Google Scholar]
  • 127.Leslie NR, Maccario H, Spinelli L, Davidson L. The significance of PTEN's protein phosphatase activity. Adv Enzyme Regul. 2009;49(1):190–6. doi: 10.1016/j.advenzreg.2008.12.002. [DOI] [PubMed] [Google Scholar]
  • 128.Maehama T, Dixon JE. The tumor suppressor, PTEN/MMAC1, dephosphorylates the lipid second messenger, phosphatidylinositol 3,4,5-trisphosphate. J Biol Chem. 1998;273(22):13375–8. doi: 10.1074/jbc.273.22.13375. [DOI] [PubMed] [Google Scholar]
  • 129.Song MS, Salmena L, Pandolfi PP. The functions and regulation of the PTEN tumour suppressor. Nature reviews Molecular cell biology. 2012;13(5):283–96. doi: 10.1038/nrm3330. [DOI] [PubMed] [Google Scholar]
  • 130.Das S, Dixon JE, Cho W. Membrane-binding and activation mechanism of PTEN. Proc Natl Acad Sci U S A. 2003;100(13):7491–6. doi: 10.1073/pnas.0932835100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Jiang BH, Liu LZ. Chapter 2 PI3K/PTEN Signaling in Angiogenesis and Tumorigenesis. 2009;102:19–65. doi: 10.1016/S0065-230X(09)02002-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Ackah E, Yu J, Zoellner S, Iwakiri Y, Skurk C, Shibata R, Ouchi N, Easton RM, Galasso G, Birnbaum MJ, Walsh K, Sessa WC. Akt1/protein kinase Balpha is critical for ischemic and VEGF-mediated angiogenesis. J Clin Invest. 2005;115(8):2119–27. doi: 10.1172/JCI24726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Lee MY, Luciano AK, Ackah E, Rodriguez-Vita J, Bancroft TA, Eichmann A, Simons M, Kyriakides TR, Morales-Ruiz M, Sessa WC. Endothelial Akt1 mediates angiogenesis by phosphorylating multiple angiogenic substrates. Proc Natl Acad Sci U S A. 2014;111(35):12865–70. doi: 10.1073/pnas.1408472111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Huang J, Kontos CD. PTEN modulates vascular endothelial growth factor-mediated signaling and angiogenic effects. J Biol Chem. 2002;277(13):10760–6. doi: 10.1074/jbc.M110219200. [DOI] [PubMed] [Google Scholar]
  • 135.Suzuki A, de la Pompa JL, Stambolic V, Elia AJ, Sasaki T, del Barco Barrantes I, Ho A, Wakeham A, Itie A, Khoo W, Fukumoto M, Mak TW. High cancer susceptibility and embryonic lethality associated with mutation of the PTEN tumor suppressor gene in mice. Curr Biol. 1998;8(21):1169–78. doi: 10.1016/s0960-9822(07)00488-5. [DOI] [PubMed] [Google Scholar]
  • 136.Di Cristofano A, Pesce B, Cordon-Cardo C, Pandolfi PP. Pten is essential for embryonic development and tumour suppression. Nat Genet. 1998;19(4):348–55. doi: 10.1038/1235. [DOI] [PubMed] [Google Scholar]
  • 137.Serra H, Chivite I, Angulo-Urarte A, Soler A, Sutherland JD, Arruabarrena-Aristorena A, Ragab A, Lim R, Malumbres M, Fruttiger M, Potente M, Serrano M, Fabra A, Vinals F, Casanovas O, Pandolfi PP, Bigas A, Carracedo A, Gerhardt H, Graupera M. PTEN mediates Notch-dependent stalk cell arrest in angiogenesis. Nat Commun. 2015;6:7935. doi: 10.1038/ncomms8935. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Hamada K, Sasaki T, Koni PA, Natsui M, Kishimoto H, Sasaki J, Yajima N, Horie Y, Hasegawa G, Naito M, Miyazaki J, Suda T, Itoh H, Nakao K, Mak TW, Nakano T, Suzuki A. The PTEN/PI3K pathway governs normal vascular development and tumor angiogenesis. Genes Dev. 2005;19(17):2054–65. doi: 10.1101/gad.1308805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Ola R, Dubrac A, Han J, Zhang F, Fang JS, Larrivée B, Lee M, Urarte AA, Kraehling JR, Genet G, Hirschi KK, Sessa WC, Vinals Canals F, Graupera M, Yan M, Young LH, Oh SP, Eichmann A. PI3 Kinase inhibition improves vascular malformations in mouse models of Hereditary Haemorrhagic Telangiectasia. Nat Commun. 2016 doi: 10.1038/ncomms13650. Accepted. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Raugei G, Ramponi G, Chiarugi P. Low molecular weight protein tyrosine phosphatases: small, but smart. Cell Mol Life Sci. 2002;59(6):941–9. doi: 10.1007/s00018-002-8481-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Alho I, Costa L, Bicho M, Coelho C. The role of low-molecular-weight protein tyrosine phosphatase (LMW-PTP ACP1) in oncogenesis. Tumour Biol. 2013;34(4):1979–89. doi: 10.1007/s13277-013-0784-1. [DOI] [PubMed] [Google Scholar]
  • 142.Maccari R, Ottana R. Low molecular weight phosphotyrosine protein phosphatases as emerging targets for the design of novel therapeutic agents. Journal of medicinal chemistry. 2012;55(1):2–22. doi: 10.1021/jm200607g. [DOI] [PubMed] [Google Scholar]
  • 143.Huang L, Sankar S, Lin C, Kontos CD, Schroff AD, Cha EH, Feng SM, Li SF, Yu Z, Van Etten RL, Blanar MA, Peters KG. HCPTPA, a protein tyrosine phosphatase that regulates vascular endothelial growth factor receptor-mediated signal transduction and biological activity. The Journal of biological chemistry. 1999;274(53):38183–8. doi: 10.1074/jbc.274.53.38183. [DOI] [PubMed] [Google Scholar]
  • 144.Wade F, Quijada P, Al-Haffar KM, Awad SM, Kunhi M, Toko H, Marashly Q, Belhaj K, Zahid I, Al-Mohanna F, Stanford SM, Alvarez R, Liu Y, Colak D, Jordan MC, Roos KP, Assiri A, Al-Habeeb W, Sussman M, Bottini N, Poizat C. Deletion of low molecular weight protein tyrosine phosphatase (Acp1) protects against stress-induced cardiomyopathy. J Pathol. 2015;237(4):482–94. doi: 10.1002/path.4594. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Qi Y, Zhao R, Cao H, Sui X, Krantz SB, Zhao ZJ. Purification and characterization of protein tyrosine phosphatase PTP-MEG2. J Cell Biochem. 2002;86(1):79–89. doi: 10.1002/jcb.10195. [DOI] [PubMed] [Google Scholar]
  • 146.Huynh H, Bottini N, Williams S, Cherepanov V, Musumeci L, Saito K, Bruckner S, Vachon E, Wang X, Kruger J, Chow CW, Pellecchia M, Monosov E, Greer PA, Trimble W, Downey GP, Mustelin T. Control of vesicle fusion by a tyrosine phosphatase. Nat Cell Biol. 2004;6(9):831–9. doi: 10.1038/ncb1164. [DOI] [PubMed] [Google Scholar]
  • 147.Wang Y, Vachon E, Zhang J, Cherepanov V, Kruger J, Li J, Saito K, Shannon P, Bottini N, Huynh H, Ni H, Yang H, McKerlie C, Quaggin S, Zhao ZJ, Marsden PA, Mustelin T, Siminovitch KA, Downey GP. Tyrosine phosphatase MEG2 modulates murine development and platelet and lymphocyte activation through secretory vesicle function. The Journal of experimental medicine. 2005;202(11):1587–97. doi: 10.1084/jem.20051108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Gu M, Warshawsky I, Majerus PW. Cloning and expression of a cytosolic megakaryocyte protein-tyrosine-phosphatase with sequence homology to retinaldehyde-binding protein and yeast SEC14p. Proc Natl Acad Sci U S A. 1992;89(7):2980–4. doi: 10.1073/pnas.89.7.2980. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Hao Q, Samten B, Ji HL, Zhao ZJ, Tang H. Tyrosine phosphatase PTP-MEG2 negatively regulates vascular endothelial growth factor receptor signaling and function in endothelial cells. American journal of physiology Cell physiology. 2012;303(5):C548–53. doi: 10.1152/ajpcell.00415.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Doody KM, Bourdeau A, Tremblay ML. T-cell protein tyrosine phosphatase is a key regulator in immune cell signaling: lessons from the knockout mouse model and implications in human disease. Immunological reviews. 2009;228(1):325–341. doi: 10.1111/j.1600-065X.2008.00743.x. [DOI] [PubMed] [Google Scholar]
  • 151.Mattila E, Auvinen K, Salmi M, Ivaska J. The protein tyrosine phosphatase TCPTP controls VEGFR2 signalling. Journal of cell science. 2008;121(Pt 21):3570–80. doi: 10.1242/jcs.031898. [DOI] [PubMed] [Google Scholar]
  • 152.Stuible M, Doody KM, Tremblay ML. PTP1B and TC-PTP: regulators of transformation and tumorigenesis. Cancer Metastasis Rev. 2008;27(2):215–30. doi: 10.1007/s10555-008-9115-1. [DOI] [PubMed] [Google Scholar]
  • 153.Bakke J, Haj FG. Protein-tyrosine phosphatase 1B substrates and metabolic regulation. Semin Cell Dev Biol. 2015;37:58–65. doi: 10.1016/j.semcdb.2014.09.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Heinonen KM, Nestel FP, Newell EW, Charette G, Seemayer TA, Tremblay ML, Lapp WS. T-cell protein tyrosine phosphatase deletion results in progressive systemic inflammatory disease. Blood. 2004;103(9):3457–64. doi: 10.1182/blood-2003-09-3153. [DOI] [PubMed] [Google Scholar]
  • 155.You-Ten KE, Muise ES, Itie A, Michaliszyn E, Wagner J, Jothy S, Lapp WS, Tremblay ML. Impaired bone marrow microenvironment and immune function in T cell protein tyrosine phosphatase-deficient mice. The Journal of experimental medicine. 1997;186(5):683–93. doi: 10.1084/jem.186.5.683. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Klee CB, Ren H, Wang X. Regulation of the calmodulin-stimulated protein phosphatase, calcineurin. The Journal of biological chemistry. 1998;273(22):13367–70. doi: 10.1074/jbc.273.22.13367. [DOI] [PubMed] [Google Scholar]
  • 157.Ho S, Clipstone N, Timmermann L, Northrop J, Graef I, Fiorentino D, Nourse J, Crabtree GR. The mechanism of action of cyclosporin A and FK506. Clin Immunol Immunopathol. 1996;80(3 Pt 2):S40–5. doi: 10.1006/clin.1996.0140. [DOI] [PubMed] [Google Scholar]
  • 158.Schulz RA, Yutzey KE. Calcineurin signaling and NFAT activation in cardiovascular and skeletal muscle development. Developmental Biology. 2004;266(1):1–16. doi: 10.1016/j.ydbio.2003.10.008. [DOI] [PubMed] [Google Scholar]
  • 159.Dawson NS, Zawieja DC, Wu MH, Granger HJ. Signaling pathways mediating VEGF165-induced calcium transients and membrane depolarization in human endothelial cells. FASEB J. 2006;20(7):991–3. doi: 10.1096/fj.05-3923fje. [DOI] [PubMed] [Google Scholar]
  • 160.Brock TA, Dvorak HF, Senger DR. Tumor-secreted vascular permeability factor increases cytosolic Ca2+ and von Willebrand factor release in human endothelial cells. The American journal of pathology. 1991;138(1):213–21. [PMC free article] [PubMed] [Google Scholar]
  • 161.Armesilla AL, Lorenzo E, Gomez del Arco P, Martinez-Martinez S, Alfranca A, Redondo JM. Vascular endothelial growth factor activates nuclear factor of activated T cells in human endothelial cells: a role for tissue factor gene expression. Mol Cell Biol. 1999;19(3):2032–43. doi: 10.1128/mcb.19.3.2032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Wu B, Wang Y, Lui W, Langworthy M, Tompkins KL, Hatzopoulos AK, Baldwin HS, Zhou B. Nfatc1 coordinates valve endocardial cell lineage development required for heart valve formation. Circ Res. 2011;109(2):183–92. doi: 10.1161/CIRCRESAHA.111.245035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Zeini M, Hang CT, Lehrer-Graiwer J, Dao T, Zhou B, Chang CP. Spatial and temporal regulation of coronary vessel formation by calcineurin-NFAT signaling. Development. 2009;136(19):3335–45. doi: 10.1242/dev.037903. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Schweighofer B, Testori J, Sturtzel C, Sattler S, Mayer H, Wagner O, Bilban M, Hofer E. The VEGF-induced transcriptional response comprises gene clusters at the crossroad of angiogenesis and inflammation. Thromb. 2009;02(3):544–54. doi: 10.1160/TH08-12-0830. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Minami T, Horiuchi K, Miura M, Abid MR, Takabe W, Noguchi N, Kohro T, Ge X, Aburatani H, Hamakubo T, Kodama T, Aird WC. Vascular endothelial growth factor- and thrombin-induced termination factor, Down syndrome critical region-1, attenuates endothelial cell proliferation and angiogenesis. The Journal of biological chemistry. 2004;279(48):50537–54. doi: 10.1074/jbc.M406454200. [DOI] [PubMed] [Google Scholar]
  • 166.Baek KH, Zaslavsky A, Lynch RC, Britt C, Okada Y, Siarey RJ, Lensch MW, Park IH, Yoon SS, Minami T, Korenberg JR, Folkman J, Daley GQ, Aird WC, Galdzicki Z, Ryeom S. Down's syndrome suppression of tumour growth and the role of the calcineurin inhibitor DSCR1. Nature. 2009;459(7250):1126–30. doi: 10.1038/nature08062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Rafiee P, Heidemann J, Ogawa H, Johnson NA, Fisher PJ, Li MS, Otterson MF, Johnson CP, Binion DG. Cyclosporin A differentially inhibits multiple steps in VEGF induced angiogenesis in human microvascular endothelial cells through altered intracellular signaling. Cell Commun Signal. 2004;2(1):3. doi: 10.1186/1478-811X-2-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Hernandez GL, Volpert OV, Iniguez MA, Lorenzo E, Martinez-Martinez S, Grau R, Fresno M, Redondo JM. Selective inhibition of vascular endothelial growth factor-mediated angiogenesis by cyclosporin A: roles of the nuclear factor of activated T cells and cyclooxygenase 2. The Journal of experimental medicine. 2001;193(5):607–20. doi: 10.1084/jem.193.5.607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Finetti F, Solito R, Morbidelli L, Giachetti A, Ziche M, Donnini S. Prostaglandin E2 regulates angiogenesis via activation of fibroblast growth factor receptor-1. The Journal of biological chemistry. 2008;283(4):2139–46. doi: 10.1074/jbc.M703090200. [DOI] [PubMed] [Google Scholar]
  • 170.Finetti F, Donnini S, Giachetti A, Morbidelli L, Ziche M. Prostaglandin E(2) primes the angiogenic switch via a synergic interaction with the fibroblast growth factor-2 pathway. Circ Res. 2009;105(7):657–66. doi: 10.1161/CIRCRESAHA.109.203760. [DOI] [PubMed] [Google Scholar]
  • 171.Salvado MD, Alfranca A, Haeggstrom JZ, Redondo JM. Prostanoids in tumor angiogenesis: therapeutic intervention beyond COX-2. Trends Mol Med. 2012;18(4):233–43. doi: 10.1016/j.molmed.2012.02.002. [DOI] [PubMed] [Google Scholar]
  • 172.Bretz CA, Savage S, Capozzi M, Penn JS. The role of the NFAT signaling pathway in retinal neovascularization. Investigative ophthalmology & visual science. 2013;54(10):7020–7. doi: 10.1167/iovs.13-12183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Johnson EN, Lee YM, Sander TL, Rabkin E, Schoen FJ, Kaushal S, Bischoff J. NFATc1 mediates vascular endothelial growth factor-induced proliferation of human pulmonary valve endothelial cells. The Journal of biological chemistry. 2003;278(3):1686–92. doi: 10.1074/jbc.M210250200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Ranger AM, Grusby MJ, Hodge MR, Gravallese EM, de la Brousse FC, Hoey T, Mickanin C, Baldwin HS, Glimcher LH. The transcription factor NF-ATc is essential for cardiac valve formation. Nature. 1998;392(6672):186–90. doi: 10.1038/32426. [DOI] [PubMed] [Google Scholar]
  • 175.de la Pompa JL, Timmerman LA, Takimoto H, Yoshida H, Elia AJ, Samper E, Potter J, Wakeham A, Marengere L, Langille BL, Crabtree GR, Mak TW. Role of the NF-ATc transcription factor in morphogenesis of cardiac valves and septum. Nature. 1998;392(6672):182–6. doi: 10.1038/32419. [DOI] [PubMed] [Google Scholar]
  • 176.Graef IA, Chen F, Chen L, Kuo A, Crabtree GR. Signals transduced by Ca(2+)/calcineurin and NFATc3/c4 pattern the developing vasculature. Cell. 2001;105(7):863–75. doi: 10.1016/s0092-8674(01)00396-8. [DOI] [PubMed] [Google Scholar]
  • 177.Wu B, Zhang Z, Lui W, Chen X, Wang Y, Chamberlain AA, Moreno-Rodriguez RA, Markwald RR, O'Rourke BP, Sharp DJ, Zheng D, Lenz J, Baldwin HS, Chang CP, Zhou B. Endocardial cells form the coronary arteries by angiogenesis through myocardial-endocardial VEGF signaling. Cell. 2012;151(5):1083–96. doi: 10.1016/j.cell.2012.10.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Sangodkar J, Farrington CC, McClinch K, Galsky MD, Kastrinsky DB, Narla G. All roads lead to PP2A: exploiting the therapeutic potential of this phosphatase. FEBS J. 2016;283(6):1004–24. doi: 10.1111/febs.13573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Liao Y, Hung MC. Physiological regulation of Akt activity and stability. Am J Transl Res. 2010;2(1):19–42. [PMC free article] [PubMed] [Google Scholar]
  • 180.Mischak H, Seitz T, Janosch P, Eulitz M, Steen H, Schellerer M, Philipp A, Kolch W. Negative regulation of Raf-1 by phosphorylation of serine 621. Mol Cell Biol. 1996;16(10):5409–18. doi: 10.1128/mcb.16.10.5409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Michell BJ, Chen Z, Tiganis T, Stapleton D, Katsis F, Power DA, Sim AT, Kemp BE. Coordinated control of endothelial nitric-oxide synthase phosphorylation by protein kinase C and the cAMP-dependent protein kinase. The Journal of biological chemistry. 2001;276(21):17625–8. doi: 10.1074/jbc.C100122200. [DOI] [PubMed] [Google Scholar]
  • 182.Mehra VC, Jackson E, Zhang XM, Jiang XC, Dobrucki LW, Yu J, Bernatchez P, Sinusas AJ, Shulman GI, Sessa WC, Yarovinsky TO, Bender JR. Ceramide-activated phosphatase mediates fatty acid-induced endothelial VEGF resistance and impaired angiogenesis. The American journal of pathology. 2014;184(5):1562–76. doi: 10.1016/j.ajpath.2014.01.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Zhang QJ, Holland WL, Wilson L, Tanner JM, Kearns D, Cahoon JM, Pettey D, Losee J, Duncan B, Gale D, Kowalski CA, Deeter N, Nichols A, Deesing M, Arrant C, Ruan T, Boehme C, McCamey DR, Rou J, Ambal K, Narra KK, Summers SA, Abel ED, Symons JD. Ceramide mediates vascular dysfunction in diet-induced obesity by PP2A-mediated dephosphorylation of the eNOS-Akt complex. Diabetes. 2012;61(7):1848–59. doi: 10.2337/db11-1399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Garcia C, Aranda J, Arnold E, Thebault S, Macotela Y, Lopez-Casillas F, Mendoza V, Quiroz-Mercado H, Hernandez-Montiel HL, Lin SH, de la Escalera GM, Clapp C. Vasoinhibins prevent retinal vasopermeability associated with diabetic retinopathy in rats via protein phosphatase 2A-dependent eNOS inactivation. J Clin Invest. 2008;118(6):2291–300. doi: 10.1172/JCI34508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Urbich C, Reissner A, Chavakis E, Dernbach E, Haendeler J, Fleming I, Zeiher AM, Kaszkin M, Dimmeler S. Dephosphorylation of endothelial nitric oxide synthase contributes to the anti-angiogenic effects of endostatin. FASEB J. 2002;16(7):706–8. doi: 10.1096/fj.01-0637fje. [DOI] [PubMed] [Google Scholar]
  • 186.Bessette DC, Wong PC, Pallen CJ. PRL-3: a metastasis-associated phosphatase in search of a function. Cells Tissues Organs. 2007;185(1-3):232–6. doi: 10.1159/000101324. [DOI] [PubMed] [Google Scholar]
  • 187.Guo K, Li J, Wang H, Osato M, Tang JP, Quah SY, Gan BQ, Zeng Q. PRL-3 initiates tumor angiogenesis by recruiting endothelial cells in vitro and in vivo. Cancer research. 2006;66(19):9625–35. doi: 10.1158/0008-5472.CAN-06-0726. [DOI] [PubMed] [Google Scholar]
  • 188.Aird WC. Molecular heterogeneity of tumor endothelium. Cell and tissue research. 2009;335(1):271–81. doi: 10.1007/s00441-008-0672-y. [DOI] [PubMed] [Google Scholar]
  • 189.Rouleau C, Roy A, St Martin T, Dufault MR, Boutin P, Liu D, Zhang M, Puorro-Radzwill K, Rulli L, Reczek D, Bagley R, Byrne A, Weber W, Roberts B, Klinger K, Brondyk W, Nacht M, Madden S, Burrier R, Shankara S, Teicher BA. Protein tyrosine phosphatase PRL-3 in malignant cells and endothelial cells: expression and function. Molecular cancer therapeutics. 2006;5(2):219–29. doi: 10.1158/1535-7163.MCT-05-0289. [DOI] [PubMed] [Google Scholar]
  • 190.St Croix B, Rago C, Velculescu V, Traverso G, Romans KE, Montgomery E, Lal A, Riggins GJ, Lengauer C, Vogelstein B, Kinzler KW. Genes expressed in human tumor endothelium. Science. 2000;289(5482):1197–202. doi: 10.1126/science.289.5482.1197. [DOI] [PubMed] [Google Scholar]
  • 191.Parker BS, Argani P, Cook BP, Liangfeng H, Chartrand SD, Zhang M, Saha S, Bardelli A, Jiang Y, St Martin TB, Nacht M, Teicher BA, Klinger KW, Sukumar S, Madden SL. Alterations in vascular gene expression in invasive breast carcinoma. Cancer research. 2004;64(21):7857–66. doi: 10.1158/0008-5472.CAN-04-1976. [DOI] [PubMed] [Google Scholar]
  • 192.Zimmerman MW, McQueeney KE, Isenberg JS, Pitt BR, Wasserloos KA, Homanics GE, Lazo JS. Protein-tyrosine phosphatase 4A3 (PTP4A3) promotes vascular endothelial growth factor signaling and enables endothelial cell motility. The Journal of biological chemistry. 2014;289(9):5904–13. doi: 10.1074/jbc.M113.480038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Caunt CJ, Keyse SM. Dual-specificity MAP kinase phosphatases (MKPs): shaping the outcome of MAP kinase signalling. FEBS J. 2013;280(2):489–504. doi: 10.1111/j.1742-4658.2012.08716.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Boerckel JD, Chandrasekharan UM, Waitkus MS, Tillmaand EG, Bartlett R, Dicorleto PE. Mitogen-activated protein kinase phosphatase-1 promotes neovascularization and angiogenic gene expression. Arteriosclerosis, thrombosis, and vascular biology. 2014;34(5):1020–31. doi: 10.1161/ATVBAHA.114.303403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Kinney CM, Chandrasekharan UM, Mavrakis L, DiCorleto PE. VEGF and thrombin induce MKP-1 through distinct signaling pathways: role for MKP-1 in endothelial cell migration, American journal of physiology. Cell physiology. 2008;294(1):C241–50. doi: 10.1152/ajpcell.00187.2007. [DOI] [PubMed] [Google Scholar]
  • 196.Kinney CM, Chandrasekharan UM, Yang L, Shen J, Kinter M, McDermott MS, DiCorleto PE. Histone H3 as a novel substrate for MAP kinase phosphatase-1, American journal of physiology. Cell physiology. 2009;296(2):C242–9. doi: 10.1152/ajpcell.00492.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Chandrasekharan UM, Yang L, Walters A, Howe P, DiCorleto PE. Role of CL-100, a dual specificity phosphatase, in thrombin-induced endothelial cell activation. The Journal of biological chemistry. 2004;279(45):46678–85. doi: 10.1074/jbc.M406441200. [DOI] [PubMed] [Google Scholar]
  • 198.Zhang J, Yang W, Luo B, Hu B, Maheshwari A, Fallon MB. The role of CX(3)CL1/CX(3)CR1 in pulmonary angiogenesis and intravascular monocyte accumulation in rat experimental hepatopulmonary syndrome. J Hepatol. 2012;57(4):752–8. doi: 10.1016/j.jhep.2012.05.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Polykratis A, Katsoris P, Courty J, Papadimitriou E. Characterization of heparin affin regulatory peptide signaling in human endothelial cells. The Journal of biological chemistry. 2005;280(23):22454–61. doi: 10.1074/jbc.M414407200. [DOI] [PubMed] [Google Scholar]
  • 200.Koutsioumpa M, Poimenidi E, Pantazaka E, Theodoropoulou C, Skoura A, Megalooikonomou V, Kieffer N, Courty J, Mizumoto S, Sugahara K, Papadimitriou E. Receptor protein tyrosine phosphatase beta/zeta is a functional binding partner for vascular endothelial growth factor. Molecular cancer. 2015;14:19. doi: 10.1186/s12943-015-0287-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Meng K, Rodriguez-Pena A, Dimitrov T, Chen W, Yamin M, Noda M, Deuel TF. Pleiotrophin signals increased tyrosine phosphorylation of beta beta-catenin through inactivation of the intrinsic catalytic activity of the receptor-type protein tyrosine phosphatase beta/zeta. Proc Natl Acad Sci U S A. 2000;97(6):2603–8. doi: 10.1073/pnas.020487997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Heroult M, Bernard-Pierrot I, Delbe J, Hamma-Kourbali Y, Katsoris P, Barritault D, Papadimitriou E, Plouet J, Courty J. Heparin affin regulatory peptide binds to vascular endothelial growth factor (VEGF) and inhibits VEGF-induced angiogenesis. Oncogene. 2004;23(9):1745–53. doi: 10.1038/sj.onc.1206879. [DOI] [PubMed] [Google Scholar]
  • 203.Ferrara N, Adamis AP. Ten years of anti-vascular endothelial growth factor therapy. Nat Rev Drug Discov. 2016 doi: 10.1038/nrd.2015.17. [DOI] [PubMed] [Google Scholar]
  • 204.Kamba T, McDonald DM. Mechanisms of adverse effects of anti-VEGF therapy for cancer. British journal of cancer. 2007;96(12):1788–95. doi: 10.1038/sj.bjc.6603813. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Simons M, Eichmann A. “On-target” cardiac effects of anticancer drugs: lessons from new biology. J Am Coll Cardiol. 2012;60(7):626–7. doi: 10.1016/j.jacc.2012.01.069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Rivera LB, Bergers G. CANCER. Tumor angiogenesis, from foe to friend. Science. 2015;349(6249):694–5. doi: 10.1126/science.aad0862. [DOI] [PubMed] [Google Scholar]
  • 207.Lee J, Baird A, Eliceiri BP. Vascular normalization in cerebral angiogenesis: friend or foe? Methods Mol Biol. 2014;1135:25–34. doi: 10.1007/978-1-4939-0320-7_3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Marti HJH, Bernaudin M, Bellail A, Schoch H, Euler M, Petit E, Risau W. Hypoxia-Induced Vascular Endothelial Growth Factor Expression Precedes Neovascularization after Cerebral Ischemia. The American journal of pathology. 2000;156(3):965–976. doi: 10.1016/S0002-9440(10)64964-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Hermann DM, Zechariah A. Implications of vascular endothelial growth factor for postischemic neurovascular remodeling. J Cereb Blood Flow Metab. 2009;29(10):1620–43. doi: 10.1038/jcbfm.2009.100. [DOI] [PubMed] [Google Scholar]
  • 210.Greenberg DA, Jin K. Vascular endothelial growth factors (VEGFs) and stroke. Cell Mol Life Sci. 2013;70(10):1753–61. doi: 10.1007/s00018-013-1282-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Zhang ZG, Zhang L, Jiang Q, Zhang R, Davies K, Powers C, Bruggen N, Chopp M. VEGF enhances angiogenesis and promotes blood-brain barrier leakage in the ischemic brain. J Clin Invest. 2000;106(7):829–38. doi: 10.1172/JCI9369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.van Bruggen N, Thibodeaux H, Palmer JT, Lee WP, Fu L, Cairns B, Tumas D, Gerlai R, Williams SP, van Lookeren Campagne M, Ferrara N. VEGF antagonism reduces edema formation and tissue damage after ischemia/reperfusion injury in the mouse brain. J Clin Invest. 1999;104(11):1613–20. doi: 10.1172/JCI8218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Simard JM, Kent TA, Chen M, Tarasov KV, Gerzanich V. Brain oedema in focal ischaemia: molecular pathophysiology and theoretical implications. The Lancet Neurology. 2007;6(3):258–268. doi: 10.1016/S1474-4422(07)70055-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Kimura R, Nakase H, Tamaki R, Sakaki T. Vascular endothelial growth factor antagonist reduces brain edema formation and venous infarction. Stroke. 2005;36(6):1259–63. doi: 10.1161/01.STR.0000165925.20413.14. [DOI] [PubMed] [Google Scholar]
  • 215.Paul R, Zhang ZG, Eliceiri BP, Jiang Q, Boccia AD, Zhang RL, Chopp M, Cheresh DA. Src deficiency or blockade of Src activity in mice provides cerebral protection following stroke. Nat Med. 2001;7(2):222–7. doi: 10.1038/84675. [DOI] [PubMed] [Google Scholar]
  • 216.Liang S, Pong K, Gonzales C, Chen Y, Ling HP, Mark RJ, Boschelli F, Boschelli DH, Ye F, Barrios Sosa AC, Mansour TS, Frost P, Wood A, Pangalos MN, Zaleska MM. Neuroprotective profile of novel SRC kinase inhibitors in rodent models of cerebral ischemia. J Pharmacol Exp Ther. 2009;331(3):827–35. doi: 10.1124/jpet.109.156562. [DOI] [PubMed] [Google Scholar]
  • 217.Roskoski R., Jr Src protein-tyrosine kinase structure, mechanism, and small molecule inhibitors. Pharmacol Res. 2015;94:9–25. doi: 10.1016/j.phrs.2015.01.003. [DOI] [PubMed] [Google Scholar]
  • 218.Adam AP. Regulation of Endothelial Adherens Junctions by Tyrosine Phosphorylation. Mediators Inflamm. 2015;2015:272858. doi: 10.1155/2015/272858. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.McCole DF. Phosphatase regulation of intercellular junctions. Tissue Barriers. 2013;1(5):e26713. doi: 10.4161/tisb.26713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Suzuki J, Shimamura M, Suda H, Wakayama K, Kumagai H, Ikeda Y, Akazawa H, Isobe M, Komuro I, Morishita R. Current therapies and investigational drugs for peripheral arterial disease. Hypertens Res. 2016;39(4):183–91. doi: 10.1038/hr.2015.134. [DOI] [PubMed] [Google Scholar]
  • 221.Giacca M, Zacchigna S. VEGF gene therapy: therapeutic angiogenesis in the clinic and beyond. Gene therapy. 2012;19(6):622–9. doi: 10.1038/gt.2012.17. [DOI] [PubMed] [Google Scholar]
  • 222.Simons M. Angiogenesis, arteriogenesis, and diabetes: paradigm reassessed? J Am Coll Cardiol. 2005;46(5):835–7. doi: 10.1016/j.jacc.2005.06.008. [DOI] [PubMed] [Google Scholar]
  • 223.Simons M, Eichmann A. Molecular controls of arterial morphogenesis. Circ Res. 2015;116(10):1712–24. doi: 10.1161/CIRCRESAHA.116.302953. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Deng Y, Larrivee B, Zhuang ZW, Atri D, Moraes F, Prahst C, Eichmann A, Simons M. Endothelial RAF1/ERK activation regulates arterial morphogenesis. Blood. 2013;121(19):3988–96. S1–9. doi: 10.1182/blood-2012-12-474601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225.Ye W. The Complexity of Translating Anti-angiogenesis Therapy from Basic Science to the Clinic. Developmental cell. 2016;37(2):114–25. doi: 10.1016/j.devcel.2016.03.015. [DOI] [PubMed] [Google Scholar]
  • 226.Jayson GC, Kerbel R, Ellis LM, Harris AL. Antiangiogenic therapy in oncology: current status and future directions. The Lancet. 2016 doi: 10.1016/S0140-6736(15)01088-0. [DOI] [PubMed] [Google Scholar]
  • 227.Ribatti D. Tumor refractoriness to anti-VEGF therapy. Oncotarget. 2016 doi: 10.18632/oncotarget.8694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.van Beijnum JR, Nowak-Sliwinska P, Huijbers EJ, Thijssen VL, Griffioen AW. The great escape; the hallmarks of resistance to antiangiogenic therapy. Pharmacol Rev. 2015;67(2):441–61. doi: 10.1124/pr.114.010215. [DOI] [PubMed] [Google Scholar]
  • 229.Nguyen LK, Matallanas D, Croucher DR, von Kriegsheim A, Kholodenko BN. Signalling by protein phosphatases and drug development: a systems-centred view. FEBS J. 2013;280(2):751–65. doi: 10.1111/j.1742-4658.2012.08522.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.Hoekstra E, Peppelenbosch MP, Fuhler GM. Meeting Report Europhosphatase 2015: Phosphatases as Drug Targets in Cancer. Cancer research. 2016;76(2):193–196. doi: 10.1158/0008-5472.CAN-15-2091. [DOI] [PubMed] [Google Scholar]
  • 231.Jain RK. Normalization of tumor vasculature: an emerging concept in antiangiogenic therapy. Science. 2005;307(5706):58–62. doi: 10.1126/science.1104819. [DOI] [PubMed] [Google Scholar]
  • 232.Arjaans M, Schroder CP, Oosting SF, Dafni U, Kleibeuker JE, de Vries EG. VEGF pathway targeting agents, vessel normalization and tumor drug uptake: from bench to bedside. Oncotarget. 2016;7(16):21247–58. doi: 10.18632/oncotarget.6918. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233.Jain RK. Antiangiogenesis strategies revisited: from starving tumors to alleviating hypoxia. Cancer Cell. 2014;26(5):605–22. doi: 10.1016/j.ccell.2014.10.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Su JC, Mar AC, Wu SH, Tai WT, Chu PY, Wu CY, Tseng LM, Lee TC, Chen KF, Liu CY, Chiu HC, Shiau CW. Disrupting VEGF-A paracrine and autocrine loops by targeting SHP-1 suppresses triple negative breast cancer metastasis. Sci Rep. 2016;6:28888. doi: 10.1038/srep28888. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235.Rincon MY, VandenDriessche T, Chuah MK. Gene therapy for cardiovascular disease: advances in vector development, targeting, and delivery for clinical translation. Cardiovasc Res. 2015;108(1):4–20. doi: 10.1093/cvr/cvv205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236.Amadio M, Govoni S, Pascale A. Targeting VEGF in eye neovascularization: What's new?: A comprehensive review on current therapies and oligonucleotide-based interventions under development. Pharmacol Res. 2016;103:253–69. doi: 10.1016/j.phrs.2015.11.027. [DOI] [PubMed] [Google Scholar]
  • 237.Taimeh Z, Loughran J, Birks EJ, Bolli R. Vascular endothelial growth factor in heart failure. Nat Rev Cardiol. 2013;10(9):519–30. doi: 10.1038/nrcardio.2013.94. [DOI] [PubMed] [Google Scholar]
  • 238.Jaipersad AS, Lip GY, Silverman S, Shantsila E. The role of monocytes in angiogenesis and atherosclerosis. J Am Coll Cardiol. 2014;63(1):1–11. doi: 10.1016/j.jacc.2013.09.019. [DOI] [PubMed] [Google Scholar]
  • 239.Brautigan DL. Protein Ser/Thr phosphatases--the ugly ducklings of cell signalling. FEBS J. 2013;280(2):324–45. doi: 10.1111/j.1742-4658.2012.08609.x. [DOI] [PubMed] [Google Scholar]
  • 240.Soulsby M, Bennett AM. Physiological signaling specificity by protein tyrosine phosphatases. Physiology (Bethesda) 2009;24:281–9. doi: 10.1152/physiol.00017.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241.He R, Zeng LF, He Y, Zhang S, Zhang ZY. Small molecule tools for functional interrogation of protein tyrosine phosphatases. FEBS J. 2013;280(2):731–50. doi: 10.1111/j.1742-4658.2012.08718.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242.Peti W, Page R. Strategies to make protein serine/threonine (PP1, calcineurin) and tyrosine phosphatases (PTP1B) druggable: achieving specificity by targeting substrate and regulatory protein interaction sites. Bioorg Med Chem. 2015;23(12):2781–5. doi: 10.1016/j.bmc.2015.02.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243.Das I, Krzyzosiak A, Schneider K, Wrabetz L, D'Antonio M, Barry N, Sigurdardottir A, Bertolotti A. Preventing proteostasis diseases by selective inhibition of a phosphatase regulatory subunit. Science. 2015;348(6231):239–42. doi: 10.1126/science.aaa4484. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 244.Chen YN, LaMarche MJ, Chan HM, Fekkes P, Garcia-Fortanet J, Acker MG, Antonakos B, Chen CH, Chen Z, Cooke VG, Dobson JR, Deng Z, Fei F, Firestone B, Fodor M, Fridrich C, Gao H, Grunenfelder D, Hao HX, Jacob J, Ho S, Hsiao K, Kang ZB, Karki R, Kato M, Larrow J, La Bonte LR, Lenoir F, Liu G, Liu S, Majumdar D, Meyer MJ, Palermo M, Perez L, Pu M, Price E, Quinn C, Shakya S, Shultz MD, Slisz J, Venkatesan K, Wang P, Warmuth M, Williams S, Yang G, Yuan J, Zhang JH, Zhu P, Ramsey T, Keen NJ, Sellers WR, Stams T, Fortin PD. Allosteric inhibition of SHP2 phosphatase inhibits cancers driven by receptor tyrosine kinases. Nature. 2016;535(7610):148–52. doi: 10.1038/nature18621. [DOI] [PubMed] [Google Scholar]
  • 245.He RJ, Yu ZH, Zhang RY, Zhang ZY. Protein tyrosine phosphatases as potential therapeutic targets. Acta Pharmacol Sin. 2014;35(10):1227–46. doi: 10.1038/aps.2014.80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 246.De Munter S, Kohn M, Bollen M. Challenges and opportunities in the development of protein phosphatase-directed therapeutics. ACS Chem Biol. 2013;8(1):36–45. doi: 10.1021/cb300597g. [DOI] [PubMed] [Google Scholar]
  • 247.Himburg HA, Harris JR, Ito T, Daher P, Russell JL, Quarmyne M, Doan PL, Helms K, Nakamura M, Fixsen E, Herradon G, Reya T, Chao NJ, Harroch S, Chute JP. Pleiotrophin regulates the retention and self-renewal of hematopoietic stem cells in the bone marrow vascular niche. Cell Rep. 2012;2(4):964–75. doi: 10.1016/j.celrep.2012.09.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248.Harroch S, Palmeri M, Rosenbluth J, Custer A, Okigaki M, Shrager P, Blum M, Buxbaum JD, Schlessinger J. No obvious abnormality in mice deficient in receptor protein tyrosine phosphatase beta. Mol Cell Biol. 2000;20(20):7706–15. doi: 10.1128/mcb.20.20.7706-7715.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249.Campochiaro PA, Khanani A, Singer M, Patel S, Boyer D, Dugel P, Kherani S, Withers B, Gambino L, Peters K, Brigell M, Group TS. Enhanced Benefit in Diabetic Macular Edema from AKB-9778 Tie2 Activation Combined with Vascular Endothelial Growth Factor Suppression. Ophthalmology. 2016;123(8):1722–30. doi: 10.1016/j.ophtha.2016.04.025. [DOI] [PubMed] [Google Scholar]
  • 250.Panzhinskiy E, Ren J, Nair S. Pharmacological inhibition of protein tyrosine phosphatase 1B: a promising strategy for the treatment of obesity and type 2 diabetes mellitus. Curr Med Chem. 2013;20(21):2609–25. doi: 10.2174/0929867311320210001. [DOI] [PubMed] [Google Scholar]
  • 251.Zinker BA, Rondinone CM, Trevillyan JM, Gum RJ, Clampit JE, Waring JF, Xie N, Wilcox D, Jacobson P, Frost L, Kroeger PE, Reilly RM, Koterski S, Opgenorth TJ, Ulrich RG, Crosby S, Butler M, Murray SF, McKay RA, Bhanot S, Monia BP, Jirousek MR. PTP1B antisense oligonucleotide lowers PTP1B protein, normalizes blood glucose, and improves insulin sensitivity in diabetic mice. Proc Natl Acad Sci U S A. 2002;99(17):11357–62. doi: 10.1073/pnas.142298199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 252.Han Y, Belley M, Bayly CI, Colucci J, Dufresne C, Giroux A, Lau CK, Leblanc Y, McKay D, Therien M, Wilson MC, Skorey K, Chan CC, Scapin G, Kennedy BP. Discovery of [(3-bromo-7-cyano-2-naphthyl)(difluoro)methyl]phosphonic acid, a potent and orally active small molecule PTP1B inhibitor. Bioorg Med Chem Lett. 2008;18(11):3200–5. doi: 10.1016/j.bmcl.2008.04.064. [DOI] [PubMed] [Google Scholar]
  • 253.Julien SG, Dube N, Read M, Penney J, Paquet M, Han Y, Kennedy BP, Muller WJ, Tremblay ML. Protein tyrosine phosphatase 1B deficiency or inhibition delays ErbB2-induced mammary tumorigenesis and protects from lung metastasis. Nat Genet. 2007;39(3):338–46. doi: 10.1038/ng1963. [DOI] [PubMed] [Google Scholar]
  • 254.Pathak MK, Yi T. Sodium stibogluconate is a potent inhibitor of protein tyrosine phosphatases and augments cytokine responses in hemopoietic cell lines. J Immunol. 2001;167(6):3391–7. doi: 10.4049/jimmunol.167.6.3391. [DOI] [PubMed] [Google Scholar]
  • 255.Kundu S, Fan K, Cao M, Lindner DJ, Zhao ZJ, Borden E, Yi T. Novel SHP-1 inhibitors tyrosine phosphatase inhibitor-1 and analogs with preclinical anti-tumor activities as tolerated oral agents. J Immunol. 2010;184(11):6529–36. doi: 10.4049/jimmunol.0903562. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 256.Mali RS, Ma P, Zeng LF, Martin H, Ramdas B, He Y, Sims E, Nabinger S, Ghosh J, Sharma N, Munugalavadla V, Chatterjee A, Li S, Sandusky G, Craig AW, Bunting KD, Feng GS, Chan RJ, Zhang ZY, Kapur R. Role of SHP2 phosphatase in KIT-induced transformation: identification of SHP2 as a druggable target in diseases involving oncogenic KIT. Blood. 2012;120(13):2669–78. doi: 10.1182/blood-2011-08-375873. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 257.Dal Ponte C, Alchera E, Follenzi A, Imarisio C, Prat M, Albano E, Carini R. Pharmacological postconditioning protects against hepatic ischemia/reperfusion injury. Liver Transpl. 2011;17(4):474–82. doi: 10.1002/lt.22256. [DOI] [PubMed] [Google Scholar]
  • 258.Keyes KT, Xu J, Long B, Zhang C, Hu Z, Ye Y. Pharmacological inhibition of PTEN limits myocardial infarct size and improves left ventricular function postinfarction. Am J Physiol Heart Circ Physiol. 2010;298(4):H1198–208. doi: 10.1152/ajpheart.00915.2009. [DOI] [PubMed] [Google Scholar]
  • 259.Caselli A, Paoli P, Santi A, Mugnaioni C, Toti A, Camici G, Cirri P. Low molecular weight protein tyrosine phosphatase: Multifaceted functions of an evolutionarily conserved enzyme. Biochim Biophys Acta. 2016;1864(10):1339–55. doi: 10.1016/j.bbapap.2016.07.001. [DOI] [PubMed] [Google Scholar]
  • 260.Zhang S, Liu S, Tao R, Wei D, Chen L, Shen W, Yu ZH, Wang L, Jones DR, Dong XC, Zhang ZY. A highly selective and potent PTP-MEG2 inhibitor with therapeutic potential for type 2 diabetes. J Am Chem Soc. 2012;134(43):18116–24. doi: 10.1021/ja308212y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 261.Mattila E, Marttila H, Sahlberg N, Kohonen P, Tahtinen S, Halonen P, Perala M, Ivaska J. Inhibition of receptor tyrosine kinase signalling by small molecule agonist of T-cell protein tyrosine phosphatase. BMC cancer. 2010;10:7. doi: 10.1186/1471-2407-10-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 262.LaMontagne K, Littlewood-Evans A, Schnell C, O'Reilly T, Wyder L, Sanchez T, Probst B, Butler J, Wood A, Liau G, Billy E, Theuer A, Hla T, Wood J. Antagonism of sphingosine-1-phosphate receptors by FTY720 inhibits angiogenesis and tumor vascularization. Cancer research. 2006;66(1):221–31. doi: 10.1158/0008-5472.CAN-05-2001. [DOI] [PubMed] [Google Scholar]
  • 263.Perrotti D, Neviani P. Protein phosphatase 2A: a target for anticancer therapy. The Lancet Oncology. 2013;14(6):e229–e238. doi: 10.1016/S1470-2045(12)70558-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 264.Campbell AM, Zhang ZY. Phosphatase of regenerating liver: a novel target for cancer therapy. Expert Opin Ther Targets. 2014;18(5):555–69. doi: 10.1517/14728222.2014.892926. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 265.Korhonen R, Moilanen E. Mitogen-activated protein kinase phosphatase 1 as an inflammatory factor and drug target. Basic Clin Pharmacol Toxicol. 2014;114(1):24–36. doi: 10.1111/bcpt.12141. [DOI] [PubMed] [Google Scholar]
  • 266.Taylor DM, Moser R, Regulier E, Breuillaud L, Dixon M, Beesen AA, Elliston L, Silva Santos Mde F, Kim J, Jones L, Goldstein DR, Ferrante RJ, Luthi-Carter R. MAP kinase phosphatase 1 (MKP-1/DUSP1) is neuroprotective in Huntington's disease via additive effects of JNK and p38 inhibition. J Neurosci. 2013;33(6):2313–25. doi: 10.1523/JNEUROSCI.4965-11.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 267.Doddareddy MR, Rawling T, Ammit AJ. Targeting mitogen-activated protein kinase phosphatase-1 (MKP-1): structure-based design of MKP-1 inhibitors and upregulators. Curr Med Chem. 2012;19(2):163–73. doi: 10.2174/092986712803414196. [DOI] [PubMed] [Google Scholar]
  • 268.Li CY, Yang LC, Guo K, Wang YP, Li YG. Mitogen-activated protein kinase phosphatase-1: a critical phosphatase manipulating mitogen-activated protein kinase signaling in cardiovascular disease (review) Int J Mol Med. 2015;35(4):1095–102. doi: 10.3892/ijmm.2015.2104. [DOI] [PubMed] [Google Scholar]
  • 269.Fujikawa A, Nagahira A, Sugawara H, Ishii K, Imajo S, Matsumoto M, Kuboyama K, Suzuki R, Tanga N, Noda M, Uchiyama S, Tomoo T, Ogata A, Masumura M, Noda M. Small-molecule inhibition of PTPRZ reduces tumor growth in a rat model of glioblastoma. Sci Rep. 2016;6:20473. doi: 10.1038/srep20473. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES