Abstract
The secreted factor Epidermal growth factor-like protein 7 (EGFL7) is involved in angiogenesis, vasculogenesis, as well as neurogenesis. Importantly, EGFL7 is also implicated in various pathological conditions, including tumor angiogenesis in human cancers. Thus, understanding the mechanisms through which EGFL7 regulates and promotes blood vessel formation is of clear practical importance. One principle means by which EGFL7’s function is investigated is via the expression and purification of the recombinant protein. This mini-review describes three methods used to produce recombinant EGFL7 protein. First, a brief overview of EGFL7’s genetics, structure, and function is provided. This is followed by an examination of the advantages and disadvantages of three common expression systems used in the production of recombinant EGFL7; (i) Escherichia coli (E. coli), (ii) human embryonic kidney (HEK) 293 cells or other mammalian cells, and (iii) a baculovirus-based Sf9 insect cell expression system. Based on the available evidence, we conclude that the baculovirus-based Sf9 insect cell expression currently has the advantages of producing active recombinant EGFL7 in the native conformation with the presence of acceptable posttranslational modifications, while providing sufficient yield and stability for experimental purposes.
1 Introduction
Secreted factors play a crucial role in regulating the formation of new blood vessels. One such secreted factor is Epidermal growth factor-like protein 7 (EGFL7), a protein involved in angiogenesis (Campagnolo et al. 2005), vasculogenesis (Nichol and Stuhlmann 2012), as well as neurogenesis (Barth et al. 2023; Bicker et al. 2017; Schmidt et al. 2009). Importantly, EGFL7 is also implicated in various pathological conditions, including tumor angiogenesis in human cancers (de Oliveira et al. 2023). As such, understanding the mechanisms through which EGFL7 regulates and promotes blood vessel formation is of clear, practical importance. One principle means by which the function of EGFL7 is studied is through the expression and purification of the recombinant protein. The goal of this mini-review is to describe and critique three common methods used to produce recombinant EGFL7 protein. We begin by providing a brief overview of EGFL7’s genetics, structure and function. We then examine the advantages and disadvantages of the three common expression systems used in the production of recombinant EGFL7; (i) Escherichia coli (E. coli), (ii) human embryonic kidney (HEK) 293 cells or other mammalian cells, and (iii) a baculovirus-based Sf9 insect cell expression system.
1.1 EGFL7 gene and protein structure
EGFL7 was first cloned as vascular endothelial station (VE statin) by Soncin and colleagues in 2003 from the 3′ end of vezf1 (Soncin et al. 2003). It is a rather small protein composed of multiple domains involved in cell-extracellular matrix (ECM) interactions (Nichol and Stuhlmann 2012; Schmidt et al. 2007a). The gene egfl7, encoding for the EGFL7 protein, is located on chromosomes 2 (2B) in mice and 9 (9q34.3-qter) in humans, with both the mouse and human egfl7 gene containing 11 exons. Human egfl7 contains two alternative exons, 1a and 1b, with transcription of the EGFL7 protein able to occur from different promoters, giving rise to alternative isoforms. However, these isoforms contain the same open reading frame, beginning at exon 3 and encompassing the rest of the gene (Fish et al. 2008). In addition, binding sites for the transcription factors ETS-related gene and GATA-2 (Bras et al. 2010) are present at the egfl7 gene locus. Of further note, egfl7 harbors the microRNA-126 within intron 7 in both humans and mice. MicroRNA-126 is a small non-coding RNA expressed, among others, in endothelial cells and implicated in modulating gene expression associated with angiogenesis and vascular integrity (Ebrahimi et al. 2014; Meister and Schmidt 2010). The expression of microRNA-126 is regulated by the transcription factor Krüppel-like factor 2 (Harris et al. 2010). The open reading frame of the human egfl7 encodes for a protein of 273 amino acids in length and a molecular weight of 29.6 kDa. The mouse homolog is approximately the same size as the human EGFL7 protein and is 275 amino acids in length with a molecular weight of 29.8 kDa. The proteins display a molecular weight of about 41 kDa upon post-translational modifications.
With regard to protein structure, EGFL7 is a modular assembled protein composed of multiple domains. From N-terminus to C-terminus, EGFL7 consists of a signal peptide, an Emilin-like domain, two EGF-like domain repeats, and a conserved coiled-coil domain at the C-terminus. The presence of the signal peptide indicates that the protein is targeted for secretion into the extracellular space (Soncin et al. 2003). The cysteine-rich Emilin-like domain is found in secreted proteins of the Emilin family and is involved in cell to cell interactions and ECM binding (Doliana et al. 2000). EGF repeats are common in extracellular proteins and are involved in numerous adhesion and ligand/receptors interactions. For example the more N-terminal EGF-like domain is implicated in non-canonical Notch receptor signaling (Schmidt et al. 2009), while an associated arginylglycylaspartic acid (RGD) motif facilitates EGFL7’s ability to bind integrins, in particular αvβ3 and α5β1 (Dudvarski Stanković et al. 2018; Nikolić et al. 2013). The more C-terminal EGF-like domain binds Ca2+ and is proposed to induce a conformational change in the protein (Fitch et al. 2004; Lindsell et al. 1995). The coiled-coil domain at the C-terminus has been suggested to act as a “retention signal”, inhibiting EGFL7 secretion from HEK-239 cells, with the expression of constructs missing the coiled-coil domain facilitated EGFL7 detection in the extracellular conditioned media (Picuric 2007). This finding indicates that post-translational modifications (e.g. proteolytic processing) may be required for secretion in the protein’s native environment. EGFL7 also has both a positively charged N-terminus and a negatively charged C-terminus, with this polarity allowing for oligomer formation in the ECM following secretion. Various ECM proteins including collagen type I and fibronectin facilitate this process (Dudvarski Stankovic et al. 2018; Nikolić et al. 2013; Schmidt et al. 2007b). Finally, the EGFL7 protein undergoes significant post-translational modification prior to secretion from expressing cells. These include post-translational modifications at the Emilin-like domain, N-terminus, and C-Terminus (see Table 1), however information regarding the function of these modifications is limited. No post-translational modifications have been reported for either of the EGF-like domains in EGFL7 in humans or mice. We note, however, that functional post-translational modifications do occur in other proteins containing EGF-like domains, such as in Notch (Wouters et al. 2005) and extracellular matrix proteins (Handford et al. 1991). Thus, when taken together, EGFL7 is a relatively small, modular assembled protein composed of multiple domains that undergoes several post-translation modifications and is ultimately secreted from expressing cells.
Post-translational modifications of EGFL7 in humans and mice.
| Species | Phosphorylation | Ubiquitylation | O-glycosylation | Reference |
|---|---|---|---|---|
| Human | Tyrosine (49, 65) | Lysine (93, 197) | Threonine (54, 55, 190) | Steentoft et al. (2013), Udeshi et al. (2013), Wagner et al. (2011) |
| Mouse | Threonine (44), glycine (27), glutamine (201) | Serine (223, 226) | – | Grimsrud et al. (2012), Sacco et al. (2016), Wiśniewski et al. (2010) |
-
Numbers in brackets reflect the amino acid residue number. Positions 44–93 reflect modifications to the Emilin-like domain. Position 27 reflects a modification at the N-terminus. Positions 190–226 reflect C-terminus modifications.
1.2 EGFL7 function in health and disease
Under physiological conditions, EGFL7 plays important roles in embryonic development, vascular homeostasis, and tissue maintenance. Regarding expression, EGFL7 is predominantly associated with the proliferating endothelium, with high levels observed during embryogenesis and organogenesis. For example, EGFL7 is shown to be involved in vascular tubulogenesis in zebrafish embryos, with knockdown disrupting this process (De Mazière et al. 2008; Parker et al. 2004). In addition, knockdown of EGFL7 in mice disrupts feto-placental vascularization (Lacko et al. 2017). In adult tissues, EGFL7 expression is significantly lower around non-proliferating vessels compared to the developing embryo (Campagnolo et al. 2005; Parker et al. 2004), with increased levels of EGFL7 observed in the proliferating, migrating, or remodeling endothelium in adults, for example, in tissues following vascular injury or during pregnancy (Campagnolo et al. 2005). EGFL7 is shown to influence smooth muscle cell migration for both blood and lymph vessels (Mahamud et al. 2019; Soncin et al. 2003), while loss of EGFL7 in human microvessels suppresses VEGF-A-induced angiogenic sprouting. This leads to an overproduction of endothelial filopodia, reduces collagen IV deposition, and impairs the barrier function of endothelial cells (Usuba et al. 2019). At a molecular level, EGFL7-mediated angiogenesis involves the Notch signaling pathways as a non-canonical ligand (Bicker and Schmidt 2010; Nichol and Stuhlmann 2012), as well as interactions with cell surface integrins, in particular αvβ3 and α5β1 (Chim et al. 2015; Dudvarski Stankovic et al. 2018; Nikolić et al. 2013).
Beyond vascular development and homeostasis, EGFL7 is involved in the regulation of the central nervous system, increasing in cells of the maturing brain and remaining at high levels into old age (Bicker and Schmidt 2010). Moreover, the source of central nervous system EGFL7 were shown to be endothelial cells and neurons. Data suggest that this protein is an endogenous inhibitor of Notch signaling in the adult brain, e.g., regulating neural stem cell proliferation and differentiation. Recent work has demonstrated that the expression of EGFL7 in the subventricular zone functions as an inhibitor of Notch signaling, which decreases the self-renewal capacity and proliferation of neural stem cells, shifting differentiation patterns towards an excess of oligodendrocytes and neurons (Schmidt et al. 2009). Additional work in the nervous system of mice has revealed that loss of EGFL7 induces an accumulation of activated neural stem cells in the subventricular zone, causes quiescence of activated neural stem cells, and promotes neuronal progeny towards differentiation via Dll4-induced Notch signaling at the blood vessel-stem cell interface (Bicker et al. 2017). This alteration in neural stem cells activity ultimately leads to a decrease in inhibitory neurons in the olfactory bulb which causes a profound disruption of neuronal network synchronicity, causing severe physiological deficits in olfaction. Interestingly, Barth and colleagues recently reported that EGFL7 is secreted by neural stem cells, neural precursor cells, and mature neurons in the other significant neurogenic niche of the adult brain, the subgranular zone of the hippocampus (Barth et al. 2023). They demonstrate that loss of EGFL7 correlates with an increased expression of VEGF-D and upregulation of neurogenesis. Behaviorally, mice with EGFL7 ablated in the hippocampus were shown to demonstrate increased spatial learning and memory. Taken together, growing evidence thus indicates that EGFL7 is an important regulator of neurogenesis and nervous system health.
Along with developmental and physiological functions, EGFL7 is also implicated in various human diseases. One of the most well studied disease contexts involving EGFL7 is human cancer. Tumor growth and metastasis often rely on the formation of new blood vessels to provide nutrients and oxygen to the growing tumor mass. Previous work has demonstrated that EGFL7 is involved in this process, facilitating tumor metastasis, proliferation, and angiogenesis (de Oliveira et al. 2023). Specifically, a marked increase in the expression of EGFL7 in the tumor microenvironment has been reported for multiple tumor types, including laryngeal (Wang et al. 2015) ovarian (Oh et al. 2014), breast (Philippin-Lauridant et al. 2013), kidney (Zhai et al. 2019), brain (Huang et al. 2010; Wang et al. 2017), colorectal (Juan et al. 2021), and gastric (Deng et al. 2016) cancers. Moreover, several investigations indicate that this increase in EGFL7 expression may provide information relevant to cancer prognosis (Hansen et al. 2017) and treatment responsiveness (Hansen et al. 2013, 2014). Mechanistically, increased EGFL7 expression is associated with the ERFR-PI3K-Akt (Huang et al. 2014; Juan et al. 2021; Luo et al. 2014) and NOTCH (Bill et al. 2020; Tang et al. 2019; J. Wang et al. 2017; Y. Wang et al. 2022) pathways, facilitating tumor angiogenesis and cancer cell metastasis (Xu et al. 2014). Previous work also indicates that regulation of adhesion molecule expression on the endothelial cell surface via EGFL7 is driven by nuclear factor kappa B (NF-κB) signaling, with EGFL7 blocking the proteasomal degradation of the NF-κB-inhibitor IκBα, subsequently reducing adhesion molecule expression (Pinte et al. 2016). In gastric cancer cells, EGFL7 has further been linked to downregulation of E-cadherin, triggering epithelial-to-mesenchymal-transition and consequentially facilitating metastasis (Luo et al. 2014). In glioma, EGFL7 was shown to be secreted by the tumor blood vessels (but not the tumor cells), which increases endothelial integrin α5β1 surface expression and promoting fibronectin-stimulated angiogenic sprouting (Dudvarski Stankovic et al. 2018).
Growing evidence thus indicates that EGFL7 is a promising target for further studies regarding cancer prognosis and treatment. Indeed, in metastatic colorectal cancer, EGFL7 was shown to be a biomarker in patients receiving combination of bevacizumab and chemotherapy, with a significant relationship between response rates and EGFL7 along with lower EGFL7 expression at the invasive front in responding patients (Hansen et al. 2014). In addition, promising evidence suggests that anti-EGFL7 antibody therapy in combination with anti-VEGF and chemotherapy could enhance stress-induced endothelial cell death, limiting progression and prolonging cancer patient survival (Johnson et al. 2013). We note however that phase II clinical trials utilizing parsatuzumab (an anti-EGFL7-antibody) together with bevacizumab (an anti-VEGF antibody) and chemotherapy in both small cell lung cancer (García-Carbonero et al. 2017) and colorectal cancer (von Pawel et al. 2018) were to date unable to show significant benefits compared to standard care. Importantly however, successful targeting of EGFL7 in cancer may depend on the tumor type. For example, treatment may be more beneficial for patients in other cancer types such as malignant brain tumors given the strong induction of angiogenesis in brain tumors or their growth in an immune-privileged area of the brain (Dudvarski Stankovic et al. 2018; Huang et al. 2010). Thus, when taken together, EGFL7 is a molecule demonstrated to be upregulated in a wide range of cancers, implicated in tumor angiogenesis, and thus representing an important emerging biomarker and target in cancer therapy. Ongoing investigations will benefit from elucidating specific mechanisms across different tumor types in order to improve the efficacy of anti-EGFL7 cancer therapy.
Beyond this role cancer, EGFL7 is also implicated in various other pathologies including stroke (Nikolić et al. 2013), preeclampsia (Lacko et al. 2014) and inflammatory disease (Pinte et al. 2016). In addition, recent work has demonstrated that EGFL7 may be an interesting target for multiple sclerosis (Larochelle et al. 2018; Uphaus et al. 2018). Specifically, Larochelle and colleagues demonstrated that the expression of EGFL7 is increased in the CNS vasculature of patients with multiple sclerosis, as well as in mice with experimental autoimmune encephalomyelitis (Larochelle et al. 2018). Furthermore, perivascular CD4 T lymphocytes were found to be in close proximity to ECM-bound EGFL7 within multiple sclerosis lesions, while activated T lymphocytes in both humans and mice demonstrated an increase in the EGFL7 ligand αvβ3 integrin expression. EGFL7-knockout mice exhibited an earlier onset of experimental autoimmune encephalomyelitis and heightened infiltration of T lymphocytes into the brain and spinal cord parenchyma, with the administration of recombinant EGFL7 ameliorating the experimental autoimmune encephalomyelitis, reducing expression of MCAM, and enhancing blood-brain barrier integrity in the EGFL7-knockout mice. These findings thus suggest a role for EGFL7 in limiting central nervous system immune infiltration, which could potentially prove a promising target for multiple sclerosis therapy.
Therefore, when taken together, the current body of evidence indicates that EGFL7 is a secreted protein, which facilitates a permissive microenvironment for blood vessel formation and growth, as well as regulating cell behavior in the adult central nervous system (see Figure 1). Importantly, based on involvement in tumor metastasis, proliferation, and angiogenesis, as well as various other pathologies, EGFL7 represents an important potential target for clinical therapeutics.

EGFL7: gene and protein structure, as well as function. Egfl7 consists of 11 introns and exons, with two alternative exons, 1a and 1b. The open reading frame begins at exon 3, continuing along the rest of the gene. Further, egfl7 encodes for microRNA-126, which is embedded within intron 7. Binding sites for the transcription factors ETS-related gene (Erg) and GATA2 are present at the 5′ end. The domain structure at the N-terminus of the EGFL7 protein includes a secretion signal peptide (SSP), facilitating cell secretion and an Emilin-like domain (EMI). Located centrally, two epithelial growth factor (EGF)-like domains are present with the more C terminal one relying on Ca2+ binding for proper folding. Among others, EGFL7 interacts with NOTCH receptors, integrins (e.g., αvβ3 and α5β1 motif) and extracellular matrix proteins. Egfl7 is functionally involved in vasculogenesis, angiogenesis and neurogenesis under physiological conditions, while also being implicated in tumor angiogenesis and neuroinflammation in pathological conditions. Created with BioRender.com.
2 Expression systems for the production of recombinant EGFL7
Since the initial identification of EGFL7 as a regulatory protein secreted by endothelial cells (Soncin et al. 2003), various expression systems have been utilized in attempts to produce and purify sufficient quantities of the recombinant protein. Importantly, because EGFL7 is identified in numerous processes in both health and disease, achieving expression and purification of the active recombinant protein has direct importance for both physiologically and clinically relevant research. Therefore, producing biologically active recombinant human and mouse EGFL7 with accurate posttranslational modifications, in the native conformation, and with high yields are essential measures for any recombinant EGFL7 expression system. However, accomplishing this has proven a challenging task, with difficulties including problems with the solution buffer, as well as issues with precipitation and yield of the purified protein (Caetano et al. 2006; Picuric et al. 2009). In the following, we compare and contrast expression systems currently described in the literature and used for the production of recombinant human and mouse EGFL7, specifically the E. coli and the baculovirus-based Sf9 insect cell expression systems (Caetano et al. 2006; Picuric et al. 2009). Additionally, we discuss our and others experience with using HEK293 and other mammalian cells as the expression system. Importantly, up to now there has been no direct experimental comparison between the recombinant EGFL7 expressions systems reported in the literature. This limits our ability to demarcate a single preferred method. Rather, our goal here is to provide a context and review of the most prominent systems to facilitate researchers’ decision making as to which recombinant EGFL7 expression system will best suit their experimental needs.
2.1 E. coli expression system
The use of E. coli bacteria for expressing recombinant human and mouse proteins is a popular choice due to its ease of use, rapid growth, and cost-effectiveness. Indeed, the earliest protocol by Caetano and colleagues describing the expression of recombinant EGFL7 utilized E. coli (Caetano et al. 2006) and a handful of studies have applied this expression system to investigate EGFL7’s function (Delfortrie et al. 2011; Lelièvre et al. 2008; Philippin-Lauridant et al. 2013). However, one drawback of isolating secreted mammalian proteins from bacteria is the difficulty in establishing proper secondary modifications. Another serious issue with using E. coli as the recombinant EGFL7 expression system is the necessity to recover the purified protein from inclusion bodies and the subsequent need to apply buffers allowing for correct post-translational folding (Caetano et al. 2006; Picuric et al. 2009). Specifically, Caetano and colleagues expressed and purified mouse EGFL7 from E. coli in the presence of urea to facilitate protein purification and extraction. However, due to the denaturing effects of urea, the EGFL7 protein required additional refolding. While Caetano and colleagues were able to successfully refold the protein and validate biological activity via the repression of human aortic smooth muscle cell migration induced by platelet-derived growth factor-BB (PDGF-B) (Soncin et al. 2003), they had difficulties establishing a buffer solution which would allow the protein to remain soluble at relatively high concentrations. After attempts with different pH, salts, organic solvents, and detergents, the only successful means of stabilizing folded, active EGFL7 in solution was in the presence of a high concentration D-arginine, in the absence of urea. Unfortunately, this high arginine concentration in the buffer solution creates a high osmotic pressure and has the potential to interfere with nitric oxide formation and signaling, thus making the purified protein most likely unusable for both in vitro and in vivo experimentation (Oess et al. 2006; Picuric et al. 2009). Our experience with recombinant EGFL7 purified from E. coli is that the protein can be applied to coat cell culture dishes and was successfully used in adhesion assays. However, to date, our attempts to apply recombinant EGFL7 from E. coli for the inhibition of PDGF-B-induced cell migration described above have failed, limiting its experimental use. Additionally, it is important to note that post-translational modifications (such as phosphorylation and glycosylation) are absent in this expression system, further limiting application. Thus, while the E. coli expression system may be useful in certain circumstances (e.g., testing cell adhesion or antibody affinity), these limitations ultimately makes this system most likely ill-suited for laboratory studies investigating the broader physiological function of EGFL7, in particular in vivo.
2.2 HEK293 and other mammalian cell expression systems
At face value, the epithelial cell line HEK293 or other mammalian cells would be ideal expression systems for active mouse or human EGFL7 production as they offer correct post-translational secondary modifications. At the least, mammalian cells expression systems come closer to the original protein than the bacterial recombinant protein, particularly due to post-translational modifications more akin to those of the protein’s native state. However, both we and others have experienced practical limitations with mammalian cells, reducing their usefulness at producing sufficient, usable yields of active recombinant EGFL7 for experimental purposes. Specifically, Schmidt and colleagues reported that they were unable to detect the EGFL7 protein in the condition medium of HEK293 and Chinese Hamster Ovary (CHO) cells, despite the cells overexpressed the full-length protein (Schmidt et al. 2007a). However, they were able to detect low levels of EGFL7 when overexpressed in fibroblast cell lines, such as chicken embryonic fibroblasts (CEF) and NIH3T3 cells. Moreover, staining for fibroblast-secreted EGFL7 revealed that the protein is located outside the cell and becomes more and more fibrous over time. They speculate that this is observed in fibroblasts compared to HEK293 or Chinese Hamster Ovary cells because of fibroblast’s role in ECM deposition, with EGFL7 being secreted from these cells and integrated into the ECM (Schmidt et al. 2007b). In other words, this evidence suggests that EGFL7 is not a classic diffusible factor but is rather an ECM component. This is further corroborated by the fact that extracellular EGFL7 significantly overlaps fibronectin when endogenously expressed by human umbilical vein endothelial cells (Schmidt et al. 2007a).
Our experience in using HEK293 cells to produce recombinant EGFL7 reflects these reported issues. Specifically, we were successfully able to detect high levels of recombinant EGFL7 utilizing HEK293 cells. However, the protein spontaneously and instantly precipitated upon secretion, profoundly limiting our ability to obtain a usable yield. Put another way, our experience with mammalian cells suggests that the higher the concertation of the purified protein, the greater the probability of precipitation. Ultimately, consistent issues with protein stability and yield may potentially limit the use of mammalian cells as viable expression vectors for recombinant EGFL7 production. Thus, while mammalian and indeed human cells would be the preferred system for expressing recombinant EGFL7, currently no successful protocol using such cells is described in the literature.
2.3 Baculovirus-based Sf9 insect cell expression
The baculovirus system is widely used for expressing recombinant human proteins (Jarvis 2009). Utilizing this expression system, Picuric and colleagues describe a protocol involving the expression and purification of active recombinant EGFL7 (Picuric et al. 2009). Specifically, Picuric and colleagues expressed human EGFL7 in the Sf9 insect cell line in an insect condition medium, purifying the protein from the cell lysate by immobilized metal affinity chromatography and gel filtration. Of note, EGFL7 was only detectable in the total cell lysate. Urea was not required throughout this process. The conformation of EGFL7 was established by circular dichroism spectrometry, which indicated that the folding of the protein correlated with theoretically calculated values of the native EGFL7 protein. Lastly, they demonstrated the biological activity of the protein via the repression of PDGF-B-induced migration of 10T1/2 myofibroblast cells, in analogy to the assay previously used as biological activity control and applied to smooth muscle cells (Soncin et al. 2003). Importantly, the buffer solution required to maintain this recombinant EGFL7 in solution did not require arginine in this protocol. Thus, Picuric and colleagues described a procedure that overcomes the issues with the arginine-containing buffer solution present with the E. coli expression system. Moreover, they were able to isolate sufficient quantities of recombinant EGFL7 to make this method viable for physiological investigations. Indeed, we and others have successfully used this expression system to investigate the function of EGFL7, both in vitro and in vivo (Larochelle et al. 2018; Salama et al. 2017; Schmidt et al. 2009). For example, we have utilized insect cell-derived recombinant EGFL7 to demonstrate that EGFL7 modulates Notch signaling and affects neural stem cell renewal (Schmidt et al. 2009).
Also of note, the use of the baculovirus-based Sf9 insect cell expression system has also been reported for a related protein within the EGFL family, EGFL6. Specifically, Oberauer and colleagues describe the production of recombinant EGFL6 via the baculovirus-based Sf9 insect cell expression system in a manner similar to recombinant EGFL7, including testing activity via a cell adhesion assay (in this case using human adipocytes). The main findings of this study demonstrates that the EGF-repeats of EGFL6 mediate binding to the cellular surface, as well as enhancing the proliferation, of human adipocytes. The authors also indicate that the EGFL6 detected in the conditioned media of human adipocytes was larger (∼65 kDa) than the recombinant protein and they suggest this may be due to differences in post-translational modifications occurring during recombinant protein production. We note that such differences in post-translational modifications may also be the case for recombinant EGFL7 and this should be taken into account when deciding on a preferred expression system.
Based on this, the baculovirus-based Sf9 insect cell expression system offers several advantages over and above the E. coli and mammalian cell expression systems. First, as described in the published protocol (Picuric et al. 2009), this expression system does not require the buffer solution to contain arginine, reducing concerns regarding disruptive, potentially biologically relevant cross-reactivity. Second, when compared to the E. coli expression system for EGFL7, baculovirus-based Sf9 insect cell expression of EGFL7 has the advantage that post-translational modifications are present, although importantly these are most probably simpler than in mammalian cells. Third, this system allows for sufficient yield and stability for experimental purposes. Disadvantages of the baculovirus-based Sf9 insect cell expression system are first and foremost the simpler post-translational modifications that are most likely present, as these may influence the recombinant proteins function compared to the native form. Second, from a practical perspective the baculovirus-based Sf9 insect cell expression system is more expensive and time consuming than the E. coli expression system. Taken together, depending on experimental goals, the baculovirus-based Sf9 insect cell expression system enables the production of active recombinant EGFL7 in its native conformation, including simple post-translational modifications, along with ensuring adequate yield and stability for experimental applications.
3 General remarks and recommendations
Taken together, the efficacy of the E.coli, mammalian cell, and baculovirus-based Sf9 insect cell recombinant protein expression systems can be summarized based on their ability to produce biologically active, usable, high-yield recombinant EGFL7 protein for experimental purposes. The expression systems discussed in this review are summarized in Figure 2. Based on the current methods papers (Caetano et al. 2006; Picuric et al. 2009), as well as successful application in several studies (Bicker et al. 2017; Dudvarski Stankovic et al. 2018; Larochelle et al. 2018; Nikolić et al. 2013; Schmidt et al. 2009), the baculovirus-based Sf9 insect cell expression system currently appears best suited for allowing sufficient, usable yield of the recombinant protein for in vitro and in vivo studies investigating the physiology of EGFL7.

Three common methods of recombinant EGFL7 expression and purification. Summary of the E.coli, mammalian cell, and baculovirus-based Sf9 insect cell recombinant protein expression systems based on their ability to produce biologically active, usable, high-yield recombinant EGFL7 protein for experimental purposes. Created with BioRender.com.
Regarding recommendations, we suggest that when purchasing recombinant EGFL7 from retailers, attention is paid to the biological activity assay reported. Specifically, adhesion assays, while useful, can also be successfully performed with the denatured EGFL7 protein. Migration assays as described above, which require the presence of the folded, active protein, are therefore the recommended means of determining biological function and thus the presence of active EGFL7 recombinant protein for experimental purposes. We also note here that numerous retailers offer recombinant EGFL7 produced via a wheat germ expression system. Unfortunately, we cannot recommend these, as we have been unable to use these products in investigations into EGFL7 due to a significant contamination from other proteins, limiting experimental use. Regarding storage, from our experience the freezing of recombinant EGFL7 resulted in a significant loss of the active protein, while refrigeration at 4 °C produces precipitation of the protein over time. We thus recommend attention is paid to the length of time the recombinant protein is stored.
4 Conclusions
EGFL7 is a secreted protein involved in numerous biological processes in health and disease and, as such, is an important potential therapeutic target. Critical to the development of EGFL7 translational therapies is a basic understanding of the proteins function in both healthy and pathological conditions. Recombinant EGFL7 protein offers an important means of examining these phenomena in a laboratory setting. Here, we have discussed the use of three common expression systems used to produce biologically active EGFL7 protein with yields allowing for basic and translational research. Ultimately, decisions regarding EGFL7 recombinant production will depend on experimental goals. Based on the above discussion, we recommend the baculovirus-based Sf9 insect cell expression system as the preferred method of producing active recombinant EGFL7 in the native conformation with acceptable posttranslational modifications, while also providing sufficient yield and stability for experimental purposes.
-
Research ethics: Not applicable.
-
Author contributions: BM and MHHS contributed to the conception and writing of this review. The authors have accepted responsibility for the entire content of this manuscript and approved its submission.
-
Competing interests: The authors state no conflict of interest.
-
Research funding: None declared.
-
Data availability: Not applicable.
References
Barth, K., Vasić, V., McDonald, B., Heinig, N., Wagner, M.-C., Schumann, U., Röhlecke, C., Bicker, F., Schumann, L., Radyushkin, K., et al.. (2023). EGFL7 loss correlates with increased VEGF-D expression, upregulating hippocampal adult neurogenesis and improving spatial learning and memory. Cell. Mol. Life Sci. 80: 54, https://doi.org/10.1007/s00018-023-04685-z.Search in Google Scholar PubMed PubMed Central
Bicker, F. and Schmidt, M.H.H. (2010). EGFL7: a new player in homeostasis of the nervous system. Cell Cycle 9: 1263–1269, https://doi.org/10.4161/cc.9.7.11091.Search in Google Scholar PubMed
Bicker, F., Vasic, V., Horta, G., Ortega, F., Nolte, H., Kavyanifar, A., Keller, S., Stankovic, N.D., Harter, P.N., Benedito, R., et al.. (2017). Neurovascular EGFL7 regulates adult neurogenesis in the subventricular zone and thereby affects olfactory perception. Nat. Commun. 8: 1, https://doi.org/10.1038/ncomms15922.Search in Google Scholar PubMed PubMed Central
Bill, M., Pathmanathan, A., Karunasiri, M., Shen, C., Burke, M.H., Ranganathan, P., Papaioannou, D., Zitzer, N.C., Snyder, K., LaRocco, A., et al.. (2020). EGFL7 antagonizes NOTCH signaling and represents a novel therapeutic target in acute myeloid leukemia. Clin. Cancer Res. 26: 669–678, https://doi.org/10.1158/1078-0432.CCR-19-2479.Search in Google Scholar PubMed
Bras, A.L., Samson, C., Trentini, M., Caetano, B., Lelievre, E., Mattot, V., Beermann, F., and Soncin, F. (2010). VE-statin/egfl7 expression in endothelial cells is regulated by a distal enhancer and a proximal promoter under the direct control of Erg and GATA-2. PLoS One 5: e12156, https://doi.org/10.1371/journal.pone.0012156.Search in Google Scholar PubMed PubMed Central
Caetano, B., Drobecq, H., and Soncin, F. (2006). Expression and purification of recombinant vascular endothelial-statin. Protein Expression Purif. 46: 136–142, https://doi.org/10.1016/j.pep.2005.07.029.Search in Google Scholar PubMed
Campagnolo, L., Leahy, A., Chitnis, S., Koschnick, S., Fitch, M.J., Fallon, J.T., Loskutoff, D., Taubman, M.B., and Stuhlmann, H. (2005). EGFL7 is a chemoattractant for endothelial cells and is up-regulated in angiogenesis and arterial injury. Am. J. Pathol. 167: 275–284, https://doi.org/10.1016/S0002-9440(10)62972-0.Search in Google Scholar PubMed PubMed Central
Chim, S.M., Kuek, V., Chow, S.T., Lim, B.S., Tickner, J., Zhao, J., Chung, R., Su, Y.-W., Zhang, G., Erber, W., et al.. (2015). EGFL7 is expressed in bone microenvironment and promotes angiogenesis via ERK, STAT3, and integrin signaling cascades. J. Cell. Physiol. 230: 82–94, https://doi.org/10.1002/jcp.24684.Search in Google Scholar PubMed
Delfortrie, S., Pinte, S., Mattot, V., Samson, C., Villain, G., Caetano, B., Lauridant-Philippin, G., Baranzelli, M.-C., Bonneterre, J., Trottein, F., et al.. (2011). Egfl7 promotes tumor escape from immunity by repressing endothelial cell activation. Cancer Res. 71: 7176–7186, https://doi.org/10.1158/0008-5472.CAN-11-1301.Search in Google Scholar PubMed
De Mazière, A., Parker, L., Van Dijk, S., Ye, W., and Klumperman, J. (2008). Egfl7 knockdown causes defects in the extension and junctional arrangements of endothelial cells during zebrafish vasculogenesis. Dev. Dyn. 237: 580–591, https://doi.org/10.1002/dvdy.21441.Search in Google Scholar PubMed
Deng, Q., Xie, L., and Li, H. (2016). Overexpressed MALAT1 promotes invasion and metastasis of gastric cancer cells via increasing EGFL7 expression. Life Sci. 157: 38–44, https://doi.org/10.1016/j.lfs.2016.05.041.Search in Google Scholar PubMed
de Oliveira, C., Gonçalves, P.G., and Bidinotto, L.T. (2023). Role of EGFL7 in human cancers: a review. J. Cell. Physiol. 238: 1756–1767, https://doi.org/10.1002/jcp.31084.Search in Google Scholar PubMed
Doliana, R., Bot, S., Bonaldo, P., and Colombatti, A. (2000). EMI, a novel cysteine-rich domain of EMILINs and other extracellular proteins, interacts with the gC1q domains and participates in multimerization. FEBS Lett. 484: 164–168, https://doi.org/10.1016/S0014-5793(00)02140-2.Search in Google Scholar
Dudvarski Stanković, N., Bicker, F., Keller, S., Jones, D.T., Harter, P.N., Kienzle, A., Gillmann, C., Arnold, P., Golebiewska, A., Keunen, O., et al.. (2018). EGFL7 enhances surface expression of integrin α5β1 to promote angiogenesis in malignant brain tumors. EMBO Mol. Med. 10: e8420, https://doi.org/10.15252/emmm.201708420.Search in Google Scholar PubMed PubMed Central
Ebrahimi, F., Gopalan, V., Smith, R.A., and Lam, A.K.-Y. (2014). miR-126 in human cancers: clinical roles and current perspectives. Exp. Mol. Pathol. 96: 98–107, https://doi.org/10.1016/j.yexmp.2013.12.004.Search in Google Scholar PubMed
Fish, J.E., Santoro, M.M., Morton, S.U., Yu, S., Yeh, R.-F., Wythe, J.D., Ivey, K.N., Bruneau, B.G., Stainier, D.Y.R., and Srivastava, D. (2008). miR-126 regulates angiogenic signaling and vascular integrity. Dev. Cell 15: 272–284, https://doi.org/10.1016/j.devcel.2008.07.008.Search in Google Scholar PubMed PubMed Central
Fitch, M.J., Campagnolo, L., Kuhnert, F., and Stuhlmann, H. (2004). Egfl7, a novel epidermal growth factor-domain gene expressed in endothelial cells. Dev. Dyn. 230: 316–324, https://doi.org/10.1002/dvdy.20063.Search in Google Scholar PubMed PubMed Central
García-Carbonero, R., van Cutsem, E., Rivera, F., Jassem, J., Gore, I., Tebbutt, N., Braiteh, F., Argiles, G., Wainberg, Z.A., Funke, R., et al.. (2017). Randomized phase II trial of Parsatuzumab (Anti-EGFL7) or placebo in combination with FOLFOX and Bevacizumab for first-line metastatic colorectal cancer. The Oncologist 22: 375-e30, https://doi.org/10.1634/theoncologist.2016-0133.Search in Google Scholar PubMed PubMed Central
Grimsrud, P.A., Carson, J.J., Hebert, A.S., Hubler, S.L., Niemi, N.M., Bailey, D.J., Jochem, A., Stapleton, D.S., Keller, M.P., Westphall, M.S., et al.. (2012). A quantitative map of the liver mitochondrial phosphoproteome reveals posttranslational control of ketogenesis. Cell Metab. 16: 672–683, https://doi.org/10.1016/j.cmet.2012.10.004.Search in Google Scholar PubMed PubMed Central
Handford, P.A., Mayhew, M., Baron, M., Winship, P.R., Campbell, I.D., and Brownlee, G.G. (1991). Key residues involved in calcium-binding motifs in EGF-like domains. Nature 351: 164–167, https://doi.org/10.1038/351164a0.Search in Google Scholar PubMed
Hansen, T.F., Christensen, R.dP., Andersen, R.F., Sørensen, F.B., Johnsson, A., and Jakobsen, A. (2013). MicroRNA-126 and epidermal growth factor-like domain 7–an angiogenic couple of importance in metastatic colorectal cancer. Results from the Nordic ACT trial. Br. J. Cancer 109: 5, https://doi.org/10.1038/bjc.2013.448.Search in Google Scholar PubMed PubMed Central
Hansen, T.F., Nielsen, B.S., Sørensen, F.B., Johnsson, A., and Jakobsen, A. (2014). Epidermal growth factor–like domain 7 predicts response to first-line chemotherapy and Bevacizumab in patients with metastatic colorectal cancer. Mol. Cancer Ther. 13: 2238–2245, https://doi.org/10.1158/1535-7163.MCT-14-0131.Search in Google Scholar PubMed
Hansen, T.F., Andersen, R.F., Olsen, D.A., Sørensen, F.B., and Jakobsen, A. (2017). Prognostic importance of circulating epidermal growth factor-like domain 7 in patients with metastatic colorectal cancer treated with chemotherapy and bevacizumab. Sci. Rep. 7: 1, https://doi.org/10.1038/s41598-017-02538-x.Search in Google Scholar PubMed PubMed Central
Harris, T.A., Yamakuchi, M., Kondo, M., Oettgen, P., and Lowenstein, C.J. (2010). Ets-1 and Ets-2 regulate the expression of microRNA-126 in endothelial cells. Arterioscler., Thromb., Vasc. Biol. 30: 1990–1997, https://doi.org/10.1161/ATVBAHA.110.211706.Search in Google Scholar PubMed PubMed Central
Huang, C., Li, X., Zhou, Y., Luo, Y., Li, C., and Yuan, X. (2010). Expression and clinical significance of EGFL7 in malignant glioma. J. Cancer Res. Clin. Oncol. 136: 1737–1743, https://doi.org/10.1007/s00432-010-0832-9.Search in Google Scholar PubMed
Huang, C., Yuan, X., Wan, Y., Liu, F., Chen, X., Zhan, X., and Li, X. (2014). VE-statin/Egfl7 expression in malignant glioma and its relevant molecular network. Int. J. Clin. Exp. Pathol. 7: 1022–1031.Search in Google Scholar
Jarvis, D.L. (2009) Chapter 14: baculovirus–insect cell expression systems. In: Burgess, R.R., and Deutscher, M.P. (Eds.). Methods in enzymology, Vol. 463. Academic Press, pp. 191–222.10.1016/S0076-6879(09)63014-7Search in Google Scholar PubMed
Johnson, L., Huseni, M., Smyczek, T., Lima, A., Yeung, S., Cheng, J.H., Molina, R., Kan, D., Mazière, A.D., Klumperman, J., et al.. (2013). Anti-EGFL7 antibodies enhance stress-induced endothelial cell death and anti-VEGF efficacy. J. Clin. Invest. 123: 3997–4009, https://doi.org/10.1172/JCI67892.Search in Google Scholar PubMed PubMed Central
Juan, Z., Dake, C., Tanaka, K., and Shuixiang, H. (2021). EGFL7 as a novel therapeutic candidate regulates cell invasion and anoikis in colorectal cancer through PI3K/AKT signaling pathway. Int. J. Clin. Oncol. 26: 1099–1108, https://doi.org/10.1007/s10147-021-01888-x.Search in Google Scholar PubMed
Lacko, L.A., Hurtado, R., Hinds, S., Poulos, M.G., Butler, J.M., and Stuhlmann, H. (2017). Altered feto-placental vascularization, feto-placental malperfusion and fetal growth restriction in mice with Egfl7 loss of function. Development 144: 2469–2479, https://doi.org/10.1242/dev.147025.Search in Google Scholar PubMed PubMed Central
Lacko, L.A., Massimiani, M., Sones, J.L., Hurtado, R., Salvi, S., Ferrazzani, S., Davisson, R.L., Campagnolo, L., and Stuhlmann, H. (2014). Novel expression of EGFL7 in placental trophoblast and endothelial cells and its implication in preeclampsia. Mech. Dev. 133: 163–176, https://doi.org/10.1016/j.mod.2014.04.001.Search in Google Scholar PubMed PubMed Central
Larochelle, C., Uphaus, T., Broux, B., Gowing, E., Paterka, M., Michel, L., Dudvarski Stankovic, N., Bicker, F., Lemaître, F., Prat, A., et al.. (2018). EGFL7 reduces CNS inflammation in mouse. Nat. Commun. 9: 1, https://doi.org/10.1038/s41467-018-03186-z.Search in Google Scholar PubMed PubMed Central
Lelièvre, E., Hinek, A., Lupu, F., Buquet, C., Soncin, F., and Mattot, V. (2008). VE-statin/egfl7 regulates vascular elastogenesis by interacting with lysyl oxidases. EMBO J. 27: 1658–1670, https://doi.org/10.1038/emboj.2008.103.Search in Google Scholar PubMed PubMed Central
Lindsell, C.E., Shawber, C.J., Boulter, J., and Weinmaster, G. (1995). Jagged: a mammalian ligand that activates notch1. Cell 80: 909–917, https://doi.org/10.1016/0092-8674(95)90294-5.Search in Google Scholar PubMed
Luo, B.-H., Xiong, F., Wang, J.-P., Li, J.-H., Zhong, M., Liu, Q.-L., Luo, G.-Q., Yang, X.-J., Xiao, N., Xie, B., et al.. (2014). Epidermal growth factor-like domain-containing protein 7 (EGFL7) enhances EGF receptor−AKT signaling, epithelial−mesenchymal transition, and metastasis of gastric cancer cells. PLoS One 9: e99922, https://doi.org/10.1371/journal.pone.0099922.Search in Google Scholar PubMed PubMed Central
Mahamud, Md. R., Geng, X., Ho, Y.-C., Cha, B., Kim, Y., Ma, J., Chen, L., Myers, G., Camper, S., Mustacich, D., et al.. (2019). GATA2 controls lymphatic endothelial cell junctional integrity and lymphovenous valve morphogenesis through miR-126. Development 146: dev184218, https://doi.org/10.1242/dev.184218.Search in Google Scholar PubMed PubMed Central
Meister, J. and Schmidt, M.H.H. (2010). miR-126 and miR-126*: new players in cancer. Sci. World J. 10: 2090–2100, https://doi.org/10.1100/tsw.2010.198.Search in Google Scholar PubMed PubMed Central
Nichol, D. and Stuhlmann, H. (2012). EGFL7: a unique angiogenic signaling factor in vascular development and disease. Blood 119: 1345–1352, https://doi.org/10.1182/blood-2011-10-322446.Search in Google Scholar PubMed PubMed Central
Nikolić, I., Stanković, N.D., Bicker, F., Meister, J., Braun, H., Awwad, K., Baumgart, J., Simon, K., Thal, S.C., Patra, C., et al.. (2013). EGFL7 ligates αvβ3 integrin to enhance vessel formation. Blood 121: 3041–3050, https://doi.org/10.1182/blood-2011-11-394882.Search in Google Scholar PubMed
Oess, S., Icking, A., Fulton, D., Govers, R., and Müller-Esterl, W. (2006). Subcellular targeting and trafficking of nitric oxide synthases. Biochem. J. 396: 401–409, https://doi.org/10.1042/BJ20060321.Search in Google Scholar PubMed PubMed Central
Oh, J., Park, S.H., Lee, T.S., Oh, H.K., Choi, J.-H., and Choi, Y.S. (2014). High expression of epidermal growth factor-like domain 7 is correlated with poor differentiation and poor prognosis in patients with epithelial ovarian cancer. J. Gynecol. Oncol. 25: 334–341, https://doi.org/10.3802/jgo.2014.25.4.334.Search in Google Scholar PubMed PubMed Central
Parker, L.H., Schmidt, M., Jin, S.-W., Gray, A.M., Beis, D., Pham, T., Frantz, G., Palmieri, S., Hillan, K., Stainier, D.Y.R., et al.. (2004). The endothelial-cell-derived secreted factor Egfl7 regulates vascular tube formation. Nature 428: 6984, https://doi.org/10.1038/nature02416.Search in Google Scholar PubMed
Philippin-Lauridant, G., Baranzelli, M.-C., Samson, C., Fournier, C., Pinte, S., Mattot, V., Bonneterre, J., and Soncin, F. (2013). Expression of Egfl7 correlates with low-grade invasive lesions in human breast cancer. Int. J. Oncol. 42: 1367–1375, https://doi.org/10.3892/ijo.2013.1820.Search in Google Scholar PubMed
Picuric, S. (2007). Biochemical characterization of the novel angiogenic factor EGFL7. Universitätsbibliothek Johann Christian Senckenberg, Available at: http://publikationen.ub.uni-frankfurt.de/frontdoor/index/index/docId/218.Search in Google Scholar
Picuric, S., Friedrich, M., and Oess, S. (2009). Expression and purification of recombinant human EGFL7 protein. Protein Expression Purif. 68: 1–6, https://doi.org/10.1016/j.pep.2009.06.006.Search in Google Scholar PubMed
Pinte, S., Caetano, B., Bras, A.L., Havet, C., Villain, G., Dernayka, R., Duez, C., Mattot, V., and Soncin, F. (2016). Endothelial cell activation is regulated by epidermal growth factor-like domain 7 (Egfl7) during inflammation. J. Biol. Chem. 291: 24017–24028, https://doi.org/10.1074/jbc.M116.731331.Search in Google Scholar PubMed PubMed Central
Sacco, F., Humphrey, S.J., Cox, J., Mischnik, M., Schulte, A., Klabunde, T., Schäfer, M., and Mann, M. (2016). Glucose-regulated and drug-perturbed phosphoproteome reveals molecular mechanisms controlling insulin secretion. Nat. Commun. 7: 1, https://doi.org/10.1038/ncomms13250.Search in Google Scholar PubMed PubMed Central
Salama, Y., Hattori, K., and Heissig, B. (2017). The angiogenic factor Egfl7 alters thymogenesis by activating Flt3 signaling. Biochem. Biophys. Res. Commun. 490: 209–216, https://doi.org/10.1016/j.bbrc.2017.06.023.Search in Google Scholar PubMed
Schmidt, M., De Mazière, A., Smyczek, T., Gray, A., Parker, L., Filvaroff, E., French, D., van Dijk, S., Klumperman, J., and Ye, W. (2007a) The role of Egfl7 in vascular morphogenesis. In: Vascular development. John Wiley & Sons, Ltd, pp. 18–36.10.1002/9780470319413.ch3Search in Google Scholar PubMed
Schmidt, M., Paes, K., De Mazière, A., Smyczek, T., Yang, S., Gray, A., French, D., Kasman, I., Klumperman, J., Rice, D.S., et al.. (2007b). EGFL7 regulates the collective migration of endothelial cells by restricting their spatial distribution. Development 134: 2913–2923, https://doi.org/10.1242/dev.002576.Search in Google Scholar PubMed
Schmidt, M.H.H., Bicker, F., Nikolic, I., Meister, J., Babuke, T., Picuric, S., Müller-Esterl, W., Plate, K.H., and Dikic, I. (2009). Epidermal growth factor-like domain 7 (EGFL7) modulates Notch signalling and affects neural stem cell renewal. Nat. Cell Biol. 11: 7, https://doi.org/10.1038/ncb1896.Search in Google Scholar PubMed
Soncin, F., Mattot, V., Lionneton, F., Spruyt, N., Lepretre, F., Begue, A., and Stehelin, D. (2003). VE-statin, an endothelial repressor of smooth muscle cell migration. EMBO J. 22: 5700–5711, https://doi.org/10.1093/emboj/cdg549.Search in Google Scholar PubMed PubMed Central
Steentoft, C., Vakhrushev, S.Y., Joshi, H.J., Kong, Y., Vester‐Christensen, M.B., Schjoldager, K.T.G., Lavrsen, K., Dabelsteen, S., Pedersen, N.B., Marcos‐Silva, L., et al.. (2013). Precision mapping of the human O‐GalNAc glycoproteome through SimpleCell technology. EMBO J. 32: 1478–1488, https://doi.org/10.1038/emboj.2013.79.Search in Google Scholar PubMed PubMed Central
Tang, H., Xiao, W.R., Liao, Y.Y., Li, L., Xiao, X., Xu, X.P., and Feng, H. (2019). EGFL7 silencing inactivates the Notch signaling pathway; enhancing cell apoptosis and suppressing cell proliferation in human cutaneous melanoma. Neoplasma 66: 187–196, https://doi.org/10.4149/neo_2018_180310N167.Search in Google Scholar PubMed
Udeshi, N.D., Svinkina, T., Mertins, P., Kuhn, E., Mani, D.R., Qiao, J.W., and Carr, S.A. (2013). Refined preparation and use of anti-diglycine remnant (K-ε-GG) antibody enables routine quantification of 10,000s of ubiquitination sites in single proteomics experiments. Mol. Cell. Proteomics 12: 825–831, https://doi.org/10.1074/mcp.O112.027094.Search in Google Scholar PubMed PubMed Central
Uphaus, T., Zipp, F., and Larochelle, C. (2018). EGFL7 – a potential therapeutic target for multiple sclerosis? Expert Opin. Ther. Targets 22: 899–902, https://doi.org/10.1080/14728222.2018.1535595.Search in Google Scholar PubMed
Usuba, R., Pauty, J., Soncin, F., and Matsunaga, Y.T. (2019). EGFL7 regulates sprouting angiogenesis and endothelial integrity in a human blood vessel model. Biomaterials 197: 305–316, https://doi.org/10.1016/j.biomaterials.2019.01.022.Search in Google Scholar PubMed
von Pawel, J., Spigel, D.R., Ervin, T., Losonczy, G., Barlesi, F., Juhász, E., Anderson, M., McCall, B., Wakshull, E., Hegde, P., et al.. (2018). Randomized phase II trial of Parsatuzumab (anti‐EGFL7) or placebo in combination with carboplatin, Paclitaxel, and Bevacizumab for first‐line nonsquamous non‐small cell lung cancer. The Oncologist 23: 654–e58, https://doi.org/10.1634/theoncologist.2017-0690.Search in Google Scholar PubMed PubMed Central
Wagner, S.A., Beli, P., Weinert, B.T., Nielsen, M.L., Cox, J., Mann, M., and Choudhary, C. (2011). A proteome-wide, quantitative survey of in vivo ubiquitylation sites reveals widespread regulatory roles. Mol. Cell. Proteomics 10, https://doi.org/10.1074/mcp.M111.013284.Search in Google Scholar PubMed PubMed Central
Wang, J., Liu, Q., Gao, H., Wan, D., Li, C., Li, Z., and Zhang, Y. (2017). EGFL7 participates in regulating biological behavior of growth hormone–secreting pituitary adenomas via Notch2/DLL3 signaling pathway. Tumor Biol. 39, https://doi.org/10.1177/1010428317706203.Search in Google Scholar PubMed
Wang, X.-X., Yao, X.-B., Qiang, Z.-S., and Zhu, H.-L. (2015). Attenuation of EGFL7 inhibits human laryngocarcinoma cells growth and invasion. Int. J. Clin. Exp. Med. 8: 3141–3155.Search in Google Scholar
Wang, Y., Chen, P., Zhao, M., Cao, H., Zhao, Y., Ji, M., Hou, P., and Chen, M. (2022). EGFL7 drives the evolution of resistance to EGFR inhibitors in lung cancer by activating NOTCH signaling. Cell Death Dis. 13: 910, https://doi.org/10.1038/s41419-022-05354-y.Search in Google Scholar PubMed PubMed Central
Wiśniewski, J.R., Nagaraj, N., Zougman, A., Gnad, F., and Mann, M. (2010). Brain phosphoproteome obtained by a FASP-based method reveals plasma membrane protein topology. J. Proteome Res. 9: 3280–3289, https://doi.org/10.1021/pr1002214.Search in Google Scholar PubMed
Wouters, M.A., Rigoutsos, I., Chu, C.K., Feng, L.L., Sparrow, D.B., and Dunwoodie, S.L. (2005). Evolution of distinct EGF domains with specific functions. Protein Sci. 14: 1091–1103, https://doi.org/10.1110/ps.041207005.Search in Google Scholar PubMed PubMed Central
Xu, H.-F., Chen, L., Liu, X.-D., Zhan, Y.-H., Zhang, H.-H., Li, Q., and Wu, B. (2014). Targeting EGFL7 expression through RNA interference suppresses renal cell carcinoma growth by inhibiting angiogenesis. Asian Pac. J. Cancer Prev. 15: 3045–3050, https://doi.org/10.7314/APJCP.2014.15.7.3045.Search in Google Scholar
Zhai, W., Zhu, R., Ma, J., Gong, D., Zhang, H., Zhang, J., Chen, Y., Huang, Y., Zheng, J., and Xue, W. (2019). A positive feed-forward loop between LncRNA-URRCC and EGFL7/P-AKT/FOXO3 signaling promotes proliferation and metastasis of clear cell renal cell carcinoma. Mol. Cancer 18: 81, https://doi.org/10.1186/s12943-019-0998-y.Search in Google Scholar PubMed PubMed Central
© 2024 Walter de Gruyter GmbH, Berlin/Boston
Articles in the same Issue
- Frontmatter
- Review
- Structure, function, and recombinant production of EGFL7
- Research Articles/Short Communications
- Protein Structure and Function
- Zinc and copper effect mechanical cell adhesion properties of the amyloid precursor protein
- The BCL11A transcription factor stimulates the enzymatic activities of the OGG1 DNA glycosylase
- Cell Biology and Signaling
- Bioprospecting hydroxylated chalcones in in vitro model of ischemia-reoxygenation and probing NOX4 interactions via molecular docking
- Carnosic acid prevents heat stress-induced oxidative damage by regulating heat-shock proteins and apoptotic proteins in mouse testis
- Novel Techniques
- Simultaneous spectral illumination of microplates for high-throughput optogenetics and photobiology
- A platform for the early selection of non-competitive antibody-fragments from yeast surface display libraries
Articles in the same Issue
- Frontmatter
- Review
- Structure, function, and recombinant production of EGFL7
- Research Articles/Short Communications
- Protein Structure and Function
- Zinc and copper effect mechanical cell adhesion properties of the amyloid precursor protein
- The BCL11A transcription factor stimulates the enzymatic activities of the OGG1 DNA glycosylase
- Cell Biology and Signaling
- Bioprospecting hydroxylated chalcones in in vitro model of ischemia-reoxygenation and probing NOX4 interactions via molecular docking
- Carnosic acid prevents heat stress-induced oxidative damage by regulating heat-shock proteins and apoptotic proteins in mouse testis
- Novel Techniques
- Simultaneous spectral illumination of microplates for high-throughput optogenetics and photobiology
- A platform for the early selection of non-competitive antibody-fragments from yeast surface display libraries