Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2019 May 11.
Published in final edited form as: Biochim Biophys Acta Mol Basis Dis. 2017 Mar 30;1863(6):1326–1335. doi: 10.1016/j.bbadis.2017.03.021

Endothelial cell calpain as a critical modulator of angiogenesis

Yixuan Zhang 1, Norika Mengchia Liu 1, Yongchen Wang 1, Ji Youn Youn 1, Hua Cai 1,*
PMCID: PMC6511073  NIHMSID: NIHMS1024738  PMID: 28366876

Abstract

Calpains are a family of calcium-dependent non-lysosomal cysteine proteases. In particular, calpains residing in the endothelial cells play important roles in angiogenesis. It has been shown that calpain activity can be increased in endothelial cells by growth factors, primarily vascular endothelial growth factor (VEGF). VEGF/VEGFR2 induces calpain 2 dependent activation of PI3K/AMPK/Akt/eNOS pathway, and consequent nitric oxide production and physiological angiogenesis. Under pathological conditions such as tumor angiogenesis, endothelial calpains can be activated by hypoxia. This review focuses on the molecular regulatory mechanisms of calpain activation, and the newly identified mechanistic roles and downstream signaling events of calpains in physiological angiogenesis, and in the conditions of pathological tumor angiogenesis and diabetic wound healing, as well as retinopathy and atherosclerosis that are also associated with an increase in calpain activity. Further discussed include the differential strategies of modulating angiogenesis through manipulating calpain expression/activity in different pathological settings. Targeted limitation of angiogenesis in cancer and targeted promotion of angiogenesis in diabetic wound healing via modulations of calpains and calpain-dependent signaling mechanisms are of significant translational potential. Emerging strategies of tissue-specific targeting, environment-dependent targeting, and genome-targeted editing may turn out to be effective regimens for targeted manipulation of angiogenesis through calpain pathways, for differential treatments including both attenuation of tumor angiogenesis and potentiation of diabetic angiogenesis.

Keywords: Angiogenesis, Calpain, Endothelial cell, VEGF, eNOS, Tumor angiogenesis, Diabetic wound healing, Atherosclerosis, Retinopathy, Shear stress

1. Introduction of calpains

Calpains belong to a family of calcium-dependent cysteine proteases. Calpains proteolytically process substrates to transform their structures to modulate activities. Calpains have been present during evolution. Isoforms of calpains exist in various organisms, ranging from fish to human [13]. To date, 15 isoforms have been identified in humans [4]. Calpain 3 is found in skeletal muscle, while calpain 6 in placenta and embryonic muscles, calpain 8 and calpain 9 in gastrointestinal tract, calpain 11 in testis and calpain 12 in hair follicles. Other calpain isoforms are ubiquitously expressed. Among all of the calpain isoforms, calpain 1 and calpain 2 have been widely studied. Of note, calpain 1 and calpain 2 are the only known calpain isoforms expressed in the endothelial cells [5]. Calpain 1 (p-calpain) and calpain 2 (m-calpain) were originally named by μmol or mmol of calcium concentration that is required for their activation in vitro. The requirements for half maximal activities are at approximately 3–50 μM and 400–800 μM for calpain 1 and calpain 2 respectively [6,7]. However, additional evidences indicate that both calpains could be activated at physiological calcium concentrations in vivo at 0.4–0.5 μM (calpain 1) and 10 μM (calpain 2) respectively, although the activation mechanisms may involve specific cellular components, such as membrane ezrin and phosphoinositides [814].

The calpain proteolytic system includes large subunits, small subunit and the endogenous calpain inhibitor calpastatin. Calpain 1 (CAPN1) and calpain 2 (CAPN2) are heterodimeric proteins, consisting of a large 78–80 kDa catalytic subunit and a common 29 kDa small regulatory subunit (CAPN4). Calpastatin has been shown to inhibit both calpain 1 and calpain 2. Upon activation, calpains cleave a broad spectrum of substrates that are involved in several fundamental cellular processes, including cell proliferation, cell migration and cytoskeletal remodeling. Several groups have shown that calpains are required for cell proliferation. Inhibition of calpains leads to reduced proliferation in different cell types, such as pulmonary artery smooth muscle cells, HeLa and WI-38 human fibroblasts [1517]. It is also reported that calpain-mediated substrate proteolysis is indispensable for cell migration and cytoskeletal remodeling [18,19]. Attenuated cell migration was observed in cells treated with calpain inhibitors [20,21]. Calpain-mediated proteolysis of focal adhesion proteins, such as paxillin, focal adhesion kinase and talin, mediates focal adhesion turnover during migration [2224]. Mouse embryonic fibroblasts obtained from CAPN4 knockout mice displayed repressed migration and impaired organization of cytoskeleton [25]. In this review, we will discuss the mechanistic roles and downstream signaling events of endothelial calpains in both physiological and pathological angiogenesis.

2. The structure of calpains

The catalytic large subunit of calpains comprises four domains of I-IV, while the small subunit has two domains V-VI. The N-terminus of domain I undergoes autolysis upon exposure to Ca2+ [26,27]. Domain II contains the catalytic sequence of Cys-His-Asn. The crystallographic structure of calpain reveals that in the absence of calcium the active site is disrupted by the separation of the two subdomains, domains Ha and lib [8,28]. Upon Ca2+ binding, however, lib and Ha form a functional catalytic center following a subtle conformational change [8,28]. The crystal structure of calpain 2 similarly reveals that during the conformational change, some residues in lib come into close contact with the membrane [8]. Domain III can bind to phospholipid in Ca2+ - dependent manner, and may play an important role in Ca2+ -dependent membrane translocation of calpains [29,30]. We have shown that Ezrin is required for the membrane specific activation of calpain [14]. Domain IV and domain VI each contains five EF-hand motifs. It has been shown that the first to third EF-hands bind Ca2 + and the fifth EF-hand motif is involved in heterodimer formation of the large subunit and the small subunit. Domain IV and domain VI are also involved in binding to calpastatin [31]. Domain V of the regulatory small subunit contains hydrophobic Gly-rich sequence [32,33]. Deletion of domain V reduced membrane binding and localization [34]. Most of this domain is cut off by autolysis, indicating no involvement in protease activity.

3. Activation of calpains in endothelial cells

Activation of calpains in endothelial cells is induced by growth factors, primarily vascular endothelial growth factor (VEGF). It has been reported that VEGF activates calpains in endothelial cells, which can be attenuated by calpain inhibitors calpeptin, ALLN and calpastatin [14,35,36]. The detailed mechanisms of calpain activation by VEGF are discussed below. The combination of VEGF and basic fibroblast growth factor (FGF) was reported to significantly elevate calpain activity in endothelial cells, as well as the cleavage of calpain substrate vimentin [37]. Another growth factor that has been shown to activate calpain 2 in endothelial cells is epidermal growth factor (EGF). It has been recently reported that EGF induces calpain 2 membrane translocation and activation in endothelial cells through phosphatidylinositol 4,5-bisphosphate [38]. Nonetheless, VEGF seems to be the primarily characterized activator of calpain signaling in endothelial cells.

Under physiological conditions, activation of calpain by VEGF results in endothelial nitric oxide synthase (eNOS) phosphorylation to produce nitric oxide (NO), which is a potent angiogenic activator [14]. It is known that VEGF activates eNOS/NO through PI3K/Akt [3941]. Our group previously identified a novel role of calpain 2 in mediating VEGF-induced PI3K/AMPK/Akt activation, and subsequent eNOS phosphorylation and NO production in endothelial cells [14]. Application of calpain inhibitors (ALLN or Calpeptin), or siRNA targeting calpain 2, abolished VEGF-induced activation of PI3K/AMPK/Akt/eNOS/NO pathway. However, calpain 1 is not involved in this response [14]. Taken together, these data indicate a unique role of calpain 2 in mediating VEGF-induced angiogenesis. Other reports also support the specific role of calpain 2 in mediating VEGF downstream signaling [42]. It has been shown that VEGF selectively activates calpain 2 in endothelial cells [42]. The activation of calpain 2 by VEGF may be caused by preferably increased calpain 2 protein expression. Su et al. demonstrated that the protein abundance of calpain 2 was increased 2 h post-VEGF stimulation [43]. However, calpain 1 expression was unchanged, suggesting that elevated calpain activity is attributed to increased calpain 2 protein content [43,44]. Our group has further revealed a calpain dependent negative feedback loop to inhibit VEGFR2 overactivation [45]. Cleavage and activation of protein tyrosine phosphatase type 1 (PTP1B) by calpain de-phosphorylates VEGFR2 [45]. Altered expression or activity of PTP1B and/or calpain modulated VEGF-induced angiogenesis and diabetic wound healing in mice [45]. Illustrations on a role of endothelial calpain 2 in VEGF-induced angiogenesis are presented in Fig. 1. Roles of calpain pathways in physiological and pathological angiogenesis are further discussed in the Section 4 below.

Fig. 1.

Fig. 1.

VEGF activates angiogenesis through calpain 2. VEGF/VEGFR2 activates calpain 2, not calpain 1, in endothelial cells. Calpain 2 activation Involves binding to Ezrln and phosphoinositides on the cell membrane. VEGFR2 Is negatively regulated by a calpain 2/PTP1B feedback loop. Calpain 2 mediates VEGF-lnduced activation of PI3K/AMPK/Akt/eNOS pathway and consequent nitric oxide (NO) production and angiogenesis.

Moreover, calpains are activated when endothelial cells are exposed to hypoxia [4649]. Zhang et al. reported that short exposure of endothelial cells to hypoxia (1–12 h) up-regulated mRNA level and activity of calpain [47]. Treatments of actinomycin D (a transcriptional inhibitor) and ALLN prevented hypoxia-induced calpain 2 transcription and activation, implicating a potential regulation at transcriptional level [47]. Though VEGF is transcriptionally regulated by hypoxia inducible factor-la (HIF-la) during hypoxia, the transcription regulation of calpain mRNA may be facilitated directly by hypoxia in short exposure. For longer exposure to hypoxia, calpain activation could be mediated by HIF-la-induced VEGF or Na+/H+ exchanger-1 (NHE1) expression [49]. Similar to VEGF, NHE1 is also a HIF-1α target. Both mRNA and protein levels of NHE1 are up-regulated by hypoxia (48 h) or adenovirus-delivered HIF-la (24 h) [49,50]. Knockdown of NHE1 selectively inhibited HIF-la-induced calpain 2 protein expression and activation, and subsequent angiogenesis [49]. These pathways are included in Fig. 2 for mechanisms of tumor angiogenesis involving calpain activation.

Fig. 2.

Fig. 2.

Calpain central signaling pathways in tumor angiogenesis. Under hypoxic conditions, von Hippel-Lindau (VHL)-mediated HIF-1α degradation is inhibited, leading to accumulation of HIF-1α in both tumor cells and endothelial cells. VEGF expression is transcriptionally up-regulated by HIF-1α. Hypoxia also increases calpain activity in tumor cells. The cleavage of filamin A, a substrate of calpain. facilitates HIF-1α nuclear translocation and enhances HIF-1α transactivation, resulting in up-regulation of VEGF expression and secretion. On the other hand, hypoxia induces calpain 2 activation in endothelial cells. During acute exposure, hypoxia directly upregulates calpain 2 mRNA, while prolonged exposure to hypoxia increases calpain 2 activity through HIF-la-induced VEGF and NHE1 expression. Other growth factors secreted by tumor cells, such as EGF and bFGF, are able to activate endothelial calpain 2 as well. Down-regulation of calpastatin causes calpain 1-dependent SOCS3 cleavage and VEGF-C production through STAT3 phosphorylation. Activation of endothelial calpain 2 leads to enhanced tumor angiogenesis. Another calpain-dependent up-regulation of angiogenesis involves VASH1. Hypoxia inhibits VEGF-induced expression of VASH1 in endothelial cells. Secreted VASH1 undergoes cleavage by tumor calpain. which impairs its anti-angiogenic function.

In addition to selective induction of calpain 2 expression, calpain activation is subjected to spatial regulations. We have shown that in response to VEGF stimulation, only the membrane-localized calpain is activated and inhibited by calpain inhibitors (ALLN and calpeptin). Furthermore, we also found that VEGF induces direct binding of calpain and ezrin [14]. Membrane colocalization of these two proteins can be detected within 10 min of VEGF stimulation. The activity of calpain in the membrane fraction was greatly decreased by ezrin siRNA [14]. These results suggest that ezrin mediates calpain membrane translocalization and activation as a novel mechanism of regulating calpain in endothelial cells. Another mechanism of calpain membrane activation involves phosphoinositides. Studies from different groups have shown that phosphoinositides, components of the membrane, interact with calpain 1 and 2 through calpain domain III [10,30,51]. Sphingosine 1-phosphate (SIP) induces calpain membrane translocation and activation without changing calpain expression level [52]. More evidence of calpain membrane localization was shown by the study of the crystal structure of calpain 2 [8].

Furthermore, calpain activation in endothelial cells involves calpastatin, the endogenous calpain inhibitor. It was reported that calpastatin binds to calpains in response to calcium to prevent calpains from activation [53]. Calpastatin has four inhibitory domains and each one of those is able to bind to one calpain molecule. Each inhibitory domain contains three subdomains, A, B and C. Subdomains A and C bind to calpain domain IV and domain VI, respectively [31,54]. A recent study has shown that calpastatin binds to the active cleft of calpains by looping out around the active site, so that calpastatin blocks the active site without being cleaved [54]. Treatment of endothelial cells with VEGF potently down-regulated calpastatin expression, leading to increased calpain activity [44]. The combined regulations of calpastatin and calpain enable maximal activation of calpain and its downstream signaling in response to VEGF.

Therefore, calpain 2 activation in endothelial cells can be induced through at least three different mechanisms: 1) selective up-regulation of calpain 2 protein abundance and activity [14,43,55]; 2) spatial regulation of calpain subcellular localization to promote membrane translocation and activation [14,56]; and 3) down-regulation of calpain inhibitor calpastatin [44]. The mechanistic pathways of calpain 2 activation by VEGF to mediate angiogenesis are summarized in Fig. 1. Calpain 2 inhibition suppresses multiple features of angiogenesis, such as proliferation [56,57], cell migration [42,55,58] and tube formation [42,43,55,58].

4. Role of calpains in angiogenesis

4.1. General introduction of angiogenesis

Angiogenesis is the process of new vessel formation from existing vessels. Angiogenesis requires a highly coordinated series of events that involve the interactions among endothelial cells, extra-cellular matrix and growth factors. Key steps of angiogenesis include endothelial cell proliferation, migration and tube formation (the formation of capillary-like tube structures). Basic physiology of angiogenesis has been discussed in several recent reviews [5961]. VEGF is one of the main initiators of angiogenesis. We and others have shown that VEGF exposure induces calpain 2 dependent activation of PI3K/AMPK/Akt/ eNOS pathway and NO production in endothelial cells through VEGFR2 [14,45]. Blocking PI3K/AMPK/Akt by pharmacological kinase inhibitors, or genetic/pharmacological abrogation of calpain 2 activation, completely attenuated VEGF-induced NO production and angiogenesis [14,6264]. Additionally, angiogenesis is promoted in conditions where ERK is activated. Studies have shown that ERK is activated in response to VEGF/VEGFR2 [65]. Abrogated ERK signaling inhibits VEGF-induced angiogenic response of endothelial cells [66].

4.2. Role of calpains in angiogenesis under physiological conditions

Under physiological conditions, angiogenesis mostly occurs during embryonic development, when it requires adequate vasculature for organ development. Normal angiogenesis in the adulthood usually happens during repair processes, such as wound healing. It has been reported that deletion of the small subunit (CAPN4) of calpain results in elimination of both calpain 1 and calpain 2 activities and embryonic death at day Ell.5, implicating a role of calpain in embryonic development [67]. Arthur et al. reported that CAPN4 knockout embryos had reduced yolk sac vasculature at E10.5. The endothelial cells lining the atria were found rounding up at E10.5, and eventually delaminated at El 1.5, indicating the indispensable role of calpains in normal vascular development [67]. Using global and conditional knockout strategies, Takano and colleagues have shown that calpain 2 deficiencies caused embryonic death on day 15, due to placental dysfunction-induced apoptosis [68]. Interestingly, calpain 1 knockout alone appeared mostly harmless (viable and fertile) [69]. Double knockout of calpain 1 and calpain 2 potentiated embryonic lethality (— 3 days earlier) however, suggesting that the two isoforms may additively modulate some common developmental pathways in vivo, at least during the developmental stage [68].

Calpains play important roles in wound healing, a physiological process highly dependent on angiogenesis. It has been shown that transgenic mice globally over-expressing calpastatin displayed a striking delay in skin wound healing through impaired angiogenesis, indicating that calpain activity is required for wound healing [70]. We have recently demonstrated that application of calpain inhibitor ALLN to wound bed significantly delayed VEGF-induced wound healing in diabetic mice [45]. However, wound healing is not a process that only involves enhanced angiogenesis. In the late stage of wound healing, a mechanism of vessel dissociation/regression is activated. Interestingly, vessel regression is mediated by calpain 1. Bodnar and colleagues reported that activation of calpain 1 in endothelial cells leads to vessel regression and reduced angiogenesis during middle and late stage of wound healing [71,72]. Calpain 1-induced dissociation of newly formed vessels is a mechanism to maintain a regular vascular network near the wounded area by eliminating excessive vessels. Moreover, a separate study reported that cells with calpain 1 siRNA transfection showed stabilized tube formation at late stages (6–24 h) [58]. Of note, the role of calpain 1 during later stage of angiogenesis is established in experiments of exposing endothelial cells to CXCL10 (IP-10), a known ligand for the resolving stage of the wound, rather than VEGF. A potential role of calpain 1 in VEGF-induced wound healing remains to be investigated.

4.3. Role of calpains in angiogenesis under pathological conditions

4.3.1. Calpains in tumor angiogenesis

The constant growth of solid tumor requires large quantity of oxygen and nutrients. Therefore, tumor cells have developed the ability to establish their own blood supply by the induction of angiogenesis. By secreting angiogenic factors, tumor cells induce angiogenesis around them to achieve nutrient delivery and removal of metabolic wastes through the newly formed vessels. One of the most important angiogenic factors produced by tumor cells is VEGF. Blockade of VEGF or VEGF receptors dramatically inhibited tumor growth via abrogation of angiogenesis [73]. As discussed in the previous section, VEGF activates calpain 2 in endothelial cells [14,42,43,55]. We and others have demonstrated that inhibition of calpain 2 (with siRNA or calpain inhibitors) abolished VEGF-induced endothelial NO production and angiogenesis [14,43,45,74]. Moreover, the fast growth of tumor cell results in hypoxia, exposure to which upregulates calpain expression and activity in endothelial cells [4649,75,76].

Under hypoxic conditions, calpains are involved in the crosstalk between tumor cells and endothelial cells. VEGF is secreted by tumor cells to influence endothelial cells. Interestingly, calpain in tumor cells serves as a newly identified regulator of the HIF-la/VEGF pathway [77]. Zheng et al. have shown that hypoxia induces filamin A proteolysis by calpain in melanoma cells, which in turn facilitates HIF-1α nuclear translocation. Calpeptin inhibition of calpain however attenuated HIF-1α nuclear accumulation and transactivation [77]. It is known that VEGF is transcriptionally up-regulated by HIF-1α. Overexpression of filamin A increased recruitment of HIF-1α to VEGF promoter and augmented angiogenesis in a tumor xenograft model [77]. Another crosstalk involves vasohibin-1 (VASH1), an angiogenesis inhibitor generated by VEGF-stimulated endothelial cells [78,79]. It was reported that hypoxia inhibits VEGF-induced VASH1 expression [79]. Interestingly, tumor cells inactivate VASH1 through calpain-dependent cleavage of VASH1 in EC-tumor cell co-culture experiments [78]. These calpain central tumor cell-EC crosstalks to facilitate angiogenesis are summarized in Fig. 2.

To study the involvement of endothelial calpains in tumor angiogenesis, Miyazaki et al. collected tumors and nearby normal tissues from patients with malignant astrocytoma, colon and lung adenocarcinomas. Immunostaining of calpastatin illustrated that the expression level of calpastatin was significantly reduced in endothelial cells of tumor vessels compared to nearby normal vessels [44]. They further generated transgenic mice that harbor endothelial cell specific transgene of calpastatin. In these animals, tumor angiogenesis was attenuated in a Lewis lung carcinoma allograft transplantation model. It turns out that calpastatin inhibits VEGF-C production through calpain/ SOCS3/STAT3 [44]. These results provide more evidences that regulation of calpain pathway is important in tumor angiogenesis (summarized in Fig. 2).

4.3.2. Calpains in diabetic wound healing

Diabetic food ulcer, one of the most common complications of diabetes mellitus, affects 15% of people with diabetes [80]. It is also the leading cause of amputations among diabetic patients [81,82]. One of the major causes of diabetic foot ulcer is impaired wound healing, which is characterized by impaired growth factor production and defective angiogenesis [8285]. It is known that the expression of growth factors and their receptors (such as VEGF, platelet-derived growth factor (PDGF)/PDGF receptor, FGF/FGF receptor, EGF) is up-regulated in the wounded area during physiological repair [83]. However, the up-regulation of growth factors and their receptors is absent in diabetic wounds. The synthesis of PDGF and FGF like growth factors was down-regulated in STZ-induced diabetic wound [83,86]. The abundance of VEGF was decreased in wounded area throughout wound healing process in the db/db mice [87]. Galkowska et al. have compared the expression of growth factors and their receptors in the margin skin tissue of diabetic foot ulcers with normal non-diabetic foot skin by immunohistochemistry [84]. They reported down-regulation of PDGF receptor and TGF-pl. There was a similar trend for VEGF/ VEGFR2, EGF and FGF [84]. In accordance with the impaired production of growth factors, angiogenesis process is also delayed in diabetic wound [8890]. To promote wound healing through angiogenesis, multiple strategies have been tested. Administration of growth factors has been shown to be effective [45,91]. Others and we have shown that VEGF activates PI3K/AMPK/Akt/eNOS cascade to induce NO production, which in turn mediates angiogenesis and wound healing [14,92,93]. Topical application of VEGF accelerated skin wound healing in both type 1 and type 2 diabetic models [45,80]. This strategy and other approaches discussed below to improve diabetic wound healing are summarized in Fig. 3.

Fig. 3.

Fig. 3.

Mechanisms of impaired angiogenesis in diabetic wound healing. Normal wound healing can be facilitated by VEGF via VEGFR2/calpain 2/eNOS/angiogenesis axis (details see Fig. 1). In diabetes, impaired wound healing is characterized by reduced growth factors expression (VEGF, FGF, and EGF), and increased PTP1B activity to inhibit VEGFR2. To promote angiogenesis in diabetic wound healing, the following strategies can be employed; a) supplementation of VEGF; b) genetic or pharmacological approaches to inhibit PTP1B to increase VEGFR2-dependent angiogenic signaling; or c) genetic or pharmacological approaches to activate calpain.

To further accelerate wound healing, an alternative approach is to facilitate angiogenesis by targeting downstream pathways of growth factors. We have recently shown that VEGF signaling is regulated by a calpain/PTPlB/VEGFR2 feedback mechanism, which can be employed to enhance VEGF signaling to facilitate therapeutic angiogenesis [45]. PTP1B activity is up-regulated in diabetic wound to constrain VEGFR2, while application of PTP1B inhibitor accelerated VEGF-dependent diabetic wound healing [45]. To test the effect of calpain in wound healing, we directly applied plasmids that encode human calpain cDNA to the wound bed. As expected, we found that calpain overexpression in wound bed accelerated VEGF-induced wound healing in STZ-induced diabetic mice [45]. Data from calpastatin transgenic mice confirmed the importance of calpain in angiogenesis and wound healing. Global calpastatin transgenic mice showed impaired angiogenesis and a striking delay in wound healing [70]. Moreover, calpain 2 is indispensable to lymphangiogenesis [58], enhancement of which contributed to accelerated diabetic wound healing [80,94,95].

Studies examining roles and mechanisms of calpains in pathophysiological angiogenesis, including models and approaches employed, are summarized in Table 1.

Table 1.

Roles and mechanisms of calpains in pathophysiological angiogenesis.

Pathophysiologic al conditions In vitro model In vivo model Way of intervention Experimental results Signaling pathways References
Wound healing BAECs. STZ-induced diabetic mice. In vitro and in vivo: application of calpain inhibitor ALLN. In vitro and in vivo: delayed VEGF-induced wound closure. VEGF/calpain/angiogenesis/wound healing [45]
Monolayer wound scratch assay. Skin wound healing assay. In vivo: over expression of calpain 1 and calpain 2. In vivo: accelerated VEGF-induced wound closure.
Wound healing N/A Global calpastatin transgenic mice;
Skin wound healing assay.
In vivo: calpastatin transgene. In vivo: calpastatin Tg mice have impaired cell proliferation, and decreased blood vessel density. Calpastatin Tg mice showed delayed angiogenesis and wound healing. Calpastatin/calpain/angiogenesis/wound healing [70]
Wound healing Human microvascular cell line (HMEC-1); N/A In vitro: IP-10 treatment. In vitro: leads to β3 integrin cleavage and tube dissociation. IP-10/CXCR3/calpain 1/(β3 integrin/tube dissociation (non-VEGF). [72]
Tube formation. In vitro: application of CI-1 (a pan-calpain inhibitor), CI-IV (calpain 2 inhibitor), BAPTA/AM. In vitro: IP-10-induced tube dissociation was prevented by BAPTA/AM and CI-1, but not CI-IV.
In vitro: IP-10-induced β3 integrin cleavage was blocked by BAPTA/AM and CI-1.
Tumor angiogenesis Melanoma cells. Tumor xenograft model for angiogenesis. In vitro and in vivo: over expression of filamin A (calpain substrate). In vitro: overexpression of filamin A increased HIF-1α transactivation and VEGF expression (mRNA and secretion) in hypoxia. Calpain/filamin A/HIF-1α activity/VEGF/angiogenesis 77
Exposure to hypoxia.
In vivo: increased HIF-1α activity (expected increased VEGF production) and angiogenesis in a tumor xenograft model derived from filamin A overexpression.
In vitro: application of calpain inhibitor calpeptin. In vitro: calpeptin inhibited filamin A cleavage, and HIF-lα nuclear accumulation and transactivation in melanoma cells (VEGF is transcriptionally regulated by HIF-lα).
Tumor angiogenesis Human aortic endothelial cells. Endothelial-specific calpastatin Tg mice. In vitro: calpastatin siRNA transfection and IL-6 treatment. In vitro: calpastatin siRNA facilitated calpain 1-induced cleavage of SOCS3, leading to VEGF-C production through amplified IL-6-driven STAT3 signals. Calpastatin/calpain 1/SOCS3/STAT3 (induced by IL-6)/VEGF-C/angiogenesis [44]
Tube formation, cell mobility, proliferation. Lewis lung carcinoma xenograft model.
Calpastatin siRNA also improved angiogenic responses (tube formation, cell mobility, proliferation) in the presence of IL-6.
In vivo: calpastatin transgene. In vivo: calpastatin Tg declined phosphorylation of STAT3 in tumors, up-regulated SOCS3 protein expression, and decreased VEGF-C production in the tumor neovessels. Calpastatin Tg suppressed tumor angiogenesis.
Atherosclerosis HMEC-1 N/A In vitro: treatments of oxLDL and calpain inhibitor calpeptin. In vitro: calpeptin inhibited oxLDL-induced calpain activation, Bid cleavage, cytochrome C release, caspase-3 activation, and cytotoxicity. OxLDL/calcium/calpain/Bid/cytochrome C/caspase-3/apoptosis [97]
Atherosclerosis HMEC-1 N/A In vitro: treatments of oxLDL and calpain inhibitor PD 151746. (PD 151746 has more than 20-fold selectivity for calpain 1 over calpain 2.) In vitro: PD 151746 decreased oxLDL-induced cytotoxicity. OxLDL-induced Bid cleavage was prevented by PD 151746. OxLDL/calpain/Bid/apoptosis [99]
Atherosclerosis HUVEC LDLR-KO or apoE-KO mice, fed with high cholesterol food (HCD). In vitro: transfection of calpain 2 siRNA and treatments of LPC and calpeptin. In vitro: LPC-induced VE-cadherin cleavage and hyperpermeability can be blocked by calpeptin or calpain 2 siRNA. LPC-induced dissociation of beta-catenin/VE-cadherin was attenuated by calpeptin. Calpain 2 directly cleaves VE-cadherin. HCD or LPC/calpain 2/VE-cadher in/beta-catenin/endothelial adherence junctions/atherosclerosis [100]
In vivo: administration of calpain inhibitors (ALLM and calpeptin). In vivo: long-term administration of calpain inhibitors attenuated VE-cadherin disorganization and atherosclerotic lesion development.
In vivo transfection of calpain 2 siRNA. In vivo: prevented disorganization of VE-cadherin and proatherogenic hyperpermeability in aortic endothelial cells.
Ischemic retinopathy Human dermal microvascular endothelial cells (HDMEC). Endothelial-specific calpastatin Tg mice. In vitro: calpastatin siRNA. IL-6 treatment. In vitro: calpastatin siRNA increased IL-6-induced tube formation. calpastatin/calpain 1/SOCS3/STAT3/VEGF-C/angiogenesis [44]
Tube formation. Oxygen-induced retinopathy (OIR) model. In vivo: endothelial-specific calpastatin transgene. In vivo: calpastatin Tg abolished OIR-induced pathological angiogenesis (vascular tufts formation), up-regulated SOCS3 expression and STAT3 phosphorylation, and diminished VEGF-C expression (mRNA).
Ischemic retinopathy Human retinal microvascular endothelial cells (RMVEC). Oxygen-induced retinopathy model. In vitro: application of calpain inhibitors: MDL28170, calpastatin. In vitro: hypoxia induces calpain activation in RMVECs, along with disrupted actin cytoskeleton. Modest inhibition of calpain by MDL28170 or calpastatin improved actin cytoskeletal organization and capillary morphogenesis in a 3D collagen matrix assay. Calpain/cell cytoskeleton (organization and alignment of actin cables)/angiogenesis [46]
Capillary-like tube growth in 3D collagen matrix.
Exposure to hypoxia. In vivo: application of MDL28170, PD150606, ALLN. In vivo: moderate calpain inhibition (by MDL28170, PD150606 or ALLN) improved organization and alignment of actin cytoskeleton and promoted formation of functional neovasculature.

5. Other mechanisms that regulate calpain activity in endothelial cells

5.1. Oxidized LDL, phospholipids and mechanical stress

In addition to growth factors and hypoxia, calpains can be activated in endothelial cells by oxidized low-density lipoprotein (oxLDL), phospholipids or mechanical stress. OxLDL induces calpain activation in endothelial cells through elevated calcium concentration [96,97]. In human atherosclerotic plaques, calpain activity was identified in apoptotic cells [98]. In addition, oxLDL-induced endothelial cell apoptosis can be partially blocked by PD151746, a calpain 1 inhibitor [99]. These results suggest that at least calpain 1 activation is involved in ox-LDL-induced endothelial apoptosis, which has long been considered as an important regulator of the initiation and progression of atherosclerotic lesions. On the other hand, calpain 2 has been reported to regulate endothelial adherence junctions (through cleavage of VE-cadherin) and promote atherogenesis. Recently, Miyazaki et al. have shown that endothelial calpain 2 up-regulation is associated with more severe atherosclerotic lesions in patients [100]. In LDL receptor (LDLR) deficient mice, high cholesterol fed animals showed elevated expression of endothelial calpain 2. Administration of calpain inhibitors (calpeptin, ALLM) on the other hand significantly limited lesion formation in high cholesterol diet-treated LDLR knockout or Apolipoprotein E deficient mice [100]. Similarly, lysophosphatidylcholine, the major lipid constituent of oxLDL, can activate calpain 2 and induce cleavage of VE-cadherin [100,101]. Taken together, activation of calpain 1 and calpain 2 by oxLDL contributes to atherogenesis. Though a causal role of angiogenesis in atherosclerosis has not been established, there is strong evidence that the development of plaques is associated with neovascularization within the plaque [102105].

It has also been reported that calpain 2 is activated by physiological shear stress through elevated intracellular calcium [106,107]. Calpeptin impaired shear stress-induced focal adhesion polarization and cell alignment under shear conditions [107]. Shear stress, combined with sphingosine 1-phosphate, induces calpain membrane translocation and MTP-MMP1 activation in endothelial cells [52]. On the other hand, Mayazaki et al. have demonstrated that calpain 2 antagonizes RhoA overactivation and endothelial barrier dysfunction in response to disturbed flow [108]. Overall calpain 2 activation under different flow patterns seem to exert protective signaling via differential mechanisms.

5.2. Ischemic retinopathy

Calpains have been reported to contribute to hypoxia-derived retina cell death in ischemic retinopathy [109,110]. Using an oxygen-induced retinopathy (OIR) model, Hoang et al. demonstrated that ischemic hypoxia activates calpain in retinal endothelial cells and disrupts actin cytoskeleton in human retinal microvascular endothelial cells [46]. Moderate inhibition (30–35%) of calpain activity results in formation of functional neovasculature. Further investigation revealed that calpain inhibitor MDL28170 and calpastatin peptide improved organization and alignment of actin cytoskeleton both in vitro and in vivo [46]. The role of calpains in ischemic retinopathy was also reported by another independent group using the OIR model [44]. Endothelial specific transgene of calpastatin abolished OIR-induced vascular tufts through down-regulation of calpain 1-dependent cleavage of SOCS3, followed by inhibition of STAT3 and VEGF-C expression [44]. These results suggest that targeting calpain system (either by application of calpain inhibitor or overexpression of calpastatin) is beneficial for the normalization of angiogenesis in ischemic retinopathy.

6. Differential strategies targeting endothelial calpain and calpain-dependent pathways for different pathophysiological conditions

Calpain 1 and calpain 2 are ubiquitously expressed in human tissues. For therapeutic purposes related to angiogenesis, endothelial targeted strategies of modulating calpain activity may be beneficial. Restrain of calpain activity inhibits tumor growth and atherogenesis, and promotes formation of normal vasculature in ischemic retinopathy. On the other hand, activation of calpain is aimed to treat diabetic wound healing and protect endothelial function under disturbed flow. Therefore, differential strategies of regulating endothelial calpain/ angiogenesis are necessary for therapeutic control of different pathophysiological conditions.

One of the options enabling endothelial cell-targeted delivery involves generating genetically modified vectors. Plasmids or viral vectors-based gene targeting could specifically modulate gene expression in certain type of cells, with the help of tissue-specific promoters/ enhancers, and sequences with physiological sensing elements (such as those responsive to hypoxia, shear stress) [111,112]. Localized intraspinal injection into rats of lentiviral vector encoding calpain 1 resulted in sustained ability of proteolysis (activated NF-κBp65 after IκB being cleaved by calpain 1) up to 7 weeks after injection [113,114]. It has been reported that hypoxia response element from the promoters of multiple genes (such as erythropoietin, phosphoglycerate kinase-1, and VEGF) has been used as a hypoxia sensitive enhancer to promote transcription of the delivered gene of interest [111,115117]. Very recently, lentiviruses that are pseudotyped with endothelial-specific envelopes recognizing endothelial cell surface marker CD105 have been shown to be efficient and specific in endothelial delivery after systemic injection [118]. Tumor endothelial cells were specifically targeted upon intratumoral injection in mice carrying a vascularized human tumor xenograft [118]. Most recently, the emergence of CRISPR/Cas9 gene editing system further increases the possibility of in vivo genome editing [119121]. Yin et al. recently reported that systemic delivery of nanoparticle-conjugated Cas9 mRNA and sgRNA/HDR sequences by AAV provided efficient genome editing and less off-target editing in diseased mice [122].

Especially, tumor cells are known to have lower pH (6.2–6.9) than normal tissues (7.3–7.4) due to the glycolysis under anaerobic environment [123]. Modified nanoparticles can be used as the vehicle of delivery which is capable of pH-dependent drug release and has been shown to effectively inhibit xenografted tumor growth [124126]. Another approach to achieve local regulation is to apply treatment in situ. In a diabetic wound healing model, topical administration of recombinant VEGF and calpain plasmids accelerated wound closure [45]. This local application is transient and restrained only to wounded area. This approach is especially applicable to dermal treatment.

In conclusion, calpain 1 and calpain 2 play important roles in VEGF-induced angiogenesis in endothelial cells. Blockade of calpain 2 impairs, while activation of calpain 2 promotes, VEGF-induced angiogenesis. In animal models, inhibition of calpain 1 and calpain 2 by calpastatin transgene specifically expressed in endothelial cells efficiently attenuated tumor angiogenesis and tumor growth [44]. Abrogation of calpain activity reduced progression of atherosclerosis and ischemic retinopathy [46,100,127]. On the other hand, activation of calpain pathway accelerated diabetic wound healing, and is dispensable to protect from disturb flow-induced endothelial cell barrier dysfunction [45,108]. Therefore, disease-specific modulations of calpain, combined with endothelial cell-targeted delivery techniques, may prove to be promising strategies for the treatments of pathophysiological conditions associated with angiogenesis.

Acknowledgements

This work was supported by National Institute of Health National Heart, Lung and Blood Institute (NHLBI) Grants HL077440 (HC), HL088975 (HC), HL108701 (HC, DGH), and HL119968 (HC), an American Heart Association Established Investigator Award (EIA) 12EIA8990025 (HC) and an AHA Postdoctoral Fellowship Award 14POST20380995 (YXZ).

Footnotes

Conflict of interest declaration

None of the authors of this manuscript have a conflict of interest.

Transparency document

The http://dx.doi.Org/10.1016/j.bbadis.2017.03.021 associated with this article can be found, in online version.

References

  • [1].Salmeron C, Garcia de la serrana D, Jimenez-Amilburu V, Fontanillas R, Navarro I, Johnston IA, Gutierrez J, Capilla E, Characterisation and expression of calpain family members in relation to nutritional status, diet composition and flesh texture in gilthead sea bream (Sparus aurata), PLoS One 8 (2013) e75349. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [2].Rawlings ND, Bacterial calpains and the evolution of the calpain (C2) family of peptidases, Biol. Direct 10 (2015) 66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [3].Macqueen DJ, Wilcox AH, Characterization of the definitive classical calpain family of vertebrates using phylogenetic, evolutionary and expression analyses, Open Biol. 4 (2014) 130219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [4].Sorimachi H, Ono Y, Regulation and physiological roles of the calpain system in muscular disorders, Cardiovasc. Res. 96 (2012) 11–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [5].Fujitani K, Kambayashi J, Sakon M, Ohmi SI, Kawashima S, Yukawa M, Yano Y, Miyoshi H, Ikeda M, Shinoki N, Monden M, Identification of mu-,m-calpains and calpastatin and capture of mu-calpain activation in endothelial cells, J. Cell. Biochem. 66 (1997) 197–209. [DOI] [PubMed] [Google Scholar]
  • [6].Goll DE, Thompson VF, Li H, Wei W, Cong J, The calpain system, Physiol. Rev. 83 (2003) 731–801. [DOI] [PubMed] [Google Scholar]
  • [7].Cong J, Goll DE, Peterson AM, Kapprell HP, The role of autolysis in activity of the Ca2+ -dependent proteinases (mu-calpain and m-calpain), J. Biol. Chem. 264 (1989) 10096–10103. [PubMed] [Google Scholar]
  • [8].Strobl S, Fernandez-Catalan C, Braun M, Huber R, Masumoto H, Nakagawa K, Irie A, Sorimachi H, Bourenkow G, Bartunik H, Suzuki K, Bode W, The crystal structure of calcium-free human m-calpain suggests an electrostatic switch mechanism for activation by calcium, Proc. Natl. Acad. Sci. U. S. A. 97 (2000) 588–592. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [9].Inserte J, Hernando V, Garcia-Dorado D, Contribution of calpains to myocardial ischaemia/reperfusion injury, Cardiovasc. Res. 96 (2012) 23–31. [DOI] [PubMed] [Google Scholar]
  • [10].Saido TC, Shibata M, Takenawa T, Murofushi H, Suzuki K, Positive regulation of mu-calpain action by polyphosphoinositides, J. Biol. Chem. 267 (1992) 24585–24590. [PubMed] [Google Scholar]
  • [11].Matsumura Y, Saeki E, Otsu K, Morita T, Takeda H, Kuzuya T, Hori M, Kusuoka H, Intracellular calcium level required for calpain activation in a single myocardial cell, J. Mol. Cell. Cardiol. 33 (2001) 1133–1142. [DOI] [PubMed] [Google Scholar]
  • [12].Melloni E, Michetti M, Salamino F, Pontremoli S, Molecular and functional properties of a calpain activator protein specific for mu-isoforms, J. Biol. Chem. 273 (1998) 12827–12831. [DOI] [PubMed] [Google Scholar]
  • [13].Melloni E, Averna M, Salamino F, Sparatore B, Minafra R, Pontremoli S, Acyl-CoA-binding protein is a potent m-calpain activator, J. Biol. Chem. 275 (2000) 82–86. [DOI] [PubMed] [Google Scholar]
  • [14].Youn JY, Wang T, Cai H, An ezrin/calpain/PI3K/AMPK/eNOSs1179 signaling cascade mediating VEGF-dependent endothelial nitric oxide production, Circ. Res.104 (2009) 50–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [15].Abeyrathna P, Kovacs L, Han W, Su Y, Calpain-2 activates Akt via TGF-beta1-mTORC2 pathway in pulmonary artery smooth muscle cells, Am. J. Physiol. Cell Physiol. 311 (2016) C24–C34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [16].Zhang W, Lane RD, Mellgren RL, The major calpain isozymes are long-lived proteins. Design of an antisense strategy for calpain depletion in cultured cells, J. Biol. Chem. 271 (1996) 18825–18830. [DOI] [PubMed] [Google Scholar]
  • [17].Zhang W, Lu Q, Xie ZJ, Mellgren RL, Inhibition of the growth of WI-38 fibroblasts by benzyloxycarbonyl-Leu-Leu-Tyr diazomethyl ketone: evidence that cleavage of p53 by a calpain-like protease is necessary for G1 to S-phase transition, Oncogene 14 (1997) 255–263. [DOI] [PubMed] [Google Scholar]
  • [18].Perrin BJ, Amann KJ, Huttenlocher A, Proteolysis of cortactin by calpain regulates membrane protrusion during cell migration, Mol. Biol. Cell 17 (2006) 239–250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [19].Jeong SY, Martchenko M, Cohen SN, Calpain-dependent cytoskeletal rearrangement exploited for anthrax toxin endocytosis, Proc. Natl. Acad. Sci. U. S. A. 110 (2013) E4007–E4015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [20].Saraiva N, Prole DL, Carrara G, Johnson BF, Taylor CW, Parsons M, Smith GL, hGAAP promotes cell adhesion and migration via the stimulation of store-operated Ca2+ entry and calpain 2, J. Cell Biol. 202 (2013) 699–713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [21].Svensson L, McDowall A, Giles KM, Stanley P, Feske S, Hogg N, Calpain 2 controls turnover of LFA-1 adhesions on migrating T lymphocytes, PLoS One 5 (2010) e15090. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [22].Cortesio CL, Boateng LR, Piazza TM, Bennin DA, Huttenlocher A, Calpain-mediated proteolysis of paxillin negatively regulates focal adhesion dynamics and cell migration, J. Biol. Chem. 286 (2011) 9998–10006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [23].Chan KT, Bennin DA, Huttenlocher A, Regulation of adhesion dynamics by calpain-mediated proteolysis of focal adhesion kinase (FAK), J. Biol. Chem. 285 (2010) 11418–11426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [24].Franco SJ, Rodgers MA, Perrin BJ, Han J, Bennin DA, Critchley DR, Huttenlocher A, Calpain-mediated proteolysis of talin regulates adhesion dynamics, Nat. Cell Biol. 6 (2004) 977–983. [DOI] [PubMed] [Google Scholar]
  • [25].Dourdin N, Bhatt AK, Dutt P, Greer PA, Arthur JS, Elce JS, Huttenlocher A, Reduced cell migration and disruption of the actin cytoskeleton in calpain-deficient embryonic fibroblasts, J. Biol. Chem. 276 (2001) 48382–48388. [DOI] [PubMed] [Google Scholar]
  • [26].Goll DE, Thompson VF, Taylor RG, Zalewska T, Is calpain activity regulated by membranes and autolysis or by calcium and calpastatin? Bioessays 14 (1992) 549–556. [DOI] [PubMed] [Google Scholar]
  • [27].Huang Y, Wang KK, The calpain family and human disease, Trends Mol. Med. 7 (2001) 355–362. [DOI] [PubMed] [Google Scholar]
  • [28].Moldoveanu T, Hosfield CM, Lim D, Elce JS, Jia Z, Davies PL, A Ca(2+0) switch aligns the active site of calpain, Cell 108 (2002) 649–660. [DOI] [PubMed] [Google Scholar]
  • [29].Carragher NO, Calpain inhibition: a therapeutic strategy targeting multiple disease states, Curr. Pharm. Des. 12 (2006) 615–638. [DOI] [PubMed] [Google Scholar]
  • [30].Shao H, Chou J, Baty CJ, Burke NA, Watkins SC, Stolz DB, Wells A, Spatial localization of m-calpain to the plasma membrane by phosphoinositide biphosphate binding during epidermal growth factor receptor-mediated activation, Mol. Cell. Biol. 26 (2006) 5481–5496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [31].Wendt A, Thompson VF, Goll DE, Interaction of calpastatin with calpain: a review, Biol. Chem. 385 (2004) 465–472. [DOI] [PubMed] [Google Scholar]
  • [32].Hosfield CM, Elce JS, Davies PL, Jia Z, Crystal structure of calpain reveals the structural basis for Ca(2+)-dependent protease activity and a novel mode of enzyme activation, EMBO J. 18 (1999) 6880–6889. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [33].Jia Z, Hosfield CM, Davies PL, Elce JS, Crystal structure of calpain and insights into Ca(2+)-dependent activation, Methods Mol. Biol. 172 (2002) 51–67. [DOI] [PubMed] [Google Scholar]
  • [34].Fernandez-Montalvan A, Assfalg-Machleidt I, Pfeiler D, Fritz H, Jochum M, Machleidt W, Mu-calpain binds to lipid bilayers via the exposed hydrophobic surface of its Ca2+-activated conformation, Biol. Chem. 387 (2006) 617–627. [DOI] [PubMed] [Google Scholar]
  • [35].Maki M, Bagci H, Hamaguchi K, Ueda M, Murachi T, Hatanaka M, Inhibition of calpain by a synthetic oligopeptide corresponding to an exon of the human calpastatin gene, J. Biol. Chem. 264 (1989) 18866–18869. [PubMed] [Google Scholar]
  • [36].Hoang MV, Nagy JA, Fox JE, Senger DR, Moderation of calpain activity promotes neovascular integration and lumen formation during VEGF-induced pathological angiogenesis, PLoS One 5 (2010) e13612. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [37].Kwak HI, Kang H, Dave JM, Mendoza EA, Su SC, Maxwell SA, Bayless KJ, Calpain-mediated vimentin cleavage occurs upstream of MT1-MMP membrane translocation to facilitate endothelial sprout initiation, Angiogenesis 15 (2012)287–303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [38].Leloup L, Shao HS, Bae YH, Deasy B, Stolz D, Roy P, Wells A, M-calpain activation is regulated by its membrane localization and by its binding to phosphatidylinositol 4,5-bisphosphate, J. Biol. Chem. 285 (2010) 33549–33566. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [39].Dimmeler S, Fleming I, Fisslthaler B, Hermann C, Busse R, Zeiher AM, Activation of nitric oxide synthase in endothelial cells by Akt-dependent phosphorylation, Nature 399 (1999) 601–605. [DOI] [PubMed] [Google Scholar]
  • [40].Fulton D, Gratton JP, McCabe TJ, Fontana J, Fujio Y, Walsh K, Franke TF, Papapetropoulos A, Sessa WC, Regulation of endothelium-derived nitric oxide production by the protein kinase Akt, Nature 399 (1999) 597–601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [41].Papapetropoulos A, Garcia-Cardena G, Madri JA, Sessa WC, Nitric oxide production contributes to the angiogenic properties of vascular endothelial growth factor in human endothelial cells, J. Clin. Invest. 100 (1997) 3131–3139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [42].Bodnar RJ, Yates CC, Wells A, IP-10 blocks vascular endothelial growth factor-induced endothelial cell motility and tube formation via inhibition of calpain, Circ. Res. 98 (2006) 617–625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [43].Su Y, Cui Z, Li Z, Block ER, Calpain-2 regulation of VEGF-mediated angiogenesis, FASEB J. 20 (2006) 1443–1451. [DOI] [PubMed] [Google Scholar]
  • [44].Miyazaki T, Taketomi Y, Saito Y, Hosono T, Lei XF, Kim-Kaneyama JR, Arata S, Takahashi H, Murakami M, Miyazaki A, Calpastatin counteracts pathological angiogenesis by inhibiting suppressor of cytokine signaling 3 degradation in vascular endothelial cells, Circ. Res. 116 (2015) 1170–1181. [DOI] [PubMed] [Google Scholar]
  • [45].Zhang Y, Li Q, Youn JY, Cai H, Protein phosphotyrosine phosphatase 1B (PTP1B) in calpain-dependent feedback regulation of vascular endothelial growth factor receptor (VEGFR2) in endothelial cells: implications in VEGF-dependent angiogenesis and diabetic wound healing, J. Biol. Chem. 292 (2017) 407–416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [46].Hoang MV, Smith LE, Senger DR, Calpain inhibitors reduce retinal hypoxia in ischemic retinopathy by improving neovascular architecture and functional perfusion, Biochim. Biophys. Acta 2011 (1812) 549–557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [47].Zhang J, Patel JM, Block ER, Hypoxia-specific upregulation of calpain activity and gene expression in pulmonary artery endothelial cells, Am. J. Phys. 275 (1998) L461–L468. [DOI] [PubMed] [Google Scholar]
  • [48].Su Y, Block ER, Acute hypoxia increases intracellular L-arginine content in cultured porcine pulmonary artery endothelial cells, J. Cell. Physiol. 167 (1996)349–353. [DOI] [PubMed] [Google Scholar]
  • [49].Mo XG, Chen QW, Li XS, Zheng MM, Ke DZ, Deng W, Li GQ, Jiang J, Wu ZQ, Wang L, Wang P, Yang Y, Cao GY, Suppression of NHE1 by small interfering RNA inhibits HIF-1alpha-induced angiogenesis in vitro via modulation of calpain activity, Microvasc. Res. 81 (2011) 160–168. [DOI] [PubMed] [Google Scholar]
  • [50].Shimoda LA, Fallon M, Pisarcik S, Wang J, Semenza GL, HIF-1 regulates hypoxic induction of NHE1 expression and alkalinization of intracellular pH in pulmonary arterial myocytes, Am. J. Physiol. Lung Cell. Mol. Physiol. 291 (2006) L941–L949. [DOI] [PubMed] [Google Scholar]
  • [51].Tompa P, Emori Y, Sorimachi H, Suzuki K, Friedrich P, Domain III of calpain is a Ca2+-regulated phospholipid-binding domain, Biochem. Biophys. Res. Commun. 280 (2001) 1333–1339. [DOI] [PubMed] [Google Scholar]
  • [52].Kang H, Kwak HI, Kaunas R, Bayless KJ, Fluid shear stress and sphingosine 1-phosphate activate calpain to promote membrane type 1 matrix metalloproteinase (MT1-MMP) membrane translocation and endothelial invasion into three-dimensional collagen matrices, J. Biol. Chem. 286 (2011) 42017–42026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [53].Averna M, De Tullio R, Capini P, Salamino F, Pontremoli S, Melloni E, Changes in calpastatin localization and expression during calpain activation: a new mechanism for the regulation of intracellular Ca(2+)-dependent proteolysis, Cell. Mol. Life Sci. 60 (2003) 2669–2678. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [54].Hanna RA, Campbell RL, Davies PL, Calcium-bound structure of calpain and its mechanism of inhibition by calpastatin, Nature 456 (2008) 409–412. [DOI] [PubMed] [Google Scholar]
  • [55].Ma H, Tochigi A, Shearer TR, Azuma M, Calpain inhibitor SNJ-1945 attenuates events prior to angiogenesis in cultured human retinal endothelial cells, J. Ocul. Pharmacol. Ther. 25 (2009) 409–414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [56].Letavernier B, Zafrani L, Nassar D, Perez J, Levi C, Bellocq A, Mesnard L, Sachon E, Haymann JP, Aractingi S, Faussat AM, Baud L, Letavernier E,Calpains contribute to vascular repair in rapidly progressive form of glomerulonephritis: potential role of their externalization, Arterioscler. Thromb. Vasc. Biol. 32 (2012) 335–342. [DOI] [PubMed] [Google Scholar]
  • [57].Su Y, Cao W, Han Z, Block ER, Cigarette smoke extract inhibits angiogenesis of pulmonary artery endothelial cells: the role of calpain, Am. J. Physiol. Lung Cell. Mol. Physiol. 287 (2004) L794–L800. [DOI] [PubMed] [Google Scholar]
  • [58].Prangsaengtong O, Senda K, Doki Y, Park JY, Jo M, Sakurai H, Shibahara N, Saiki I, Koizumi K, Calpain 1 and −2 play opposite roles in cord formation of lymphatic endothelial cells via eNOS regulation, Hum. Cell 25 (2012) 36–44. [DOI] [PubMed] [Google Scholar]
  • [59].Adams RH, Alitalo K, Molecular regulation of angiogenesis and lymphangiogenesis, Nat. Rev. Mol. Cell Biol. 8 (2007) 464–478. [DOI] [PubMed] [Google Scholar]
  • [60].Otrock ZK, Mahfouz RA, Makarem JA, Shamseddine AI, Understanding the biology of angiogenesis: review of the most important molecular mechanisms, Blood Cells Mol. Dis. 39 (2007) 212–220. [DOI] [PubMed] [Google Scholar]
  • [61].Potente M, Gerhardt H, Carmeliet P, Basic and therapeutic aspects of angiogenesis, Cell 146 (2011) 873–887. [DOI] [PubMed] [Google Scholar]
  • [62].Liu LZ, Jiang Y, Carpenter RL, Jing Y, Peiper SC, Jiang BH, Role and mechanism of arsenic in regulating angiogenesis, PLoS One 6 (2011) e20858. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [63].Radisavljevic Z, Avraham H, Avraham S, Vascular endothelial growth factor upregulates ICAM-1 expression via the phosphatidylinositol 3 OH-kinase/AKT/nitric oxide pathway and modulates migration of brain microvascular endothelial cells, J. Biol. Chem. 275 (2000) 20770–20774. [DOI] [PubMed] [Google Scholar]
  • [64].Matsunaga T, Weihrauch DW, Moniz MC, Tessmer J, Warltier DC, Chilian WM, Angiostatin inhibits coronary angiogenesis during impaired production of nitric oxide, Circulation 105 (2002) 2185–2191. [DOI] [PubMed] [Google Scholar]
  • [65].Ridnour LA, Isenberg JS, Espey MG, Thomas DD, Roberts DD, Wink DA, Nitric oxide regulates angiogenesis through a functional switch involving thrombospondin-1, Proc. Natl. Acad. Sci. U. S. A. 102 (2005) 13147–13152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [66].Zhang Y, Jiang X, Qin X, Ye D, Yi Z, Liu M, Bai O, Liu W, Xie X, Wang Z, Fang J, Chen Y, RKTG inhibits angiogenesis by suppressing MAPK-mediated autocrine VEGF signaling and is downregulated in clear-cell renal cell carcinoma,Oncogene 29 (2010) 5404–5415. [DOI] [PubMed] [Google Scholar]
  • [67].Arthur JS, Elce JS, Hegadorn C, Williams K, Greer PA, Disruption of the murine calpain small subunit gene, Capn4: calpain is essential for embryonic development but not for cell growth and division, Mol. Cell. Biol. 20 (2000) 4474–4481. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [68].Takano J, Mihira N, Fujioka R, Hosoki E, Chishti AH, Saido TC, Vital role of the calpain-calpastatin system for placental-integrity-dependent embryonic survival, Mol. Cell. Biol. 31 (2011) 4097–4106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [69].Azam M, Andrabi SS, Sahr KE, Kamath L, Kuliopulos A, Chishti AH, Disruption of the mouse mu-calpain gene reveals an essential role in platelet function, Mol. Cell. Biol. 21 (2001) 2213–2220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [70].Nassar D, Letavernier E, Baud L, Aractingi S, Khosrotehrani K, Calpain activity is essential in skin wound healing and contributes to scar formation, PLoS One 7 (2012) e37084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [71].Bodnar RJ, Rodgers ME, Chen WC, Wells A, Pericyte regulation of vascular remodeling through the CXC receptor 3, Arterioscler. Thromb. Vasc. Biol. 33(2013) 2818–2829. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [72].Bodnar RJ, Yates CC, Rodgers ME, Du X, Wells A, IP-10 induces dissociation of newly formed blood vessels, J. Cell Sci. 122 (2009) 2064–2077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [73].Warren RS, Yuan H, Matli MR, Gillett NA, Ferrara N, Regulation by vascular endothelial growth factor of human colon cancer tumorigenesis in a mouse model of experimental liver metastasis, J. Clin. Invest. 95 (1995) 1789–1797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [74].Hein TW, Rosa RH Jr., Ren Y, Xu W, Kuo L, VEGF receptor-2-linked PI3K/ calpain/SIRT1 activation mediates retinal arteriolar dilations to VEGF and shear stress, Invest. Ophthalmol. Vis. Sci. 56 (2015) 5381–5389. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [75].Arnould T, Michiels C, Alexandre I, Remacle J, Effect of hypoxia upon intracellular calcium concentration of human endothelial cells, J. Cell. Physiol. 152 (1992) 215–221. [DOI] [PubMed] [Google Scholar]
  • [76].Aono Y, Ariyoshi H, Tsuji Y, Ueda A, Tokunaga M, Sakon M, Monden M, Localized activation of m-calpain in human umbilical vein endothelial cells upon hypoxia, Thromb. Res. 102 (2001) 353–361. [DOI] [PubMed] [Google Scholar]
  • [77].Zheng X, Zhou AX, Rouhi P, Uramoto H, Boren J, Cao Y, Pereira T, Akyurek LM, Poellinger L, Hypoxia-induced and calpain-dependent cleavage of filamin A regulates the hypoxic response, Proc. Natl. Acad. Sci. U. S. A. 111 (2014) 2560–2565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [78].Saito M, Suzuki Y, Yano S, Miyazaki T, Sato Y, Proteolytic inactivation of anti-angiogenic vasohibin-1 by cancer cells, J. Biochem. 160 (2016) 227–232. [DOI] [PubMed] [Google Scholar]
  • [79].Watanabe K, Hasegawa Y, Yamashita H, Shimizu K, Ding Y, Abe M, Ohta H, Imagawa K, Hojo K, Maki H, Sonoda H, Sato Y, Vasohibin as an endothelium-derived negative feedback regulator of angiogenesis, J. Clin. Invest. 114 (2004) 898–907. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [80].Saaristo A, Tammela T, Farkkila A, Karkkainen M, Suominen E, Yla-Herttuala S, Alitalo K, Vascular endothelial growth factor-C accelerates diabetic wound healing, Am. J. Pathol. 169 (2006) 1080–1087. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [81].Brem H, Sheehan P, Rosenberg HJ, Schneider JS, Boulton AJ, Evidence-based protocol for diabetic foot ulcers, Plast. Reconstr. Surg. 117 (2006) 193S–209S (discussion 210S-211S). [DOI] [PubMed] [Google Scholar]
  • [82].Brem H, Tomic-Canic M, Cellular and molecular basis of wound healing in diabetes, J. Clin. Invest. 117 (2007) 1219–1222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [83].Shukla A, Dubey MP, Srivastava R, Srivastava BS, Differential expression of proteins during healing of cutaneous wounds in experimental normal and chronic models, Biochem. Biophys. Res. Commun. 244 (1998) 434–439. [DOI] [PubMed] [Google Scholar]
  • [84].Galkowska H, Wojewodzka U, Olszewski WL, Chemokines, cytokines, and growth factors in keratinocytes and dermal endothelial cells in the margin of chronic diabetic foot ulcers, Wound Repair Regen. 14 (2006) 558–565. [DOI] [PubMed] [Google Scholar]
  • [85].Falanga V, Wound healing and its impairment in the diabetic foot, Lancet 366 (2005) 1736–1743. [DOI] [PubMed] [Google Scholar]
  • [86].Cianfarani F, Zambruno G, Brogelli L, Sera F, Lacal PM, Pesce M, Capogrossi MC, Failla CM, Napolitano M, Odorisio T, Placenta growth factor in diabetic wound healing: altered expression and therapeutic potential, Am. J. Pathol. 169 (2006) 1167–1182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [87].Frank S, Hubner G, Breier G, Longaker MT, Greenhalgh DG, Werner S, Regulation of vascular endothelial growth factor expression in cultured keratinocytes. Implications for normal and impaired wound healing, J. Biol. Chem. 270(1995) 12607–12613. [DOI] [PubMed] [Google Scholar]
  • [88].Okizaki S, Ito Y, Hosono K, Oba K, Ohkubo H, Kojo K, Nishizawa N, Shibuya M, Shichiri M, Majima M, Vascular endothelial growth factor receptor type 1 signaling prevents delayed wound healing in diabetes by attenuating the production of IL-1beta by recruited macrophages, Am. J. Pathol. 186 (2016) 1481–1498. [DOI] [PubMed] [Google Scholar]
  • [89].Jing L, Li S, Li Q, Akt/hypoxia-inducible factor-1alpha signaling deficiency compromises skin wound healing in a type 1 diabetes mouse model, Exp. Ther. Med. 9 (2015) 2141–2146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [90].Katagiri S, Park K, Maeda Y, Rao TN, Khamaisi M, Li Q, Yokomizo H, Mima A, Lancerotto L, Wagers A, Orgill DP, King GL, Overexpressing IRS1 in endothelial cells enhances angioblast differentiation and wound healing in diabetes and insulin resistance, Diabetes 65 (2016) 2760–2771. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [91].Wieman TJ, Smiell JM, Su Y, Efficacy and safety of a topical gel formulation of recombinant human platelet-derived growth factor-BB (becaplermin) in patients with chronic neuropathic diabetic ulcers. A phase III randomized placebo-controlled double-blind study, Diabetes Care 21 (1998) 822–827. [DOI] [PubMed] [Google Scholar]
  • [92].Somanath PR, Chen J, Byzova TV, Akt1 is necessary for the vascular maturation and angiogenesis during cutaneous wound healing, Angiogenesis 11 (2008) 277–288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [93].Lee PC, Salyapongse AN, Bragdon GA, Shears LL 2nd, Watkins SC, Edington HD, Billiar TR, Impaired wound healing and angiogenesis in eNOS-deficient mice, Am. J. Phys. 277 (1999) H1600–H1608. [DOI] [PubMed] [Google Scholar]
  • [94].Maruyama K, Asai J, Ii M, Thorne T, Losordo DW, D’Amore PA, Decreased macrophage number and activation lead to reduced lymphatic vessel formation and contribute to impaired diabetic wound healing, Am. J. Pathol. 170 (2007) 1178–1191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [95].Cho CH, Sung HK, Kim KT, Cheon HG, Oh GT, Hong HJ, Yoo OJ, Koh GY, COMP-angiopoietin-1 promotes wound healing through enhanced angiogenesis, lymphangiogenesis, and blood flow in a diabetic mouse model (vol 103, pg 4946, 2006), Proc. Natl. Acad. Sci. U. S. A. 103 (2006) 10146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [96].Sanson M, Ingueneau C, Vindis C, Thiers JC, Glock Y, Rousseau H, Sawa Y, Bando Y, Mallat Z, Salvayre R, Negre-Salvayre A, Oxygen-regulated protein-150 prevents calcium homeostasis deregulation and apoptosis induced by oxidized LDL in vascular cells, Cell Death Differ. 15 (2008) 1255–1265. [DOI] [PubMed] [Google Scholar]
  • [97].Vindis C, Elbaz M, Escargueil-Blanc I, Auge N, Heniquez A, Thiers JC, Negre-Salvayre A, Salvayre R, Two distinct calcium-dependent mitochondrial pathways are involved in oxidized LDL-induced apoptosis, Arterioscler. Thromb. Vasc. Biol. 25 (2005) 639–645. [DOI] [PubMed] [Google Scholar]
  • [98].Goncalves I, Nitulescu M, Saido TC, Dias N, Pedro LM, JF EF, Ares MP and Porn-Ares I., Activation of calpain-1 in human carotid artery atherosclerotic lesions, BMC Cardiovasc. Disord. 9 (2009) 26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [99].Porn-Ares MI, Saido TC, Andersson T, Ares MP, Oxidized low-density lipoprotein induces calpain-dependent cell death and ubiquitination of caspase 3 in HMEC-1 endothelial cells, Biochem. J. 374 (2003) 403–411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [100].Miyazaki T, Taketomi Y, Takimoto M, Lei XF, Arita S, Kim-Kaneyama JR, Arata S, Ohata H, Ota H, Murakami M, Miyazaki A, m-Calpain induction in vascular endothelial cells on human and mouse atheromas and its roles in VE-cadherin disorganization and atherosclerosis, Circulation 124 (2011) 2522–2532. [DOI] [PubMed] [Google Scholar]
  • [101].Chaudhuri P, Colles SM, Damron DS, Graham LM, Lysophosphatidylcholine inhibits endothelial cell migration by increasing intracellular calcium and activating calpain, Arterioscler. Thromb. Vasc. Biol. 23 (2003) 218–223. [DOI] [PubMed] [Google Scholar]
  • [102].Williams JK, Armstrong ML, Heistad DD, Vasa vasorum in atherosclerotic coronary arteries: responses to vasoactive stimuli and regression of atherosclerosis,Circ. Res. 62 (1988) 515–523. [DOI] [PubMed] [Google Scholar]
  • [103].Zhang Y, Cliff WJ, Schoefl GI, Higgins G, Immunohistochemical study of intimal microvessels in coronary atherosclerosis, Am. J. Pathol. 143 (1993)164–172. [PMC free article] [PubMed] [Google Scholar]
  • [104].Napoli C, Ignarro LJ, Nitric oxide and atherosclerosis, Nitric Oxide Biol. Chem. 5(2001) 88–97. [DOI] [PubMed] [Google Scholar]
  • [105].Napoli C, de Nigris F, Williams-Ignarro S, Pignalosa O, Sica V, Ignarro LJ, Nitric oxide and atherosclerosis: an update, Nitric Oxide Biol. Chem. 15 (2006) 265–279. [DOI] [PubMed] [Google Scholar]
  • [106].Ariyoshi H, Yoshikawa N, Aono Y, Tsuji Y, Ueda A, Tokunaga M, Sakon M, Monden M, Localized activation of m-calpain in migrating human umbilical vein endothelial cells stimulated by shear stress, J. Cell. Biochem. 81 (2001) 184–192. [PubMed] [Google Scholar]
  • [107].Miyazaki T, Honda K, Ohata H, Requirement of Ca2+ influx- and phosphatidylinositol 3-kinase-mediated m-calpain activity for shear stress-induced endothelial cell polarity, Am. J. Physiol. Cell Physiol. 293 (2007) C1216–C1225. [DOI] [PubMed] [Google Scholar]
  • [108].Miyazaki T, Honda K, Ohata H, m-Calpain antagonizes RhoA overactivation and endothelial barrier dysfunction under disturbed shear conditions, Cardiovasc. Res. 85 (2010) 530–541. [DOI] [PubMed] [Google Scholar]
  • [109].Nakajima E, David LL, Bystrom C, Shearer TR, Azuma M, Calpain-specific proteolysis in primate retina: contribution of calpains in cell death, Invest. Ophthalmol. Vis. Sci. 47 (2006) 5469–5475. [DOI] [PubMed] [Google Scholar]
  • [110].Azuma M, Shearer TR, The role of calcium-activated protease calpain in experimental retinal pathology, Surv. Ophthalmol. 53 (2008) 150–163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [111].Modlich U, Pugh CW, Bicknell R, Increasing endothelial cell specific expression by the use of heterologous hypoxic and cytokine-inducible enhancers, Gene Ther. 7 (2000) 896–902. [DOI] [PubMed] [Google Scholar]
  • [112].Houston P, White BP, Campbell CJ, Braddock M, Delivery and expression of fluid shear stress-inducible promoters to the vessel wall: applications for cardiovascular gene therapy, Hum. Gene Ther. 10 (1999) 3031–3044. [DOI] [PubMed] [Google Scholar]
  • [113].Lin YC, Brown K, Siebenlist U, Activation of Nf-kappa-B requires proteolysis of the inhibitor I-kappa-alpha — signal-induced phosphorylation of I-kappa-B-alpha alone does not release active Nf-kappa-B, Proc. Natl. Acad. Sci. U. S. A. 92 (1995) 552–556. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [114].Yu X, Bondada V, Rogers C, Meyer CA, Yu C, Targeting ERK½-calpain 1-NF-κB signal transduction in secondary tissue damage and astrogliosis after spinal cord injury, Front. Biol. 10 (2015) 427–438. [Google Scholar]
  • [115].Lee M, Rentz J, Bikram M, Han S, Bull DA, Kim SW, Hypoxia-inducible VEGF gene delivery to ischemic myocardium using water-soluble lipopolymer, Gene Ther. 10 (2003) 1535–1542. [DOI] [PubMed] [Google Scholar]
  • [116].Binley K, Askham Z, Martin L, Spearman H, Day D, Kingsman S, Naylor S, Hypoxia-mediated tumour targeting, Gene Ther. 10 (2003) 540–549. [DOI] [PubMed] [Google Scholar]
  • [117].Pin RH, Reinblatt M, Fong Y, Employing tumor hypoxia to enhance oncolytic viral therapy in breast cancer, Surgery 136 (2004) 199–204. [DOI] [PubMed] [Google Scholar]
  • [118].Abel T, El Filali E, Waern J, Schneider IC, Yuan Q, Munch RC, Hick M, Warnecke G, Madrahimov N, Kontermann RE, Schuttrumpf J, Muller UC, Seppen J, Ott M, Buchholz CJ, Specific gene delivery to liver sinusoidal and artery endothelial cells, Blood 122 (2013) 2030–2038. [DOI] [PubMed] [Google Scholar]
  • [119].Haurwitz RE, Jinek M, Wiedenheft B, Zhou K, Doudna JA, Sequence- and structure-specific RNA processing by a CRISPR endonuclease, Science 329 (2010) 1355–1358. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [120].Jinek M, Chylinski K, Fonfara I, Hauer M, Doudna JA, Charpentier E, A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity, Science 337 (2012) 816–821. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [121].Cong L, Ran FA, Cox D, Lin SL, Barretto R, Habib N, Hsu PD, Wu XB, Jiang WY, Marraffini LA, Zhang F, Multiplex genome engineering using CRISPR/Cas systems, Science 339 (2013) 819–823. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [122].Yin H, Song CQ, Dorkin JR, Zhu LJ, Li Y, Wu Q, Park A, Yang J, Suresh S, Bizhanova A, Gupta A, Bolukbasi MF, Walsh S, Bogorad RL, Gao G, Weng Z, Dong Y, Koteliansky V, Wolfe SA, Langer R, Xue W, Anderson DG, Therapeutic genome editing by combined viral and non-viral delivery of CRISPR system components in vivo, Nat. Biotechnol. 34 (2016) 328–333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [123].Cardone RA, Casavola V, Reshkin SJ, The role of disturbed pH dynamics and the Na+/H+ exchanger in metastasis, Nat. Rev. Cancer 5 (2005) 786–795. [DOI] [PubMed] [Google Scholar]
  • [124].Lee ES, Na K, Bae YH, Polymeric micelle for tumor pH and folate-mediated targeting, J. Control. Release 91 (2003) 103–113. [DOI] [PubMed] [Google Scholar]
  • [125].Lee ES, Na K, Bae YH, Doxorubicin loaded pH-sensitive polymeric micelles for reversal of resistant MCF-7 tumor, J. Control. Release 103 (2005) 405–418. [DOI] [PubMed] [Google Scholar]
  • [126].Sethuraman VA, Bae YH, TAT peptide-based micelle system for potential active targeting of anti-cancer agents to acidic solid tumors, J. Control. Release 118 (2007) 216–224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [127].Tang F, Chan E, Lu M, Zhang X, Dai C, Mei M, Zhang S, Wang H, Song Q, Calpain-1 mediated disorder of pyrophosphate metabolism contributes to vascular calcification induced by oxLDL, PLoS One 10 (2015) e0129128. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES