Next Article in Journal
Acute ACAT1/SOAT1 Blockade Increases MAM Cholesterol and Strengthens ER-Mitochondria Connectivity
Next Article in Special Issue
Role of Muscarinic Acetylcholine Receptors in Intestinal Epithelial Homeostasis: Insights for the Treatment of Inflammatory Bowel Disease
Previous Article in Journal
Origin of Elevated S-Glutathionylated GAPDH in Chronic Neurodegenerative Diseases
Previous Article in Special Issue
Identification of Novel Core Genes Involved in Malignant Transformation of Inflamed Colon Tissue Using a Computational Biology Approach and Verification in Murine Models
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Contribution of Blood Vessel Activation, Remodeling and Barrier Function to Inflammatory Bowel Diseases

by
Nathalie Britzen-Laurent
1,2,*,
Carl Weidinger
3 and
Michael Stürzl
2,4
1
Division of Surgical Research, Department of Surgery, Translational Research Center, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
2
Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), 91054 Erlangen, Germany
3
Department of Gastroenterology, Infectious Diseases and Rheumatology, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, 12203 Berlin, Germany
4
Division of Molecular and Experimental Surgery, Translational Research Center, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(6), 5517; https://doi.org/10.3390/ijms24065517
Submission received: 15 February 2023 / Revised: 9 March 2023 / Accepted: 10 March 2023 / Published: 14 March 2023
(This article belongs to the Special Issue Solving the Puzzle: Molecular Research in Inflammatory Bowel Diseases)

Abstract

:
Inflammatory bowel diseases (IBDs) consist of a group of chronic inflammatory disorders with a complex etiology, which represent a clinical challenge due to their often therapy-refractory nature. In IBD, inflammation of the intestinal mucosa is characterized by strong and sustained leukocyte infiltration, resulting in the loss of epithelial barrier function and subsequent tissue destruction. This is accompanied by the activation and the massive remodeling of mucosal micro-vessels. The role of the gut vasculature in the induction and perpetuation of mucosal inflammation is receiving increasing recognition. While the vascular barrier is considered to offer protection against bacterial translocation and sepsis after the breakdown of the epithelial barrier, endothelium activation and angiogenesis are thought to promote inflammation. The present review examines the respective pathological contributions of the different phenotypical changes observed in the microvascular endothelium during IBD, and provides an overview of potential vessel-specific targeted therapy options for the treatment of IBD.

1. Introduction

Inflammatory bowel diseases (IBD) are a group of intestinal chronic inflammatory disorders characterized by cyclic flares of destructive inflammation that comprise two major forms, Crohn’s disease (CD) and ulcerative colitis (UC) [1]. Although inflammation in UC is restricted to the colon and only extends to the mucosal layers, transmural inflammation can be observed in CD, which can manifest at any site of the gut. The etiology of IBD is thought to be multifactorial and to involve the patient’s genetics and immune response, the intestinal microbiome, and environmental factors [2]. IBD is considered to result from an inappropriate immune response to the intestinal microflora and environmental triggers in genetically susceptible individuals [1]. The resulting inflammation induces tissue damage and, notably, a disruption of the epithelial barrier, leading to the perturbation of the intestinal microenvironment and the relationship between the mucosal surface and the commensal microbiota. This disequilibrium not only affects the maintenance, function, and repair of the epithelial barrier, it also results in the perturbation of the commensal microbiota composition, leading to dysbiosis [3,4].
The intestinal microvasculature lies in close vicinity to the epithelial layer and represents a second barrier to the penetration and dissemination of commensals and microbial products. In IBD, the microvascular endothelium is strongly affected by inflammation [5]. Endothelial cells (ECs) of the gut microvasculature are activated to allow leukocyte recruitment and infiltration, whereas the vascular barrier function is compromised and local bursts of angiogenesis are observed. Despite its well-defined function in inflammation, the contribution of the endothelium to the development and maintenance of IBD has been rather overlooked, in particular as a potential therapeutic target. In the present review, we summarize the respective contributions of blood vessel activation, remodeling, and barrier function to the pathogenesis of IBD, and discuss the status and perspectives of vessel-directed therapies.

2. The Intestinal Vasculature in Homeostasis

The intestine is vascularized by arterioles from the submucosa, which divide into capillary networks in the mucosa and muscle layers, with the mucosal layers receiving 80% of the total blood flow [6]. The anatomy of the mucosal vasculature differs between the small and large intestine (colon) due to their different tissue architectures. In the small intestine, the epithelium builds villi and crypts. Each villus contains a single arteriole going to the tip, forming a tuft-like network of capillaries which are located directly under the epithelial monolayer, and the blood is collected into a single central venule. The crypts are infused with a capillary network which drains into the venule as well. In contrast to the small intestine, the colon epithelium does not have villi. The arterioles and their capillary branches are arranged along the colonic crypts, developing into a capillary honeycomb-like network around the crypts that is in very close proximity to the epithelial layer (1 μm) [6,7]. Intestinal post-capillary veins are devoid of smooth-muscle cells and represent the most reactive segment of the microvasculature [8]. The intestinal microcirculation regulates oxygen and nutrient exchange, tissue fluid homeostasis, and leucocyte abundance [9,10,11].
Under physiological conditions, the single layer of ECs lining the vessel lumen provides an anti-adhesive and selectively permeable exchange barrier. In the past few years, the general opinion about the role of the intestinal microvasculature has evolved, and it is now viewed as an integral component of the intestinal barrier [12]. The intestinal barrier is tightly regulated to allow the absorption of essential nutrients, electrolytes, and water from the intestinal lumen into the circulation, while preventing the entry of microbiota through different layers of protection. The first barrier is formed by a tight epithelial monolayer covered by a thick layer of mucus produced by specialized enterocytes, the goblet cells. In addition, another population, the Paneth cells, secretes antimicrobial peptides. This intestinal epithelial barrier prevents the penetration of microbes or microbe-derived molecules into the tissue. A second barrier, the gut vascular barrier (GVB), has been identified in humans and mice, and provides a second layer of protection, which blocks microbial dissemination into the systemic circulation (Figure 1) [12,13,14].
In contrast to the blood–brain barrier (BBB), which has a size exclusion threshold of 500 Da, the GVB is permeable to molecules as large as 4 kDa, allowing the passage of nutrients and antigens for tolerance induction [14]. Spadoni et al. demonstrated that endothelial cells in the intestine are closely associated with pericytes and enteric glial cells to form what they termed gut–vascular units (Figure 1) [13]. Enteric glial cells seem to be crucial for the development and maintenance of the GVB, as it has been shown that transgenic mice lacking enteric glial cells feature increased epithelial permeability and microvascular disturbances, resulting in the uncontrolled spread of bacteria into the blood circulation and the subsequent death of the affected animals (Table 1) [15,16]. Interestingly, the interplay of the GVB with the commensal microbiota was found to increase angiogenesis, endothelial coverage, and the formation of the enteric glial cell network in the lamina propria using human ECs and murine models [17,18,19,20], suggesting that the microbiota supports the formation and maintenance of the GVB.
Within the gut–vascular units, endothelial cells form tight cell–cell contacts, which are enhanced through interaction with enteric glial cells and pericytes. Adherens junctions (AJs) and tight junctions (TJs) found in human and mouse intestinal microvascular ECs regulate paracellular trafficking of molecules and leukocytes and express different classes of transporters (such as ATP-binding cassette transporters and sugar transporters) [12]. TJs control permeability for ions and small molecules (<800 Da), whereas AJs are primarily responsible for the maintenance of vascular barrier function and control its permeability for molecules of high molecular weight [21]. In intestinal ECs, AJs are composed of vascular endothelial cadherin (VE-cadherin), α- and β-catenin, and p120, all of which are expressed homogenously throughout the intestines and the vascular beds. The building of VE-cadherin adhesions is considered to be the primary event during vascular development [13,21]. It precedes TJ formation and is required for TJ maintenance. Disruption of AJs leads to disassembly of TJs [13,21]. Although numerous TJs can be found in small arterioles, the level of TJs is reduced in capillaries and post-capillary veins. As a result, AJs are predominantly found in capillaries and post-capillary venules [21]. In intestinal ECs, TJs are formed by occludin, zonula occludens-1 (ZO-1), cingulin, junctional adhesion molecule-A (JAM-A), and claudins [13,22]. The expression of endothelial claudins has been extensively studied in the mouse intestine and varies between gut areas and cell types. While claudin-1 is expressed at similar levels in ECs throughout the intestines, the channel-forming claudins-7, -12, and -15 are exclusively expressed in the colon [22]. Outside of the BBB and the blood–retinal barrier, where a high expression of claudin-5 prevents the passage of small molecules, claudin-5 exhibits diminishing expression along the arteriovenous axis [23]. In the gut, claudin-5 expression is restricted to lymphatic ECs, high endothelial venules (HEVs), and certain capillary ECs [13,24]. These different patterns of expression are thought to be responsible for variations in permeability and to reflect the site-specific physiological function of the GVB. In humans, claudins working as channels show a higher expression in the colon, where they regulate solute paracellular transport, compared to the small intestine or the BBB [25,26].
Kalucka et al. performed a single-cell transcriptome analysis of murine ECs across 11 tissues, revealing heterogeneity between tissues and vascular beds [24]. Overall, they found that colon and small-intestine ECs are characterized by the high expression of genes involved in vascular barrier integrity and maintenance. In addition, two specific EC-fractions were found in the intestine. First, a subset of capillary ECs was described, which display an elevated expression of genes involved in the uptake and metabolism of glycerol and fatty acids. Cells in this cluster were notably characterized by a high expression of aquaglyceroporin 7 (Aqp7), a pore-forming transmembrane protein involved in glycerol transport across cell membranes, and were therefore termed Aqp7+ capillary ECs. Second, a subset of intestinal venous ECs showing enriched expression of HEV markers (Madcam1, Lrg1, Ackr1) was identified. HEVs represent a specialized subtype of post-capillary venules mediating the recruiting and trafficking of lymphocytes from blood to lymph nodes and secondary lymphoid organs, which take their name from the cuboidal appearance of their endothelial cells (Table 1).
The investigation of isolated human intestinal mucosa-derived endothelial cells (HIMECs) has revealed unique functional features when compared to human umbilical veins (HUVECs) (Table 1). For instance, exposure to lipopolysaccharide (LPS) induces a transient increase in the presence of adhesion molecules in HIMECs, compared to a long-lasting increase in the presence of adhesion molecules in HUVECs [27]. This reflects the relative tolerance of intestinal microvascular cells to bacterial products from the gut microbiota. In addition, HIMECs produce different cytokines, including IL-3 and IL-6, as HUVECs upon activation with inflammatory cytokines [28]. Furthermore, HIMECs constitutively express the (otherwise) inducible nitric oxide synthase (iNOS) in addition to endothelial NOS (eNOS) [29]. Under physiological conditions, endothelial-derived NO maintains the anti-adhesive state of the endothelium by limiting leukocyte and platelet adhesion, and regulates vasodilatation [29,30].
Overall, in the state of homeostasis, intestinal ECs are involved in barrier function maintenance, nutrient uptake, and immune tolerance.

3. The Intestinal Vasculature in IBD

The intestinal microvasculature plays a crucial role during inflammation by regulating tissue recruitment of inflammatory cells and wound healing. However, uncontrolled inflammation induces a sustained EC activation (Figure 1), causing an increase in leakiness (edema), adhesiveness (leukocyte recruitment), pro-coagulant activity (thrombus), and angiogenesis (immature vessels) [5,31]. As a result, inflammation is enhanced, ultimately leading to sustained tissue and vessel damage.
The histopathological analysis of inflamed human and murine intestinal tissues has revealed massive changes in the blood microvasculature, including vasodilatation, vasocongestion, edema, flares of angiogenesis, microvascular occlusions, and abnormal vessel architecture characterized by tortuous vessels of varying diameter (Figure 2) [32,33,34]. These profound alterations have been considered to be an early event since they precede the development of mucosal ulceration, and to significantly enhance inflammation in IBD [32]. Hence, the intestinal microvasculature can be seen both as a regulating factor and as a target of inflammation. In the following, we examine the respective contributions of intestinal vascular changes to IBD pathogenesis (Figure 2).

3.1. Endothelial Cell Activation and Leukocyte Recruitment

The recruitment of circulating leukocytes into tissues is an early and central event during inflammation. It starts with the activation of the microvascular endothelium by inflammatory mediators, including cytokines such as IFN-γ, IL-1β, and TNF-α (Table 2). The activated endothelium regulates the leukocyte extravasation cascade in a tightly coordinated sequence, including tethering and rolling, activation, adhesion, spreading, and the transmigration of leukocytes [35]. Activated ECs are characterized by an elevation in the level of cell adhesion molecules (CAMs), the production of chemokines, and the expression of costimulatory molecules, which further amplify the recruitment and activation of leukocytes (Figure 1). Leukocyte hyper-adhesion has been observed in the intestinal ECs of patients with IBD. For instance, Binion et al. described a significant increase in leukocyte binding in HIMECs isolated from inflamed regions of IBD patients compared to HIMECs that were obtained from non-inflamed intestinal sites or from the guts of control subjects [36].
At the molecular level, the MAPK pathway has been shown to play an important role in the upregulation of CAMs and the production of chemokines by activated HIMECs, as well as lymphocyte extravasation [37]. In the inflamed mucosa of IBD patients, increased levels of phosphorylated MAPK have been detected in the microvasculature [37].
Among the various CAMs expressed in activated ECs, P- and E-selectins are glycoproteins involved in the rolling and recruitment of leukocytes. Although P-selectin is constitutively available as a pool that can be mobilized upon activation, E-selectin expression is induced in response to inflammatory stimuli. A notable increase in P-selectin levels has been observed in the colons of UC patients compared to controls, whereas serum levels of the decoy soluble P-selectin were decreased [37,38,39].
An important class of adhesion molecules expressed in ECs is the immunoglobin CAM superfamily, which includes intracellular cell adhesion molecule 1 (ICAM-1), vascular cell adhesion molecule 1 (VCAM-1), platelet endothelial cell adhesion molecule (PECAM-1, also known as CD31), as well as the gut-specific mucosal addressin cell adhesion molecule 1 (MadCAM-1). Upon EC activation, ICAM-1 is recruited from the EC junctions to the apical surface [27]. The microvascular expression of ICAM-1 is increased in IBD patients, and ICAM-1 has been shown to be crucial to T-cell recruitment in the T-cell transfer murine colitis model [40]. ICAM-1 is constitutively expressed in HIMECs and can be upregulated by inflammatory cytokines and vascular endothelial growth factor-A (VEGF-A) [27,41,42]. VCAM-1, which mediates adhesion to lymphocytes expressing integrin α4β1 or α4β7, is also inducible in HIMECs, notably by VEGF-A. The expression of VCAM-1 in the mucosal vasculature is increased in patients with IBD and murine colitis models, where its expression correlates with disease severity [43,44]. PECAM-1/CD31 is also inducible by inflammatory cytokines and is involved in leukocyte rolling and firm adhesion during IBD [45]. A large amount of attention has been paid to MadCAM-1, a gut-specific homing molecule mediating the recruitment of T and B cells expressing integrin α4β7 [46]. High MadCAM-1 expression has been observed in the inflamed intestinal endothelium during IBD [36]. In HIMECs, MadCAM-1 expression can be induced by inflammatory cytokines (IFN-γ, IL-1β, TNF-α) [47,48]. Interestingly, the expression of MadCAM-1 in HIMECs is inversely correlated to cellular density, suggesting that high MadCAM-1 expression might be a marker of proliferating vessels [47,48]. Recently, MadCAM-1 has been found to be critical for the recruitment of antibody-producing B cells into the intestinal mucosa in the IL-10-knockout colitis model [49].

3.2. Pathological Angiogenesis in IBD

Angiogenesis is a hallmark of chronic inflammation [50]. In human IBD and in several murine models of colitis, the microvascular density is increased and directly correlates with disease severity (Figure 1 and Figure 2) [51,52,53]. Angiogenic ECs exhibit increased proliferation and migration, as well as a unique cell-surface molecular pattern. For instance, the integrins ανβ3 and ανβ5 are specifically expressed at the surface of ECs from newly formed vessels, and an increased expression of ανβ3 has been observed in the inflamed mucosa of IBD patients [51]. The blockade of ανβ3 was found to reduce disease activity in the IL-10 knockout colitis model, suggesting that angiogenesis contributes to IBD pathogenesis [54]. The angiogenic expansion of the vascular bed is assumed to physically increase blood supply through the increased endothelial surface, and therefore to enhance the leukocyte supply to the tissue. However, the newly formed vessels in the context of chronic inflammation show an immature phenotype in mouse and human tissues [34,55,56]. They are leaky, have less or no coverage by pericytes, and are hypoperfused and often hyperthrombic [32,33,34]. Stenoses are also frequently observed [32]. Hence, pathological angiogenesis appears to contribute to the intense vascular remodeling observed in IBD.
The fact that mucosal extracts of IBD patients could induce dose-dependent HIMEC migration in vitro supported the presence of a pro-angiogenic microenvironment in the inflamed gut mucosa [57]. Two main mechanisms of angiogenesis have been proposed to occur during IBD, namely, extension from existing vessels (sprouting) and vessel splitting (intussusception). Angiogenesis through the recruitment of endothelial progenitor cells seems less likely to occur during colitis since the number of bone-marrow-derived endothelial progenitor cells is reduced in UC [58,59,60].
The induction of sprouting angiogenesis during IBD has been partly attributed to inflammation-related hypoxia, with hypoxia-inducible factor-1 and -2 transcriptionally activating the expression of vascular endothelial growth factor A (VEGF-A), the major angiogenic growth factor (AGF) [61]. In agreement with this hypothesis, the human inflamed intestinal epithelium was found to represent an important source of VEGF-A, which is also produced by leukocytes [62,63,64,65]. Furthermore, increased expression of VEGF and other AGFs including basic fibroblast growth factor (bFGF) and platelet-derived growth factor (PDGF) has been detected in mucosal extracts and in the serum of IBD patients as compared to controls (Table 3) [51,53,66,67,68,69,70], although this increase was more evident for UC than for CD, especially for VEGF [61,71,72,73]. However, experimental colitis models have provided conflicting results regarding the contribution of sprouting angiogenesis to disease activity. The inhibition of angiogenesis via the neutralization of VEGF-A improved the course of intestinal inflammation in mice [34,64,74,75] and modestly in rats [76]. In contrast, the knock-out of placental growth factor, a VEGF homolog, also caused decreased angiogenesis, but lead to an aggravation of colonic injury in the mouse dextran sodium sulfate (DSS)-induced colitis model [77]. These results are reflected in the responses of cancer patients treated with anti-angiogenic treatment. Rare adverse effects of bevacizumab, a humanized monoclonal antibody against VEGF, include intestinal perforation, gastrointestinal bleeding, and ulcerative colitis [78,79,80]. However, anti-VEGF therapy is well tolerated by most patients with quiescent and moderately active IBD [81]. The frequency of inflammatory side effects is higher when the PDFG pathway is involved. For instance, an exaggeration of UC has been observed during treatment with the angiogenic inhibitors sunitinib and sorafenib, which target both the VEGF and PDGF pathways [82,83,84].
Several reasons might explain the contradictory effects of angiogenesis in IBD. Firstly, while angiogenic and inflammatory vessels can synchronously co-exist in the inflamed mucosa during mouse colitis [32], the two phenotypes are mutually exclusive in a single EC. In particular, inflammatory cytokines (ICs) such as IFN-γ, TNF-α, and IL1-β can inhibit AGF-induced proliferation and the migration of human ECs in vitro (Table 2) [85,86,87]. In addition to ICs, several anti-angiogenic factors are upregulated during intestinal inflammation, including the chemokine CXCL-10, thrombospondin, angiostatin—a cleaved fragment of plasminogen, and endostatin—a cleaved fragment of collagen XVIII [63,88,89,90,91,92]. Hence, the balance between angiogenic and inflammatory-associated angiostatic stimuli might explain the relative contribution of angiogenesis to IBD pathogenesis.
Secondly, there is a strong interplay between angiogenesis, inflammatory vessel activation, and barrier function during inflammation. For instance, human and murine VEGF-A stimulates angiogenesis and increases vessel permeability, while reducing vessel coverage [41,42,93]. In murine colitis models, the permeability marker CD146/MUC18 was shown to induce angiogenesis, lymphangiogenesis, and leukocyte recruitment [94,95,96]. In a similar manner, mice that are deficient in CD40 or CD40L display both a decrease in leukocyte and platelet recruitment and impaired angiogenesis in the gut [97,98,99].
Thirdly, the importance of intussusceptive angiogenesis during IBD might have been underestimated. The vessel splitting that is characteristic of intussusceptive angiogenesis occurs through intraluminal endothelial cell rearrangements rather than endothelial cell proliferation [100]. These are triggered by increased blow flow and are regulated by the nitric oxide (NO), endoglin, and ephrinB2/EphB4 signaling pathways [101,102,103,104,105]. Intussusceptive angiogenesis has been observed in murine colitis [100,106,107], where it was induced through mechanical forces and changes in the intraluminal blood flow, and was regulated by MT1-MMP [100,106]. EC-specific knockout of MT1-MMP ameliorated dextran sodium sulfate (DSS)-induced colitis in mice [107]. The predominance of intussusceptive angiogenesis compared to sprouting angiogenesis during colitis might explain the relative success of classical anti-angiogenic approaches in animal models of IBD.
Finally, angiogenesis not only sustains inflammation, it also plays an essential role during mucosal healing. For instance, VEGF-A is involved in UC healing and angiogenesis via the recruitment of cells expressing vascular endothelial growth factor receptor 1 (VEGFR1), including monocytes, Tregs, and bone-marrow derived stem cells, to ulcerated tissues [108]. Furthermore, the expression of the Wnt pathway member adenomatous polyposis coli (APC) in murine intestinal ECs has been shown to mediate mucosal repair following colonic inflammation through angiogenesis [109]. Hence, treatment with angiogenesis inhibitors might result in wound healing complications. Despite the activation of angiogenic signals, impaired mucosal healing is observed and represents a major issue in IBD, notably in UC. Pathological angiogenesis or an increase in anti-angiogenic signals such as endostatin and angiostatin might explain why mucosal lesions are slow to repair in UC [91]. In addition, the decrease in the number of bone-marrow-derived endothelial progenitor cells (BMD-EPC) observed in UC, whether it is due to a decreased release from the bone marrow and/or impaired homing in colonic lesions, participates in mucosal healing impairment [59,60]. In addition, there is crosstalk between VEGF and transforming growth factor-beta (TGF-β), which is essential for wound healing, tissue repair, and the resolution of inflammation. A dysregulation of the TGF-β pathway, as seen in case of the impairment of endoglin, the endothelial-specific co-receptor for TGF-β, caused the levels of VEGF to spike in the acute DSS colitis model [110,111]. This resulted in enhanced and chronic intestinal inflammation, characterized by higher angiogenesis and MAdCAM-1 vascular expression [110,111]. Hence, both the therapeutic inhibition of angiogenesis and the presence of exacerbated or pathological angiogenesis seem to impair wound healing in IBD and experimental colitis models.
Another example of the interplay between vascular remodeling and inflammation in IBD is given by HEVs (Table 1). Subsequently to the ectopic formation of tertiary lymphoid organs observed in the inflamed gut mucosa, the density of HEVs has been found to increase in the mucosa of IBD patients [112,113]. However, HEVs not only regulate the lymph drainage of antigen-presenting cells, they also regulate the homing of T-cells. Extrafollicular HEV formation has been observed in the intestinal mucosa of IBD patients during active inflammation, where it correlates with T-cell infiltration and disease activity [113,114]. Human intestinal HEVs express high levels of MadCAM-1, and in UC, the O-glycosylation of MadCAM1 is increased, which induces a higher binding to L-selectin, expressed on leukocytes [112,115]. Taken together, pathological angiogenesis in IBD seems to both potentiate inflammation and impair mucosal healing.

3.3. The Gut–Vascular Barrier in IBD

During IBD, the epithelial barrier is compromised, as shown by an increased permeability and mucosal damage such as erosions or ulcers [116,117]. In consequence, the passage of antigens, bacteria, and bacterial products into the submucosa is increased [117]. This further enhances the local inflammation, resulting in the release of large amounts of inflammatory mediators from—but not restricted to—immune cells, which can in turn affect the gut–vascular barrier [118].
IBD is associated with increased gut vascular permeability (Figure 1), which is indicative of a loss of intestinal vascular barrier function, and results in edema and tissue damage [33,34,69,119,120,121]. For instance, human CD146 (MUC18), a cell junction molecule constitutively expressed in ECs, and its soluble form (sCD146), which is considered a marker for vascular permeability, are upregulated in intestinal ECs and serum of IBD patients, respectively [96,122]. Endothelial damage and increased colonic vascular permeability have been observed early during the development of experimental ulcerative colitis in rats and mice [123]. Post-capillary venules, the most reactive part of the vascular tree, were also shown to be the major site of vascular leakage [124]. The increased vascular permeability of inflamed post-capillary venules has been attributed to endothelial cell-cell contact disruption, to the contraction of activated ECs, to EC death and detachment, and/or to plasma protein extravasation at the site of leukocyte transendothelial migration [125,126]. A variety of inflammation mediators found in IBD can induce vascular permeability, including histamine, serotonin, substance P, bradykinin, and ICs, notably IFN-γ and TNF-α (Table 2) [121]. In addition, the reduction of anti-inflammatory cytokines such as IL-10 can further amplify intestinal vascular permeability induced by IFN-γ in experimental colitis [121]. Vascular permeability is also increased during angiogenesis, notably through a direct effect of VEGF [127,128,129]. Intussusceptive angiogenesis has also been associated with an enhanced permeability, likely because it induces holes in the vascular layer. In particular, endothelial cell-specific MT1-MMP knockout mice, which are characterized by a lower intussusceptive angiogenesis, also display a reduced vessel permeability during DSS-induced colitis [107].
In a study comparing the respective effects of IFN-γ and VEGF on disease development, both endothelial-specific knockout of the IFN-γ receptor (IFNγR) and VEGF blockade inhibited DSS-induced colitis in mice [34]. In agreement with the angiostatic properties of IFN-γ and the pro-angiogenic effect of VEGF, angiogenesis was increased in the case of IFNγR knockout, whereas it was decreased after VEGF blockade as compared to controls. Both approaches, however, led to a strong decrease in vascular permeability [34,74]. These results suggested that the induction of vascular permeability by VEGF might contribute more to colitis pathogenesis than the mere induction of angiogenesis. Several reports further support the promoting role of GVB disruption in IBD. Vascular permeability is associated with disease activity in UC and CD [34]. The inhibition of vascular permeability with the RTK inhibitor imatinib restores VE-cadherin junctions, increases vascular coverage, and inhibits DSS-induced colitis [34]. Fibrinogen, which is upregulated in UC and mouse colitis, was shown to promote DSS-induced colitis by enhancing vascular permeability [130]. Moreover, transient receptor potential vanilloid 4 (TRPV4) channels were found to enhance DSS-induced colonic inflammation in mice through an increase in vascular permeability [131]. Taken together, these studies have established the pathogenic contribution of vascular barrier breakdown to IBD.

3.3.1. VE-Cadherin and Vascular Barrier Regulation in IBD

VE-cadherin, the major component of endothelial adherens junctions and the master regulator of vascular barrier function, is regarded as the primary target during inflammation-induced vascular permeability [132,133,134]. In IBD patients, membrane VE-cadherin expression is significantly reduced in blood vessels found in inflamed areas compared to uninvolved intestinal tissues [34]. Several mechanisms of VE-cadherin junction disruption have been described. Inflammation-induced proteases such as matrix metalloproteinases (MMPs) or elastase, which are produced by leukocytes, smooth muscle cells, and ECs, can promote vascular permeability through the degradation of VE-cadherin AJs and TJs [5,34,135,136,137,138]. In IBD, vascular smooth muscle cells and pericytes express MMP-1 and MMP-9 [137], and MMP-9 serum levels are increased [139,140,141]. This is supported by the fact that MMP9 deficiency was found to attenuate intestinal injury in animal colitis models [142,143,144]. In addition, MMP-catalyzed VE-cadherin cleavage results in the generation of soluble VE-cadherin, which itself can further destabilize the vascular barrier by impairing the binding of VE-cadherin molecules, as shown in human rheumatoid arthritis, systemic inflammation, and sepsis [145,146,147].
The vascular hyperpermeability induced by VEGF and inflammatory cytokines also involves the direct disassembly of VE-cadherin junctions. Binding of VEGF to VEGFR2 at the surface of human ECs activates the Src kinase, resulting in VE-cadherin phosphorylation and internalization, which occurs via clathrin-dependent endocytosis and is mediated by neuropilin and Rac [127,129,147,148,149,150]. In a similar manner, TNF-α was shown to increase tyrosine phosphorylation of VE-cadherin and to open the paracellular pathway in the human lung endothelium through the activation of the Fyn kinase [138]. The effect of IFN-γ on the disruption of the VE-cadherin junction was found to be even stronger and longer lasting than the effect of VEGF in human ECs and mouse intestinal endothelial cells (MIECs) [33,34,151]. However, the molecular mechanism by which IFN-γ dismantles VE-cadherin junctions remains to be elucidated.
VE-cadherin disruption can also result from EC contraction, since the VE-cadherin complex and the actin cytoskeleton are functionally connected [134,152,153]. For instance, treatment of ECs with TNF-α was found to induce an almost immediate rise in mechanical substrate traction force and internal monolayer tension [154]. VEGF also induces actin reorganization and the migration of endothelial cells via the serine/threonine kinase Akt [155]. A similar disruption of actin has been observed in human ECs following exposure to IFN-γ, and might explain, at least partially, the IFN-γ-induced destabilization of VE-cadherin [156]. Such a mechanism was also observed during experimental colitis, where fibrinogen was shown to induce vascular permeability through activation of AKT and depolymerization of actin microfilaments [130].
Downregulation of AJ and TJ adhesion molecule expression has also been observed upon exposure to ICs and VEGF. For instance, VE-cadherin expression decreases in MIECs after treatment with IFN-γ [34]. Using an in-vitro intestinal endothelial barrier model composed of rat intestinal microvascular endothelial cells, Liu et al. found that TNF-α decreases the expression of TJ proteins, including ZO-1, occludin, and claudin-1, while increasing the expression of pore-forming claudin-2 [157]. Nevertheless, several reports showed no difference in the TJ-associated protein expression of zonula occludens-1 (ZO-1) in MIECs or intestinal endothelial cells during DSS-induced colitis [34,158]. In this model, ZO-1 was notably only reduced in epithelial cells but not in ECs [158], supporting the predominant role of VE-cadherin junctions in the regulation of the GVB.

3.3.2. Vessel Coverage and Permeability

During intestinal inflammation, vessel coverage with adventitial support cells, the so-called mural cells (pericytes and smooth muscle cells), is reduced in mouse models of colitis [159,160]. Mural cell recruitment is regulated by the PDGF-B-PDGFR-β pathway. PDGF-B is secreted by sprouting ECs and signals through PDGFR-β at the surface of mural cells [159]. This induces the proliferation and migration of mural cells. PDGF-B/PDGFR-β knockout models in mice showed reduced mural cell coverage and increased vessel permeability [159,161,162]. In the DSS colitis model, vessel coverage has been found to promote the stabilization of the vascular barrier, decreasing vessel permeability and inflammation [163,164]. Endothelial IFN-γ receptor knockout leads to increased vessel coverage during DSS-induced colitis in mice [33,34]. This could be attributed to the fact that IFN-γ inhibits PDGF-like protein release and decreases the PDGF-B chain mRNA level in HUVECs [165]. Similarly, excess VEGF-A disrupts pericyte recruitment and VEGF blockade was found to increase vessel coverage in the DSS-colitis model [34,166].

3.4. Microvascular Dysfunction in IBD

In IBD, the mucosal vasculature displays pathological traits, including tortuous structures, edema, arteriolar dilatation, hypercoagulation, and vascular damage (Figure 2) [32]. These vascular defects are multifactorial. For instance, chronic high levels of angiogenic factors and inflammatory cytokines can alter microvascular structure and function [167]. More precisely, structural changes in vessels have been shown to result from pathological angiogenesis and inflammation-driven GVB dysfunction, including the loss of mural cell coverage and an immature phenotype [167]. Another example is edema formation, which results both from increased afferent blood flow (hyperemia) due to arteriolar dilatation and from hyperpermeability. The imbalances in vascular function observed during IBD lead to complex and sometimes contradictory effects. In IBD patients, for instance, hyperemia has been observed in submucosal arterioles enlarged by increased afferent blood flow, whereas decreased perfusion was found in the intestinal mucosa [168].

3.4.1. The Role of Nitric Oxide in Vascular Dysfunction during IBD

Among the numerous mediators involved in microvascular function, nitric oxide (NO) has attracted a large amount of interest due to its broad range of functions. Under physiological conditions, NO counteracts leukocyte and platelet adhesion to ECs, regulates vasodilatation and endothelial permeability, and acts as radical scavenger [30]. As mentioned above, HIMECs express both the endogenous endothelial and the inducible form of NO-synthase (eNOS and iNOS), indicating the high tolerance of the gut microvasculature towards inflammatory activation [29]. The resulting high NO levels can, for instance, inhibit the expression of endothelial CAMs and MMPs induced by ICs (Table 3). During IBD, the production of NO by ECs is reduced due to the loss of iNOS and eNOS in HIMECs, and leukocyte adhesion is increased [169,170]. Furthermore, an upregulation of arginase expression and activity has been observed in inflamed HIMECs. Arginase competes with NOS for L-arginine, and therefore can limit NO production due to reduced substrate availability [170]. The decreased production of NO in ECs during IBD results in a loss of NO-mediated vasodilatation and an increase in ROS production in the microvessels of affected intestinal areas [171,172]. In parallel, NO also plays a role in VEGF-driven angiogenesis, as well as in intussusceptive angiogenesis [107], hence contributing both to the perpetuation of inflammation and to wound healing. Finally, NO has been shown to regulate endothelial barrier function in human, murine, and bovine ECs, notably by promoting VEGF-induced permeability through targeting of the VE-cadherin/β-catenin and Rho pathways [173,174]. However, NO can also protect ECs from hypoxia-induced barrier dysfunction [175]. In line with these contradictory results, the deficiency of eNOS and iNOS has been associated with either a better or a more severe course of disease in mouse models of colitis [176,177,178,179]. These differences might be attributed to the different roles played by NO in different cell compartments. The expression of eNOS by intestinal endothelial cells has been shown to specifically maintain mucosal integrity and prevent bacterial translocation in an TNBS-colitis model in mice [179]. Overall, the impairment of NO production during IBD increases the inflammatory activation of ECs and impairs vasodilatation, which might ultimately lead to pathological vasoconstriction, reduced mucosal perfusion, impaired wound healing, and the maintenance of chronic inflammation.

3.4.2. Coagulation

In mucosal tissues from patients with IBD or murine colitis models, the presence of thrombi due to increased platelet activation and binding to the EC surface has been observed [57]. This, in turn, can result in ischemic inflammation in the intestinal microvasculature, further enhancing tissue damage. In the DSS-colitis model and in IBD patient samples, accumulation of platelets in mucosal venules is linked to an increased leukocyte binding and disease activity [57,180,181]. The increase in thrombus formation associated with IBD has been attributed to several factors (Table 3). At the endothelial level, the activation of ECs through ICs and the decrease in NO levels upregulate the surface expression of adhesion molecules. Activated platelets found in the general circulation and in the intestinal mucosa of IBD patients can activate ECs, notably through the expression of P-selectin or CD40L, as well as the release of the soluble form of CD40L (sCD40L) [182,183]. In turn, P-selectin and CD40L/sCD40L activate HIMECs, resulting in increased adhesion molecule expression, the secretion of cytokines such as IL-8, and increased binding to leukocytes, as well as to platelets themselves [97,183]. Increased coagulation has also been attributed to a reduced expression of thrombomodulin and protein C receptor (PCR) in the mucosal microvasculature of IBD patients and in colitic mice [184,185,186,187]. Protein C activation is also impaired in HIMECs under inflammatory conditions. This dampening of the PC system is correlated with increased adhesiveness of ECs, thereby promoting leukocyte recruitment and inflammation.

3.5. Regulatory Role of the Vasculature during Mucosal Inflammation

3.5.1. Vasculature and Innate Immunity

Following the rupture of the gut–epithelial barrier, mucosal microvascular ECs are exposed to bacteria and bacterial products. ECs can then launch an innate immune reaction through the engagement of toll-like receptors (TLRs) via pathogen-associated molecular patterns (PAMPs). Gut microvascular ECs exhibit a particular TLR response pattern compared to ECs from other origins (Table 3). For instance, tolerance to lipopolysaccharide (LPS), an activator of TLR4, has been observed in HIMECs but not in HUVECs after repeated exposure [188]. TLR3 and TLR5 are expressed at the surfaces of intestinal ECs. TLR3 is involved in the anti-viral response and is constitutively expressed in HIMECs, where it can be further upregulated by IFN-γ [189]. TLR5, a receptor for flagellin, was shown to play an important role in the EC innate immune response [190,191]. Activation of TLR3 and TLR5 in HIMECs induces the upregulation of inflammatory mediators and ICAM-1, leading to leukocyte recruitment [189,192]. TLRs expressed by intestinal ECs serve as a second barrier in the case of epithelial barrier breakdown. In particular, flagellin has been described as a dominant antigen in CD [193,194,195]. In agreement with this protective function of the GVB, TLR3 and TLR5 expression on ECs is protective against colitis in mice [196,197]. Taken together, the propensity of HIMECs to develop endotoxin tolerance might prevent an excessive immune reaction from occurring in the case of luminal bacterial penetration into the mucosa, while the response to viruses and flagellin remains intact, protecting against systemic propagation.

3.5.2. Paracrine Effects of the Inflamed Vasculature

ECs are highly reactive and can themselves express and secrete inflammatory mediators upon activation [192,198,199]. Those factors can either further promote or dampen inflammation through local or systemic effects (Table 3). For example, CX3CL1 (fractalkine), a chemokine that is upregulated in ECs via the MAPK pathway upon stimulation by TNF-α, IL-1, LPS, and IFN-γ, is highly upregulated in the mucosal endothelia of IBD patients [200,201]. CX3CL1 released from ECs stimulates the adhesion and transmigration of leukocytes expressing the CX3CR1 receptor. Higher levels of circulating and infiltrating CX3CL1+ T cells have been observed during IBD [202]. Under inflammatory conditions, HIMECs are also able to express monocyte chemotactic protein 1 (MCP-1) [199]. In addition, intestinal vessels might exert angiocrine activity on epithelial cells during IBD through the release of TIMP1 and CXCL10, the latter being able to increase crypt survival [199].
Vascular ECs also secrete the C-C chemokine ligands 5 (CCL5) and 25 (CCL25) during inflammation [5]. CCL25 leads to the recruitment of immune cells expressing CCR9 [203]. Both CCR9 and CCL25 are upregulated during DSS-induced colitis, in which the CCL25-CCR9 axis exerts a protective anti-colitic effect in the intestinal mucosa by balancing different dendritic cell subsets [203]. Furthermore, during experimental colitis, NO of endothelial origin protects the intestinal mucosa of mice against inflammation by increasing the number of goblet cells and mucin production, thereby preventing luminal bacteria translocation [179].

3.6. Role of the Mesenteric Lymphatic Vasculature in IBD

Intestinal lymphatic vessels are involved in the removal of excess interstitial fluids, immune regulation, and the absorption of fatty acids by lacteal lymphatic endothelial cells [204]. During inflammation, lymphatic endothelial cells participate in the regulation of the adaptive inflammatory response and promote the resolution of inflammation through the clearance of immune cells and mediators [204]. Lymphangiogenesis is frequently observed during inflammation [205]. An increase in lymphatic vessel density has been observed in UC and CD throughout the mucosa, including in non-inflamed areas [206,207,208]. This development of the lymphatic system is thought to be a reaction to the inflammation and edema in the mucosa, aiming to dampen tissue damage by draining immune cells and excess fluid. However, similarly to blood vessels, lymph vessels show abnormal architectures and are often dysfunctional [209,210]. In CD, granulomatous structures characteristic of chronic lymphangitis have been observed in mesenteric lymph nodes and lymphatic vessels of the intestinal mucosa, and these correlate with disease activity [57,209,211]. In addition, lymphangiectasia, lymphadenopathy, and lymphatic vessel obstruction occurring during IBD compromise lymph drainage and leukocyte trafficking. This ultimately leads to edema, lymph leakage, and the deposition of adipose tissue into the mucosa, further fostering inflammation [211,212,213]. These observations are supported by the fact that a notable reduction in lymphatic contractile activity has been observed during murine experimental colitis, both locally and systematically [209,210,214]. Taken together, the lymphangiogenesis observed in IBD cannot resolve inflammation due the dysfunctionality of the newly formed lymphatics. The normalization of lymphatic function might represent an additional therapeutic approach in IBD.

3.7. Vascular Function and Extra-Intestinal Manifestations of IBD

More than one third of patients with CD and UC are affected by extraintestinal manifestations in addition to intestinal inflammation (Figure 2). The most common manifestations include thromboembolisms; hepatobiliary disorders; arthropathies; and cutaneous, pulmonary, and ocular manifestations, as well as neurological and psychosocial disturbances [14,215,216]. For example, major depressive disorder and multiple sclerosis are well-described IBD comorbidities [216,217,218]. Some of these extraintestinal manifestations correlate with flares of intestinal inflammation but others occur independently [215]. It remains unclear why only a fraction of patients with IBD present extra-intestinal manifestations.

3.7.1. Systemic Vascular Barrier Dysfunction in IBD

The GVB has been shown to lie at the heart of the gut–liver–brain axis. Increased GVB leakiness and dysfunction has been observed in patients during Salmonella infection, diet-induced nonalcoholic steatohepatitis, and metastatic colorectal cancer, leading to an impaired gut–liver axis connection [13,219,220]. The breakdown of the GVB in IBD may induce widespread low-grade vascular inflammation through the uncontrolled release of microbial products or pro-inflammatory factors into the systemic circulation, which might then compromise the vascular barrier at distant organs and result in extra-intestinal manifestations [12]. A strong link between the GVB and the blood–brain barrier (BBB) has been established in IBD. Manifestations of anxiety and depression have been reported in up to 40% of patients with active IBD (14), together with deterioration in cognitive functions [216,217,218,221,222,223,224]. Similar observations have been made in DSS-colitis models where mice displayed increased anxiety- and depression-like behaviors and alterations of the limbic system [158,225,226,227]. Carloni et al. have recently shown that after DSS challenge, there is a persistent increase in intestinal vascular permeability in treated mice, even after recovery [158]. They also observed an increased absolute number and percentage of innate immune cells in the liver and the brain, suggesting that acute intestinal inflammation quickly spreads to other organs, including the brain [158]. Interestingly, vascular permeability induced by DSS treatment was only increased transiently in the brain due to the closure of the vascular barrier in the brain choroid plexus (PVB), which was then released after discontinuation of the application of DSS. In a transgenic mouse model of the inducible closure of the PVB, animals exhibited anxiety-like behavior and a deficit in short-term memory, suggesting that PVB closure may correlate with cognitive and mental disturbances [158]. These observations are further supported by the fact that a disruption of the BBB has been observed during TNBS-induced colitis in mice [228].
The translocation of bacteria into the bloodstream during IBD-related intestinal inflammation has been proposed to contribute to extra-intestinal effects. Indeed, patients with IBD have an elevated risk of sepsis [229,230]. Nevertheless, sepsis represents a rare complication of IBD, mainly occurring after surgery [230]. In DSS-induced colitis, mice treated with the glucocorticoid budesonide, bacterial translocation to the liver, and endotoxemia have been observed following massive intestinal barrier disruption [231]. However, these effects were not observed in the transfer colitis model, or in DSS-treated mice which did not receive budesonide. Together, these data suggest that the translocation of bacteria and bacterial products is limited during IBD and experimental colitis [231].
Bacterial products, on the contrary, have been frequently detected in the circulation of patients with IBD. Bacterial DNA can be found in the blood for up to 50% of IBD patients [232], and bacterial DNA translocation represents an independent risk factor of relapse at 6 months in CD patients [233]. The presence of bacterial endotoxin and LPS-binding protein can also be detected in the circulation of IBD patients [234,235,236,237,238]. Higher serum levels of LPS were observed in patients with IBD-associated spondylarthritis compared to IBD alone, indicating that an increased translocation of bacterial products is linked to the development of extra-intestinal manifestations [236]. Recently, Carloni et al. showed that the LPS blood concentration increases only transiently after DSS challenge in mice. The fact that patients with UC have elevated serum concentrations of LPS-binding protein but not of LPS supports the idea of a transient increase [158]. The authors proposed that increased scavenging by circulating inflammatory cells might explain the transient character of the serum LPS spike.
The release of inflammatory mediators by the inflamed intestinal endothelium might also lead to systemic low-grade inflammation, resulting in vascular activation and/or barrier dysfunction at distant organs. Increased levels of circulating inflammatory cytokines (IL-6, TNF-α…), for instance, can be found in IBD patients and animals with colitis [158]. There is indeed increasing evidence that circulating pro-inflammatory mediators in IBD patients may contribute to the progression of several central nervous system (CNS) disorders [158,239,240,241].

3.7.2. Endothelial Damage and Systemic Vascular Inflammation

During IBD, local endothelial dysfunction, which can spread into systemic vascular barrier defects, might also promote generalized vascular inflammation. There is evidence linking IBD with atherosclerosis, coronary dysfunction, and an increased risk of cardiovascular (CV) morbidity and mortality [242,243,244,245,246,247,248]. Furthermore, IBD patients have an elevated risk of vasculitis, which is linked to more frequent headaches and extraintestinal symptoms [249,250]. Antibodies to endothelial cells reflect vascular injury and have been detected in the serum of patients with vasculitis. Anti-endothelial cell antibody levels are elevated in UC and CD patients compared with healthy controls [251]. In addition, increased anti-EC antibody levels were found to correlate with circulating levels of Von Willebrand factor, a marker of vascular inflammation and injury, indicating the occurrence of vasculitis during IBD. Some association between anti-endothelial cell antibody levels and disease activity has been found in both UC and CD [252,253].
Increasing evidence links intestinal microbiota dysbiosis, vascular aging, and cardiovascular diseases (the “gut–heart axis”). The passage of bacterial products into the bloodstream has been associated with increased arterial stiffness, atherosclerosis, hypertension, and cardiovascular risk in human individuals [254]. For instance, the reduction of short chain fatty acids and the increased production of trimethylamine-N-oxide, which are associated with gut dysbiosis, increase cardiovascular risk. In addition, the transmigration of LPS into the bloodstream activates vascular inflammation [254].

3.7.3. Coagulation and Thrombosis in IBD

The local increase of pro-coagulant and pro-thrombotic events in the microvasculature of inflamed intestinal tissues is associated with systemic subclinical thrombosis in patients with IBD. Markers of coagulation are elevated in the serum of IBD patients, and increased extra-intestinal thrombus formation is enhanced in the DSS-colitis model [184,255]. Thrombosis represents a significant comorbidity, and thromboembolitic events have been estimated to account for up to 25% of IBD-related deaths [256]. The risk of venous thromboembolism is particularly increased in patients with IBD during a flare up or in the presence of chronically active inflammation [257,258,259,260,261,262,263]. In comparison, the overall risk of arterial disease is only modestly increased [260].

3.8. Targeting the Vasculature in IBD Therapy

The role of the microvasculature in the pathogenesis and perpetuation of IBD is receiving increasing recognition, and it represents an attractive therapy target.
Interestingly, several anti-inflammatory drugs used in the clinical management of IBD also ameliorate vascular dysfunction, suggesting that their efficacy depends in part on their endothelium-directed effects. For instance, mesalazine (5-aminosylicyclic acid), a medication used to treat mildly to moderately severe forms of IBD, has been shown to inhibit platelet activation [264]. In addition, the anti-TNF-α neutralizing antibody infliximab, which blocks TNF-induced inflammation and has been successfully used in IBD therapy, notably improves endothelial dysfunction in CD by enhancing agonist-induced vasodilatation, by reducing thrombus formation through inhibition of the CD40/CD40L/sCD40L pathway, and by inhibiting TNF-α-induced endothelial cell permeability [265,266,267].
Anti-TNF-α therapy has allowed healthcare professionals to bridge a therapeutic gap for IBD patients who are refractory or intolerant to treatment with classic immunosuppressive agents. However, a significant proportion of patients does not respond to anti-TNF-α therapy. New approaches based on the blockade of T-cell homing have shown promising results. Here, the recruitment of T-cells through the binding of α4β7 integrins to endothelial MadCAM1 is inhibited [268,269,270]. The α4β7-integrin-specific antibody vedolizumab induces long-term remission in CD and UC, and represents a good alternative for patients with refractory disease and colonic inflammation [271,272,273]. The β7-integrin-specific antibody etrolizumab as well as anti-MadCAM1 antibodies are currently being evaluated in clinical trials [269,274,275]. Hence, the specific blockade of the interaction of T-cells with the activated MadCAM1+ endothelium is increasingly being implemented in the clinical routine.
Therapeutic strategies targeting the endothelium represent an interesting way to reduce mucosal inflammation and/or extraintestinal manifestations by normalizing the vascular function. For example, heparin has been administrated to IBD patients to prevent venous thromboembolism [276,277,278]. In addition, treatment with low-molecular-weight heparin has shown therapeutic efficiency in IBD [279,280], which was not only due to the inhibition of microvascular thromboses, but also to immuno-modulating properties (such as the suppression of neutrophil recruitment) and to an increase in mucosal recovery [281,282]. A new therapeutic approach based on the delivery of heparin via nanoparticles (NPs) has provided promising results in the TNBS-induced colitis mouse model [283]. In addition, heparin-coated human serum albumin NPs have been shown to efficiently deliver drugs into the inflamed intestine in a murine model of colitis, opening up new possibilities for combinational treatment [284]. Similarly, the treatment of microvascular lesions with Panax notoginseng attenuated inflammation and disease activity in rats with colitis [285]. In another study, the targeting of phosphatidylserine externalized by stressed ECs in capillaries of the mouse colonic mucosa using annexin-V inhibited TNBS-induced colitis [286].
Another promising strategy for vessel-directed therapy of IBD is the inhibition of the loss of GVB function during IBD. The receptor tyrosine kinase imatinib has been shown to inhibit vascular dysfunction and edema in various models [162,287,288,289]. Treatment with imatinib blocks vessel permeability and alleviates DSS-induced colitis in mice [34]. Imatinib, given in the context of chronic myeloid leukemia, has been reported to induce long-standing remission of CD [290]. Sphingosine 1-phosphate (S1P), a sphingolipid mediator, represents another potential target. S1P signals through high-affinity G protein-coupled receptors S1P1 to 5 to regulate the egress of lymphocytes from lymphoid organs and the maintenance of vascular integrity [291,292], targeting both blood and lymphatic ECs. A dysfunctional S1P signaling axis leads to pathological angiogenesis and increased vascular permeability [293,294]. Several S1P agonists including ozanimod and etrasimod have shown promising results by blocking lymphocyte recruitment and improving barrier function, and are currently being tested in phase 3 clinical trials for UC and CD [293,295,296]. In conclusion, pharmacological normalization of the vasculature could not only prevent vascular co-morbidities in IBD patients, but might also complement the standard anti-inflammatory regimens.

4. Conclusions

The endothelium lies at the heart of the inflammation circle, and the manifold changes it undergoes during activation are regulated by complex mechanisms. These mechanisms can explain in part the refractory character of IBD, but might also represent complementary anti-inflammatory therapy targets. The blockade of leukocyte recruitment by endothelial cells plays an important role in that regard. Vascular damage, hyperpermeability, and the activation of the hyperthrombic state of the inflamed vasculature also represent important contributors to inflammation, whereas the role of angiogenesis appears to be less important than initially thought. In recent years, the intestinal microvascular barrier has been shown to play a decisive role as a second barrier in the gut. The loss of its function has furthermore been found to promote inflammation and has been linked to the development of extra-intestinal manifestations of IBD. Therefore, the intestinal microvascular barrier is now emerging as a promising therapeutic target for the treatment of IBD patients.

Author Contributions

Writing—original draft preparation, N.B.-L.; writing—review and editing, N.B.-L., C.W., M.S.; funding acquisition, N.B.-L., C.W., M.S. All authors have read and agreed to the published version of the manuscript.

Funding

This work has been funded by the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation)—TRR 241-375876048 (A06 to M.S. and N.B.-L.; A09 and B01 to C.W.); TRR305-429280966 (B07 to N.B.-L.); STU 238/10-1-437201724 to M.S.; KFO 257-4-190140969 to M.S.; FOR 2438-2 280163318 to M.S.—as well as by the Interdisciplinary Center for Clinical Research (IZKF) of the Clinical Center Erlangen (ELAN-Fonds project P027 to N.B.-L., Project J19 to N.B.-L., and D34 to M.S.), by the W. Lutz Stiftung, and by the Forschungsstiftung Medizin am Universi-tätsklinikum Erlangen to M.S., by the Deutsche Forschungsgemeinschaft and Frie-drich-Alexander-Universität Erlangen-Nürnberg within the funding programme "Open Access Publication Funding”.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Zhang, Y.Z.; Li, Y.Y. Inflammatory bowel disease: Pathogenesis. World J. Gastroenterol. 2014, 20, 91–99. [Google Scholar] [CrossRef]
  2. Wallace, K.L.; Zheng, L.B.; Kanazawa, Y.; Shih, D.Q. Immunopathology of inflammatory bowel disease. World J. Gastroenterol. 2014, 20, 6–21. [Google Scholar] [CrossRef] [PubMed]
  3. Ni, J.; Wu, G.D.; Albenberg, L.; Tomov, V.T. Gut microbiota and IBD: Causation or correlation? Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 573–584. [Google Scholar] [CrossRef] [Green Version]
  4. Thoo, L.; Noti, M.; Krebs, P. Keep calm: The intestinal barrier at the interface of peace and war. Cell Death Dis. 2019, 10, 849. [Google Scholar] [CrossRef] [Green Version]
  5. Cromer, W.E.; Mathis, J.M.; Granger, D.N.; Chaitanya, G.V.; Alexander, J.S. Role of the endothelium in inflammatory bowel diseases. World J. Gastroenterol. 2011, 17, 578–593. [Google Scholar] [CrossRef] [PubMed]
  6. Kvietys, P.R. Integrated Systems Physiology: From Molecule to Function. In The Gastrointestinal Circulation; Morgan & Claypool Life Sciences: San Rafael, CA, USA, 2010. [Google Scholar]
  7. Geboes, K.; Geboes, K.P.; Maleux, G. Vascular anatomy of the gastrointestinal tract. Best Pract. Res. Clin. Gastroenterol. 2001, 15, 1–14. [Google Scholar] [CrossRef]
  8. Arfors, K.E.; Rutili, G.; Svensjö, E. Microvascular transport of macromolecules in normal and inflammatory conditions. Acta Physiol. Scand. Suppl. 1979, 463, 93–103. [Google Scholar]
  9. Hasibeder, W. Gastrointestinal microcirculation: Still a mystery? Br. J. Anaesth. 2010, 105, 393–396. [Google Scholar] [CrossRef] [Green Version]
  10. Miller, M.J.; McDole, J.R.; Newberry, R.D. Microanatomy of the intestinal lymphatic system. Ann. N. Y. Acad. Sci. 2010, 1207 (Suppl. 1), E21–E28. [Google Scholar] [CrossRef] [Green Version]
  11. Cifarelli, V.; Eichmann, A. The Intestinal Lymphatic System: Functions and Metabolic Implications. Cell. Mol. Gastroenterol. Hepatol. 2019, 7, 503–513. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Brescia, P.; Rescigno, M. The gut vascular barrier: A new player in the gut-liver-brain axis. Trends Mol. Med. 2021, 27, 844–855. [Google Scholar] [CrossRef]
  13. Spadoni, I.; Zagato, E.; Bertocchi, A.; Paolinelli, R.; Hot, E.; Di Sabatino, A.; Caprioli, F.; Bottiglieri, L.; Oldani, A.; Viale, G.; et al. A gut-vascular barrier controls the systemic dissemination of bacteria. Science 2015, 350, 830–834. [Google Scholar] [CrossRef] [PubMed]
  14. Spadoni, I.; Fornasa, G.; Rescigno, M. Organ-specific protection mediated by cooperation between vascular and epithelial barriers. Nat. Rev. Immunol. 2017, 17, 761–773. [Google Scholar] [CrossRef] [PubMed]
  15. Bush, T.G.; Savidge, T.C.; Freeman, T.C.; Cox, H.J.; Campbell, E.A.; Mucke, L.; Johnson, M.H.; Sofroniew, M.V. Fulminant jejuno-ileitis following ablation of enteric glia in adult transgenic mice. Cell 1998, 93, 189–201. [Google Scholar] [CrossRef] [Green Version]
  16. Cornet, A.; Savidge, T.C.; Cabarrocas, J.; Deng, W.L.; Colombel, J.F.; Lassmann, H.; Desreumaux, P.; Liblau, R.S. Enterocolitis induced by autoimmune targeting of enteric glial cells: A possible mechanism in Crohn’s disease? Proc. Natl. Acad. Sci. USA 2001, 98, 13306–13311. [Google Scholar] [CrossRef] [Green Version]
  17. Kabouridis, P.S.; Lasrado, R.; McCallum, S.; Chng, S.H.; Snippert, H.J.; Clevers, H.; Pettersson, S.; Pachnis, V. Microbiota controls the homeostasis of glial cells in the gut lamina propria. Neuron 2015, 85, 289–295. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Reinhardt, C.; Bergentall, M.; Greiner, T.U.; Schaffner, F.; Ostergren-Lundén, G.; Petersen, L.C.; Ruf, W.; Bäckhed, F. Tissue factor and PAR1 promote microbiota-induced intestinal vascular remodelling. Nature 2012, 483, 627–631. [Google Scholar] [CrossRef] [Green Version]
  19. Schirbel, A.; Kessler, S.; Rieder, F.; West, G.; Rebert, N.; Asosingh, K.; McDonald, C.; Fiocchi, C. Pro-angiogenic activity of TLRs and NLRs: A novel link between gut microbiota and intestinal angiogenesis. Gastroenterology 2013, 144, 613–623.e9. [Google Scholar] [CrossRef] [Green Version]
  20. Stappenbeck, T.S.; Hooper, L.V.; Gordon, J.I. Developmental regulation of intestinal angiogenesis by indigenous microbes via Paneth cells. Proc. Natl. Acad. Sci. USA 2002, 99, 15451–15455. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Komarova, Y.A.; Kruse, K.; Mehta, D.; Malik, A.B. Protein Interactions at Endothelial Junctions and Signaling Mechanisms Regulating Endothelial Permeability. Circ. Res. 2017, 120, 179–206. [Google Scholar] [CrossRef] [Green Version]
  22. Scalise, A.A.; Kakogiannos, N.; Zanardi, F.; Iannelli, F.; Giannotta, M. The blood–brain and gut–vascular barriers: From the perspective of claudins. Tissue Barriers 2021, 9, 1926190. [Google Scholar] [CrossRef]
  23. Richards, M.; Nwadozi, E.; Pal, S.; Martinsson, P.; Kaakinen, M.; Gloger, M.; Sjöberg, E.; Koltowska, K.; Betsholtz, C.; Eklund, L.; et al. Claudin5 protects the peripheral endothelial barrier in an organ and vessel-type-specific manner. eLife 2022, 11, e78517. [Google Scholar] [CrossRef] [PubMed]
  24. Kalucka, J.; de Rooij, L.P.M.H.; Goveia, J.; Rohlenova, K.; Dumas, S.J.; Meta, E.; Conchinha, N.V.; Taverna, F.; Teuwen, L.-A.; Veys, K.; et al. Single—Cell Transcriptome Atlas of Murine Endothelial Cells. Cell 2020, 180, 764–779.e20. [Google Scholar] [CrossRef] [PubMed]
  25. Günzel, D.; Yu, A.S.L. Claudins and the modulation of tight junction permeability. Physiol. Rev. 2013, 93, 525–569. [Google Scholar] [CrossRef] [Green Version]
  26. Garcia-Hernandez, V.; Quiros, M.; Nusrat, A. Intestinal epithelial claudins: Expression and regulation in homeostasis and inflammation. Ann. N. Y. Acad. Sci. 2017, 1397, 66–79. [Google Scholar] [CrossRef]
  27. Haraldsen, G.; Kvale, D.; Lien, B.; Farstad, I.N.; Brandtzaeg, P. Cytokine-regulated expression of E-selectin, intercellular adhesion molecule-1 (ICAM-1), and vascular cell adhesion molecule-1 (VCAM-1) in human microvascular endothelial cells. J. Immunol. 1996, 156, 2558–2565. [Google Scholar] [CrossRef] [PubMed]
  28. Nilsen, E.M.; Johansen, F.E.; Jahnsen, F.L.; Lundin, K.E.A.; Scholz, T.; Brandtzaeg, P.; Haraldsen, G. Cytokine profiles of cultured microvascular endothelial cells from the human intestine. Gut 1998, 42, 635–642. [Google Scholar] [CrossRef]
  29. Binion, D.G.; Fu, S.; Ramanujam, K.S.; Chai, Y.C.; Dweik, R.A.; Drazba, J.A.; Wade, J.G.; Ziats, N.P.; Erzurum, S.C.; Wilson, K.T. iNOS expression in human intestinal microvascular endothelial cells inhibits leukocyte adhesion. Am. J. Physiol. 1998, 275, G592–G603. [Google Scholar] [CrossRef]
  30. Sessa, W.C. Molecular control of blood flow and angiogenesis: Role of nitric oxide. J. Thromb. Haemost. 2009, 7 (Suppl. 1), 35–37. [Google Scholar] [CrossRef]
  31. Pober, J.S.; Sessa, W.C. Evolving functions of endothelial cells in inflammation. Nat. Rev. Immunol. 2007, 7, 803–815. [Google Scholar] [CrossRef]
  32. Papa, A.; Scaldaferri, F.; Danese, S.; Guglielmo, S.; Roberto, I.; Bonizzi, M.; Mocci, G.; Felice, C.; Ricci, C.; Andrisani, G.; et al. Vascular involvement in inflammatory bowel disease: Pathogenesis and clinical aspects. Dig. Dis. 2008, 26, 149–155. [Google Scholar] [CrossRef]
  33. Haep, L.; Britzen-Laurent, N.; Weber, T.G.; Naschberger, E.; Schaefer, A.; Kremmer, E.; Foersch, S.; Vieth, M.; Scheuer, W.; Wirtz, S.; et al. Interferon Gamma Counteracts the Angiogenic Switch and Induces Vascular Permeability in Dextran Sulfate Sodium Colitis in Mice. Inflamm. Bowel Dis. 2015, 21, 2360–2371. [Google Scholar] [CrossRef]
  34. Langer, V.; Vivi, E.; Regensburger, D.; Winkler, T.H.; Waldner, M.J.; Rath, T.; Schmid, B.; Skottke, L.; Lee, S.; Jeon, N.L.; et al. IFN-γ drives inflammatory bowel disease pathogenesis through VE-cadherin-directed vascular barrier disruption. J. Clin. Investig. 2019, 129, 4691–4707. [Google Scholar] [CrossRef] [Green Version]
  35. Nourshargh, S.; Alon, R. Leukocyte Migration into Inflamed Tissues. Immunity 2014, 41, 694–707. [Google Scholar] [CrossRef] [Green Version]
  36. Binion, D.G.; West, G.A.; Volk, E.E.; Drazba, J.A.; Ziats, N.P.; Petras, R.E.; Fiocchi, C. Acquired increase in leucocyte binding by intestinal microvascular endothelium in inflammatory bowel disease. Lancet 1998, 352, 1742–1746. [Google Scholar] [CrossRef]
  37. Scaldaferri, F.; Sans, M.; Vetrano, S.; Correale, C.; Arena, V.; Pagano, N.; Rando, G.; Romeo, F.; Potenza, A.E.; Repici, A.; et al. The role of MAPK in governing lymphocyte adhesion to and migration across the microvasculature in inflammatory bowel disease. Eur. J. Immunol. 2009, 39, 290–300. [Google Scholar] [CrossRef] [PubMed]
  38. Fägerstam, J.P.; Whiss, P.A.; Ström, M.; Andersson, R.G. Expression of platelet P-selectin and detection of soluble P-selectin, NPY and RANTES in patients with inflammatory bowel disease. Inflamm. Res. 2000, 49, 466–472. [Google Scholar] [CrossRef] [PubMed]
  39. Fägerstam, J.P.; Whiss, P.A. Higher platelet P-selectin in male patients with inflammatory bowel disease compared to healthy males. World J. Gastroenterol. 2006, 12, 1270–1272. [Google Scholar] [CrossRef] [Green Version]
  40. Ostanin, D.V.; Bao, J.; Koboziev, I.; Gray, L.; Robinson-Jackson, S.A.; Kosloski-Davidson, M.; Price, V.H.; Grisham, M.B. T cell transfer model of chronic colitis: Concepts, considerations, and tricks of the trade. Am. J. Physiol. Gastrointest. Liver Physiol. 2009, 296, G135–G146. [Google Scholar] [CrossRef] [Green Version]
  41. Zittermann, S.I.; Issekutz, A.C. Endothelial growth factors VEGF and bFGF differentially enhance monocyte and neutrophil recruitment to inflammation. J. Leukoc. Biol. 2006, 80, 247–257. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Goebel, S.; Huang, M.; Davis, W.C.; Jennings, M.; Siahaan, T.J.; Alexander, J.S.; Kevil, C.G. VEGF-A stimulation of leukocyte adhesion to colonic microvascular endothelium: Implications for inflammatory bowel disease. Am. J. Physiol. Gastrointest. Liver Physiol. 2006, 290, G648–G654. [Google Scholar] [CrossRef]
  43. Sans, M.; Fuster, D.; Vázquez, A.; Setoain, F.J.; Piera, C.; Piqué, J.M.; Panés, J. 123Iodine-labelled anti-VCAM-1 antibody scintigraphy in the assessment of experimental colitis. Eur. J. Gastroenterol. Hepatol. 2001, 13, 31–38. [Google Scholar] [CrossRef]
  44. Soriano, A.; Salas, A.; Salas, A.; Sans, M.; Gironella, M.; Elena, M.; Anderson, D.C.; Piqué, J.M.; Panés, J. VCAM-1, but not ICAM-1 or MAdCAM-1, immunoblockade ameliorates DSS-induced colitis in mice. Lab. Investig. 2000, 80, 1541–1551. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Rijcken, E.; Mennigen, R.B.; Schaefer, S.D.; Laukoetter, M.G.; Anthoni, C.; Spiegel, H.U.; Bruewer, M.; Senninger, N.; Krieglstein, C.F. PECAM-1 (CD 31) mediates transendothelial leukocyte migration in experimental colitis. Am. J. Physiol. Gastrointest. Liver Physiol. 2007, 293, G446–G452. [Google Scholar] [CrossRef] [PubMed]
  46. Briskin, M.; Winsor-Hines, D.; Shyjan, A.; Cochran, N.; Bloom, S.; Wilson, J.; McEvoy, L.M.; Butcher, E.C.; Kassam, N.; Mackay, C.R.; et al. Human mucosal addressin cell adhesion molecule-1 is preferentially expressed in intestinal tract and associated lymphoid tissue. Am. J. Pathol. 1997, 151, 97–110. [Google Scholar]
  47. Ogawa, H.; Binion, D.G.; Heidemann, J.; Theriot, M.; Fisher, P.J.; Johnson, N.A.; Otterson, M.F.; Rafiee, P. Mechanisms of MAdCAM-1 gene expression in human intestinal microvascular endothelial cells. Am. J. Physiol. Cell Physiol. 2005, 288, C272–C281. [Google Scholar] [CrossRef] [PubMed]
  48. Ando, T.; Jordan, P.; Wang, Y.; Itoh, M.; Joh, T.; Sasaki, M.; Elrod, J.W.; Carpenter, A.; Jennings, M.H.; Minagar, A.; et al. MAdCAM-1 expression and regulation in murine colonic endothelial cells in vitro. Inflamm. Bowel Dis. 2005, 11, 258–264. [Google Scholar] [CrossRef]
  49. Tyler, C.J.; Guzman, M.; Lundborg, L.R.; Yeasmin, S.; Zgajnar, N.; Jedlicka, P.; Bamias, G.; Rivera-Nieves, J. Antibody secreting cells are critically dependent on integrin α4β7/MAdCAM-1 for intestinal recruitment and control of the microbiota during chronic colitis. Mucosal Immunol. 2022, 15, 109–119. [Google Scholar] [CrossRef]
  50. Jackson, J.R.; Seed, M.P.; Kircher, C.H.; Willoughby, D.A.; Winkler, J.D. The codependence of angiogenesis and chronic inflammation. FASEB J. 1997, 11, 457–465. [Google Scholar] [CrossRef]
  51. Danese, S.; Sans, M.; de la Motte, C.; Graziani, C.; West, G.; Phillips, M.H.; Pola, R.; Rutella, S.; Willis, J.; Gasbarrini, A.; et al. Angiogenesis as a novel component of inflammatory bowel disease pathogenesis. Gastroenterology 2006, 130, 2060–2073. [Google Scholar] [CrossRef] [Green Version]
  52. Chidlow, J.H., Jr.; Langston, W.; Greer, J.J.; Ostanin, D.; Abdelbaqi, M.; Houghton, J.; Senthilkumar, A.; Shukla, D.; Mazar, A.P.; Grisham, M.B.; et al. Differential angiogenic regulation of experimental colitis. Am. J. Pathol. 2006, 169, 2014–2030. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Alkim, C.; Alkim, H.; Koksal, A.R.; Boga, S.; Sen, I. Angiogenesis in Inflammatory Bowel Disease. Int. J. Inflamm. 2015, 2015, 970890. [Google Scholar] [CrossRef] [Green Version]
  54. Danese, S.; Sans, M.; Spencer, D.M.; Beck, I.; Doñate, F.; Plunkett, M.L.; de la Motte, C.; Redline, R.; Shaw, D.E.; Levine, A.D.; et al. Angiogenesis blockade as a new therapeutic approach to experimental colitis. Gut 2007, 56, 855–862. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Danese, S. Inflammation and the mucosal microcirculation in inflammatory bowel disease: The ebb and flow. Curr. Opin. Gastroenterol. 2007, 23, 384–389. [Google Scholar] [CrossRef]
  56. Binion, D.G.; Rafiee, P. Is inflammatory bowel disease a vascular disease? Targeting angiogenesis improves chronic inflammation in inflammatory bowel disease. Gastroenterology 2009, 136, 400–403. [Google Scholar] [CrossRef] [PubMed]
  57. Danese, S. Role of the vascular and lymphatic endothelium in the pathogenesis of inflammatory bowel disease: ‘Brothers in arms’. Gut 2011, 60, 998–1008. [Google Scholar] [CrossRef] [PubMed]
  58. Dulic-Sills, A.; Blunden, M.J.; Mawdsley, J.; Bastin, A.J.; McAuley, D.; Griffiths, M.; Rampton, D.S.; Yaqoob, M.M.; Macey, M.G.; Agrawal, S.G. New flow cytometric technique for the evaluation of circulating endothelial progenitor cell levels in various disease groups. J. Immunol. Methods 2006, 316, 107–115. [Google Scholar] [CrossRef] [PubMed]
  59. Garolla, A.; D’Incà, R.; Checchin, D.; Biagioli, A.; De Toni, L.; Nicoletti, V.; Scarpa, M.; Bolzonello, E.; Sturniolo, G.C.; Foresta, C. Reduced endothelial progenitor cell number and function in inflammatory bowel disease: A possible link to the pathogenesis. Am. J. Gastroenterol. 2009, 104, 2500–2507. [Google Scholar] [CrossRef]
  60. Deng, X.; Szabo, S.; Chen, L.; Paunovic, B.; Khomenko, T.; Tolstanova, G.; Tarnawski, A.S.; Jones, M.K.; Sandor, Z. New cell therapy using bone marrow-derived stem cells/endothelial progenitor cells to accelerate neovascularization in healing of experimental ulcerative colitis. Curr. Pharm. Des. 2011, 17, 1643–1651. [Google Scholar] [CrossRef]
  61. Giatromanolaki, A.; Sivridis, E.; Maltezos, E.; Papazoglou, D.; Simopoulos, C.; Gatter, K.C.; Harris, A.L.; Koukourakis, M.I. Hypoxia inducible factor 1alpha and 2alpha overexpression in inflammatory bowel disease. J. Clin. Pathol. 2003, 56, 209–213. [Google Scholar] [CrossRef] [Green Version]
  62. Alkim, C.; Savas, B.; Ensari, A.; Alkim, H.; Dagli, U.; Parlak, E.; Ulker, A.; Sahin, B. Expression of p53, VEGF, microvessel density, and cyclin-D1 in noncancerous tissue of inflammatory bowel disease. Dig. Dis. Sci. 2009, 54, 1979–1984. [Google Scholar] [CrossRef] [PubMed]
  63. Alkim, C.; Sakiz, D.; Alkim, H.; Livaoglu, A.; Kendir, T.; Demirsoy, H.; Erdem, L.; Akbayir, N.; Sokmen, M. Thrombospondin-1 and VEGF in inflammatory bowel disease. Libyan J. Med. 2012, 7, 8942. [Google Scholar] [CrossRef] [PubMed]
  64. Scaldaferri, F.; Vetrano, S.; Sans, M.; Arena, V.; Straface, G.; Stigliano, E.; Repici, A.; Sturm, A.; Malesci, A.; Panes, J.; et al. VEGF-A links angiogenesis and inflammation in inflammatory bowel disease pathogenesis. Gastroenterology 2009, 136, 585–595.e5. [Google Scholar] [CrossRef] [Green Version]
  65. Griga, T.; Gutzeit, A.; Sommerkamp, C.; May, B. Increased production of vascular endothelial growth factor by peripheral blood mononuclear cells in patients with inflammatory bowel disease. Eur. J. Gastroenterol. Hepatol. 1999, 11, 175–179. [Google Scholar] [CrossRef] [PubMed]
  66. Bousvaros, A.; Leichtner, A.; Zurakowski, D.; Kwon, J.; Law, T.; Keough, K.; Fishman, S. Elevated serum vascular endothelial growth factor in children and young adults with Crohn’s disease. Dig. Dis. Sci. 1999, 44, 424–430. [Google Scholar] [CrossRef] [PubMed]
  67. Pousa, I.D.; Maté, J.; Gisbert, J.P. Angiogenesis in inflammatory bowel disease. Eur. J. Clin. Investig. 2008, 38, 73–81. [Google Scholar] [CrossRef] [PubMed]
  68. Krzystek-Korpacka, M.; Neubauer, K.; Matusiewicz, M. Platelet-derived growth factor-BB reflects clinical, inflammatory and angiogenic disease activity and oxidative stress in inflammatory bowel disease. Clin. Biochem. 2009, 42, 1602–1609. [Google Scholar] [CrossRef] [PubMed]
  69. Macé, V.; Ahluwalia, A.; Coron, E.; Le Rhun, M.; Boureille, A.; Bossard, C.; Mosnier, J.F.; Matysiak-Budnik, T.; Tarnawski, A.S. Confocal laser endomicroscopy: A new gold standard for the assessment of mucosal healing in ulcerative colitis. J. Gastroenterol. Hepatol. 2015, 30 (Suppl. 1), 85–92. [Google Scholar] [CrossRef]
  70. Ippolito, C.; Colucci, R.; Segnani, C.; Errede, M.; Girolamo, F.; Virgintino, D.; Dolfi, A.; Tirotta, E.; Buccianti, P.; Di Candio, G.; et al. Fibrotic and Vascular Remodelling of Colonic Wall in Patients with Active Ulcerative Colitis. J. Crohn’s Colitis 2016, 10, 1194–1204. [Google Scholar] [CrossRef] [Green Version]
  71. Kapsoritakis, A.; Sfiridaki, A.; Maltezos, E.; Simopoulos, K.; Giatromanolaki, A.; Sivridis, E.; Koukourakis, M.I. Vascular endothelial growth factor in inflammatory bowel disease. Int. J. Color. Dis. 2003, 18, 418–422. [Google Scholar] [CrossRef]
  72. Konno, S.; Iizuka, M.; Yukawa, M.; Sasaki, K.; Sato, A.; Horie, Y.; Nanjo, H.; Fukushima, T.; Watanabe, S. Altered expression of angiogenic factors in the VEGF-Ets-1 cascades in inflammatory bowel disease. J. Gastroenterol. 2004, 39, 931–939. [Google Scholar] [CrossRef] [PubMed]
  73. Magro, F.; Araujo, F.; Pereira, P.; Meireles, E.; Diniz-Ribeiro, M.; Velosom, F.T. Soluble selectins, sICAM, sVCAM, and angiogenic proteins in different activity groups of patients with inflammatory bowel disease. Dig. Dis. Sci. 2004, 49, 1265–1274. [Google Scholar] [CrossRef] [PubMed]
  74. Tolstanova, G.; Khomenko, T.; Deng, X.; Chen, L.; Tarnawski, A.; Ahluwalia, A.; Szabo, S.; Sandor, Z. Neutralizing anti-vascular endothelial growth factor (VEGF) antibody reduces severity of experimental ulcerative colitis in rats: Direct evidence for the pathogenic role of VEGF. J. Pharmacol. Exp. Ther. 2009, 328, 749–757. [Google Scholar] [CrossRef] [PubMed]
  75. Cromer, W.E.; Ganta, C.V.; Patel, M.; Traylor, J.; Kevil, C.G.; Alexander, J.S.; Mathis, J.M. VEGF-A isoform modulation in an preclinical TNBS model of ulcerative colitis: Protective effects of a VEGF164b therapy. J. Transl. Med. 2013, 11, 207. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Ozsoy, Z.; Ozsoy, S.; Gevrek, F.; Demir, E.; Benli, I.; Daldal, E.; Yenidogan, E. Effect of bevacizumab on acetic acid–induced ulcerative colitis in rats. J. Surg. Res. 2017, 216, 191–200. [Google Scholar] [CrossRef]
  77. Hindryckx, P.; Waeytens, A.; Laukens, D.; Peeters, H.; Van Huysse, J.; Ferdinande, L.; Carmeliet, P.; De Vos, M. Absence of placental growth factor blocks dextran sodium sulfate-induced colonic mucosal angiogenesis, increases mucosal hypoxia and aggravates acute colonic injury. Lab. Investig. 2010, 90, 566–576. [Google Scholar] [CrossRef] [Green Version]
  78. Hapani, S.; Chu, D.; Wu, S. Risk of gastrointestinal perforation in patients with cancer treated with bevacizumab: A meta-analysis. Lancet Oncol. 2009, 10, 559–568. [Google Scholar] [CrossRef]
  79. Coriat, R.; Mir, O.; Leblanc, S.; Ropert, S.; Brezault, C.; Chaussade, S.; Goldwasser, F. Feasibility of anti-VEGF agent bevacizumab in patients with Crohn’s disease. Inflamm. Bowel Dis. 2010, 17, 1632. [Google Scholar] [CrossRef]
  80. Tanaka, M.; Ishii, H.; Azuma, K.; Saisho, C.; Matsuo, N.; Imamura, Y.; Tokito, T.; Kinoshita, T.; Yamada, K.; Takedatsu, H.; et al. Ulcerative colitis in a patient with non-small-cell lung cancer receiving bevacizumab. Investig. New Drugs 2015, 33, 1133–1135. [Google Scholar] [CrossRef]
  81. Herrera-Gómez, R.G.; Grecea, M.; Gallois, C.; Boige, V.; Pautier, P.; Pistilli, B.; Planchard, D.; Malka, D.; Ducreux, M.; Mir, O. Safety and Efficacy of Bevacizumab in Cancer Patients with Inflammatory Bowel Disease. Cancers 2022, 14, 2914. [Google Scholar] [CrossRef]
  82. Loriot, Y.; Boudou-Rouquette, P.; Billemont, B.; Ropert, S.; Goldwasser, F. Acute exacerbation of hemorrhagic rectocolitis during antiangiogenic therapy with sunitinib and sorafenib. Ann. Oncol. 2008, 19, 1975. [Google Scholar] [CrossRef] [PubMed]
  83. Fukunaga, S.; Mori, A.; Ohuchi, A.; Yoshioka, S.; Akiba, J.; Mistuyama, K.; Tsuruta, O.; Torimura, T. Gastrointestinal: Abdominal pain, diarrhea, and bloody stools in a patient treated for renal cell carcinoma with sunitinib. J. Gastroenterol. Hepatol. 2020, 35, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Gündoğdu, Y.; Deniz, O.C.; Saka, D.; Şişman, G.; Erdamar, S.; Köksal, İ. Sunitinib induced colitis manifesting as invasive diarrhea in a patient with renal cell carcinoma. J. Oncol. Pharm. Pract. 2022, 28, 516–518. [Google Scholar] [CrossRef] [PubMed]
  85. Guenzi, E.; Töpolt, K.; Cornali, E.; Lubeseder-Martellato, C.; Jörg, A.; Matzen, K.; Zietz, C.; Kremmer, E.; Nappi, F.; Schwemmle, M.; et al. The helical domain of GBP-1 mediates the inhibition of endothelial cell proliferation by inflammatory cytokines. EMBO J. 2001, 20, 5568–5577. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Guenzi, E.; Töpolt, K.; Lubeseder-Martellato, C.; Jörg, A.; Naschberger, E.; Benelli, R.; Albini, A.; Stürzl, M. The guanylate binding protein-1 GTPase controls the invasive and angiogenic capability of endothelial cells through inhibition of MMP-1 expression. EMBO J. 2003, 22, 3772–3782. [Google Scholar] [CrossRef]
  87. Lubeseder-Martellato, C.; Guenzi, E.; Jörg, A.; Töpolt, K.; Naschberger, E.; Kremmer, E.; Zietz, C.; Tschachler, E.; Hutzler, P.; Schwemmle, M.; et al. Guanylate-binding protein-1 expression is selectively induced by inflammatory cytokines and is an activation marker of endothelial cells during inflammatory diseases. Am. J. Pathol. 2002, 161, 1749–1759. [Google Scholar] [CrossRef] [Green Version]
  88. Cornelius, L.A.; Nehring, L.C.; Harding, E.; Bolanowski, M.; Welgus, H.G.; Kobayashi, D.K.; Pierce, R.A.; Shapiro, S.D. Matrix metalloproteinases generate angiostatin: Effects on neovascularization. J. Immunol. 1998, 161, 6845–6852. [Google Scholar] [CrossRef]
  89. Danese, S. Negative regulators of angiogenesis in inflammatory bowel disease: Thrombospondin in the spotlight. Pathobiology 2008, 75, 22–24. [Google Scholar] [CrossRef]
  90. Punekar, S.; Zak, S.; Kalter, V.G.; Dobransky, L.; Punekar, I.; Lawler, J.W.; Gutierrez, L.S. Thrombospondin 1 and its mimetic peptide ABT-510 decrease angiogenesis and inflammation in a murine model of inflammatory bowel disease. Pathobiology 2008, 75, 9–21. [Google Scholar] [CrossRef]
  91. Sandor, Z.; Deng, X.M.; Khomenko, T.; Tarnawski, A.S.; Szabo, S. Altered angiogenic balance in ulcerative colitis: A key to impaired healing? Biochem. Biophys. Res. Commun. 2006, 350, 147–150. [Google Scholar] [CrossRef]
  92. Singh, U.P.; Singh, N.P.; Murphy, E.A.; Price, R.L.; Fayad, R.; Nagarkatti, M.; Nagarkatti, P.S. Chemokine and cytokine levels in inflammatory bowel disease patients. Cytokine 2016, 77, 44–49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Ozawa, C.R.; Banfi, A.; Glazer, N.L.; Thurston, G.; Springer, M.L.; Kraft, P.E.; McDonald, D.M.; Blau, H.M. Microenvironmental VEGF concentration, not total dose, determines a threshold between normal and aberrant angiogenesis. J. Clin. Investig. 2004, 113, 516–527. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Xing, S.; Luo, Y.; Liu, Z.; Bu, P.; Duan, H.; Liu, D.; Wang, P.; Yang, J.; Song, L.; Feng, J.; et al. Targeting endothelial CD146 attenuates colitis and prevents colitis-associated carcinogenesis. Am. J. Pathol. 2014, 184, 1604–1616. [Google Scholar] [CrossRef] [PubMed]
  95. Yan, H.; Zhang, C.; Wang, Z.; Tu, T.; Duan, H.; Luo, Y.; Feng, J.; Liu, F.; Yan, X. CD146 is required for VEGF-C-induced lymphatic sprouting during lymphangiogenesis. Sci. Rep. 2017, 7, 7442. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Tsiolakidou, G.; Koutroubakis, I.E.; Tzardi, M.; Kouroumalis, E.A. Increased expression of VEGF and CD146 in patients with inflammatory bowel disease. Dig. Liver Dis. 2008, 40, 673–679. [Google Scholar] [CrossRef]
  97. Danese, S.; de la Motte, C.; Sturm, A.; Vogel, J.D.; West, G.A.; Strong, S.A.; Katz, J.A.; Fiocchi, C. Platelets trigger a CD40-dependent inflammatory response in the microvasculature of inflammatory bowel disease patients. Gastroenterology 2003, 124, 1249–1264. [Google Scholar] [CrossRef]
  98. Vogel, J.D.; West, G.A.; Danese, S.; De La Motte, C.; Phillips, M.H.; Strong, S.A.; Willis, J.; Fiocchi, C. CD40-mediated immune-nonimmune cell interactions induce mucosal fibroblast chemokines leading to T-cell transmigration. Gastroenterology 2004, 126, 63–80. [Google Scholar] [CrossRef] [Green Version]
  99. Danese, S.; Scaldaferri, F.; Vetrano, S.; Stefanelli, T.; Graziani, C.; Repici, A.; Ricci, R.; Straface, G.; Sgambato, A.; Malesci, A.; et al. Critical role of the CD40 CD40-ligand pathway in regulating mucosal inflammation-driven angiogenesis in inflammatory bowel disease. Gut 2007, 56, 1248–1256. [Google Scholar] [CrossRef] [Green Version]
  100. Konerding, M.A.; Turhan, A.; Ravnic, D.J.; Lin, M.; Fuchs, C.; Secomb, T.W.; Tsuda, A.; Mentzer, S.J. Inflammation-induced intussusceptive angiogenesis in murine colitis. Anat. Rec. 2010, 293, 849–857. [Google Scholar] [CrossRef] [Green Version]
  101. Djonov, V.; Baum, O.; Burri, P.H. Vascular remodeling by intussusceptive angiogenesis. Cell Tissue Res. 2003, 314, 107–117. [Google Scholar] [CrossRef]
  102. Burri, P.H.; Hlushchuk, R.; Djonov, V. Intussusceptive angiogenesis: Its emergence, its characteristics, and its significance. Dev. Dyn. 2004, 231, 474–488. [Google Scholar] [CrossRef]
  103. Styp-Rekowska, B.; Hlushchuk, R.; Pries, A.R.; Djonov, V. Intussusceptive angiogenesis: Pillars against the blood flow. Acta Physiol. 2011, 202, 213–223. [Google Scholar] [CrossRef] [PubMed]
  104. Hlushchuk, R.; Styp-Rekowska, B.; Dzambazi, J.; Wnuk, M.; Huynh-Do, U.; Makanya, A.; Djonov, V. Endoglin inhibition leads to intussusceptive angiogenesis via activation of factors related to COUP-TFII signaling pathway. PLoS ONE 2017, 12, e0182813. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Groppa, E.; Brkic, S.; Uccelli, A.; Wirth, G.; Korpisalo-Pirinen, P.; Filippova, M.; Dasen, B.; Sacchi, V.; Muraro, M.G.; Trani, M.; et al. EphrinB2/EphB4 signaling regulates non-sprouting angiogenesis by VEGF. EMBO Rep. 2018, 19, e45054. [Google Scholar] [CrossRef] [PubMed]
  106. Ackermann, M.; Tsuda, A.; Secomb, T.W.; Mentzer, S.J.; Konerding, M.A. Intussusceptive remodeling of vascular branch angles in chemically-induced murine colitis. Microvasc. Res. 2013, 87, 75–82. [Google Scholar] [CrossRef] [Green Version]
  107. Esteban, S.; Clemente, C.; Koziol, A.; Gonzalo, P.; Rius, C.; Martínez, F.; Linares, P.M.; Chaparro, M.; Urzainqui, A.; Andrés, V.; et al. Endothelial MT1-MMP targeting limits intussusceptive angiogenesis and colitis via TSP1/nitric oxide axis. EMBO Mol. Med. 2020, 12, e10862. [Google Scholar] [CrossRef]
  108. Betto, T.; Amano, H.; Ito, Y.; Eshima, K.; Yoshida, T.; Matsui, Y.; Yamane, S.; Inoue, T.; Otaka, F.; Kobayashi, K.; et al. Vascular endothelial growth factor receptor 1 tyrosine kinase signaling facilitates healing of DSS-induced colitis by accumulation of Tregs in ulcer area. Biomed. Pharm. 2019, 111, 131–141. [Google Scholar] [CrossRef]
  109. Yoshimi, K.; Tanaka, T.; Serikawa, T.; Kuramoto, T. Tumor suppressor APC protein is essential in mucosal repair from colonic inflammation through angiogenesis. Am. J. Pathol. 2013, 182, 1263–1274. [Google Scholar] [CrossRef]
  110. Jerkic, M.; Peter, M.; Ardelean, D.; Fine, M.; Konerding, M.A.; Letarte, M. Dextran sulfate sodium leads to chronic colitis and pathological angiogenesis in endoglin heterozygous mice. Inflamm. Bowel Dis. 2010, 16, 1859–1870. [Google Scholar] [CrossRef] [Green Version]
  111. Ardelean, D.S.; Yin, M.; Jerkic, M.; Peter, M.; Ngan, B.; Kerbel, R.S.; Foster, F.S.; Letarte, M. Anti-VEGF therapy reduces intestinal inflammation in Endoglin heterozygous mice subjected to experimental colitis. Angiogenesis 2014, 17, 641–659. [Google Scholar] [CrossRef]
  112. Kobayashi, M.; Fukuda, M.; Nakayama, J. Role of sulfated O-glycans expressed by high endothelial venule-like vessels in pathogenesis of chronic inflammatory gastrointestinal diseases. Biol. Pharm. Bull. 2009, 32, 774–779. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Horjus Talabur Horje, C.S.; Smids, C.; Meijer, J.W.; Groenen, M.J.; Rijnders, M.K.; van Lochem, E.G.; Wahab, P.J. High endothelial venules associated with T cell subsets in the inflamed gut of newly diagnosed inflammatory bowel disease patients. Clin. Exp. Immunol. 2017, 188, 163–173. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Roosenboom, B.; Lochem, E.G.V.; Meijer, J.; Smids, C.; Nierkens, S.; Brand, E.C.; Erp, L.W.V.; Kemperman, L.; Groenen, M.J.M.; Horje, C.; et al. Development of Mucosal PNAd+ and MAdCAM-1+ Venules during Disease Course in Ulcerative Colitis. Cells 2020, 9, 891. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Kobayashi, M.; Hoshino, H.; Masumoto, J.; Fukushima, M.; Suzawa, K.; Kageyama, S.; Suzuki, M.; Ohtani, H.; Fukuda, M.; Nakayama, J. GlcNAc6ST-1-mediated decoration of MAdCAM-1 protein with L-selectin ligand carbohydrates directs disease activity of ulcerative colitis. Inflamm. Bowel Dis. 2009, 15, 697–706. [Google Scholar] [CrossRef] [Green Version]
  116. Nakai, D.; Miyake, M. The change of the electrophysiological parameters using human intestinal tissues from ulcerative colitis and Crohn’s disease. J. Pharmacol. Sci. 2022, 150, 90–93. [Google Scholar] [CrossRef]
  117. Jergens, A.E.; Parvinroo, S.; Kopper, J.; Wannemuehler, M.J. Rules of Engagement: Epithelial—Microbe Interactions and Inflammatory Bowel Disease. Front. Med. 2021, 8, 669913. [Google Scholar] [CrossRef]
  118. Neurath, M.F. Cytokines in inflammatory bowel disease. Nat. Rev. Immunol. 2014, 14, 329–342. [Google Scholar] [CrossRef]
  119. Buda, A.; Hatem, G.; Neumann, H.; D’Incà, R.; Mescoli, C.; Piselli, P.; Jackson, J.; Bruno, M.; Sturniolo, G.C. Confocal laser endomicroscopy for prediction of disease relapse in ulcerative colitis: A pilot study. J. Crohn’s Colitis 2014, 8, 304–311. [Google Scholar] [CrossRef] [Green Version]
  120. Taniguchi, T.; Inoue, A.; Okahisa, T.; Kimura, T.; Gohji, T.; Niki, M.; Kitamura, S.; Takeuchi, H.; Okamoto, K.; Kaji, M.; et al. Increased Angiogenesis and Vascular Permeability in Patient with Ulcerative Colitis. Gastrointest. Endosc. 2009, 69, AB365. [Google Scholar] [CrossRef]
  121. Oshima, T.; Laroux, F.S.; Coe, L.L.; Morise, Z.; Kawachi, S.; Bauer, P.; Grisham, M.B.; Specian, R.D.; Carter, P.; Jennings, S.; et al. Interferon-gamma and interleukin-10 reciprocally regulate endothelial junction integrity and barrier function. Microvasc. Res. 2001, 61, 130–143. [Google Scholar] [CrossRef]
  122. Bardin, N.; Reumaux, D.; Geboes, K.; Colombel, J.F.; Blot-Chabaud, M.; Sampol, J.; Duthilleul, P.; Dignat-George, F. Increased expression of CD146, a new marker of the endothelial junction in active inflammatory bowel disease. Inflamm. Bowel Dis. 2006, 12, 16–21. [Google Scholar] [CrossRef] [PubMed]
  123. Tolstanova, G.; Deng, X.; French, S.W.; Lungo, W.; Paunovic, B.; Khomenko, T.; Ahluwalia, A.; Kaplan, T.; Dacosta-Iyer, M.; Tarnawski, A.; et al. Early endothelial damage and increased colonic vascular permeability in the development of experimental ulcerative colitis in rats and mice. Lab. Investig. 2012, 92, 9–21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Laroux, F.S.; Grisham, M.B. Immunological basis of inflammatory bowel disease: Role of the microcirculation. Microcirculation 2001, 8, 283–301. [Google Scholar] [CrossRef] [PubMed]
  125. Vestweber, D.; Wessel, F.; Nottebaum, A.F. Similarities and differences in the regulation of leukocyte extravasation and vascular permeability. Semin. Immunopathol. 2014, 36, 177–192. [Google Scholar] [CrossRef] [PubMed]
  126. Wautier, J.L.; Wautier, M.P. Vascular Permeability in Diseases. Int. J. Mol. Sci. 2022, 23, 3645. [Google Scholar] [CrossRef] [PubMed]
  127. Tolstanova, G.; Khomenko, T.; Deng, X.; Szabo, S.; Sandor, Z. New molecular mechanisms of the unexpectedly complex role of VEGF in ulcerative colitis. Biochem. Biophys. Res. Commun. 2010, 399, 613–616. [Google Scholar] [CrossRef] [PubMed]
  128. Weis, S.M.; Cheresh, D.A. Pathophysiological consequences of VEGF-induced vascular permeability. Nature 2005, 437, 497–504. [Google Scholar] [CrossRef]
  129. Gavard, J.; Gutkind, J.S. VEGF controls endothelial-cell permeability by promoting the beta-arrestin-dependent endocytosis of VE-cadherin. Nat. Cell Biol. 2006, 8, 1223–1234. [Google Scholar] [CrossRef]
  130. Zhang, C.; Chen, H.; He, Q.; Luo, Y.; He, A.; Tao, A.; Yan, J. Fibrinogen/AKT/Microfilament Axis Promotes Colitis by Enhancing Vascular Permeability. Cell. Mol. Gastroenterol. Hepatol. 2021, 11, 683–696. [Google Scholar] [CrossRef]
  131. Matsumoto, K.; Yamaba, R.; Inoue, K.; Utsumi, D.; Tsukahara, T.; Amagase, K.; Tominaga, M.; Kato, S. Transient receptor potential vanilloid 4 channel regulates vascular endothelial permeability during colonic inflammation in dextran sulphate sodium-induced murine colitis. Br. J. Pharmacol. 2018, 175, 84–99. [Google Scholar] [CrossRef] [Green Version]
  132. Dejana, E.; Orsenigo, F.; Lampugnani, M.G. The role of adherens junctions and VE-cadherin in the control of vascular permeability. J. Cell Sci. 2008, 121, 2115–2122. [Google Scholar] [CrossRef] [Green Version]
  133. Vestweber, D. VE-cadherin: The major endothelial adhesion molecule controlling cellular junctions and blood vessel formation. Arter. Thromb. Vasc. Biol. 2008, 28, 223–232. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Giannotta, M.; Trani, M.; Dejana, E. VE-cadherin and endothelial adherens junctions: Active guardians of vascular integrity. Dev. Cell 2013, 26, 441–454. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Xiao, K.; Allison, D.F.; Kottke, M.D.; Summers, S.; Sorescu, G.P.; Faundez, V.; Kowalczyk, A.P. Mechanisms of VE-cadherin processing and degradation in microvascular endothelial cells. J. Biol. Chem. 2003, 278, 19199–19208. [Google Scholar] [CrossRef] [Green Version]
  136. Schulz, B.; Pruessmeyer, J.; Maretzky, T.; Ludwig, A.; Blobel, C.P.; Saftig, P.; Reiss, K. ADAM10 regulates endothelial permeability and T-Cell transmigration by proteolysis of vascular endothelial cadherin. Circ. Res. 2008, 102, 1192–1201. [Google Scholar] [CrossRef] [Green Version]
  137. Arihiro, S.; Ohtani, H.; Hiwatashi, N.; Torii, A.; Sorsa, T.; Nagura, H. Vascular smooth muscle cells and pericytes express MMP-1, MMP-9, TIMP-1 and type I procollagen in inflammatory bowel disease. Histopathology 2001, 39, 50–59. [Google Scholar] [CrossRef] [PubMed]
  138. Angelini, D.J.; Hyun, S.W.; Grigoryev, D.N.; Garg, P.; Gong, P.; Singh, I.S.; Passaniti, A.; Hasday, J.D.; Goldblum, S.E. TNF-alpha increases tyrosine phosphorylation of vascular endothelial cadherin and opens the paracellular pathway through fyn activation in human lung endothelia. Am. J. Physiol. Lung Cell. Mol. Physiol. 2006, 291, L1232–L1245. [Google Scholar] [CrossRef]
  139. Baugh, M.D.; Perry, M.J.; Hollander, A.P.; Davies, D.R.; Cross, S.S.; Lobo, A.J.; Taylor, C.J.; Evans, G.S. Matrix metalloproteinase levels are elevated in inflammatory bowel disease. Gastroenterology 1999, 117, 814–822. [Google Scholar] [CrossRef]
  140. Matusiewicz, M.; Neubauer, K.; Mierzchala-Pasierb, M.; Gamian, A.; Krzystek-Korpacka, M. Matrix metalloproteinase-9: Its interplay with angiogenic factors in inflammatory bowel diseases. Dis. Mark. 2014, 2014, 643645. [Google Scholar] [CrossRef] [Green Version]
  141. Meijer, M.J.; Mieremet-Ooms, M.A.; van der Zon, A.M.; van Duijn, W.; van Hogezand, R.A.; Sier, C.F.; Hommes, D.W.; Lamers, C.B.; Verspaget, H.W. Increased mucosal matrix metalloproteinase-1, -2, -3 and -9 activity in patients with inflammatory bowel disease and the relation with Crohn’s disease phenotype. Dig. Liver Dis. 2007, 39, 733–739. [Google Scholar] [CrossRef] [PubMed]
  142. Santana, A.; Medina, C.; Paz-Cabrera, M.C.; Díaz-Gonzalez, F.; Farré, E.; Salas, A.; Radomski, M.W.; Quintero, E. Attenuation of dextran sodium sulphate induced colitis in matrix metalloproteinase-9 deficient mice. World J. Gastroenterol. 2006, 12, 6464–6472. [Google Scholar] [CrossRef] [PubMed]
  143. Medina, C.; Santana, A.; Paz, M.C.; Díaz-Gonzalez, F.; Farre, E.; Salas, A.; Radomski, M.W.; Quintero, E. Matrix metalloproteinase-9 modulates intestinal injury in rats with transmural colitis. J. Leukoc. Biol. 2006, 79, 954–962. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Nighot, P.; Al-Sadi, R.; Rawat, M.; Guo, S.; Watterson, D.M.; Ma, T. Matrix metalloproteinase 9-induced increase in intestinal epithelial tight junction permeability contributes to the severity of experimental DSS colitis. Am. J. Physiol. Gastrointest. Liver Physiol. 2015, 309, G988–G997. [Google Scholar] [CrossRef] [Green Version]
  145. Sidibé, A.; Mannic, T.; Arboleas, M.; Subileau, M.; Gulino-Debrac, D.; Bouillet, L.; Jan, M.; Vandhuick, T.; Le Loet, X.; Vittecoq, O.; et al. Soluble VE-cadherin in rheumatoid arthritis patients correlates with disease activity: Evidence for tumor necrosis factor alpha-induced VE-cadherin cleavage. Arthritis Rheum. 2012, 64, 77–87. [Google Scholar] [CrossRef] [PubMed]
  146. Flemming, S.; Burkard, N.; Renschler, M.; Vielmuth, F.; Meir, M.; Schick, M.A.; Wunder, C.; Germer, C.T.; Spindler, V.; Waschke, J.; et al. Soluble VE-cadherin is involved in endothelial barrier breakdown in systemic inflammation and sepsis. Cardiovasc. Res. 2015, 107, 32–44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Polena, H.; Creuzet, J.; Dufies, M.; Sidibé, A.; Khalil-Mgharbel, A.; Salomon, A.; Deroux, A.; Quesada, J.L.; Roelants, C.; Filhol, O.; et al. The tyrosine-kinase inhibitor sunitinib targets vascular endothelial (VE)-cadherin: A marker of response to antitumoural treatment in metastatic renal cell carcinoma. Br. J. Cancer 2018, 118, 1179–1188. [Google Scholar] [CrossRef] [PubMed]
  148. Wallez, Y.; Cand, F.; Cruzalegui, F.; Wernstedt, C.; Souchelnytskyi, S.; Vilgrain, I.; Huber, P. Src kinase phosphorylates vascular endothelial-cadherin in response to vascular endothelial growth factor: Identification of tyrosine 685 as the unique target site. Oncogene 2007, 26, 1067–1077. [Google Scholar] [CrossRef] [Green Version]
  149. Nottebaum, A.F.; Cagna, G.; Winderlich, M.; Gamp, A.C.; Linnepe, R.; Polaschegg, C.; Filippova, K.; Lyck, R.; Engelhardt, B.; Kamenyeva, O.; et al. VE-PTP maintains the endothelial barrier via plakoglobin and becomes dissociated from VE-cadherin by leukocytes and by VEGF. J. Exp. Med. 2008, 205, 2929–2945. [Google Scholar] [CrossRef] [Green Version]
  150. Gioelli, N.; Neilson, L.J.; Wei, N.; Villari, G.; Chen, W.; Kuhle, B.; Ehling, M.; Maione, F.; Willox, S.; Brundu, S.; et al. Neuropilin 1 and its inhibitory ligand mini-tryptophanyl-tRNA synthetase inversely regulate VE-cadherin turnover and vascular permeability. Nat. Commun. 2022, 13, 4188. [Google Scholar] [CrossRef]
  151. Wong, R.K.; Baldwin, A.L.; Heimark, R.L. Cadherin-5 redistribution at sites of TNF-alpha and IFN-gamma-induced permeability in mesenteric venules. Am. J. Physiol. 1999, 276, H736–H748. [Google Scholar]
  152. Gavard, J. Endothelial permeability and VE-cadherin: A wacky comradeship. Cell Adhes. Migr. 2013, 7, 455–461. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Abu Taha, A.; Schnittler, H.J. Dynamics between actin and the VE-cadherin/catenin complex: Novel aspects of the ARP2/3 complex in regulation of endothelial junctions. Cell Adhes. Migr. 2014, 8, 125–135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Brandt, M.; Gerke, V.; Betz, T. Human endothelial cells display a rapid tensional stress increase in response to tumor necrosis factor-α. PLoS ONE 2022, 17, e0270197. [Google Scholar] [CrossRef] [PubMed]
  155. Morales-Ruiz, M.; Fulton, D.; Sowa, G.; Languino, L.R.; Fujio, Y.; Walsh, K.; Sessa, W.C. Vascular endothelial growth factor-stimulated actin reorganization and migration of endothelial cells is regulated via the serine/threonine kinase Akt. Circ. Res. 2000, 86, 892–896. [Google Scholar] [CrossRef] [PubMed]
  156. Ostler, N.; Britzen-Laurent, N.; Liebl, A.; Naschberger, E.; Lochnit, G.; Ostler, M.; Forster, F.; Kunzelmann, P.; Ince, S.; Supper, V.; et al. Gamma interferon-induced guanylate binding protein 1 is a novel actin cytoskeleton remodeling factor. Mol. Cell. Biol. 2014, 34, 196–209. [Google Scholar] [CrossRef] [Green Version]
  157. Liu, P.; Bian, Y.; Fan, Y.; Zhong, J.; Liu, Z. Protective Effect of Naringin on In Vitro Gut-Vascular Barrier Disruption of Intestinal Microvascular Endothelial Cells Induced by TNF-α. J. Agric. Food Chem. 2020, 68, 168–175. [Google Scholar] [CrossRef]
  158. Carloni, S.; Bertocchi, A.; Mancinelli, S.; Bellini, M.; Erreni, M.; Borreca, A.; Braga, D.; Giugliano, S.; Mozzarelli, A.M.; Manganaro, D.; et al. Identification of a choroid plexus vascular barrier closing during intestinal inflammation. Science 2021, 374, 439–448. [Google Scholar] [CrossRef]
  159. Armulik, A.; Abramsson, A.; Betsholtz, C. Endothelial/pericyte interactions. Circ. Res. 2005, 97, 512–523. [Google Scholar] [CrossRef] [Green Version]
  160. Ganta, V.C.; Cromer, W.; Mills, G.L.; Traylor, J.; Jennings, M.; Daley, S.; Clark, B.; Mathis, J.M.; Bernas, M.; Boktor, M.; et al. Angiopoietin-2 in experimental colitis. Inflamm. Bowel Dis. 2010, 16, 1029–1039. [Google Scholar] [CrossRef] [Green Version]
  161. Sweeney, M.; Foldes, G. It Takes Two: Endothelial-Perivascular Cell Cross-Talk in Vascular Development and Disease. Front. Cardiovasc. Med. 2018, 5, 154. [Google Scholar] [CrossRef]
  162. Armulik, A.; Genove, G.; Mae, M.; Nisancioglu, M.H.; Wallgard, E.; Niaudet, C.; He, L.; Norlin, J.; Lindblom, P.; Strittmatter, K.; et al. Pericytes regulate the blood-brain barrier. Nature 2010, 468, 557–561. [Google Scholar] [CrossRef] [Green Version]
  163. Alexander, J.S.; Chaitanya, G.V.; Grisham, M.B.; Boktor, M. Emerging roles of lymphatics in inflammatory bowel disease. Ann. N. Y. Acad. Sci. 2010, 1207 (Suppl. 1), E75–E85. [Google Scholar] [CrossRef] [PubMed]
  164. Regensburger, D.; Tenkerian, C.; Pürzer, V.; Schmid, B.; Wohlfahrt, T.; Stolzer, I.; López-Posadas, R.; Günther, C.; Waldner, M.J.; Becker, C.; et al. Matricellular Protein SPARCL1 Regulates Blood Vessel Integrity and Antagonizes Inflammatory Bowel Disease. Inflamm. Bowel Dis. 2021, 27, 1491–1502. [Google Scholar] [CrossRef] [PubMed]
  165. Suzuki, H.; Shibano, K.; Okane, M.; Kono, I.; Matsui, Y.; Yamane, K.; Kashiwagi, H. Interferon-gamma modulates messenger RNA levels of c-sis (PDGF-B chain), PDGF-A chain, and IL-1 beta genes in human vascular endothelial cells. Am. J. Pathol. 1989, 134, 35–43. [Google Scholar] [PubMed]
  166. Darden, J.; Payne, L.B.; Zhao, H.; Chappell, J.C. Excess vascular endothelial growth factor-A disrupts pericyte recruitment during blood vessel formation. Angiogenesis 2019, 22, 167–183. [Google Scholar] [CrossRef] [PubMed]
  167. Sprague, A.H.; Khalil, R.A. Inflammatory cytokines in vascular dysfunction and vascular disease. Biochem. Pharmacol. 2009, 78, 539–552. [Google Scholar] [CrossRef] [Green Version]
  168. Hatoum, O.A.; Binion, D.G.; Gutterman, D.D. Paradox of simultaneous intestinal ischaemia and hyperaemia in inflammatory bowel disease. Eur. J. Clin. Investig. 2005, 35, 599–609. [Google Scholar] [CrossRef]
  169. Binion, D.G.; Rafiee, P.; Ramanujam, K.S.; Fu, S.; Fisher, P.J.; Rivera, M.T.; Johnson, C.P.; Otterson, M.F.; Telford, G.L.; Wilson, K.T. Deficient iNOS in inflammatory bowel disease intestinal microvascular endothelial cells results in increased leukocyte adhesion. Free Radic. Biol. Med. 2000, 29, 881–888. [Google Scholar] [CrossRef]
  170. Horowitz, S.; Binion, D.G.; Nelson, V.M.; Kanaa, Y.; Javadi, P.; Lazarova, Z.; Andrekopoulos, C.; Kalyanaraman, B.; Otterson, M.F.; Rafiee, P. Increased arginase activity and endothelial dysfunction in human inflammatory bowel disease. Am. J. Physiol. Gastrointest. Liver Physiol. 2007, 292, G1323–G1336. [Google Scholar] [CrossRef] [Green Version]
  171. Hatoum, O.A.; Binion, D.G.; Otterson, M.F.; Gutterman, D.D. Acquired microvascular dysfunction in inflammatory bowel disease: Loss of nitric oxide-mediated vasodilation. Gastroenterology 2003, 125, 58–69. [Google Scholar] [CrossRef]
  172. Hatoum, O.A.; Gauthier, K.M.; Binion, D.G.; Miura, H.; Telford, G.; Otterson, M.F.; Campbell, W.B.; Gutterman, D.D. Novel mechanism of vasodilation in inflammatory bowel disease. Arter. Thromb. Vasc. Biol. 2005, 25, 2355–2361. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Thibeault, S.; Rautureau, Y.; Oubaha, M.; Faubert, D.; Wilkes, B.C.; Delisle, C.; Gratton, J.P. S-nitrosylation of beta-catenin by eNOS-derived NO promotes VEGF-induced endothelial cell permeability. Mol. Cell 2010, 39, 468–476. [Google Scholar] [CrossRef] [PubMed]
  174. Di Lorenzo, A.; Lin, M.I.; Murata, T.; Landskroner-Eiger, S.; Schleicher, M.; Kothiya, M.; Iwakiri, Y.; Yu, J.; Huang, P.L.; Sessa, W.C. eNOS-derived nitric oxide regulates endothelial barrier function through VE-cadherin and Rho GTPases. J. Cell Sci. 2013, 126, 5541–5552. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Seerapu, H.; Subramaniam, G.P.; Majumder, S.; Sinha, S.; Bisana, S.; Mahajan, S.; Kolluru, G.K.; Muley, A.; Siamwala, J.H.; Illavazagan, G.; et al. Inhibition of dynamin-2 confers endothelial barrier dysfunctions by attenuating nitric oxide production. Cell Biol. Int. 2010, 34, 755–761. [Google Scholar] [CrossRef] [PubMed]
  176. Beck, P.L.; Xavier, R.; Wong, J.; Ezedi, I.; Mashimo, H.; Mizoguchi, A.; Mizoguchi, E.; Bhan, A.K.; Podolsky, D.K. Paradoxical roles of different nitric oxide synthase isoforms in colonic injury. Am. J. Physiol. Gastrointest. Liver Physiol. 2004, 286, G137–G147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Hokari, R.; Kato, S.; Matsuzaki, K.; Kuroki, M.; Iwai, A.; Kawaguchi, A.; Nagao, S.; Miyahara, T.; Itoh, K.; Sekizuka, E.; et al. Reduced sensitivity of inducible nitric oxide synthase-deficient mice to chronic colitis. Free Radic. Biol. Med. 2001, 31, 153–163. [Google Scholar] [CrossRef]
  178. Sasaki, M.; Bharwani, S.; Jordan, P.; Elrod, J.W.; Grisham, M.B.; Jackson, T.H.; Lefer, D.J.; Alexander, J.S. Increased disease activity in eNOS-deficient mice in experimental colitis. Free Radic. Biol. Med. 2003, 35, 1679–1687. [Google Scholar] [CrossRef]
  179. Vallance, B.A.; Dijkstra, G.; Qiu, B.; van der Waaij, L.A.; van Goor, H.; Jansen, P.L.; Mashimo, H.; Collins, S.M. Relative contributions of NOS isoforms during experimental colitis: Endothelial-derived NOS maintains mucosal integrity. Am. J. Physiol. Gastrointest. Liver Physiol. 2004, 287, G865–G874. [Google Scholar] [CrossRef]
  180. Collins, C.E.; Rampton, D.S.; Rogers, J.; Williams, N.S. Platelet aggregation and neutrophil sequestration in the mesenteric circulation in inflammatory bowel disease. Eur. J. Gastroenterol. Hepatol. 1997, 9, 1213–1217. [Google Scholar]
  181. Tekelioglu, Y.; Uzun, H.; Sisman, G. Activated platelets in patients suffering from inflammatory bowel disease. Bratisl. Lek. Listy 2014, 115, 83–85. [Google Scholar] [CrossRef] [Green Version]
  182. Koutroubakis, I.E.; Theodoropoulou, A.; Xidakis, C.; Sfiridaki, A.; Notas, G.; Kolios, G.; Kouroumalis, E.A. Association between enhanced soluble CD40 ligand and prothrombotic state in inflammatory bowel disease. Eur. J. Gastroenterol. Hepatol. 2004, 16, 1147–1152. [Google Scholar] [CrossRef] [PubMed]
  183. Danese, S.; Katz, J.A.; Saibeni, S.; Papa, A.; Gasbarrini, A.; Vecchi, M.; Fiocchi, C. Activated platelets are the source of elevated levels of soluble CD40 ligand in the circulation of inflammatory bowel disease patients. Gut 2003, 52, 1435–1441. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Yoshida, H.; Russell, J.; Stokes, K.Y.; Yilmaz, C.E.; Esmon, C.T.; Granger, D.N. Role of the protein C pathway in the extraintestinal thrombosis associated with murine colitis. Gastroenterology 2008, 135, 882–888. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Scaldaferri, F.; Sans, M.; Vetrano, S.; Graziani, C.; De Cristofaro, R.; Gerlitz, B.; Repici, A.; Arena, V.; Malesci, A.; Panes, J.; et al. Crucial role of the protein C pathway in governing microvascular inflammation in inflammatory bowel disease. J. Clin. Investig. 2007, 117, 1951–1960. [Google Scholar] [CrossRef]
  186. Faioni, E.M.; Ferrero, S.; Fontana, G.; Gianelli, U.; Ciulla, M.M.; Vecchi, M.; Saibeni, S.; Biguzzi, E.; Cordani, N.; Franchi, F.; et al. Expression of endothelial protein C receptor and thrombomodulin in the intestinal tissue of patients with inflammatory bowel disease. Crit. Care Med. 2004, 32, S266–S270. [Google Scholar] [CrossRef]
  187. Vetrano, S.; Ploplis, V.A.; Sala, E.; Sandoval-Cooper, M.; Donahue, D.L.; Correale, C.; Arena, V.; Spinelli, A.; Repici, A.; Malesci, A.; et al. Unexpected role of anticoagulant protein C in controlling epithelial barrier integrity and intestinal inflammation. Proc. Natl. Acad. Sci. USA 2011, 108, 19830–19835. [Google Scholar] [CrossRef] [Green Version]
  188. Ogawa, H.; Rafiee, P.; Heidemann, J.; Fisher, P.J.; Johnson, N.A.; Otterson, M.F.; Kalyanaraman, B.; Pritchard, K.A., Jr.; Binion, D.G. Mechanisms of endotoxin tolerance in human intestinal microvascular endothelial cells. J. Immunol. 2003, 170, 5956–5964. [Google Scholar] [CrossRef] [Green Version]
  189. Heidemann, J.; Domschke, W.; Kucharzik, T.; Maaser, C. Intestinal microvascular endothelium and innate immunity in inflammatory bowel disease: A second line of defense? Infect. Immun. 2006, 74, 5425–5432. [Google Scholar] [CrossRef] [Green Version]
  190. Maaser, C.; Heidemann, J.; von Eiff, C.; Lugering, A.; Spahn, T.W.; Binion, D.G.; Domschke, W.; Lugering, N.; Kucharzik, T. Human intestinal microvascular endothelial cells express Toll-like receptor 5: A binding partner for bacterial flagellin. J. Immunol. 2004, 172, 5056–5062. [Google Scholar] [CrossRef] [Green Version]
  191. Vijay-Kumar, M.; Aitken, J.D.; Gewirtz, A.T. Toll like receptor-5: Protecting the gut from enteric microbes. Semin. Immunopathol. 2008, 30, 11–21. [Google Scholar] [CrossRef]
  192. Heidemann, J.; Rüther, C.; Kebschull, M.; Domschke, W.; Brüwer, M.; Koch, S.; Kucharzik, T.; Maaser, C. Expression of IL-12-related molecules in human intestinal microvascular endothelial cells is regulated by TLR3. Am. J. Physiol. Gastrointest. Liver Physiol. 2007, 293, G1315–G1324. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Lodes, M.J.; Cong, Y.; Elson, C.O.; Mohamath, R.; Landers, C.J.; Targan, S.R.; Fort, M.; Hershberg, R.M. Bacterial flagellin is a dominant antigen in Crohn disease. J. Clin. Investig. 2004, 113, 1296–1306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Sitaraman, S.V.; Klapproth, J.M.; Moore, D.A., 3rd; Landers, C.; Targan, S.; Williams, I.R.; Gewirtz, A.T. Elevated flagellin-specific immunoglobulins in Crohn’s disease. Am. J. Physiol. Gastrointest. Liver Physiol. 2005, 288, G403–G406. [Google Scholar] [CrossRef] [PubMed]
  195. Targan, S.R.; Landers, C.J.; Yang, H.; Lodes, M.J.; Cong, Y.; Papadakis, K.A.; Vasiliauskas, E.; Elson, C.O.; Hershberg, R.M. Antibodies to CBir1 flagellin define a unique response that is associated independently with complicated Crohn’s disease. Gastroenterology 2005, 128, 2020–2028. [Google Scholar] [CrossRef]
  196. Vijay-Kumar, M.; Sanders, C.J.; Taylor, R.T.; Kumar, A.; Aitken, J.D.; Sitaraman, S.V.; Neish, A.S.; Uematsu, S.; Akira, S.; Williams, I.R.; et al. Deletion of TLR5 results in spontaneous colitis in mice. J. Clin. Investig. 2007, 117, 3909–3921. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  197. Vijay-Kumar, M.; Wu, H.; Aitken, J.; Kolachala, V.L.; Neish, A.S.; Sitaraman, S.V.; Gewirtz, A.T. Activation of toll-like receptor 3 protects against DSS-induced acute colitis. Inflamm. Bowel Dis. 2007, 13, 856–864. [Google Scholar] [CrossRef]
  198. Hu, G.; Xue, J.; Duan, H.; Yang, Z.; Gao, L.; Luo, H.; Mu, X.; Cui, S. IFN-γ induces IFN-α and IFN-β expressions in cultured rat intestinal mucosa microvascular endothelial cells. Immunopharmacol. Immunotoxicol. 2010, 32, 656–662. [Google Scholar] [CrossRef]
  199. Stürzl, M.; Kunz, M.; Krug, S.M.; Naschberger, E. Angiocrine Regulation of Epithelial Barrier Integrity in Inflammatory Bowel Disease. Front. Med. 2021, 8, 643607. [Google Scholar] [CrossRef]
  200. Imaizumi, T.; Yoshida, H.; Satoh, K. Regulation of CX3CL1/fractalkine expression in endothelial cells. J. Atheroscler. Thromb. 2004, 11, 15–21. [Google Scholar] [CrossRef] [Green Version]
  201. Nishimura, M.; Kuboi, Y.; Muramoto, K.; Kawano, T.; Imai, T. Chemokines as novel therapeutic targets for inflammatory bowel disease. Ann. N. Y. Acad. Sci. 2009, 1173, 350–356. [Google Scholar] [CrossRef]
  202. Sans, M.; Danese, S.; de la Motte, C.; de Souza, H.S.; Rivera-Reyes, B.M.; West, G.A.; Phillips, M.; Katz, J.A.; Fiocchi, C. Enhanced recruitment of CX3CR1+ T cells by mucosal endothelial cell-derived fractalkine in inflammatory bowel disease. Gastroenterology 2007, 132, 139–153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  203. Wu, X.; Sun, M.; Yang, Z.; Lu, C.; Wang, Q.; Wang, H.; Deng, C.; Liu, Y.; Yang, Y. The Roles of CCR9/CCL25 in Inflammation and Inflammation-Associated Diseases. Front. Cell Dev. Biol. 2021, 9, 686548. [Google Scholar] [CrossRef] [PubMed]
  204. Bernier-Latmani, J.; Petrova, T.V. Intestinal lymphatic vasculature: Structure, mechanisms and functions. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 510–526. [Google Scholar] [CrossRef]
  205. Kim, H.; Kataru, R.P.; Koh, G.Y. Inflammation-associated lymphangiogenesis: A double-edged sword? J. Clin. Investig. 2014, 124, 936–942. [Google Scholar] [CrossRef]
  206. Geleff, S.; Schoppmann, S.F.; Oberhuber, G. Increase in podoplanin-expressing intestinal lymphatic vessels in inflammatory bowel disease. Virchows Arch. 2003, 442, 231–237. [Google Scholar] [CrossRef] [PubMed]
  207. Fogt, F.; Pascha, T.L.; Zhang, P.J.; Gausas, R.E.; Rahemtulla, A.; Zimmerman, R.L. Proliferation of D2-40-expressing intestinal lymphatic vessels in the lamina propria in inflammatory bowel disease. Int. J. Mol. Med. 2004, 13, 211–214. [Google Scholar] [CrossRef]
  208. Rahier, J.F.; De Beauce, S.; Dubuquoy, L.; Erdual, E.; Colombel, J.F.; Jouret-Mourin, A.; Geboes, K.; Desreumaux, P. Increased lymphatic vessel density and lymphangiogenesis in inflammatory bowel disease. Aliment. Pharmacol. Ther. 2011, 34, 533–543. [Google Scholar] [CrossRef] [PubMed]
  209. Wu, T.F.; MacNaughton, W.K.; von der Weid, P.Y. Lymphatic vessel contractile activity and intestinal inflammation. Mem. Inst. Oswaldo Cruz 2005, 100 (Suppl. 1), 107–110. [Google Scholar] [CrossRef] [Green Version]
  210. Von Der Weid, P.Y.; Rehal, S. Lymphatic pump function in the inflamed gut. Ann. N. Y. Acad. Sci. 2010, 1207 (Suppl. 1), E69–E74. [Google Scholar] [CrossRef]
  211. Nikolakis, D.; de Voogd, F.A.E.; Pruijt, M.J.; Grootjans, J.; van de Sande, M.G.; D’Haens, G.R. The Role of the Lymphatic System in the Pathogenesis and Treatment of Inflammatory Bowel Disease. Int. J. Mol. Sci. 2022, 23, 1854. [Google Scholar] [CrossRef]
  212. Zhang, L.; Ocansey, D.K.W.; Liu, L.; Olovo, C.V.; Zhang, X.; Qian, H.; Xu, W.; Mao, F. Implications of lymphatic alterations in the pathogenesis and treatment of inflammatory bowel disease. Biomed Pharm. 2021, 140, 111752. [Google Scholar] [CrossRef]
  213. von der Weid, P.Y.; Rainey, K.J. Review article: Lymphatic system and associated adipose tissue in the development of inflammatory bowel disease. Aliment. Pharmacol. Ther. 2010, 32, 697–711. [Google Scholar] [CrossRef] [PubMed]
  214. Agollah, G.D.; Wu, G.; Peng, H.L.; Kwon, S. Dextran sulfate sodium-induced acute colitis impairs dermal lymphatic function in mice. World J. Gastroenterol. 2015, 21, 12767–12777. [Google Scholar] [CrossRef] [PubMed]
  215. Ott, C.; Schölmerich, J. Extraintestinal manifestations and complications in IBD. Nat. Rev. Gastroenterol. Hepatol. 2013, 10, 585–595. [Google Scholar] [CrossRef] [PubMed]
  216. Navabi, S.; Gorrepati, V.S.; Yadav, S.; Chintanaboina, J.; Maher, S.; Demuth, P.; Stern, B.; Stuart, A.; Tinsley, A.; Clarke, K.; et al. Influences and Impact of Anxiety and Depression in the Setting of Inflammatory Bowel Disease. Inflamm. Bowel Dis. 2018, 24, 2303–2308. [Google Scholar] [CrossRef] [PubMed]
  217. Gupta, G.; Gelfand, J.M.; Lewis, J.D. Increased Risk for Demyelinating Diseases in Patients With Inflammatory Bowel Disease. Gastroenterology 2005, 129, 819–826. [Google Scholar] [CrossRef]
  218. Ferro, J.M.; Oliveira, S.N.; Correia, L. Neurologic manifestations of inflammatory bowel diseases. Handb. Clin. Neurol. 2014, 120, 595–605. [Google Scholar] [CrossRef]
  219. Mouries, J.; Brescia, P.; Silvestri, A.; Spadoni, I.; Sorribas, M.; Wiest, R.; Mileti, E.; Galbiati, M.; Invernizzi, P.; Adorini, L.; et al. Microbiota-driven gut vascular barrier disruption is a prerequisite for non-alcoholic steatohepatitis development. J. Hepatol. 2019, 71, 1216–1228. [Google Scholar] [CrossRef] [Green Version]
  220. Bertocchi, A.; Carloni, S.; Ravenda, P.S.; Bertalot, G.; Spadoni, I.; Lo Cascio, A.; Gandini, S.; Lizier, M.; Braga, D.; Asnicar, F.; et al. Gut vascular barrier impairment leads to intestinal bacteria dissemination and colorectal cancer metastasis to liver. Cancer Cell 2021, 39, 708–724.e11. [Google Scholar] [CrossRef]
  221. Golan, D.; Gross, B.; Miller, A.; Klil-Drori, S.; Lavi, I.; Shiller, M.; Honigman, S.; Almog, R.; Segol, O. Cognitive Function of Patients with Crohn’s Disease is Associated with Intestinal Disease Activity. Inflamm. Bowel Dis. 2016, 22, 364–371. [Google Scholar] [CrossRef] [Green Version]
  222. Byrne, G.; Rosenfeld, G.; Leung, Y.; Qian, H.; Raudzus, J.; Nunez, C.; Bressler, B. Prevalence of Anxiety and Depression in Patients with Inflammatory Bowel Disease. Can. J. Gastroenterol. Hepatol. 2017, 2017, 6496727. [Google Scholar] [CrossRef] [Green Version]
  223. Tadin Hadjina, I.; Zivkovic, P.M.; Matetic, A.; Rusic, D.; Vilovic, M.; Bajo, D.; Puljiz, Z.; Tonkic, A.; Bozic, J. Impaired neurocognitive and psychomotor performance in patients with inflammatory bowel disease. Sci. Rep. 2019, 9, 13740. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  224. Cluny, N.L.; Nyuyki, K.D.; Almishri, W.; Griffin, L.; Lee, B.H.; Hirota, S.A.; Pittman, Q.J.; Swain, M.G.; Sharkey, K.A. Recruitment of α4β7 monocytes and neutrophils to the brain in experimental colitis is associated with elevated cytokines and anxiety-like behavior. J. Neuroinflammation 2022, 19, 73. [Google Scholar] [CrossRef] [PubMed]
  225. Do, J.; Woo, J. From Gut to Brain: Alteration in Inflammation Markers in the Brain of Dextran Sodium Sulfate-induced Colitis Model Mice. Clin. Psychopharmacol. Neurosci. 2018, 16, 422–433. [Google Scholar] [CrossRef] [PubMed]
  226. Nyuyki, K.D.; Cluny, N.L.; Swain, M.G.; Sharkey, K.A.; Pittman, Q.J. Altered Brain Excitability and Increased Anxiety in Mice With Experimental Colitis: Consideration of Hyperalgesia and Sex Differences. Front. Behav. Neurosci. 2018, 12, 58. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  227. Dempsey, E.; Abautret-Daly, Á.; Docherty, N.G.; Medina, C.; Harkin, A. Persistent central inflammation and region specific cellular activation accompany depression- and anxiety-like behaviours during the resolution phase of experimental colitis. Brain Behav. Immun. 2019, 80, 616–632. [Google Scholar] [CrossRef]
  228. Natah, S.S.; Mouihate, A.; Pittman, Q.J.; Sharkey, K.A. Disruption of the blood-brain barrier during TNBS colitis. Neurogastroenterol. Motil. 2005, 17, 433–446. [Google Scholar] [CrossRef]
  229. Bernstein, C.N.; Nugent, Z.; Targownik, L.E.; Singh, H.; Lix, L.M. Predictors and risks for death in a population-based study of persons with IBD in Manitoba. Gut 2015, 64, 1403–1411. [Google Scholar] [CrossRef]
  230. Ludvigsson, J.F.; Holmgren, J.; Grip, O.; Halfvarson, J.; Askling, J.; Sachs, M.C.; Olén, O. Adult-onset inflammatory bowel disease and rate of serious infections compared to the general population: A nationwide register-based cohort study 2002-2017. Scand. J. Gastroenterol. 2021, 56, 1152–1162. [Google Scholar] [CrossRef]
  231. Ocón, B.; Aranda, C.J.; Gámez-Belmonte, R.; Suárez, M.D.; Zarzuelo, A.; Martínez-Augustin, O.; Sánchez de Medina, F. The glucocorticoid budesonide has protective and deleterious effects in experimental colitis in mice. Biochem. Pharm. 2016, 116, 73–88. [Google Scholar] [CrossRef]
  232. Gutiérrez, A.; Francés, R.; Amorós, A.; Zapater, P.; Garmendia, M.; Ndongo, M.; Cano, R.; Jover, R.; Such, J.; Perez-Mateo, M. Cytokine association with bacterial DNA in serum of patients with inflammatory bowel disease. Inflamm. Bowel Dis. 2009, 15, 508–514. [Google Scholar] [CrossRef] [PubMed]
  233. Gutiérrez, A.; Zapater, P.; Juanola, O.; Sempere, L.; Garcia, M.; Laveda, R.; Martinez, A.; Scharl, M.; Gonzalez-Navajas, J.M.; Such, J.; et al. Gut Bacterial DNA Translocation is an Independent Risk Factor of Flare at Short Term in Patients With Crohn’s Disease. Am. J. Gastroenterol. 2016, 111, 529–540. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  234. Pasternak, B.A.; D’Mello, S.; Jurickova, I.I.; Han, X.; Willson, T.; Flick, L.; Petiniot, L.; Uozumi, N.; Divanovic, S.; Traurnicht, A.; et al. Lipopolysaccharide exposure is linked to activation of the acute phase response and growth failure in pediatric Crohn’s disease and murine colitis. Inflamm. Bowel Dis. 2010, 16, 856–869. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  235. Rojo, Ó.P.; Román, A.L.S.; Arbizu, E.A.; de la Hera Martínez, A.; Sevillano, E.R.; Martínez, A.A. Serum lipopolysaccharide-binding protein in endotoxemic patients with inflammatory bowel disease. Inflamm. Bowel Dis. 2006, 13, 269–277. [Google Scholar] [CrossRef] [PubMed]
  236. Luchetti, M.M.; Ciccia, F.; Avellini, C.; Benfaremo, D.; Rizzo, A.; Spadoni, T.; Svegliati, S.; Marzioni, D.; Santinelli, A.; Costantini, A.; et al. Gut epithelial impairment, microbial translocation and immune system activation in inflammatory bowel disease-associated spondyloarthritis. Rheumatology 2021, 60, 92–102. [Google Scholar] [CrossRef]
  237. Pastorelli, L.; Dozio, E.; Pisani, L.F.; Boscolo-Anzoletti, M.; Vianello, E.; Munizio, N.; Spina, L.; Tontini, G.E.; Peyvandi, F.; Corsi Romanelli, M.M.; et al. Procoagulatory state in inflammatory bowel diseases is promoted by impaired intestinal barrier function. Gastroenterol. Res. Pract. 2015, 2015, 189341. [Google Scholar] [CrossRef] [Green Version]
  238. Tulkens, J.; Vergauwen, G.; Van Deun, J.; Geeurickx, E.; Dhondt, B.; Lippens, L.; De Scheerder, M.A.; Miinalainen, I.; Rappu, P.; De Geest, B.G.; et al. Increased levels of systemic LPS-positive bacterial extracellular vesicles in patients with intestinal barrier dysfunction. Gut 2020, 69, 191–193. [Google Scholar] [CrossRef] [Green Version]
  239. Marques, F.; Sousa, J.C.; Coppola, G.; Falcao, A.M.; Rodrigues, A.J.; Geschwind, D.H.; Sousa, N.; Correia-Neves, M.; Palha, J.A. Kinetic profile of the transcriptome changes induced in the choroid plexus by peripheral inflammation. J. Cereb. Blood Flow Metab. 2009, 29, 921–932. [Google Scholar] [CrossRef] [Green Version]
  240. Marques, F.; Sousa, J.C.; Coppola, G.; Geschwind, D.H.; Sousa, N.; Palha, J.A.; Correia-Neves, M. The choroid plexus response to a repeated peripheral inflammatory stimulus. BMC Neurosci. 2009, 10, 135. [Google Scholar] [CrossRef] [Green Version]
  241. Neuendorf, R.; Harding, A.; Stello, N.; Hanes, D.; Wahbeh, H. Depression and anxiety in patients with Inflammatory Bowel Disease: A systematic review. J. Psychosom. Res. 2016, 87, 70–80. [Google Scholar] [CrossRef]
  242. Aloi, M.; Tromba, L.; Di Nardo, G.; Dilillo, A.; Del Giudice, E.; Marocchi, E.; Viola, F.; Civitelli, F.; Berni, A.; Cucchiara, S. Premature subclinical atherosclerosis in pediatric inflammatory bowel disease. J. Pediatr. 2012, 161, 589–594.e1. [Google Scholar] [CrossRef] [PubMed]
  243. Aloi, M.; Tromba, L.; Rizzo, V.; D’Arcangelo, G.; Dilillo, A.; Blasi, S.; Civitelli, F.; Kiltzanidi, D.; Redler, A.; Viola, F. Aortic Intima-Media Thickness as an Early Marker of Atherosclerosis in Children With Inflammatory Bowel Disease. J. Pediatr. Gastroenterol. Nutr. 2015, 61, 41–46. [Google Scholar] [CrossRef] [PubMed]
  244. Caliskan, Z.; Gokturk, H.S.; Caliskan, M.; Gullu, H.; Ciftci, O.; Ozgur, G.T.; Guven, A.; Selcuk, H. Impaired coronary microvascular and left ventricular diastolic function in patients with inflammatory bowel disease. Microvasc. Res. 2015, 97, 25–30. [Google Scholar] [CrossRef]
  245. Cappello, M.; Licata, A.; Calvaruso, V.; Bravatà, I.; Aiello, A.; Torres, D.; Della Corte, V.; Tuttolomondo, A.; Perticone, M.; Licata, G.; et al. Increased expression of markers of early atherosclerosis in patients with inflammatory bowel disease. Eur. J. Intern. Med. 2017, 37, 83–89. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  246. Erolu, E.; Polat, E. Cardiac functions and aortic elasticity in children with inflammatory bowel disease: Effect of age at disease onset. Cardiol. Young 2020, 30, 313–317. [Google Scholar] [CrossRef]
  247. Triantafyllou, C.; Nikolaou, M.; Ikonomidis, I.; Bamias, G.; Papaconstantinou, I. Endothelial and Cardiac Dysfunction in Inflammatory Bowel Diseases: Does Treatment Modify the Inflammatory Load on Arterial and Cardiac Structure and Function? Curr. Vasc. Pharmacol. 2020, 18, 27–37. [Google Scholar] [CrossRef]
  248. Kakuta, K.; Dohi, K.; Yamamoto, T.; Fujimoto, N.; Shimoyama, T.; Umegae, S.; Ito, M. Coronary Microvascular Dysfunction Restored After Surgery in Inflammatory Bowel Disease: A Prospective Observational Study. J. Am. Heart Assoc. 2021, 10, e019125. [Google Scholar] [CrossRef]
  249. Sy, A.; Khalidi, N.; Dehghan, N.; Barra, L.; Carette, S.; Cuthbertson, D.; Hoffman, G.S.; Koening, C.L.; Langford, C.A.; McAlear, C.; et al. Vasculitis in patients with inflammatory bowel diseases: A study of 32 patients and systematic review of the literature. Semin. Arthritis Rheum. 2016, 45, 475–482. [Google Scholar] [CrossRef] [Green Version]
  250. Ho, T.; Orenstein, L.A.V.; Boos, M.D.; White, K.P.; Fett, N. Cutaneous Small-Vessel Vasculitis in Two Children with Inflammatory Bowel Disease: Case Series and Review of the Literature. Pediatr. Dermatol. 2017, 34, e235–e240. [Google Scholar] [CrossRef]
  251. Romas, E.; Paspaliaris, B.; d’Apice, A.J.; Elliott, P.R. Autoantibodies to neutrophil cytoplasmic (ANCA) and endothelial cell surface antigens (AECA) in chronic inflammatory bowel disease. Aust. N. Z. J. Med. 1992, 22, 652–659. [Google Scholar] [CrossRef]
  252. Stevens, T.R.; Harley, S.L.; Groom, J.S.; Cambridge, G.; Leaker, B.; Blake, D.R.; Rampton, D.S. Anti-endothelial cell antibodies in inflammatory bowel disease. Dig. Dis. Sci. 1993, 38, 426–432. [Google Scholar] [CrossRef] [PubMed]
  253. Aldebert, D.; Notteghem, B.; Reumaux, D.; Lassalle, P.; Lion, G.; Desreumaux, P.; Duthilleul, P.; Colombel, J.F. Anti-endothelial cell antibodies in sera from patients with inflammatory bowel disease. Gastroenterol. Clin. Biol. 1995, 19, 867–870. [Google Scholar] [PubMed]
  254. Agnoletti, D.; Piani, F.; Cicero, A.F.G.; Borghi, C. The Gut Microbiota and Vascular Aging: A State-of-the-Art and Systematic Review of the Literature. J. Clin. Med. 2022, 11, 3557. [Google Scholar] [CrossRef] [PubMed]
  255. Yoshida, H.; Russell, J.; Granger, D.N. Thrombin mediates the extraintestinal thrombosis associated with experimental colitis. Am. J. Physiol. Gastrointest. Liver Physiol. 2008, 295, G904–G908. [Google Scholar] [CrossRef]
  256. Papay, P.; Miehsler, W.; Tilg, H.; Petritsch, W.; Reinisch, W.; Mayer, A.; Haas, T.; Kaser, A.; Feichtenschlager, T.; Fuchssteiner, H.; et al. Clinical presentation of venous thromboembolism in inflammatory bowel disease. J. Crohn’s Colitis 2013, 7, 723–729. [Google Scholar] [CrossRef] [Green Version]
  257. Di Fabio, F.; Obrand, D.; Satin, R.; Gordon, P.H. Intra-abdominal venous and arterial thromboembolism in inflammatory bowel disease. Dis. Colon Rectum 2009, 52, 336–342. [Google Scholar] [CrossRef]
  258. Cognat, E.; Crassard, I.; Denier, C.; Vahedi, K.; Bousser, M.G. Cerebral venous thrombosis in inflammatory bowel diseases: Eight cases and literature review. Int. J. Stroke 2011, 6, 487–492. [Google Scholar] [CrossRef]
  259. Awab, A.; Elahmadi, B.; Elmoussaoui, R.; Elhijri, A.; Alilou, M.; Azzouzi, A. Cerebral venous thrombosis and inflammatory bowel disease: Reflections on pathogenesis. Colorectal. Dis. 2012, 14, 1153–1154. [Google Scholar] [CrossRef]
  260. Tan, V.P.; Chung, A.; Yan, B.P.; Gibson, P.R. Venous and arterial disease in inflammatory bowel disease. J. Gastroenterol. Hepatol. 2013, 28, 1095–1113. [Google Scholar] [CrossRef]
  261. Principi, M.; Mastrolonardo, M.; Scicchitano, P.; Gesualdo, M.; Sassara, M.; Guida, P.; Bucci, A.; Zito, A.; Caputo, P.; Albano, F.; et al. Endothelial function and cardiovascular risk in active inflammatory bowel diseases. J. Crohn’s Colitis 2013, 7, e427–e433. [Google Scholar] [CrossRef] [Green Version]
  262. Orfei, M.; Gasparetto, M.; Torrente, F. Headache and inflammatory bowel disease: Think cerebral vein! BMJ Case Rep. 2019, 12, e227228. [Google Scholar] [CrossRef] [PubMed]
  263. Rohani, P.; Taraghikhah, N.; Nasehi, M.M.; Alimadadi, H.; Assadzadeh Aghdaei, H. Cerebrovascular Events in Pediatric Inflammatory Bowel Disease: A Review of Published Cases. Pediatr. Gastroenterol. Hepatol. Nutr. 2022, 25, 180–193. [Google Scholar] [CrossRef] [PubMed]
  264. Carty, E.; MacEy, M.; Rampton, D.S. Inhibition of platelet activation by 5-aminosalicylic acid in inflammatory bowel disease. Aliment. Pharmacol. Ther. 2000, 14, 1169–1179. [Google Scholar] [CrossRef] [PubMed]
  265. Hommes, D.W.; van Dullemen, H.M.; Levi, M.; van der Ende, A.; Woody, J.; Tytgat, G.N.; van Deventer, S.J. Beneficial effect of treatment with a monoclonal anti-tumor necrosis factor-alpha antibody on markers of coagulation and fibrinolysis in patients with active Crohn’s disease. Haemostasis 1997, 27, 269–277. [Google Scholar] [CrossRef]
  266. Schinzari, F.; Armuzzi, A.; De Pascalis, B.; Mores, N.; Tesauro, M.; Melina, D.; Cardillo, C. Tumor necrosis factor-alpha antagonism improves endothelial dysfunction in patients with Crohn’s disease. Clin. Pharmacol. Ther. 2008, 83, 70–76. [Google Scholar] [CrossRef]
  267. Danese, S.; Sans, M.; Scaldaferri, F.; Sgambato, A.; Rutella, S.; Cittadini, A.; Piqué, J.M.; Panes, J.; Katz, J.A.; Gasbarrini, A.; et al. TNF-alpha blockade down-regulates the CD40/CD40L pathway in the mucosal microcirculation: A novel anti-inflammatory mechanism of infliximab in Crohn’s disease. J. Immunol. 2006, 176, 2617–2624. [Google Scholar] [CrossRef] [Green Version]
  268. Löwenberg, M.; D’Haens, G. Next-Generation Therapeutics for IBD. Curr. Gastroenterol. Rep. 2015, 17, 21. [Google Scholar] [CrossRef] [Green Version]
  269. Neurath, M.F. Current and emerging therapeutic targets for IBD. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 269–278. [Google Scholar] [CrossRef] [Green Version]
  270. Park, S.C.; Jeen, Y.T. Anti-integrin therapy for inflammatory bowel disease. World J. Gastroenterol. 2018, 24, 1868–1880. [Google Scholar] [CrossRef] [Green Version]
  271. Feagan, B.G.; Rutgeerts, P.; Sands, B.E.; Hanauer, S.; Colombel, J.F.; Sandborn, W.J.; Van Assche, G.; Axler, J.; Kim, H.J.; Danese, S.; et al. Vedolizumab as induction and maintenance therapy for ulcerative colitis. N. Engl. J. Med. 2013, 369, 699–710. [Google Scholar] [CrossRef] [Green Version]
  272. Sandborn, W.J.; Feagan, B.G.; Rutgeerts, P.; Hanauer, S.; Colombel, J.F.; Sands, B.E.; Lukas, M.; Fedorak, R.N.; Lee, S.; Bressler, B.; et al. Vedolizumab as induction and maintenance therapy for Crohn’s disease. N. Engl. J. Med. 2013, 369, 711–721. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  273. Zundler, S.; Schillinger, D.; Fischer, A.; Atreya, R.; Lopez-Posadas, R.; Watson, A.; Neufert, C.; Atreya, I.; Neurath, M.F. Blockade of alphaEbeta7 integrin suppresses accumulation of CD8+ and Th9 lymphocytes from patients with IBD in the inflamed gut in vivo. Gut 2017, 66, 1936–1948. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  274. Picardo, S.; Panaccione, R. Anti-MADCAM therapy for ulcerative colitis. Expert Opin. Biol. Ther. 2020, 20, 437–442. [Google Scholar] [CrossRef] [PubMed]
  275. Reinisch, W.; Sandborn, W.J.; Danese, S.; Hébuterne, X.; Kłopocka, M.; Tarabar, D.; Vaňásek, T.; Greguš, M.; Hellstern, P.A.; Kim, J.S.; et al. Long-term Safety and Efficacy of the Anti-MAdCAM-1 Monoclonal Antibody Ontamalimab [SHP647] for the Treatment of Ulcerative Colitis: The Open-label Study TURANDOT II. J. Crohn’s Colitis 2021, 15, 938–949. [Google Scholar] [CrossRef]
  276. Lightner, A.L.; Sklow, B.; Click, B.; Regueiro, M.; McMichael, J.J.; Jia, X.; Vaidya, P.; Delaney, C.P.; Cohen, B.; Wexner, S.D.; et al. Venous Thromboembolism in Admitted Patients with Inflammatory Bowel Disease: An Enterprise—Wide Experience of 86,000 Hospital Encounters. Dis. Colon Rectum 2022, 66, 410–418. [Google Scholar] [CrossRef]
  277. Chande, N.; Wang, Y.; McDonald, J.W.; MacDonald, J.K. Unfractionated or low-molecular weight heparin for induction of remission in ulcerative colitis. Cochrane Database Syst. Rev. 2015, 8, Cd006774. [Google Scholar] [CrossRef]
  278. Hong, L.; Chen, G.; Cai, Z.; Liu, H.; Zhang, C.; Wang, F.; Xiao, Z.; Zhong, J.; Wang, L.; Wang, Z.; et al. Balancing Microthrombosis and Inflammation via Injectable Protein Hydrogel for Inflammatory Bowel Disease. Adv. Sci. 2022, 9, e2200281. [Google Scholar] [CrossRef]
  279. Prajapati, D.N.; Newcomer, J.R.; Emmons, J.; Abu-Hajir, M.; Binion, D.G. Successful treatment of an acute flare of steroid-resistant Crohn’s colitis during pregnancy with unfractionated heparin. Inflamm. Bowel Dis. 2002, 8, 192–195. [Google Scholar] [CrossRef]
  280. Chande, N.; MacDonald, J.K.; Wang, J.J.; McDonald, J.W. Unfractionated or low molecular weight heparin for induction of remission in ulcerative colitis: A Cochrane inflammatory bowel disease and functional bowel disorders systematic review of randomized trials. Inflamm. Bowel Dis. 2011, 17, 1979–1986. [Google Scholar] [CrossRef]
  281. Day, R.; Forbes, A. Heparin, cell adhesion, and pathogenesis of inflammatory bowel disease. Lancet 1999, 354, 62–65. [Google Scholar] [CrossRef]
  282. Papa, A.; Danese, S.; Gasbarrini, A.; Gasbarrini, G. Review article: Potential therapeutic applications and mechanisms of action of heparin in inflammatory bowel disease. Aliment. Pharmacol. Ther. 2000, 14, 1403–1409. [Google Scholar] [CrossRef] [PubMed]
  283. Yazeji, T.; Moulari, B.; Beduneau, A.; Stein, V.; Dietrich, D.; Pellequer, Y.; Lamprecht, A. Nanoparticle-based delivery enhances anti-inflammatory effect of low molecular weight heparin in experimental ulcerative colitis. Drug Deliv. 2017, 24, 811–817. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  284. Zhang, S.; Cho, W.J.; Jin, A.T.; Kok, L.Y.; Shi, Y.; Heller, D.E.; Lee, Y.L.; Zhou, Y.; Xie, X.; Korzenik, J.R.; et al. Heparin-Coated Albumin Nanoparticles for Drug Combination in Targeting Inflamed Intestine. Adv. Healthc. Mater. 2020, 9, e2000536. [Google Scholar] [CrossRef]
  285. Wang, S.Y.; Tao, P.; Hu, H.Y.; Yuan, J.Y.; Zhao, L.; Sun, B.Y.; Zhang, W.J.; Lin, J. Effects of initiating time and dosage of Panax notoginseng on mucosal microvascular injury in experimental colitis. World J. Gastroenterol. 2017, 23, 8308–8320. [Google Scholar] [CrossRef]
  286. Zhang, X.; Song, L.; Li, L.; Zhu, B.; Huo, L.; Hu, Z.; Wang, X.; Wang, J.; Gao, M.; Zhang, J.; et al. Phosphatidylserine externalized on the colonic capillaries as a novel pharmacological target for IBD therapy. Signal Transduct. Target. Ther. 2021, 6, 235. [Google Scholar] [CrossRef] [PubMed]
  287. Adzemovic, M.V.; Zeitelhofer, M.; Eriksson, U.; Olsson, T.; Nilsson, I. Imatinib ameliorates neuroinflammation in a rat model of multiple sclerosis by enhancing blood-brain barrier integrity and by modulating the peripheral immune response. PLoS ONE 2013, 8, e56586. [Google Scholar] [CrossRef]
  288. Aman, J.; van Bezu, J.; Damanafshan, A.; Huveneers, S.; Eringa, E.C.; Vogel, S.M.; Groeneveld, A.B.; Vonk Noordegraaf, A.; van Hinsbergh, V.W.; van Nieuw Amerongen, G.P. Effective treatment of edema and endothelial barrier dysfunction with imatinib. Circulation 2012, 126, 2728–2738. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  289. Hosaka, K.; Yang, Y.; Seki, T.; Nakamura, M.; Andersson, P.; Rouhi, P.; Yang, X.; Jensen, L.; Lim, S.; Feng, N.; et al. Tumour PDGF-BB expression levels determine dual effects of anti-PDGF drugs on vascular remodelling and metastasis. Nat. Commun. 2013, 4, 2129. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  290. Magro, F.; Costa, C. Long-standing remission of Crohn’s disease under imatinib therapy in a patient with Crohn’s disease. Inflamm. Bowel Dis. 2006, 12, 1087–1089. [Google Scholar] [CrossRef]
  291. Gonzalez-Cabrera, P.J.; Brown, S.; Studer, S.M.; Rosen, H. S1P signaling: New therapies and opportunities. F1000Prime Rep. 2014, 6, 109. [Google Scholar] [CrossRef]
  292. Verstockt, B.; Vetrano, S.; Salas, A.; Nayeri, S.; Duijvestein, M.; Vande Casteele, N. Sphingosine 1-phosphate modulation and immune cell trafficking in inflammatory bowel disease. Nat. Rev. Gastroenterol. Hepatol. 2022, 19, 351–366. [Google Scholar] [CrossRef] [PubMed]
  293. Obinata, H.; Hla, T. Sphingosine 1-phosphate and inflammation. Int. Immunol. 2019, 31, 617–625. [Google Scholar] [CrossRef] [PubMed]
  294. Oo, M.L.; Chang, S.H.; Thangada, S.; Wu, M.T.; Rezaul, K.; Blaho, V.; Hwang, S.I.; Han, D.K.; Hla, T. Engagement of S1P₁-degradative mechanisms leads to vascular leak in mice. J. Clin. Investig. 2011, 121, 2290–2300. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  295. Jeya Paul, J.; Weigel, C.; Müller, T.; Heller, R.; Spiegel, S.; Gräler, M.H. Inflammatory Conditions Disrupt Constitutive Endothelial Cell Barrier Stabilization by Alleviating Autonomous Secretion of Sphingosine 1-Phosphate. Cells 2020, 9, 928. [Google Scholar] [CrossRef]
  296. Ziegler, A.C.; Gräler, M.H. Barrier maintenance by S1P during inflammation and sepsis. Tissue Barriers 2021, 9, 1940069. [Google Scholar] [CrossRef]
Figure 1. In the healthy intestine (depicted here, the colon mucosa), the mucosal microvasculature participates in homeostasis by regulating the absorption of essential nutrients, electrolytes, and water, while building a second barrier towards luminal microbes. During IBD, inflammation activates the intestinal microvasculature through release of cytokines and growth factors, leading to adhesion molecule expression, leukocyte extravasation, vascular hyperpermeability, and an increase in both sprouting and intussusceptive angiogenesis.
Figure 1. In the healthy intestine (depicted here, the colon mucosa), the mucosal microvasculature participates in homeostasis by regulating the absorption of essential nutrients, electrolytes, and water, while building a second barrier towards luminal microbes. During IBD, inflammation activates the intestinal microvasculature through release of cytokines and growth factors, leading to adhesion molecule expression, leukocyte extravasation, vascular hyperpermeability, and an increase in both sprouting and intussusceptive angiogenesis.
Ijms 24 05517 g001
Figure 2. (A) The colonic microvascular architecture is massively remodeled in the presence of inflammation. The vessel structure was analyzed in mouse colon samples in the absence (left) or presence (right) of colitis. Vessels were visualized via the staining of CD31 (2-photon microscopy) or via lectin-staining (light sheet microscopy and ultramicroscopy). (B). Vascular changes and dysfunction observed in IBD participate in the initiation and perpetuation of mucosal inflammation and extra-cellular manifestations. PAMP: pathogen-associated molecular pattern.
Figure 2. (A) The colonic microvascular architecture is massively remodeled in the presence of inflammation. The vessel structure was analyzed in mouse colon samples in the absence (left) or presence (right) of colitis. Vessels were visualized via the staining of CD31 (2-photon microscopy) or via lectin-staining (light sheet microscopy and ultramicroscopy). (B). Vascular changes and dysfunction observed in IBD participate in the initiation and perpetuation of mucosal inflammation and extra-cellular manifestations. PAMP: pathogen-associated molecular pattern.
Ijms 24 05517 g002
Table 1. Different mesenteric vascular cell subtypes in homeostasis and in IBD.
Table 1. Different mesenteric vascular cell subtypes in homeostasis and in IBD.
Cell TypeIn HomeostasisIn IBD
HIMECsTolerance to bacterial products from the gut microbiota
Constitutive iNOS expression
Leukocyte hyperadhesion
Angiogenesis
↑ ανβ3 integrin expression
↑ vessel permeability
↓ iNOS and eNOS expression
↓ protein C system activation
↑ secretion of inflammatory mediators
HEVsRecruitment and trafficking of lymphocytes from blood to lymph nodes and secondary lymphoid organs↑ density
↑ leukocyte binding
Formation of extrafollicular HEVs
Lymphatic ECsAbsorption of fatty acids
Immune regulation
↑ density
↓ contractile activity
Lymphangitis
Mural cellsDevelopment and maintenance of the GVB↓ vessel coverage
↑ MMP expression
Enteric glial cellsPart of gut–vascular units
Development and maintenance of the GVB
Sensing of bacterial translocation
Closure of the PVB
Abbreviations: ECs: endothelial cells; HIMECs: human intestinal microvascular ECs; HEVs: high endothelial venules; TJs: tight junctions; GVB: gut–vascular barrier; PVB: plexus–vascular barrier; ↑: increase; ↓: decrease.
Table 2. Vessel-directed effects of key inflammatory cytokines involved in the pathogenesis of IBD.
Table 2. Vessel-directed effects of key inflammatory cytokines involved in the pathogenesis of IBD.
CytokinesEffect
IFN-γEC activation, ↑ CAM expression (notably MadCAM-1)
↑ vascular permeability
Disassembly of VE-cadherin junctions, ↓ VE-cadherin expression
↓ EC proliferation and migration, ↓ angiogenesis
↓ vascular coverage, ↓ PDGF-B
↑ TLR3 expression
↑ CX3CL1 (fractalkine)
TNF-αEC activation, ↑ CAM expression (notably MadCAM-1)
↑ vascular permeability, ↑ Phosphorylation of VE-cadherin,
↑ monolayer tension
↓ TJ protein expression in EC
↑ circulating levels, ↑ vascular dysfunction
↓ EC proliferation and migration, ↓ angiogenesis
↑ CX3CL1 (fractalkine)
IL-1βEC activation, ↑ CAM expression (notably MadCAM-1)
↑ CX3CL1 (fractalkine)
Abbreviations: ↑: increase; ↓: decrease.
Table 3. Pathogenic effects of vascular factors in IBD.
Table 3. Pathogenic effects of vascular factors in IBD.
FactorEffect on the Intestinal Vasculature
Angiogenic growth factors
VEGFExpression increased in IBD
↑ sprouting angiogenesis
↑ EC proliferation and migration
↑ ICAM-1 and VCAM-1 expression
↑ recruitment of VEGFR- expressing immune cells
↑ vascular permeability, disassembly of VE-cadherin junctions
↓ vascular coverage
↑ wound healing
bFGFExpression increased in IBD
↑ sprouting angiogenesis
PDGFExpression increased in IBD
↑ sprouting angiogenesis
↑ vascular coverage
Protective effect in UC
Nitric oxide (NO)Decreased constitutive expression of eNOS and iNOS in intestinal EC during IBD
↓ CAM expression
↓ ROS production
↑ intestinal endothelial barrier function
↑ vasodilatation
Potentiates VEGF-mediated effects
Coagulation factorsIncreased platelet activation and thrombi in IBD
↓ thrombomodulin expression in IBD
↓ protein C receptor expression in IBD
Impaired protein C activation in activated intestinal EC
Toll-like receptors (TLR)Tolerance to endotoxin in intestinal EC
Expression of TLR3 and TLR5 by intestinal EC, protective against colitis in mice
↑ endothelial barrier function
Angiocrine factors↑ CX3CL1 (fractalkine) → ↑ adhesion and activation of CX3CR1+ leukocytes
↑ CL25 → recruitment of CCR9+ immune cells → protective effect
↑ NO
↑ CXCL10 → epithelial cell survival
Abbreviations: ↑: increase; ↓: decrease.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Britzen-Laurent, N.; Weidinger, C.; Stürzl, M. Contribution of Blood Vessel Activation, Remodeling and Barrier Function to Inflammatory Bowel Diseases. Int. J. Mol. Sci. 2023, 24, 5517. https://doi.org/10.3390/ijms24065517

AMA Style

Britzen-Laurent N, Weidinger C, Stürzl M. Contribution of Blood Vessel Activation, Remodeling and Barrier Function to Inflammatory Bowel Diseases. International Journal of Molecular Sciences. 2023; 24(6):5517. https://doi.org/10.3390/ijms24065517

Chicago/Turabian Style

Britzen-Laurent, Nathalie, Carl Weidinger, and Michael Stürzl. 2023. "Contribution of Blood Vessel Activation, Remodeling and Barrier Function to Inflammatory Bowel Diseases" International Journal of Molecular Sciences 24, no. 6: 5517. https://doi.org/10.3390/ijms24065517

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop