Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2019 Oct 1.
Published in final edited form as: Eur J Immunol. 2018 Sep 21;48(10):1607–1620. doi: 10.1002/eji.201646789

Pathogen interactions with endothelial cells and the induction of innate and adaptive immunity.

Christoph Konradt 1, Christopher A Hunter 1,*
PMCID: PMC6172149  NIHMSID: NIHMS987379  PMID: 30160302

Abstract

There are over 10 trillion endothelial cells (EC) that line the vasculature of the human body. These cells not only have specialized functions in the maintenance of homeostasis within the circulation and various tissues but they also have a major role in immune function. EC also represent an important replicative niche for a subset of viral, bacterial and parasitic organisms that are present in the blood or lymph; however, there are major gaps in our knowledge regarding how pathogens interact with EC and how this influences disease outcome. In this article, we review the literature on EC-pathogen interactions and their role in innate and adaptive mechanisms of resistance to infection and highlight opportunities to address prominent knowledge gaps.

Graphical Abstract

graphic file with name nihms-987379-f0001.jpg

Through their ability to express cytokine and toll like receptors Endothelial cells can sense microbial threats and respond to inflammatory stimuli. This can lead to the release of chemokines, expression of adhesion molecules and upregulation of MHC molecules, which can have a crucial impact on the immune response to infection.

A. Introduction

The circulatory system in vertebrates consists of a network of vessels that can be broadly divided into vascular and lymphatic arms. The vascular system has an important role in thermoregulation, fluid balance and provides a conduit for the delivery of oxygen and nutrition to, and waste products and carbon dioxide away from, organs [1]. The lymphatic system functions to remove interstitial fluid from tissues and provides a conduit for antigen transport and immune-cell trafficking from peripheral tissues to secondary lymphoid organs. It is typically assumed that the lymphatics represent the primary route for pathogen dissemination after initial breach of mucosal or surface barriers, but there are few studies that directly address this issue. For vector-borne pathogens, the trauma and anti-coagulants associated with insect feeding will likely provide infectious stages with direct access to the blood. For example, the motile sporozoites of malaria that are deposited in the skin by mosquitoes can directly access the blood [2]. In contrast, for pathogens that enter their hosts through mucosal surfaces in the absence of obvious trauma, the involvement of the lymphatics for dissemination would seem likely; however, following oral challenge with Yersinia enterocolitica, the lymphatics were found not to be a major route for bacterial spread, rather small scale breaches of the intestine allowed systemic dissemination [3]. Another study has highlighted the importance of a gut-vascular barrier that limits entry of commensals into the blood, but Salmonella typhimurium, for example, utilizes its type III secretion system to cross this barrier [4]. These examples illustrate bacterial pathogens that have evolved strategies to directly access the vascular compartment and bypass the lymphatics, which presumably delays their recognition by the immune system. While, mainly due to technical challenges, the interactions of EC with pathogens is certainly understudied, these reports highlight that vascular EC are involved in many of the earliest events associated with infections that access the blood.

There are approximately 10 – 60 trillion endothelial cells (EC) in the human body that cover approximately 4000 m2 [1]; they line the blood and lymphatic vessels with the exception of the placenta where EC are replaced by trophoblasts. EC provide a physical barrier between the circulation and tissues, and have functional specializations related to tissue location and activation status that affect function [1],[5]. The endothelium can be categorized into the macrovasculature composed of arteries and veins, and the microvasculature that includes arterioles, capillaries and venules. The macrovascular endothelium is non-fenestrated (lack pores) and continuous with limited vascular permeability whereas the microvasculature can be either continuous, fenestrated or discontinuous depending on the type of capillary bed. Fenestrated endothelium allows the rapid exchange, uptake and secretion of fluids, solutes and molecules and is present in tissues involved in filtration and secretion such as exocrine and endocrine glands, kidney glomeruli and the intestinal mucosa. Discontinuous endothelium is found in sinusoidal vascular beds such as those in the liver and bone marrow where the ability of cells to readily enter and exit the circulation is relevant [6].

EC in the blood-brain barrier (BBB) exhibit unique features such as intercellular tight junctions, the absence of fenestrae and a reduced level of pinocytic activity, asymmetrically-localized enzymes and carrier-mediated transport systems that distinguish them from peripheral EC [1],[7]. These specialized features protect the CNS from pathogens, toxic molecules and limits access of antibodies and immune cells (discussed in more detail in Section CIV, EC of the BBB and infection). Thus, EC are a heterogeneous group of cells that are responsive to signals from the microenvironment, which include metabolic and mechanical stimuli, and interactions with other cells. The basis for this heterogeneity is the subject of other reviews [6],[8] and will not be discussed further here.

Given the inherent heterogeneity of EC populations in different tissues, it would be expected that EC in distinct compartments had unique responses and functions associated with diverse infections. For example, EC in the bone marrow that are involved in platelet formation would have a role in vivo distinct from those that form the BBB, but there is a lack of studies that systematically compare how EC in different tissues respond to infection. This represents a major knowledge gap and is, in part, a reflection of several technical issues. Cultures of EC (most notably Human Umbilical Vein Endothelial cells, HUVECs) have been used extensively to study how these cells respond to different microbes, but EC tend to lose their specialized properties in culture and obtaining differentiated EC from human adults is a challenge. The ability to use induced pluripotent stem cells to generate EC that resemble mature primary vascular endothelium and which can adopt an activated phenotype that supports cytokine production, leukocyte adhesion and transmigration will help address these issues [9]. This approach should also be useful to assess the impact of host genetics on EC responses to infection and provide the opportunity to study the influence of different EC subtypes on the immune response. There are also relevant concerns that while it is straightforward to expose EC to pathogens in vitro, often under non-physiological conditions, the in vivo relevance can be uncertain. The application of intravital imaging approaches that allow the visualization of EC-pathogen interactions in real time has been important but extended live imaging of deep tissues in the context of infection is not trivial. Despite these challenges, there continues to be remarkable progress in addressing various aspects of EC-pathogen interactions as discussed in this review, which concentrates on the response of the endothelium system as a whole to infection, calling out sub-location differences where known and focusing on micro-organisms where the pathophysiology of the infection is closely linked to EC biology.

B. Homeostatic and Inflammatory Functions of EC

While EC are a key structural component of the vascular system they also have functions that revolve broadly around the maintenance of immunological homeostasis versus the initiation and amplification of the inflammatory response to vascular insults such as infection or trauma. EC also have mechanisms that act to prevent aberrant inflammation and their ability to produce prostaglandin I2 and constitutive expression of endothelial nitric oxide synthase (eNOS) both antagonize cytokine mediated upregulation of adhesion molecules [10]–[12]. Similarly, basal expression of tissue factor pathway inhibitors (TFPIs) block the initiation of the coagulation cascade and inhibit platelet adhesion and aggregation (Figure 1A) [13]. In addition, during infection with a human pathogenic strain of influenza virus, EC expression of the S1P receptor limits the early innate immune response and results in reduced mortality [14]. This finding indicates that the ability of EC to respond to bioactive sphingolipid sphingosine 1-phosphate is important to limit systemic pathological responses [14]. EC also synthesize Weibel–Palade bodies (WPB), which are specialized storage vesicles containing von Willebrand factor, P-selectin, Angiopoetin-2 and chemokines which can mediate an immediate response to inflammatory signals. However, the number of WPBs and their precise content vary between endothelial tissues [15]. Thus, at sites of discrete vascular injury, the ability of EC to release von Willebrand factor supports the local recruitment of platelets that plug damaged vessels and thereby avoids the induction of systemic pro-coagulant events (Figure 1B) [16].

Figure 1: EC functions associated with homeostasis and resistance to infection.

Figure 1:

(A) In steady state EC have mechanisms that act to prevent inflammation through their ability to produce prostaglandin I2, which together with the constitutive expression of endothelial nitric oxide synthase (eNOS), antagonizes cytokine mediated upregulation of adhesion molecules. Basal expression of tissue factor pathway inhibitors (TFPIs) block the initiation of the coagulation cascade and inhibit platelet adhesion and aggregation. (B) EC contain Weibel–Palade bodies (WPB), which are cytoplasmic storage vesicles that contain von Willebrand factor (vWF) and P-selectin, in response to vascular injury, complement activation via the C1qRs and C5aR, EC release von Willebrand factor that supports the local recruitment of platelets and coagulation to prevent blood loss. EC damage can also promotes expression of adhesion molecules. (C) EC express cytokine receptors (eg IFN-γR and TNFR) and many classes of PRRs (e.g. TLR2, TLR4, TLR9, RIG-1, and STING) relevant to bacterial and viral infection, which allow EC to respond to infection by the activation of NF-κB and MAPK. Further, EC activation by pathogen leads to the up-regulation of adhesion molecules such as ICAM1, VCAM1, Selectins and CX3CL1, and the release of chemokines and pro-inflammatory cytokines such as IL-1/6/8, MCP, and CXCL9/10/11. (D) EC activation by IFN-γ, TNF-α or Type I IFNs induce a number of mechanisms that can restrict the growth of micro-organisms. IFN-γ and TNF-α activate EC to upregulate indoleamine 2,3-dioxygenase (IDO) leading to tryptophan degradation and starvation of T. gondii, S. aureus and Rickettsia. Moreover, EC production of nitric oxide (NO) inhibits microbial growth of Rickettsia and M. tuberculosis. Further, Type I IFNs induced expression of interferon-stimulated genes (ISG) inhibit viral replication during HCMV infection.

In steady state, EC express basal levels of adhesion molecules, such as intercellular adhesion molecule 1 (ICAM1) and, additionally, E-selectin in the skin, and vascular cell-adhesion molecule 1 (VCAM1) in the brain and bone marrow; however, during inflammation activated EC universally upregulate the expression of adhesion molecules (Figure 1C). EC can also bind chemokines on their luminal surface via cell surface heparan sulphate proteoglycans [17]. The activation of EC in response to damage, infection or immune stimuli (Figure 1B-D) also has profound physiological consequences that include increased blood flow and the formation of gaps between EC which allows leakage of plasma proteins [5]. The complement system represents an important mechanism for pathogen control [18], and the binding of C1q to the C1qRs promotes EC expression of adhesion molecules [19], and the release of cytokines and chemokines such as interleukin (IL)-6, IL-8 and monocyte chemoattractant peptide-1 (MCP-1) [20]. The binding of C5a to C5aR also stimulates EC to express P-selectin, release von Willebrand factor [21],[22], and to upregulate expression of adhesion molecules, vascular endothelial growth factor (VEGF)-C, IL-1β, IL-8 and RANTES [22], which promote immune cell access to damaged tissues. Furthermore, EC express receptors for the cytokines interferon (IFN)-γ, tumor necrosis factor (TNF), IL-1 and IL-6 [5],[23], which allows them to sense inflammatory signals derived from cells in the vascular compartment or inflamed tissues. EC responses to these signals are characterized by the up-regulation of ICAM1, VCAM1, E- and P-selectin and the secretion of pro-inflammatory cytokines and chemokines that promote leukocyte rolling and adherence to the vessel wall. These events are typically associated with extravasation into inflamed tissues to control infection [24], but it is unclear if these processes are directly relevant for the clearance of infected EC.

C. The vascular system and pathogens

For many pathogens, after initial invasion, the ability to access the vascular compartment is essential not only for dissemination in the new host but also for transmission to insect vectors or blood borne transmission. To limit systemic spread of infection, there are multiple anti-microbial immune effectors in the blood that include macrophages, lipoproteins as well as complement- and coagulation-mediated pathways [18],[25],[26]. The importance of complement in resistance to infection is illustrated by the increased incidence of Neisseria meningitidis in patients with primary genetic deficiencies in the complement pathway [27]. Further, it is well accepted that the presence of pathogen-specific antibodies that activate complement or promote phagocytosis can curtail infections in the blood. The significance of these immune-mediated mechanisms of resistance is apparent by the evolution of vascular pathogens that express surface coats that mitigate the effects of complement activation or which have proteins that deactivate complement [18],[28]. For a sub-set of micro-organisms, the ability to invade EC not only allows them to evade humoral immunity but also provides ready access to the vasculature. In some cases, EC represent a site of microbial persistence and a recent study has highlighted lymphatic EC as a niche for Mycobacterium tuberculosis [29]. Similarly, the ability of Chlamydia pneumoniae to persist in EC may be involved in the induction and acceleration of atherosclerosis [30], a process with an immune component characterized by the formation of plaques in arteries. Nevertheless, it is relevant to recognize that EC are not simply a replicative niche for diverse organisms but there is a growing appreciation that EC influence the outcome of infections that affect the circulatory system. The examples provided below illustrate some of the most important pathogens, which infect or interact with EC and highlights the potential role of EC in microbial detection, as well as illustrating how EC-pathogen interactions affect disease manifestation (Figure 2).

Figure 2: EC-pathogen interactions that lead to disease.

Figure 2:

(A) Infection of the endothelium by Bartonella sp. and Kaposi sarcoma–associated herpesvirus (KSHV) are associated with the influx of monocytes/lymphocytes that secrete pro-angiogenic factors that can lead to vaso-proliferation and the formation of vascular tumors. (B) For N. meningitidis, the adhesion of the meningococci to EC is accompanied by host cell cytoskeletal modifications and the formation of microvilli-like protrusions; these changes are associated with increased coagulation in small blood vessels. Plasmodium matures in erythrocytes and within these cells can adhere to EC. These events provoke localized inflammation within the capillaries and can lead to cerebral malaria. (C) For Human cytomegalovirus (HCMV), Dengue virus (DV), Nipah, Ebola and West Nile Virus there may be a viral tropism for EC and this viral presence is associated with a number of pathological symptoms; for some of these pathogens, this might allow their access to the CNS. (D) EC can serve as an important replicative niche for a subset of bacterial, parasitic and fungal pathogens. The infection of, replication in and lysis of EC can be critical for pathogen access to tissues like the CNS.

CI. Interactions of Endothelial Cells with Pathogens.

Pathogen Detection

Given that the appearance of micro-organisms within the vascular system can have life threatening consequences, there is a need to mount vascular-specific anti-pathogen responses. The location of EC at the interface between the blood flow and tissues, and EC expression of diverse pattern recognition receptors (PRRs) that recognize pathogen-associated molecular pattern molecules (PAMPs) and damage-associated molecular pattern molecules (DAMPs) makes them ideal sentinel cells [31]–[33] (Figure 1). This concept becomes more relevant with the recognition that some pathogens such as S. typhimurium and Y. enterocolitica appear capable of directly accessing the vasculature [3],[4]. However, EC are highly polarized cells with significant differences in membrane composition on the apical and abluminal sides and it is not clear whether the distribution of PRRs allow EC to distinguish microbial threats present in tissues or the vascular lumen. A more likely scenario is that for those organisms in the blood the direct interaction with EC engages conserved pro-inflammatory pathways, dominated by the activation of NF-κB signaling (summarized in Table 1), and it is assumed that this process contributes to pathogen control. Indeed, there are multiple illustrations that support this concept. For example, Listeriolysin O, a pore forming toxin from Listeria monocytogenes, induces increased EC expression of surface E-selectin and ICAM1 [34]. VirB is a Bartonella sp virulence factor that not only inhibits apoptosis of infected EC, but also activates NFκB that leads to increased EC adhesion molecule expression and chemokine secretion [35],[36]. Chlamydia pneumoniae can infect microsvascular EC and the TLR4-MD2 complex provides a PRR that recognizes Chlamydial heat shock protein 60 (cHSP60) and activates NF-κB, which promotes inflammatory responses similar to those that contribute to atherogenesis [37]. Similarly, EC recognition of Candida albicans through TLR3 results in the activation of NF-κB and the p38 MAPK pathway which engages a pro-inflammatory transcriptome associated with resistance to this organism [38].

Table 1:

This table summarizes some of the literature on diverse pathogens which infect or interact with EC and their response. The recognition of PAMPs or DAMPs by EC leads to their activation and the initiation of conserved pro-inflammatory pathways, dominated by the activation of NF-κB signaling. This leads to the production of chemokines and the upregulation of adhesion molecules that promotes adherence of immune cells to EC and extravasation of neutrophils, monocytes and lymphocytes into inflamed tissue.

Pathogen Recognition Signaling Increased
expression
Induction Suppression References
Dengue Virus RIG-1 NFκB ICAM-1 IFN-β BAFF,
CXCL9/10/11,
RANTES
IL-6/7/8
[43]-[46]
West Nile Virus TLR3 ICAM1,
VCAM1, E-
selectin
TNF-α [107]
Ebola virus MHC-I, IL-6,
ICAM1, PKR,
IRF-1
[78]
HCMV STING Type I IFNs [39]
Listeria
monocytogenes
ICAM1,
VCAM1, E-
selectin
[34],[139]
Bartonella sp NFκB ICAM1 IL-8, MCP-1 [35],[36]
Staphylococcus
aureus
ICAM1,
VCAM1
IL-1β, IL-6/8,
MCP-1
[140]-[143]
Rickettsia sp NFκB E-selectin IL-1/6/8, [144]
Chlamydia
pneumoniae
TLR4, MD2 NFκB, MAPK ICAM1,
VCAM1, E-
selectin
IL-8, MCP-1 [145]-[147]
Toxoplasma
gondii
ICAM1,
VCAM1, E-
selectin, P-
selectin,
CX3CL1
IL-6, MCP-1 [148],[149]
Trypanosoma
cruzi
NFκB ICAM1,
VCAM1, E-
selectin
IL-1β, IL-6,
CSF-1
[150]-[152]
Candida albicans TLR3 NFκB, MAPK IL-8, CXCL8 [38]

EC also express cytosolic PRRs which allows them to respond to viral invasion and the DNA sensor Stimulator of interferon genes (STING) is required in EC for optimal HCMV-induced Type I IFN production and inhibition of viral replication [39]. Other relevant cytosolic receptors include Retinoic acid-inducible gene 1 (RIG-1) and melanoma differentiation-associated gene 5 (MDA5) that can detect viral products such as genomic RNA [40],[41]. In EC, double-stranded RNA (dsRNA) induces the upregulation of Type I IFN, MHC-I genes and the antiviral genes protein kinase R (PKR) and 2’,5’-oligoadenylate synthetase (2’,5’-OAS), as well as cytokines and adhesion molecules [42], which together restrain viral replication. Dengue Virus (DV) infection of human EC in vitro causes apoptosis but also the expression of a range of cytokines including IFN-β [43]-[45]. In human brain micro-vascular endothelial cells (HBMECs), RIG-1 is required for DV-induced production of type I IFN and proinflammatory cytokines [46]. The importance of these mechanisms is illustrated by the observation that the blockade of IFN-β during infection in vitro leads to increased DV replication in EC [47]. The relevance of these sensors is not restricted to viral products, and EC stimulated with LPS can utilize RIG-1, and MAVS to induce NF-κB–mediated production of adhesion molecules and cytokines [48], Overall, these examples highlight the existence of conserved responses of EC to infection that form the basis for the idea that EC may not just be a substrate to recruit inflammatory cells to sites of infection, but rather have a crucial role in sensing as well as in combating infection.

EC control of pathogen replication

In vitro studies have established that EC can support the replication of a wide array of pathogens, but EC also have a number of mechanisms that can restrict the growth of micro-organisms. Thus, the ability of EC to detect the presence of pathogens (described above) is usually accompanied by enhanced anti-microbial activities as a consequence of EC-intrinsic pathways or by secondary signals derived from immune cells. Multiple in vitro studies demonstrate that EC can be activated by IFN-γ or the type I IFNs to limit replication of pathogens such as T. gondii, S. aureus and HCMV [39],[49],[50] (Figure 1D). Indeed, the combination of IFN-γ alone or in combination with TNF can activate EC to upregulate indoleamine 2,3-dioxygenase (IDO) which in turn leads to tryptophan degradation and starvation of T. gondii [49], S. aureus [50] and Rickettsia [51]. For Rickettsia and M. tuberculosis, EC production of nitric oxide (NO) also inhibits microbial growth [29],[51] and, in IFN-γ–activated EC, M. tuberculosis and eNOS co-localize [29]. More recent studies with EC have also highlighted that autophagy is a component of the cell intrinsic pathways that EC use to control T. gondii [52]. Thus, EC engage multiple mechanisms to limit pathogen replication, but the extent to which these vary with EC subtype and across host species is unclear. Indeed, because of the lack of tractable model systems to target EC-specific pathways in vivo, there remains a significant knowledge gap in defining the core anti-microbial pathways utilized by different EC cell types and their relevance to different micro-organisms. The field would benefit from the application of standardized approaches for in vitro studies, combined with a way to reliably and specifically target EC in vivo, in order to identify core EC responses that mediate resistance to different classes of pathogens.

Microbial influence on EC homeostasis

Interestingly, EC not only respond to microbial threats but there are examples of how the microbiome can influence EC activity, reinforcing the role of EC as sentinels that monitor their environment. In germ free mice, the BBB has reduced expression of tight junction proteins and increased permeability which can be reversed by the colonization of these mice with a pathogen-free microbiota [53]. Interestingly, cerebral cavernous malformations (CCM) are vascular abnormalities that have a genetic basis, likely are formed during development and are prone to leakage. When this occurs in the CNS, it predisposes to haemorrhagic stroke and seizures. The observation that the bacterial microbiome is the primary source of TLR4 ligand that stimulates CCM formation in mice [54] highlights the complex interplay between EC, PRR and microbes that can result either in disease or promote normal developmental processes.

CII. EC interactions with infections that underlie pathogenesis

Most of the infections discussed in this review are associated with the ability to injure EC as a result of direct invasion and, given that EC are prominently infected in vivo with Human cytomegalovirus (HCMV), Dengue virus (DV), Nipah, Ebola or West Nile Virus, it suggests that there may be a viral tropism for these host cells. However, there can also be bystander effects that lead to disease and here, we will focus on some examples where the involvement of EC appear to directly contribute to distinct pathologies. HCMV is a ubiquitous herpesvirus with a broad host cell range and the HCMV genes UL128 to UL150 facilitate adsorption and entry into EC via endocytosis followed by fusion with endosomal membranes [55]. In vitro, HCMV replication in EC promotes the secretion of von Willebrand factor accompanied by platelet adherence and aggregation. These processes may explain the links of HCMV to the development of transplant vascular sclerosis, restenosis following angioplasty, and atherosclerosis [56]. Similarly, the Rickettsia genus is composed of Gram-negative, obligate, intracellular bacteria that are associated with generalized vascular inflammation [57]. In vitro, Rickettsia replicates in human and mouse EC and the increased permeability of endothelial monolayers infected with Rickettsia is associated with disruption of intercellular adherens junctions [57]-[59]. EC stimulation with Rickettsia induces a rapid up-regulation of the inducible isoenzyme cyclooxygenase (CoX) which catalyses the production of prostaglandins and leukotrienes which in turn influence inflammatory responses and vascular permeability [58]. These observations may provide a mechanistic explanation into how rickettsial infection of the endothelium leads to local dermal and epidermal necrosis, vasodilation and fluid leakage into the interstitial spaces [59].

The ability of Gram-negative Bartonella sp. to infect EC is associated with multiple effects which includes the activation of the host GTPases Rho, Rac and Cdc42 which mediate rearrangement of the actin cytoskeleton leading to the uptake of the bacteria [60],[61]. Bartonella sp. also encode two type IV secretion systems (T4SS) and in EC one of these inhibits apoptosis of infected cells [62], and induces activation of NFκB which leads to adhesion molecule expression and chemokine secretion [35],[36]. In humans, infection of the endothelium with Bartonella sp. can lead to vaso-proliferation, and the formation of vascular tumors is associated with the influx of monocytes/lymphocytes that secrete pro-angiogenic factors that support vasoproliferative growth [60]. Since Kaposi sarcoma–associated herpesvirus is also linked with the formation of vascular tumors [63], it suggests that aberrant inflammation may provide a common mechanism that underlies tumor formation.

There are also several eukaryotic pathogens that can cause significant disease in the vasculature and there are a select group of fungi whose interactions with EC are well studied. For example, during the hematogenous spread of Candida albicans the ability to adhere to EC is important for transmigration into tissues where C. albicans causes disease. Consequently, the molecular basis for C. albicans adherence to, and invasion of, EC through candidal adhesins is well characterized [64]. It is also known that the uptake of C. albicans can lead to EC death through a process dependent on EC iron metabolism [65]. Similar to the studies described for S. typhimurium and Yersinia [3],[4] the hyphae of Aspergillus fumigatus within the lung parenchyma can enter EC from the abluminal side and then transverse into the vascular compartment for hematogenous dissemination. Interestingly, abluminal invasion causes less EC damage than luminal invasion but also greater induction of EC genes encoding cytokines and adhesion molecules [66]. For Plasmodium falciparum, a causative agent of malaria, as the parasite matures within erythrocytes these cells can adhere to EC. These events allow the mature parasite to avoid passage through the spleen, where macrophages remove infected RBC [67]. At the molecular level, it is the ability of infected erythrocytes to form ‘knobs’ at their surface, where parasite-derived surface receptors such as P. falciparum erythrocyte membrane protein 1 (PfEMP1) are concentrated [68],[69], that facilitates the adhesion of infected RBCs to EC via EC surface binding molecules including ICAM-1, CD31, CD36, CSA, gC1qR and the Protein C receptor [68],[70]. One consequence of this evasion strategy is that these events provoke localized inflammation within the capillaries of many tissues and, when this occurs in the CNS, it leads to cerebral malaria. Cytoadherence of infected cells to EC is also seen with other species of malaria and related parasites, which indicates that this is an evolutionary conserved strategy to avoid immune-mediated clearance mechanisms in the spleen.

CIII. Infections and coagulation

The coagulation process (discussed earlier in the section “Homeostatic and inflammatory functions of EC”) has an important role in the repair of local damage in the vascular system and resistance to infection. This is illustrated by the essential role for fibrin in limiting infection-induced blood loss in mice infected with T. gondii [71] and also by examples where fibrin deposition has anti-microbial activities e.g. in listeriosis [72]. However, extensive EC damage can result in systemic induction of the coagulation cascade which, in the context of sepsis, leads to disseminated intravascular coagulation (DIC), widespread micro-vascular thrombosis and hemorrhage [25]. Similarly, a mutation in the protein C gene, which encodes a protein that regulates coagulation by degradation of factor Va and factor VIIIa, results in increased bleeding or thrombosis during acquired disturbances such as infectious disease or cancer [73],[74].

For N. meningitidis, a crucial step in the pathogenesis of this infection is the adhesion of meningococci to EC located in regions of low blood flow. The meningococci interact with the human receptor CD147 which is expressed by EC in capillaries [75] and, after initial attachment, Neisseria forms micro-colonies. This attachment is accompanied by EC cytoskeletal modifications and the formation of microvilli-like protrusions that stabilize bacterial adhesion, and altered localization of junctional proteins allows these bacteria to penetrate into tissues [76]. This intimate interaction of meningococci with EC engages local inflammatory and coagulation processes which lead to skin associated purpura fulminans, a disorder characterized by coagulation in small blood vessels that can result in necrosis and DIC.

Although DV can infect EC in vitro, EC are not considered a major cellular target in humans. Nonetheless this infection is responsible for dengue hemorrhagic fever (DHF) and dengue shock syndrome (DSS), the cardinal signs of which include hemorrhage, vascular leakage, and shock, accompanied by severe thrombocytopenia and systemic complement activation. In mouse models, DV infection of EC is linked to release of oxygen free radicals that mediate vascular leakage [47],[77]. Ebola virus (EV) also infects EC and causes severe disease characterized by hypotension, lymphopenia, coagulative disorders, and hemorrhage with high mortality rates in humans and nonhuman primates [78],[79]. Several EV-derived proteins can induce a decrease in EC barrier function or are cytotoxic to EC and thus may contribute to endothelial damage [79]. However, there is evidence that EV-induced coagulopathy results primarily from vascular disruption induced by factors secreted from infected monocytes/macrophages and dendritic cells, and that virus-induced EC damage has a secondary role in the increased susceptibility to hemorrhage [80]. Given that changes in coagulation and thrombosis formation are a common feature of the examples discussed above understanding the events that underlie the development of the clinical diseases associated with these hemorrhagic viruses (and other pathogens) may provide the opportunity to restore haemostasis and better manage these conditions.

CIV. EC of the Blood Brain Barrier and infection

Systemic inflammation-induced changes in EC may allow microbial access to tissues and, although there is considerable EC heterogeneity, there are no systematic comparisons of how different EC sub-types respond to various infections. The major exception may be the EC of the brain, which are highly specialized and provide a barrier function that limits microbial access to the CNS. Certain micro-organisms appear to have a bona fide tropism for neural tissues [81] and there has been a significant focus on pathogen interactions with respect to EC as a component of the BBB. Cryptococcus neoformans is an encapsulated budding yeast that can invade EC and causes meningoencephalitis in humans and animals. Several microbial factors, including urease and phospholipase B1 [82], as well as host plasmin [83], have been reported to be involved in adherence of C. neoformans to brain EC. The fungal inositol transporters Itr1a and Itr3c mediate increases in inositol which promotes Cryptococcus production of hyaluronic acid [84], which is a ligand for host CD44. The interactions of C. neoformans with microvascular EC membrane lipid rafts promotes invasion in a CD44-dependent manner [85]-[87]. Adherence of C. neoformans to EC leads to EC structural changes that ultimately result in EC necrosis and which may facilitate C. neoformans CNS invasion [88]. The protozoan T. gondii is clinically relevant in humans where it can cause toxoplasmic encephalitis. Mice are natural hosts for this parasite and acute infection results in a transient parasitemia that precedes access to the CNS [89],[90]. There are reports that cells infected with T. gondii can cross the BBB [89],[91] or that infected cells release parasites when they make contact with EC [92]. In vitro experiments have shown that extracellular parasites can readily infect EC under flow conditions and can perform paracellular migration across EC monolayers [93]. In vivo, EC are readily infected and replication in, and lysis of, EC is required for the ability of T. gondii to access the CNS [90]. This mechanism is likely relevant for other CNS-tropic pathogens such as L. monocytogenes that can accesses EC either by direct invasion or by cell-to-cell spread from infected mononuclear phagocytes or other EC [94]-[97].

There are also examples where the host receptors that allow pathogen attachment and entry to EC of the BBB have been identified and these may provide targets for therapies. Thus, Nipah virus (NiV) initially infects epithelial cells in the lungs but its ability to disseminate and infect EC in the small blood vessels supplying the CNS is the basis for the development of fatal encephalitis in humans [98],[99]. Consistent with its cellular tropism, the attachment glycoprotein of NiV binds to ephrinB2, the membrane bound ligand for the EphB class of receptor tyrosine kinases (RTKs), which is expressed by EC. In EC, the fusion and attachment proteins of NiV mediate syncytia formation, and multinucleated giant EC are a characteristic feature of affected tissues [100]. For Escherichia coli K1 (the most common Gram-negative cause of neonatal meningitis), the ability to adhere to and invade EC is required for this bacterium to cross the BBB [101],[102]. E. coli adherence to EC is initiated by the Type 1 fimbrial adhesive protein FimH, which can interact with CD48 on EC, and by the outer membrane protein OmpA, which interacts with the glycoprotein gp96 present on the surface of human brain micro-vascular EC and which may influence neuro-tropism [103],[104]. Regarding Streptococcus pneumoniae, the analysis of brain biopsies from patients who died of pneumococcal meningitis revealed that pneumococci co-localize with the polymeric immunoglobulin receptor (pIgR) and with platelet endothelial cell adhesion molecule (PECAM-1) [105]. Moreover, through the use of a murine model, it was shown that antibodies against these molecules synergize with anti-bacterial drugs to reduce bacterial invasion of the brain [105]. This is an important proof of concept for a treatment strategy that is relevant to other pathogens with well-defined host receptors that mediate entry to the brain.

West Nile virus (WNV) is an important human pathogen that targets neurons and can cause encephalitis characterized by disruption of the BBB, enhanced infiltration of immune cells into the CNS, microglial activation and eventual loss of neurons. WNV can infect EC in vitro, which in turn up-regulate claudin-1, ICAM1, VCAM1 and E-selectin expression and multiple mechanisms have been proposed for how it crosses the BBB [106]. In vivo during infection, TLR3 engagement leads to the secretion of TNF and this induces a transient change in the permeability of the BBB [107], which may allow virus to cross into the CNS [108],[109]. Similarly, African trypanosomes, which exist as motile extracellular forms present in the blood, can access the brain, most likely through the post-capillary venules or via the Virchow Robbins space (fluid filled perivascular gap) [110],[111]. While a parasite-derived protease is implicated in this process, host IFN-γ and G protein–coupled receptors (GPCR) contribute to the ability of trypanosomes to cross the BBB, presumably through loosening of the EC tight junctions [112],[113]. Thus, infection-induced systemic inflammation can alter BBB permeability and thereby increase microbial access to the CNS

D. EC interaction with Immune Cells During Infection.

Monocytes

Monocytes play an important role in microbial control and help to remove dead cells or damaged tissues. As noted above (Table 1, Figure 1), a common innate response of EC to infection is the production of chemokines and the upregulation of adhesion molecules that promotes adherence of immune cells to EC and extravasation of neutrophils, monocytes and lymphocytes into inflamed tissue. EC also constitutively express the chemokine fractalkine (CX3CL1) and, while fractalkine levels are upregulated in response to inflammation, constitutive levels enable rolling of monocytes along EC surfaces. Intravital imaging studies have shown that monocytes which express the fractalkine receptor (CX3CR1) adhere to and patrol the luminal side of the entire microvasculature at homeostasis and, in CX3CR1-KO mice, monocyte adherence is reduced six-fold [114]. The authors proposed that this behavior provides a mechanism that allows the monocytes resident in the blood to rapidly access the site of an infection where they mediate anti-microbial activities but it also seems possible that monocytes may interact with infected EC. The same group have suggested a role for a sub-population of these patrolling monocytes as intravascular housekeepers which phagocytose EC that have been damaged by local neutrophil responses [115]. Whether this subset of monocytes is involved in the resolution of infection-induced damage is uncertain but, given that many of the EC chemokines that are upregulated by C. albicans are involved in the recruitment of neutrophils, which play a pivotal role in the resolution of fungal infections [116], EC damage by neutrophils may be widespread.

T cells and NK cells

Although it is recognized that there are subsets of tissue-resident T cells that are specialized for many different tissues, it is not yet clear whether there are T cells that are specialized for surveying the vascular compartment. Nonetheless, EC do express MHC class I and II, are able to present class I and II restricted antigens and can interact with cells of the adaptive immune system [5],[24],[117]. EC are also implicated in cross-presentation of antigen [118],[119] but whether these antigen-restricted EC events impact on the trafficking, activation, and differentiation of lymphocytes during infection is understudied but of interest. There is also growing interest in the role of the lymphatic endothelium as an archive of antigen following vaccination or infection that can be accessed by DC and used to maintain memory T-cell responses [120]-[122]. Interestingly, a subset of activated T and NK cells express the chemokine receptor CX3CR1 and there is in vitro evidence that the ability of NK cells to injure EC is CX3CR1-dependent [123]. In response to several infections (Listeria, LCMV, TB) a subset of pathogen-specific effector or memory T cells located within the vascular compartment express CX3CR1 [124],[125]. Current paradigms suggest that this pathway is likely important for T cells to extravasate and access inflamed tissues; however, it is also possible that CX3CR1 expression might be involved in T-cell surveillance of the vasculature lumen for infected ECs or to promote T-cell interactions with the lymphatic endothelium.

While it is not clear if there is specific immune surveillance of the endothelium, it provides a potential mechanism for resistance to infection but such a mechanism can also have adverse consequences. In a model of cerebral malaria, although EC are not infected with Plasmodium, the production of IFN-γ enables brain EC to cross-present parasite antigens and become susceptible to killing by CD8+ T cells [118]. In other reports, malaria specific CD8+ T cell responses lead to changes in tight junction function and breakdown of the BBB that leads to cerebral herniation [126],[127], with pathology analogous to the human disease [128]. Similarly, following intra-cerebral challenge with LCMV, the interaction of CD8+ T cells with EC leads to vascular injury in the meningeal compartment and the recruitment of neutrophils and monocytes that mediate the breakdown of the BBB [129].

An additional mechanism that allows EC to exert a regulatory activity on T-cell activities includes the presentation of viral antigen by liver sinusoidal EC to CD8+ T cells, thereby inducing tolerance [130]. In addition, IFN-γ stimulation of EC leads to expression of the inhibitory receptors PD-L1 and PD-L2 which can induce PD-1+ CD8+ T cells to limit their production of IFN-γ and cytolytic activity. Indeed, after systemic LCMV infection in the absence of PD-1, CD8+ T cells killed infected vascular EC and compromised the vascular integrity [131]. Thus, EC expression of PD-L1 and PD-L2 may be a mechanism that allows the activation and extravasation of T cells without excessive vessel damage [132].

F. Conclusions

The sections above have highlighted the significant impact of EC on a variety of immune processes, such as innate recognition, anti-microbial activity, antigen presentation and immune regulation, that govern the outcome of EC-pathogen interactions; however, one of the major challenges of studying EC during infection in vivo is the ability to alter gene expression in EC specifically in order to test which cell-intrinsic immune pathways are involved in mediating resistance to infection versus contributing to the development of disease. For example, the use of promoters for Tie2 and Cdh5 (the gene for VE-cadherin) have proven useful to target EC but Tie2 is also expressed in hematopoietic cells [133]. Further, our own experience with the Cdh5 promoter is that it drives Cre expression in approximately 30% of hematopoietic cells, presumably because Cdh5 is expressed early in development. These technical challenges make it difficult to isolate the immunological activities of EC in vivo in systems where canonical immune populations have a role. While bone marrow chimeras can help address this problem, there remains a need to apply inducible Cre systems to target immunologically relevant pathways in EC of adult mice that have minimal off-target effects. Ideally, these genetic approaches could be combined with intravital imaging to detect rare encounters of EC with pathogens or immune cells. Advances in this field include the development of approaches that allow long-term imaging of the vasculature, in combination with the ability to visualize monocytes, T cells or micro-organisms engineered to express fluorescent reporters, lacZ, Cre or calcium signaling reporters. These approaches have already changed how we view the patrolling behavior of immune cells and their ability to monitor EC integrity and have allowed the visualization of pathogen interactions with EC in real time [90],[134]-[138]. The continued application of these technologies should help to distinguish the circumstances in which EC act either as innate sensors of pathogens or regulators of protective and pathological immune responses that dictate the outcome of infection.

Acknowledgements

This work was supported by NIAID R01 AI110201, NIAID R01 AI 41158 and the Commonwealth of Pennsylvania (all to CAH).

Footnotes

Conflict of interest

The authors declare no financial or commercial conflict of interest.

References.

  • 1.Aird WC. Phenotypic heterogeneity of the endothelium: I. Structure, function, and mechanisms. Circulation Research 2007; 100:158–173. 10.1161/01.RES.0000255691.76142.4a. [DOI] [PubMed] [Google Scholar]
  • 2.Amino R, Thiberge S, Martin B, Celli S, Shorte S, Frischknecht F, Ménard R. Quantitative imaging of Plasmodium transmission from mosquito to mammal. Nature Medicine 2006; 12:220–224. 10.1038/nm1350. [DOI] [PubMed] [Google Scholar]
  • 3.Barnes PD, Bergman MA, Mecsas J, Isberg RR. Yersinia pseudotuberculosis disseminates directly from a replicating bacterial pool in the intestine. J. Exp. Med 2006; 203:1591–1601. 10.1084/jem.20060905. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Spadoni I, Zagato E, Bertocchi A, Paolinelli R, Hot E, Di Sabatino A, Caprioli F, et al. A gut-vascular barrier controls the systemic dissemination of bacteria. Science 2015; 350:830–834. 10.1126/science.aad0135. [DOI] [PubMed] [Google Scholar]
  • 5.Pober JS, Sessa WC. Evolving functions of endothelial cells in inflammation. Nat Rev Immunol 2007; 7:803–815. 10.1038/nri2171. [DOI] [PubMed] [Google Scholar]
  • 6.Potente M, Mäkinen T. Vascular heterogeneity and specialization in development and disease. Nat. Rev. Mol. Cell Biol 2017; 18:477–494. 10.1038/nrm.2017.36. [DOI] [PubMed] [Google Scholar]
  • 7.Macdonald JA, Murugesan N, Pachter JS. Endothelial cell heterogeneity of blood-brain barrier gene expression along the cerebral microvasculature. J. Neurosci. Res 2010; 88:1457–1474. 10.1002/jnr.22316. [DOI] [PubMed] [Google Scholar]
  • 8.Nolan DJ, Ginsberg M, Israely E, Palikuqi B, Poulos MG, James D, Ding B-S, et al. Molecular signatures of tissue-specific microvascular endothelial cell heterogeneity in organ maintenance and regeneration. Dev. Cell 2013; 26:204–219. 10.1016/j.devcel.2013.06.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Adams WJ, Zhang Y, Cloutier J, Kuchimanchi P, Newton G, Sehrawat S, Aird WC, et al. Functional vascular endothelium derived from human induced pluripotent stem cells. Stem Cell Reports 2013; 1:105–113. 10.1016/j.stemcr.2013.06.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Sessa WC. eNOS at a glance. Journal of Cell Science 2004; 117:2427–2429. 10.1242/jcs.01165. [DOI] [PubMed] [Google Scholar]
  • 11.Jaiswal N, Diz DI, Chappell MC, Khosla MC, Ferrario CM. Stimulation of endothelial cell prostaglandin production by angiotensin peptides. Characterization of receptors. Hypertension 1992; 19:II49–55. [DOI] [PubMed] [Google Scholar]
  • 12.Khan BV, Harrison DG, Olbrych MT, Alexander RW, Medford RM. Nitric oxide regulates vascular cell adhesion molecule 1 gene expression and redox-sensitive transcriptional events in human vascular endothelial cells. Proc. Natl. Acad. Sci. U.S.A 1996; 93:9114–9119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Wood JP, Bunce MW, Maroney SA, Tracy PB, Camire RM, Mast AE. Tissue factor pathway inhibitor-alpha inhibits prothrombinase during the initiation of blood coagulation. Proc. Natl. Acad. Sci. U.S.A 2013; 110:17838–17843. 10.1073/pnas.1310444110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Teijaro JR, Walsh KB, Cahalan S, Fremgen DM, Roberts E, Scott F, Martinborough E, et al. Endothelial cells are central orchestrators of cytokine amplification during influenza virus infection. Cell 2011; 146:980–991. 10.1016/j.cell.2011.08.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Bonfanti R, Furie BC, Furie B, Wagner DD. PADGEM (GMP140) is a component of Weibel-Palade bodies of human endothelial cells. Blood 1989; 73:1109–1112. [PubMed] [Google Scholar]
  • 16.van Hinsbergh VWM. Endothelium--role in regulation of coagulation and inflammation. Semin Immunopathol 2012; 34:93–106. 10.1007/s00281-011-0285-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Hillyer P, Male D. Expression of chemokines on the surface of different human endothelia. Immunol. Cell Biol 2005; 83:375–382. 10.1111/j.1440-1711.2005.01345.x. [DOI] [PubMed] [Google Scholar]
  • 18.Lambris JD, Ricklin D, Geisbrecht BV. Complement evasion by human pathogens. Nat. Rev. Microbiol 2008; 6:132–142. 10.1038/nrmicro1824. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Lozada C, Levin RI, Huie M, Hirschhorn R, Naime D, Whitlow M, Recht PA, et al. Identification of C1q as the heat-labile serum cofactor required for immune complexes to stimulate endothelial expression of the adhesion molecules E-selectin and intercellular and vascular cell adhesion molecules 1. Proc. Natl. Acad. Sci. U.S.A 1995; 92:8378–8382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.van den Berg RH, Faber-Krol MC, Sim RB, Daha MR. The first subcomponent of complement, C1q, triggers the production of IL-8, IL-6, and monocyte chemoattractant peptide-1 by human umbilical vein endothelial cells. J. Immunol 1998; 161:6924–6930. [PubMed] [Google Scholar]
  • 21.Foreman KE, Vaporciyan AA, Bonish BK, Jones ML, Johnson KJ, Glovsky MM, Eddy SM, et al. C5a-induced expression of P-selectin in endothelial cells. J. Clin. Invest 1994; 94:1147–1155. 10.1172/JCI117430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Albrecht EA, Chinnaiyan AM, Varambally S, Kumar-Sinha C, Barrette TR, Sarma JV, Ward PA. C5a-induced gene expression in human umbilical vein endothelial cells. AJPA 2004; 164:849–859. 10.1016/S0002-9440(10)63173-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Romano M, Sironi M, Toniatti C, Polentarutti N, Fruscella P, Ghezzi P, Faggioni R, et al. Role of IL-6 and its soluble receptor in induction of chemokines and leukocyte recruitment. Immunity 1997; 6:315–325. [DOI] [PubMed] [Google Scholar]
  • 24.Carman CV, Martinelli R. T Lymphocyte-Endothelial Interactions: Emerging Understanding of Trafficking and Antigen-Specific Immunity. Front Immunol 2015; 6:603 10.3389/fimmu.2015.00603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Engelmann B, Massberg S. Thrombosis as an intravascular effector of innate immunity. Nat Rev Immunol 2013; 13:34–45. 10.1038/nri3345. [DOI] [PubMed] [Google Scholar]
  • 26.Thomson R, Samanovic M, Raper J. Activity of trypanosome lytic factor: a novel component of innate immunity. Future Microbiology 2009; 4:789–796. 10.2217/fmb.09.57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Lewis LA, Ram S. Meningococcal disease and the complement system. virulence 2014; 5:98–126. 10.4161/viru.26515. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Fernández FJ, Gómez S, Vega MC. Pathogens’ toolbox to manipulate human complement. Semin. Cell Dev. Biol 2017. 10.1016/j.semcdb.2017.12.001. [DOI] [PubMed] [Google Scholar]
  • 29.Lerner TR, de Souza Carvalho-Wodarz C, Repnik U, Russell MRG, Borel S, Diedrich CR, Rohde M, et al. Lymphatic endothelial cells are a replicative niche for Mycobacterium tuberculosis. J. Clin. Invest 2016; 126:1093–1108. 10.1172/JCI83379. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Campbell LA, Rosenfeld ME. Persistent C. pneumoniae infection in atherosclerotic lesions: rethinking the clinical trials. Front Cell Infect Microbiol 2014; 4:34 10.3389/fcimb.2014.00034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Dauphinee SM, Karsan A. Lipopolysaccharide signaling in endothelial cells. Lab. Invest 2006; 86:9–22. 10.1038/labinvest.3700366. [DOI] [PubMed] [Google Scholar]
  • 32.Dunzendorfer S, Lee H-K, Tobias PS. Flow-dependent regulation of endothelial Toll-like receptor 2 expression through inhibition of SP1 activity. Circulation Research 2004; 95:684–691. 10.1161/01.RES.0000143900.19798.47. [DOI] [PubMed] [Google Scholar]
  • 33.Li J, Ma Z, Tang Z-L, Stevens T, Pitt B, Li S. CpG DNA-mediated immune response in pulmonary endothelial cells. Am. J. Physiol. Lung Cell Mol. Physiol 2004; 287:L552–8. 10.1152/ajplung.00436.2003. [DOI] [PubMed] [Google Scholar]
  • 34.Drevets DA. Listeria monocytogenes virulence factors that stimulate endothelial cells. Infection and Immunity 1998; 66:232–238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Fuhrmann O, Arvand M, Göhler A, Schmid M, Krüll M, Hippenstiel S, Seybold J, et al. Bartonella henselae induces NF-kappaB-dependent upregulation of adhesion molecules in cultured human endothelial cells: possible role of outer membrane proteins as pathogenic factors. Infection and Immunity 2001; 69:5088–5097. 10.1128/IAI.69.8.5088-5097.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.McCord AM, Burgess AWO, Whaley MJ, Anderson BE. Interaction of Bartonella henselae with endothelial cells promotes monocyte/macrophage chemoattractant protein 1 gene expression and protein production and triggers monocyte migration. Infection and Immunity 2005; 73:5735–5742. 10.1128/IAI.73.9.5735-5742.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Bulut Y, Faure E, Thomas L, Karahashi H, Michelsen KS, Equils O, Morrison SG, et al. Chlamydial heat shock protein 60 activates macrophages and endothelial cells through Toll-like receptor 4 and MD2 in a MyD88-dependent pathway. J. Immunol 2002; 168:1435–1440. 10.4049/jimmunol.168.3.1435. [DOI] [PubMed] [Google Scholar]
  • 38.Müller V, Viemann D, Schmidt M, Endres N, Ludwig S, Leverkus M, Roth J, et al. Candida albicans triggers activation of distinct signaling pathways to establish a proinflammatory gene expression program in primary human endothelial cells. J. Immunol 2007; 179:8435–8445. 10.4049/jimmunol.179.12.8435. [DOI] [PubMed] [Google Scholar]
  • 39.Lio C- WJ, McDonald B, Takahashi M, Dhanwani R, Sharma N, Huang J, Pham E, et al. cGAS-STING Signaling Regulates Initial Innate Control of Cytomegalovirus Infection. J. Virol 2016; 90:7789–7797. 10.1128/JVI.01040-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Loo Y- M, Fornek J, Crochet N, Bajwa G, Perwitasari O, Martinez-Sobrido L, Akira S, et al. Distinct RIG-I and MDA5 signaling by RNA viruses in innate immunity. J. Virol 2008; 82:335–345. 10.1128/JVI.01080-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Asdonk T, Steinmetz M, Krogmann A, Ströcker C, Lahrmann C, Motz I, Paul-Krahe K, et al. MDA-5 activation by cytoplasmic double-stranded RNA impairs endothelial function and aggravates atherosclerosis. J. Cell. Mol. Med 2016; 20:1696–1705. 10.1111/jcmm.12864. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Harcourt JL, Hagan MK, Offermann MK. Modulation of double-stranded RNA-mediated gene induction by interferon in human umbilical vein endothelial cells. J. Interferon Cytokine Res 2000; 20:1007–1013. 10.1089/10799900050198453. [DOI] [PubMed] [Google Scholar]
  • 43.Avirutnan P, Malasit P, Seliger B, Bhakdi S, Husmann M. Dengue virus infection of human endothelial cells leads to chemokine production, complement activation, and apoptosis. J. Immunol 1998; 161:6338–6346. [PubMed] [Google Scholar]
  • 44.Dalrymple NA, Mackow ER. Endothelial cells elicit immune-enhancing responses to dengue virus infection. J. Virol 2012; 86:6408–6415. 10.1128/JVI.00213-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Huang YH, Lei HY, Liu HS, Lin YS, Liu CC, Yeh TM. Dengue virus infects human endothelial cells and induces IL-6 and IL-8 production. Am. J. Trop. Med. Hyg 2000; 63:71–75. [DOI] [PubMed] [Google Scholar]
  • 46.da Conceição TM, Rust NM, Berbel ACER, Martins NB, do Nascimento Santos CA, Da Poian AT, de Arruda LB. Essential role of RIG-I in the activation of endothelial cells by dengue virus. Virology 2013; 435:281–292. 10.1016/j.virol.2012.09.038. [DOI] [PubMed] [Google Scholar]
  • 47.Calvert JK, Helbig KJ, Dimasi D, Cockshell M, Beard MR, Pitson SM, Bonder CS, et al. Dengue Virus Infection of Primary Endothelial Cells Induces Innate Immune Responses, Changes in Endothelial Cells Function and Is Restricted by Interferon-Stimulated Responses. Journal of Interferon & Cytokine Research 2015; 35:654–665. 10.1089/jir.2014.0195. [DOI] [PubMed] [Google Scholar]
  • 48.Moser J, Heeringa P, Jongman RM, Zwiers PJ, Niemarkt AE, Yan R, de Graaf IA, et al. Intracellular RIG-I Signaling Regulates TLR4-Independent Endothelial Inflammatory Responses to Endotoxin. The Journal of Immunology 2016; 196:4681–4691. 10.4049/jimmunol.1501819. [DOI] [PubMed] [Google Scholar]
  • 49.Daubener W, Spors B, Hucke C, Adam R, Stins M, Kim KS, Schroten H. Restriction of Toxoplasma gondii growth in human brain microvascular endothelial cells by activation of indoleamine 2,3-dioxygenase. Infection and Immunity 2001; 69:6527–6531. 10.1128/IAI.69.10.6527-6531.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Schroten H, Spors B, Hucke C, Stins M, Kim KS, Adam R, Daubener W. Potential role of human brain microvascular endothelial cells in the pathogenesis of brain abscess: inhibition of Staphylococcus aureus by activation of indoleamine 2,3-dioxygenase. Neuropediatrics 2001; 32:206–210. 10.1055/s-2001-17375. [DOI] [PubMed] [Google Scholar]
  • 51.Feng HM, Walker DH. Mechanisms of intracellular killing of Rickettsia conorii in infected human endothelial cells, hepatocytes, and macrophages. Infection and Immunity 2000; 68:6729–6736. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Van Grol J, Muniz-Feliciano L, Portillo J-AC, Bonilha VL, Subauste CS. CD40 induces anti-Toxoplasma gondii activity in nonhematopoietic cells dependent on autophagy proteins. Infection and Immunity 2013; 81:2002–2011. 10.1128/IAI.01145-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Braniste V, Al-Asmakh M, Kowal C, Anuar F, Abbaspour A, Tóth M, Korecka A, et al. The gut microbiota influences blood-brain barrier permeability in mice. Sci Transl Med 2014; 6:263ra158 10.1126/scitranslmed.3009759. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Tang AT, Choi JP, Kotzin JJ, Yang Y, Hong CC, Hobson N, Girard R, et al. Endothelial TLR4 and the microbiome drive cerebral cavernous malformations. Nature 2017; 545:305–310. 10.1038/nature22075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Ryckman BJ, Jarvis MA, Drummond DD, Nelson JA, Johnson DC. Human cytomegalovirus entry into epithelial and endothelial cells depends on genes UL128 to UL150 and occurs by endocytosis and low-pH fusion. J. Virol 2006; 80:710–722. 10.1128/JVI.80.2.710-722.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Rahbar A, Söderberg-Nauclér C. Human cytomegalovirus infection of endothelial cells triggers platelet adhesion and aggregation. J. Virol 2005; 79:2211–2220. 10.1128/JVI.79.4.2211-2220.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Sahni SK, Rydkina E. Host-cell interactions with pathogenic Rickettsia species. Future Microbiology 2009; 4:323–339. 10.2217/fmb.09.6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Rydkina E, Sahni A, Baggs RB, Silverman DJ, Sahni SK. Infection of human endothelial cells with spotted Fever group rickettsiae stimulates cyclooxygenase 2 expression and release of vasoactive prostaglandins. Infection and Immunity 2006; 74:5067–5074. 10.1128/IAI.00182-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Walker DH, Ismail N. Emerging and re-emerging rickettsioses: endothelial cell infection and early disease events. Nat. Rev. Microbiol 2008; 6:375–386. 10.1038/nrmicro1866. [DOI] [PubMed] [Google Scholar]
  • 60.Dehio C Bartonella-host-cell interactions and vascular tumour formation. Nat. Rev. Microbiol 2005; 3:621–631. 10.1038/nrmicro1209. [DOI] [PubMed] [Google Scholar]
  • 61.Verma A, Davis GE, Ihler GM. Infection of human endothelial cells with Bartonella bacilliformis is dependent on Rho and results in activation of Rho. Infection and Immunity 2000; 68:5960–5969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Schmid MC, Schulein R, Dehio M, Denecker G, Carena I, Dehio C. The VirB type IV secretion system of Bartonella henselae mediates invasion, proinflammatory activation and antiapoptotic protection of endothelial cells. Molecular Microbiology 2004; 52:81–92. 10.1111/j.1365-2958.2003.03964.x. [DOI] [PubMed] [Google Scholar]
  • 63.Hong Y-K, Foreman K, Shin JW, Hirakawa S, Curry CL, Sage DR, Libermann T, et al. Lymphatic reprogramming of blood vascular endothelium by Kaposi sarcoma-associated herpesvirus. Nat. Genet 2004; 36:683–685. 10.1038/ng1383. [DOI] [PubMed] [Google Scholar]
  • 64.Fu Y, Rieg G, Fonzi WA, Belanger PH, Edwards JE, Filler SG. Expression of the Candida albicans gene ALS1 in Saccharomyces cerevisiae induces adherence to endothelial and epithelial cells. Infection and Immunity 1998; 66:1783–1786. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Fratti RA, Belanger PH, Ghannoum MA, Edwards JE, Filler SG. Endothelial cell injury caused by Candida albicans is dependent on iron. Infection and Immunity 1998; 66:191–196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Kamai Y, Lossinsky AS, Liu H, Sheppard DC, Filler SG. Polarized response of endothelial cells to invasion by Aspergillus fumigatus. Cellular Microbiology 2009; 11:170–182. 10.1093/infdis/152.6.1264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Miller LH, Baruch DI, Marsh K, Doumbo OK. The pathogenic basis of malaria. Nature 2002; 415:673–679. 10.1038/415673a. [DOI] [PubMed] [Google Scholar]
  • 68.Baruch DI, Pasloske BL, Singh HB, Bi X, Ma XC, Feldman M, Taraschi TF, et al. Cloning the P. falciparum gene encoding PfEMP1, a malarial variant antigen and adherence receptor on the surface of parasitized human erythrocytes. Cell 1995; 82:77–87. [DOI] [PubMed] [Google Scholar]
  • 69.Pasternak ND, Dzikowski R. PfEMP1: an antigen that plays a key role in the pathogenicity and immune evasion of the malaria parasite Plasmodium falciparum. Int. J. Biochem. Cell Biol 2009; 41:1463–1466. 10.1016/j.biocel.2008.12.012. [DOI] [PubMed] [Google Scholar]
  • 70.Shabani E, Opoka RO, Bangirana P, Park GS, Vercellotti GM, Guan W, Hodges JS, et al. The endothelial protein C receptor rs867186-GG genotype is associated with increased soluble EPCR and could mediate protection against severe malaria. Sci. Rep 2016; 6:27084 10.1038/srep27084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Johnson LL, Berggren KN, Szaba FM, Chen W, Smiley ST. Fibrin-mediated protection against infection-stimulated immunopathology. J. Exp. Med 2003; 197:801–806. 10.1084/jem.20021493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Mullarky IK, Szaba FM, Berggren KN, Parent MA, Kummer LW, Chen W, Johnson LL, et al. Infection-stimulated fibrin deposition controls hemorrhage and limits hepatic bacterial growth during listeriosis. Infection and Immunity 2005; 73:3888–3895. 10.1128/IAI.73.7.3888-3895.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Fijnvandraat K, Derkx B, Peters M, Bijlmer R, Sturk A, Prins MH, van Deventer SJ, et al. Coagulation activation and tissue necrosis in meningococcal septic shock: severely reduced protein C levels predict a high mortality. Thromb Haemost 1995; 73:15–20. [PubMed] [Google Scholar]
  • 74.Taylor FB, Peer GT, Lockhart MS, Ferrell G, Esmon CT. Endothelial cell protein C receptor plays an important role in protein C activation in vivo. Blood 2001; 97:1685–1688. [DOI] [PubMed] [Google Scholar]
  • 75.Bernard SC, Simpson N, Join-Lambert O, Federici C, Laran-Chich M- P, Maïssa N, Bouzinba-Ségard H, et al. Pathogenic Neisseria meningitidis utilizes CD147 for vascular colonization. Nature Medicine 2014; 20:725–731. 10.1038/nm.3563. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Coureuil M, Lécuyer H, Scott MGH, Boularan C, Enslen H, Soyer M, Mikaty G, et al. Meningococcus Hijacks a β2-adrenoceptor/β-Arrestin pathway to cross brain microvasculature endothelium. Cell 2010; 143:1149–1160. 10.1016/j.cell.2010.11.035. [DOI] [PubMed] [Google Scholar]
  • 77.Chen H-C, Hofman FM, Kung JT, Lin Y-D, Wu-Hsieh BA. Both virus and tumor necrosis factor alpha are critical for endothelium damage in a mouse model of dengue virus-induced hemorrhage. J. Virol 2007; 81:5518–5526. 10.1128/JVI.02575-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Harcourt BH, Sanchez A, Offermann MK. Ebola virus inhibits induction of genes by double-stranded RNA in endothelial cells. Virology 1998; 252:179–188. 10.1006/viro.1998.9446. [DOI] [PubMed] [Google Scholar]
  • 79.Wahl-Jensen VM, Afanasieva TA, Seebach J, Ströher U, Feldmann H, Schnittler H-J. Effects of Ebola virus glycoproteins on endothelial cell activation and barrier function. J. Virol 2005; 79:10442–10450. 10.1128/JVI.79.16.10442-10450.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Geisbert TW, Young HA, Jahrling PB, Davis KJ, Larsen T, Kagan E, Hensley LE. Pathogenesis of Ebola hemorrhagic fever in primate models: evidence that hemorrhage is not a direct effect of virus-induced cytolysis of endothelial cells. AJPA 2003; 163:2371–2382. 10.1016/S0002-9440(10)63592-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Klein RS, Hunter CA. Protective and Pathological Immunity during Central Nervous System Infections. Immunity 2017; 46:891–909. 10.1016/j.immuni.2017.06.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Maruvada R, Zhu L, Pearce D, Zheng Y, Perfect J, Kwon-Chung KJ, Kim KS. Cryptococcus neoformans phospholipase B1 activates host cell Rac1 for traversal across the blood-brain barrier. Cellular Microbiology 2012; 14:1544–1553. 10.1111/j.1462-5822.2012.01819.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Stie J, Fox D. Blood-brain barrier invasion by Cryptococcus neoformans is enhanced by functional interactions with plasmin. Microbiology 2012; 158:240–258. 10.1099/mic.0.051524-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Liu T-B, Kim J-C, Wang Y, Toffaletti DL, Eugenin E, Perfect JR, Kim KJ, et al. Brain Inositol Is a Novel Stimulator for Promoting Cryptococcus Penetration of the Blood-Brain Barrier. PLoS Pathog 2013; 9:e1003247 10.1371/journal.ppat.1003247.s005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Jong A, Wu C-H, Shackleford GM, Kwon-Chung KJ, Chang YC, Chen H-M, Ouyang Y, et al. Involvement of human CD44 during Cryptococcus neoformans infection of brain microvascular endothelial cells. Cellular Microbiology 2008; 10:1313–1326. 10.1111/j.1462-5822.2008.01128.x. [DOI] [PubMed] [Google Scholar]
  • 86.Jong A, Wu C-H, Gonzales-Gomez I, Kwon-Chung KJ, Chang YC, Tseng H-K, Cho W-L, et al. Hyaluronic acid receptor CD44 deficiency is associated with decreased Cryptococcus neoformans brain infection. J. Biol. Chem 2012; 287:15298–15306. 10.1074/jbc.M112.353375. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Huang S-H, Long M, Wu C-H, Kwon-Chung KJ, Chang YC, Chi F, Lee S, et al. Invasion of Cryptococcus neoformans into human brain microvascular endothelial cells is mediated through the lipid rafts-endocytic pathway via the dual specificity tyrosine phosphorylation-regulated kinase 3 (DYRK3). J. Biol. Chem 2011; 286:34761–34769. 10.1074/jbc.M111.219378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Vu K, Eigenheer RA, Phinney BS, Gelli A. Cryptococcus neoformans promotes its transmigration into the central nervous system by inducing molecular and cellular changes in brain endothelial cells. Infection and Immunity 2013; 81:3139–3147. 10.1128/IAI.00554-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Courret N, Darche S, Sonigo P, Milon G, Buzoni-Gâtel D, Tardieux I. CD11c- and CD11b-expressing mouse leukocytes transport single Toxoplasma gondii tachyzoites to the brain. Blood 2006; 107:309–316. 10.1182/blood-2005-02-0666. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Konradt C, Ueno N, Christian DA, Delong JH, Pritchard GH, Herz J, Bzik DJ, et al. Endothelial cells are a replicative niche for entry of Toxoplasma gondii to the central nervous system. Nat Microbiol 2016; 1:16001 10.1038/nmicrobiol.2016.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Ueno N, Harker KS, Clarke EV, McWhorter FY, Liu WF, Tenner AJ, Lodoen MB. Real-time imaging of Toxoplasma-infected human monocytes under fluidic shear stress reveals rapid translocation of intracellular parasites across endothelial barriers. Cellular Microbiology 2014; 16:580–595. 10.1111/cmi.12239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Baba M, Batanova T, Kitoh K, Takashima Y. Adhesion of Toxoplasma gondii tachyzoite-infected vehicle leukocytes to capillary endothelial cells triggers timely parasite egression. Sci. Rep 2017; 7:5675 10.1038/s41598-017-05956-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Harker KS, Jivan E, McWhorter FY, Liu WF, Lodoen MB. Shear forces enhance Toxoplasma gondii tachyzoite motility on vascular endothelium. mBio 2014; 5:e01111–13. 10.1128/mBio.01111-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Drevets DA, Sawyer RT, Potter TA, Campbell PA. Listeria monocytogenes infects human endothelial cells by two distinct mechanisms. Infection and Immunity 1995; 63:4268–4276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Greiffenberg L, Goebel W, Kim KS, Weiglein I, Bubert A, Engelbrecht F, Stins M, et al. Interaction of Listeria monocytogenes with human brain microvascular endothelial cells: InlB-dependent invasion, long-term intracellular growth, and spread from macrophages to endothelial cells. Infection and Immunity 1998; 66:5260–5267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Parida SK, Domann E, Rohde M, Müller S, Darji A, Hain T, Wehland J, et al. Internalin B is essential for adhesion and mediates the invasion of Listeria monocytogenes into human endothelial cells. Molecular Microbiology 1998; 28:81–93. [DOI] [PubMed] [Google Scholar]
  • 97.Greiffenberg L, Sokolovic Z, Schnittler HJ, Spory A, Böckmann R, Goebel W, Kuhn M. Listeria monocytogenes-infected human umbilical vein endothelial cells: internalin-independent invasion, intracellular growth, movement, and host cell responses. FEMS Microbiol. Lett 1997; 157:163–170. [DOI] [PubMed] [Google Scholar]
  • 98.Maisner A, Neufeld J, Weingartl H. Organ- and endotheliotropism of Nipah virus infections in vivo and in vitro. Thromb Haemost 2009; 102:1014–1023. 10.1160/TH09-05-0310. [DOI] [PubMed] [Google Scholar]
  • 99.Negrete OA, Levroney EL, Aguilar HC, Bertolotti-Ciarlet A, Nazarian R, Tajyar S, Lee B. EphrinB2 is the entry receptor for Nipah virus, an emergent deadly paramyxovirus. Nature 2005; 436:401–405. 10.1038/nature03838. [DOI] [PubMed] [Google Scholar]
  • 100.Wong KT, Shieh W-J, Kumar S, Norain K, Abdullah W, Guarner J, Goldsmith CS, et al. Nipah virus infection: pathology and pathogenesis of an emerging paramyxoviral zoonosis. AJPA 2002; 161:2153–2167. 10.1016/S0002-9440(10)64493-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Prasadarao NV, Wass CA, Weiser JN, Stins MF, Huang SH, Kim KS. Outer membrane protein A of Escherichia coli contributes to invasion of brain microvascular endothelial cells. Infection and Immunity 1996; 64:146–153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Huang SH, Wass C, Fu Q, Prasadarao NV, Stins M, Kim KS. Escherichia coli invasion of brain microvascular endothelial cells in vitro and in vivo: molecular cloning and characterization of invasion gene ibe10. Infection and Immunity 1995; 63:4470–4475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Prasadarao NV, Srivastava PK, Rudrabhatla RS, Kim KS, Huang S-H, Sukumaran SK. Cloning and expression of the Escherichia coli K1 outer membrane protein A receptor, a gp96 homologue. Infection and Immunity 2003; 71:1680–1688. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Teng C-H, Cai M, Shin S, Xie Y, Kim KJ, Khan NA, Di Cello F, et al. Escherichia coli K1 RS218 interacts with human brain microvascular endothelial cells via type 1 fimbria bacteria in the fimbriated state. Infection and Immunity 2005; 73:2923–2931. 10.1128/IAI.73.5.2923-2931.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Iovino F, Engelen-Lee J-Y, Brouwer M, van de Beek D, van der Ende A, Valls Seron M, Mellroth P, et al. pIgR and PECAM-1 bind to pneumococcal adhesins RrgA and PspC mediating bacterial brain invasion. Journal of Experimental Medicine 2017; 214:1619–1630. 10.1084/jem.20161668. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Roe K, Orillo B, Verma S. West Nile virus-induced cell adhesion molecules on human brain microvascular endothelial cells regulate leukocyte adhesion and modulate permeability of the in vitro blood-brain barrier model. PLoS ONE 2014; 9:e102598 10.1371/journal.pone.0102598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Wang T, Town T, Alexopoulou L, Anderson JF, Fikrig E, Flavell RA. Toll-like receptor 3 mediates West Nile virus entry into the brain causing lethal encephalitis. Nature Medicine 2004; 10:1366–1373. 10.1038/nm1140. [DOI] [PubMed] [Google Scholar]
  • 108.Verma S, Lo Y, Chapagain M, Lum S, Kumar M, Gurjav U, Luo H, et al. West Nile virus infection modulates human brain microvascular endothelial cells tight junction proteins and cell adhesion molecules: Transmigration across the in vitro blood-brain barrier. Virology 2009; 385:425–433. 10.1016/j.virol.2008.11.047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Suthar MS, Diamond MS, Gale M. West Nile virus infection and immunity. Nat. Rev. Microbiol 2013; 11:115–128. 10.1038/nrmicro2950. [DOI] [PubMed] [Google Scholar]
  • 110.Frevert U, Movila A, Nikolskaia OV, Raper J, Mackey ZB, Abdulla M, McKerrow J, et al. Early invasion of brain parenchyma by African trypanosomes. PLoS ONE 2012; 7:e43913 10.1371/journal.pone.0043913. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Mogk S, Meiwes A, Boßelmann CM, Wolburg H, Duszenko M. The lane to the brain: how African trypanosomes invade the CNS. Trends in Parasitology 2014; 30:470–477. 10.1016/j.pt.2014.08.002. [DOI] [PubMed] [Google Scholar]
  • 112.Grab DJ, Garcia-Garcia JC, Nikolskaia OV, Kim YV, Brown A, Pardo CA, Zhang Y, et al. Protease activated receptor signaling is required for African trypanosome traversal of human brain microvascular endothelial cells. PLoS Negl Trop Dis 2009; 3:e479 10.1371/journal.pntd.0000479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Masocha W, Robertson B, Rottenberg ME, Mhlanga J, Sorokin L, Kristensson K. Cerebral vessel laminins and IFN-gamma define Trypanosoma brucei brucei penetration of the blood-brain barrier. J. Clin. Invest 2004; 114:689–694. 10.1172/JCI22104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Auffray C, Fogg D, Garfa M, Elain G, Join-Lambert O, Kayal S, Sarnacki S, et al. Monitoring of blood vessels and tissues by a population of monocytes with patrolling behavior. Science 2007; 317:666–670. 10.1126/science.1142883. [DOI] [PubMed] [Google Scholar]
  • 115.Carlin LM, Stamatiades EG, Auffray C, Hanna RN, Glover L, Vizcay-Barrena G, Hedrick CC, et al. Nr4a1-dependent Ly6C(low) monocytes monitor endothelial cells and orchestrate their disposal. Cell 2013; 153:362–375. 10.1016/j.cell.2013.03.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Gazendam RP, van de Geer A, Roos D, van den Berg TK, Kuijpers TW. How neutrophils kill fungi. Immunol. Rev 2016; 273:299–311. 10.1111/imr.12454. [DOI] [PubMed] [Google Scholar]
  • 117.Kambayashi T, Laufer TM. Atypical MHC class II-expressing antigen-presenting cells: can anything replace a dendritic cell? Nat Rev Immunol 2014; 14:719–730. 10.1038/nri3754. [DOI] [PubMed] [Google Scholar]
  • 118.Howland SW, Poh CM, Rénia L. Activated Brain Endothelial Cells Cross-Present Malaria Antigen. PLoS Pathog 2015; 11:e1004963 10.1371/journal.ppat.1004963. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Böttcher JP, Schanz O, Garbers C, Zaremba A, Hegenbarth S, Kurts C, Beyer M, et al. IL-6 trans-signaling-dependent rapid development of cytotoxic CD8+ T cell function. CellReports 2014; 8:1318–1327. 10.1016/j.celrep.2014.07.008. [DOI] [PubMed] [Google Scholar]
  • 120.Kedl RM, Tamburini BA. Antigen archiving by lymph node stroma: A novel function for the lymphatic endothelium. Eur. J. Immunol 2015; 45:2721–2729. 10.1002/eji.201545739. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Tamburini BA, Burchill MA, Kedl RM. Antigen capture and archiving by lymphatic endothelial cells following vaccination or viral infection. Nature Communications 5:3989 10.1038/ncomms4989. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Kedl RM, Lindsay RS, Finlon JM, Lucas ED, Friedman RS, Tamburini BAJ. Migratory dendritic cells acquire and present lymphatic endothelial cell-archived antigens during lymph node contraction. Nature Communications 2017; 8:2034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Yoneda O, Imai T, Goda S, Inoue H, Yamauchi A, Okazaki T, Imai H, et al. Fractalkine-mediated endothelial cell injury by NK cells. J. Immunol 2000; 164:4055–4062. 10.4049/jimmunol.164.8.4055. [DOI] [PubMed] [Google Scholar]
  • 124.Gerlach C, Moseman EA, Loughhead SM, Alvarez D, Zwijnenburg AJ, Waanders L, Garg R, et al. The Chemokine Receptor CX3CR1 Defines Three Antigen-Experienced CD8 T Cell Subsets with Distinct Roles in Immune Surveillance and Homeostasis. Immunity 2016; 45:1270–1284. 10.1016/j.immuni.2016.10.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Sakai S, Kauffman KD, Schenkel JM, McBerry CC, Mayer-Barber KD, Masopust D, Barber DL. Cutting edge: control of Mycobacterium tuberculosis infection by a subset of lung parenchyma-homing CD4 T cells. The Journal of Immunology 2014; 192:2965–2969. 10.4049/jimmunol.1400019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Swanson PA, Hart GT, Russo MV, Nayak D, Yazew T, Peña M, Khan SM, et al. CD8+ T Cells Induce Fatal Brainstem Pathology during Cerebral Malaria via Luminal Antigen-Specific Engagement of Brain Vasculature. PLoS Pathog 2016; 12:e1006022 10.1371/journal.ppat.1006022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Huggins MA, Johnson HL, Jin F, N Songo A, Hanson LM, LaFrance SJ, Butler NS, et al. Perforin Expression by CD8 T Cells Is Sufficient To Cause Fatal Brain Edema during Experimental Cerebral Malaria. Infection and Immunity 2017; 85 10.1128/IAI.00985-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Seydel KB, Kampondeni SD, Valim C, Potchen MJ, Milner DA, Muwalo FW, Birbeck GL, et al. Brain swelling and death in children with cerebral malaria. N. Engl. J. Med 2015; 372:1126–1137. 10.1056/NEJMoa1400116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Kim JV, Kang SS, Dustin ML, McGavern DB. Myelomonocytic cell recruitment causes fatal CNS vascular injury during acute viral meningitis. Nature 2009; 457:191–195. 10.1038/nature07591. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Limmer A, Ohl J, Kurts C, Ljunggren HG, Reiss Y, Groettrup M, Momburg F, et al. Efficient presentation of exogenous antigen by liver endothelial cells to CD8+ T cells results in antigen-specific T-cell tolerance. Nature Medicine 2000; 6:1348–1354. 10.1038/82161. [DOI] [PubMed] [Google Scholar]
  • 131.Frebel H, Nindl V, Schuepbach RA, Braunschweiler T, Richter K, Vogel J, Wagner CA, et al. Programmed death 1 protects from fatal circulatory failure during systemic virus infection of mice. Journal of Experimental Medicine 2012; 209:2485–2499. 10.1084/jem.20121015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Rodig N, Ryan T, Allen JA, Pang H, Grabie N, Chernova T, Greenfield EA, et al. Endothelial expression of PD-L1 and PD-L2 down-regulates CD8+ T cell activation and cytolysis. Eur. J. Immunol 2003; 33:3117–3126. 10.1002/eji.200324270. [DOI] [PubMed] [Google Scholar]
  • 133.Tang Y, Harrington A, Yang X, Friesel RE, Liaw L. The contribution of the Tie2+ lineage to primitive and definitive hematopoietic cells. Genesis 2010; 48:563–567. 10.1002/dvg.20654. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Chtanova T, Han S-J, Schaeffer M, van Dooren GG, Herzmark P, Striepen B, Robey EA. Dynamics of T cell, antigen-presenting cell, and pathogen interactions during recall responses in the lymph node. Immunity 2009; 31:342–355. 10.1016/j.immuni.2009.06.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Germain RN, Robey EA, Cahalan MD. A decade of imaging cellular motility and interaction dynamics in the immune system. Science 2012; 336:1676–1681. 10.1126/science.1221063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Coombes JL, Robey EA. Dynamic imaging of host-pathogen interactions in vivo. Nat Rev Immunol 2010; 10:353–364. 10.1038/nri2746. [DOI] [PubMed] [Google Scholar]
  • 137.Köberle M, Klein-Günther A, Schütz M, Fritz M, Berchtold S, Tolosa E, Autenrieth IB, et al. Yersinia enterocolitica targets cells of the innate and adaptive immune system by injection of Yops in a mouse infection model. PLoS Pathog 2009; 5:e1000551 10.1371/journal.ppat.1000551. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Massberg S, Gawaz M, Gruner S, Schulte V, Konrad I, Zohlnhofer D, Heinzmann U, et al. A Crucial Role of Glycoprotein VI for Platelet Recruitment to the Injured Arterial Wall In Vivo. Journal of Experimental Medicine 2002; 197:41–49. 10.1084/jem.20020945. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Wilson SL, Drevets DA. Listeria monocytogenes infection and activation of human brain microvascular endothelial cells. J. Infect. Dis 1998; 178:1658–1666. [DOI] [PubMed] [Google Scholar]
  • 140.Menzies BE, Kourteva I. Internalization of Staphylococcus aureus by endothelial cells induces apoptosis. Infection and Immunity 1998; 66:5994–5998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Pöhlmann-Dietze P, Ulrich M, Kiser KB, Döring G, Lee JC, Fournier JM, Botzenhart K, et al. Adherence of Staphylococcus aureus to endothelial cells: influence of capsular polysaccharide, global regulator agr, and bacterial growth phase. Infection and Immunity 2000; 68:4865–4871. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Matussek A, Strindhall J, Stark L, Rohde M, Geffers R, Buer J, Kihlstrom E, et al. Infection of human endothelial cells with Staphylococcus aureus induces transcription of genes encoding an innate immunity response. Scand. J. Immunol 2005; 61:536–544. 10.1111/j.1365-3083.2005.01597.x. [DOI] [PubMed] [Google Scholar]
  • 143.Tekstra J, Beekhuizen H, Van De Gevel JS, Van Benten IJ, Tuk CW, Beelen RH. Infection of human endothelial cells with Staphylococcus aureus induces the production of monocyte chemotactic protein-1 (MCP-1) and monocyte chemotaxis. Clin. Exp. Immunol 1999; 117:489–495. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Bechah Y, Capo C, Raoult D, Mege JL. Infection of endothelial cells with virulent Rickettsia prowazekii increases the transmigration of leukocytes. J. Infect. Dis 2008; 197:142–147. 10.1086/523649. [DOI] [PubMed] [Google Scholar]
  • 145.Vielma SA. Chlamydophila pneumoniae Induces ICAM-1 Expression in Human Aortic Endothelial Cells via Protein Kinase C-Dependent Activation of Nuclear Factor-kappaB. Circulation Research 2003; 92:1130–1137. 10.1161/01.RES.0000074001.46892.1C. [DOI] [PubMed] [Google Scholar]
  • 146.Dechend R, Maass M, Gieffers J, Dietz R. Chlamydia pneumoniae infection of vascular smooth muscle and endothelial cells activates NF-κB and induces tissue factor and PAI-1 expression. Circulation 1999. [DOI] [PubMed]
  • 147.Krüll M, Kramp J, Petrov T, Klucken AC, Hocke AC, Walter C, Schmeck B, et al. Differences in Cell Activation by Chlamydophila pneumoniae and Chlamydia trachomatis Infection in Human Endothelial Cells. Infection and Immunity 2004; 72:6615–6621. 10.1128/IAI.72.11.6615-6621.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Lachenmaier SM, Deli MA, Meissner M, Liesenfeld O. Intracellular transport of Toxoplasma gondii through the blood–brain barrier. Journal of Neuroimmunology 2010:1–12. 10.1016/j.jneuroim.2010.10.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Taubert A, Krüll M, Zahner H, Hermosilla C. Toxoplasma gondii and Neospora caninum infections of bovine endothelial cells induce endothelial adhesion molecule gene transcription and subsequent PMN adhesion. Veterinary Immunology and Immunopathology 2006; 112:272–283. 10.1016/j.vetimm.2006.03.017. [DOI] [PubMed] [Google Scholar]
  • 150.Todorov AG, Andrade D, Pesquero JB, Araujo R de C, Bader M, Stewart J, Gera L, et al. Trypanosoma cruzi induces edematogenic responses in mice and invades cardiomyocytes and endothelial cells in vitro by activating distinct kinin receptor (B1/B2) subtypes. FASEB J 2003; 17:73–75. 10.1096/fj.02-0477fje. [DOI] [PubMed] [Google Scholar]
  • 151.Tanowitz HB, Gumprecht JP, Spurr D, Calderon TM, Ventura MC, Raventos-Suarez C, Kellie S, et al. Cytokine gene expression of endothelial cells infected with Trypanosoma cruzi. J. Infect. Dis 2017; 166:598–603. [DOI] [PubMed] [Google Scholar]
  • 152.Huang H, Calderon TM, Berman JW, Braunstein VL, Weiss LM, Wittner M, Tanowitz HB. Infection of endothelial cells with Trypanosoma cruzi activates NF-kappaB and induces vascular adhesion molecule expression. Infection and Immunity 1999; 67:5434–5440. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES