key: cord-0856259-gfs9pdjz authors: Eustaquio Do Imperio, Guinever; Lye, Phetcharawan; Bloise, Enrrico; Matthews, Stephen G. title: Function of Multidrug Resistance Transporters is Disrupted by Infection Mimics in Human Brain Endothelial Cells date: 2021-01-11 journal: Tissue barriers DOI: 10.1080/21688370.2020.1860616 sha: e59d07b27995816ed553767dc2a9a8c14c66714d doc_id: 856259 cord_uid: gfs9pdjz P-glycoprotein (P-gp/ABCB1) and breast cancer resistance protein (BCRP/ABCG2) modulate the distribution of drugs and toxins across the blood-brain barrier (BBB). Animal studies reported that infection-induced disruption of these transporters in the developing BBB impairs fetal brain protection. However, the impact of infection mimics on P-gp/BCRP function in human brain endothelium is less well understood. We hypothesized that Toll-like receptor ligands mimicking bacterial and viral infection would modify the expression and function of P-gp and BCRP in human brain endothelial cells (BECs). Human cerebral microvascular endothelial cells (hCMEC/D3) were challenged with bacterial [Lipopolysaccharide (LPS)] and viral-mimics [polyinosinic:polycytidylic acid (PolyI:C) or single-stranded RNA (ssRNA)], or pro-inflammatory cytokines interleukin (IL)-6, tumor necrosis factor (TNF)-α and interferon gamma (IFN)-ɣ. P-gp and BCRP function was assessed after 4 or 24 h, using Calcein-AM and Chlorin-6 assays, respectively. Western blot and qPCR quantified P-gp/ABCB1 and BCRP/ABCG2 expression following treatments. Infection mimics are potent modulators of drug transporters in human BECs in vitro. LPS and PolyI:C increased, while ssRNA exposure reduced P-gp activity. In contrast, LPS and PolyI:C decreased, while ssRNA increased BCRP activity (P < .05). There was little correlation between drug transporter function, gene expression and total protein level. Altered plasma membrane BCRP may suggest modified intracellular trafficking induced by infection in human BECs. Bacterial and viral infection mimics modify P-gp and BCRP transport function in human BECs, in vitro. This knowledge may contribute and have important implications for human brain protection and possible altered biodistribution of drugs and xenobiotics in the brain following exposure to TLR agonists. The blood-brain barrier (BBB) is a selective barrier formed by specialized brain endothelial cells (BECs) that protect the brain from invasive pathogens and penetration of toxic compounds from the bloodstream. 1 The multidrug-resistance transporters, P-glycoprotein (P-gp; encoded by ABCB1) and breast cancer resistance protein (BCRP; ABCG2), belong to the ATP-binding cassette (ABC) transporters superfamily of efflux transporters and are central protective components of the BBB. 2 They are present at the luminal surface of the cerebral microvascular endothelium, where they actively efflux endogenous (steroid hormones, cytokines, chemokines) and exogenous substrates (analgesics, antiretrovirals, antiepileptics, antibiotics, synthetic glucocorticoids, selective serotonin reuptake inhibitor (SSRIs), agrochemicals), back into the bloodstream. As such, they act to control accumulation of their substrates in the central nervous system (CNS). 2 The development of the mammalian brain is highly vulnerable to xenobiotics and environmental toxins. 1, 3 Using animal models, our group demonstrated that BBB drug transport function is regulated by early exposure to several factors, including the proinflammatory cytokines interleukin (IL)-1β, IL-6, and tumor necrosis factor (TNF)-α. These cytokines inhibit P-gp function in BECs, while exposure to glucocorticoids and transforming growth factor-β1 (TGF-β1) up-regulates P-gp function. 1,4-7 Therefore, P-gp activity in BECs (the major component of the BBB) may either be reduced or increased depending on the nature of stimuli, resulting in altered accumulation of xenobiotics in the brain. No studies have investigated the impact of bacterial or viral infection on drug transport function in BECs in adulthood or during development in humans. In the present study, we sought to investigate the impact of infection mimics on P-gp and BCRP transport function using an in vitro human brain endothelium model. We hypothesized that bacterial and viral mimics alter P-gp and BCRP function in the human brain endothelium. We challenged human cerebral microvascular endothelial cells (hCMEC/D3) with bacterial-and viralmimics and key infection-associated cytokines and demonstrated that control (i.e. inhibition or activation) of human BEC function is dependent on the nature of infective/inflammatory stimuli. The human cerebral microvascular endothelial cell line (hCMEC/D3, Cedarlane Labs #CLU512, Burlington, ON, Canada) was selected as a model to investigate drug transporters function since it has been extensively used and characterized to have a brain endothelial phenotype and is, thus a widely used model to investigate human BBB function. [8] [9] [10] The cells have similar morphology and protein expression levels to primary human cerebral microvascular endothelial cells, and they are well adapted for drug transport studies. 11 The cells were purchased between passages 25 and 26 (P25/26), and cultured at 37°C in 5% CO 2 with EndoGRO TM -MV Complete Culture Media Kit® (Millipore, ON, Canada, #SCME004), consisting of EndoGRO Basal Medium and EndoGRO Supplement kit containing recombinant human epidermal growth factor (5 ng/mL), L-glutamine (10 mM), hydrocortisone hemisuccinate (1.0 µg/ mL), heparin sulfate (0.75 U/mL), ascorbic acid (50 µg/mL) and fetal bovine serum (5%; FBS). The media was supplemented with human basic fibroblast growth factor (1 ng/mL; Sigma, #F0291) and penicillin-streptomycin (1%; 10000 units -10000 μg/mL, Life Technologies, #15140-122). Cells were expanded through P29, frozen in 5% DMSO in FBS for 24 h (−80°C), and then stored in liquid nitrogen. All procedures, treatments, and analyses were performed with hCMEC/D3 cells at P30. Lipopolysaccharide (LPS, #L4391), polyinosinic:polycytidylic acid (PolyI: C, #P9582), and calcein-acetoxymethyl ester (Ca-AM, #177831) were purchased from Sigma-Aldrich (Oakville, ON, Canada). Single-stranded RNA (ssRNA, #tlr-lrna40) and its vehicle Lyovec (#lyec-12) were purchased from InvivoGen (San Diego, California, USA), and the cytokines IL-6 (#PHC0065), TNF-α (#PHC3015) and interferongamma (IFN)-ɣ (#PHC4031) were purchased from Gibco (Grand Island, New York, USA). Immunohistochemical analysis was performed to confirm the phenotype of the human BEC cell line. Cells were rinsed twice with cold PBS, fixed with icecold Methanol for 10 min, and then permeabilized with 0.2% Triton X-100 (5 min; room temperature). Cells were then blocked with 0.1% BSA in PBS (1 h), and incubated with primary antibodies anti-P-gp (Santa Cruz Biotechnology, Dallas, Texas, USA, #SC-55510, 1:200), anti-BCRP (Millipore, #MAB146, 1:200), anti-Claudin-5 (Invitrogen #352588, 1:200), anti-PECAM-1 (Dako, Burloak, ON, Canada, #M0823, 1:200) and anti-GFAP (Cell Signaling Technology, Whitby, ON, Canada, #3670, 1:300) overnight at 4°C. The primary antibodies anti-rabbit IgG (Abcam, Toronto, ON, Canada, #ab171870), anti-mouse IgG1 (Dako, #X0931), antimouse IgG2a (Dako, #X0943), and anti-mouse IgG2b (Dako, #X0944) were used as isotype controls. Subsequently, cells were washed (3X) and incubated with fluorochrome-labeled secondary antibody Alexa Fluor 488 (Molecular Probes Inc., Eugene, OR, USA, 1:300) and counterstained with DAPI (1 mg/mL, 60 min). The area and intensity of antibody staining were analyzed, and representative images were obtained using a spinning disc confocal fluorescent microscope (Leica DMI6000 B, Concord, ON, Canada) at 40X magnification. hCMEC/D3 cells (P30) were plated in clear flat bottom 6-well or 96-well TC-treated polystyrene culture plates (Costar, Kennebunk, ME, USA, #3516; or Falcon, Mississauga, ON, Canada, #353072, respectively) at a density of 20000 cells/ cm 2 with EndoGRO TM , and 5% charcoal-stripped (CS)-FBS in DMEM. After 24 h of incubation, cells were challenged with the Toll-like receptor (TLR)-4 ligand, LPS (10 3−6 pg/mL bacterial mimic); the TLR-3 ligand PolyI:C (10 3−6 pg/mL; doublestranded viral mimic) or the TLR-7/8 ligand ssRNA (10 3−6 pg/mL; single-stranded viral mimic), to mimic bacterial and viral infection on human BECs, in vitro. The cytokines IL-6 (10 1−4 pg/mL), TNF-α (10 1−4 pg/mL), and IFN-ɣ (10 1−4 pg/mL) were used to determine the action of downstream mediators induced by infection/inflammation. Functional drug transport assays were undertaken or cells were harvested for total/plasma membrane protein or mRNA analysis 4-and 24 h after treatments. Dose and time of exposure regimens were selected based on previous studies from our group. 4, 6 Total RNA was extracted using the RNeasy Mini Kit (Qiagen #74104, Toronto, ON, Canada), according to the manufacturer's instructions. RNA concentration and purity were determined using a NanoDrop1000 Spectrophotometer (Thermo Scientific, Wilmington, DE, USA) and total RNA (1 μg) was reverse transcribed to cDNA using iScript TM Reverse Transcription Supermix (Bio-Rad #1708840, Mississauga, ON, Canada). The sequences of the primers used are listed in Table 1 . SYBR Green reagent (Sigma #S9194) was used to run the qPCR, using the CFX 380 Real-Time system C1000 TM Thermal Cycler (Bio-Rad). The parameters used were: one cycle (95°C, 2 min), followed by 40 cycles (95°C, 5s) and 60°C for 20s. RNA (10 ng) was used per reaction and each sample was run in triplicate. Hypoxanthine-guanine phosphoribosyltransferase (HPRT), TATA-box binding protein (TBP), and tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein zeta (YWHAZ) were used as reference genes and their geometric mean was used to normalize expression of our genes of interest. The relative mRNA expression was calculated by comparing the PCR cycle threshold (CT) between groups using the 2 −ΔΔCT Method, 14 and the control group was normalized to 1. P-gp activity was assessed as described previously by our group with adaptations. [4] [5] [6] [7] 15 Briefly, charcoalstripped (CS)-FBS (5%) was withdrawn and cells were washed twice with warm Tyrode salts' solution (Sigma, #T2145) supplemented with sodium bicarbonate (1 g/L; Sigma, #S6014). Cells were incubated with 10 −6 M Ca-AM at 37°C and 5% CO 2 (1 h). Ca-AM is intracellularly cleaved by endogenous esterases, producing fluorescent calcein that can be used as an indirect measure of P-gp activity, since it is transported out of cells by P-gp. After incubation with Ca-AM, the plates were placed on ice and the cells were washed twice with ice-cold PBS, followed by cell lysis with 1% Triton X-100 (Sigma #X100) lysis buffer. The BCRP activity assay was adapted from a previous study 16 using Chlorin 6 (Ce6, Santa Cruz Biotechnology, #SC-263067) as a specific BCRP substrate. 17 Cells were incubated with 2 nM Ce6 (37° C; 5% CO 2 ; 1 h). Plates were placed on ice, cells were washed (2X) with cold Tyrode salt solution and lysis buffer added. Calcein and Ce6 accumulation were quantified using a fluorescent microplate reader, at excitation/emission wavelengths of 485/510 nm and 407/667 nm, respectively. The uptake of Ca-AM and Ce6 by P-gp and BCRP was confirmed under the Figure 1 ). N = 6-8 was used per group; assays were repeated 2-4 times. Mean background readings were subtracted and P-gp and BCRP activity following treatment were expressed as a percentage of controls. Cells were detached and lysed using lysis buffer (1 mol/L Tris-HCL pH 6. Immunohistochemical analysis confirmed the presence of P-gp and BCRP in hCMEC/D3 cells Figure 1 . hCMEC/D3 cells also expressed claudin-5 and PECAM protein, consistent with a BEC phenotype. The astrocyte marker GFAP was not present. Mouse IgG1 was used as a negative control for GFAP and PECAM, and mouse IgG2a and 2b (insets) in Figure 1 were used as negative controls for BCRP, P-gp and Claudin-5, respectively. No staining in negative controls was detected. Bacterial-and viral-mimics evoked a proinflammatory response in human BECs 18 activating an antiviral response mediated by nuclear factor-kB (NF-kB) and IFN regulatory factors (IRFs). 19 LPS increased RIG-I (p < .001) and MDA5 (p < .05 and p < .001) mRNA levels 4 h and 24 h after exposure. PolyI: C increased MDA5 (p < .05) mRNA expression 24 h posttreatment, while ssRNA had no effect on RIG-I or MDA5 mRNA. As expected, exposure of hCMEC/D3 cells to Verapamil and Ko143 significantly increased Ca-AM and Ce6 accumulation, respectively. This confirms correct functionof the activity assays (Supplementary Figure 1 ). LPS (10 5 pg/mL) treatment increased (p < .01) P-gp activity at 24 h, whereas BCRP activity was decreased in response to LPS (10 6 pg/mL, p < .05) at 24 h Figure 2 . PolyI:C treatment (10 6 pg/mL) increased (p < .05) P-gp activity at 24 h, while PolyI: C (10 4 and 10 5 pg/mL) decreased (p < .05, p < .01) BCRP activity 24 h after exposure. ssRNA treatment led to a marked decrease in P-gp function 4 h (10 4 pg/ mL, p < .01; 10 5 pg/mL, p < .05) and 24 h (10 4 pg/mL, p < .05; 10 5 and 10 6 pg/mL, p < .01) after treatment. In contrast, ssRNA exposure resulted in a pronounced increase in BCRP function at 24 h (10 4 pg/mL, p < .01; 10 5 pg/mL, p < .05; Figure 2 ). The effects of LPS on ABCB1 and ABCG2 mRNA levels and P-gp and BCRP total protein are illustrated in Table 2 and Figure 3 , respectively. LPS decreased both ABCB1 and ABCG2 mRNA levels at 24 h (p < .05; Table 2 ), and reduced P-gp protein expression at 4 h (p < .05), however BCRP protein levels were increased by LPS 4 h post-treatment (p < .05). PolyI:C had no effect on ABCB1 or ABCG2 mRNA or P-gp protein at either time-point, but increased BCRP expression at 4 h of treatment (p < .05). ssRNA exposure upregulated ABCB1 mRNA at 24 h, and ABCG2 at 4 h post-treatment, respectively (p < .01). While there was no effect of ssRNA on total P-gp, total BCRP protein levels were increased at 4 h (p < .05). The inconsistency between total cellular P-gp and BCRP protein levels and P-gp and BCRP function following exposure to bacterial and viral mimics, led us to undertake analysis of P-gp and BCRP levels in the plasma membrane of human BEC cells following treatment Figure 4 . LPS had no effect on plasma membrane P-gp at 4 or 24 h; however, it reduced plasma membrane BCRP at 24 h post-treatment (p < .05). PolyI:C treatment increased P-gp levels in the plasma membrane at 4 h of exposure (p < .001), with no changes after 24 h. PolyI:C did not affect BCRP levels in the plasma membrane. ssRNA had no effect on P-gp in the plasma membrane but increased BCRP 4 h after treatment (p < .05). Cytokine regulation of P-gp/ABCB1 and BCRP/ ABCG2 mRNA expression and functionP-gp and BCRP function and ABCB1 and ABCG2 mRNA levels were assessed in BECs following 4 h and 24 h exposure to key infection-associated pro-inflammatory cytokines, IL-6, TNF-α and IFN-ɣ Figure 5 and Table 3 . Exposure to IL-6 inhibited P-gp activity after 24 h at all concentrations investigated (10 1−4 pg/mL; p < .05), though no effects were observed at 4 h. Similarly, IL-6 (10 3 pg/mL) down-regulated ABCB1 mRNA levels (p < .001) 24 h after treatment. IL-6 also decreased BCRP function (10 3 and 10 4 pg/mL; p < .01) and ABCG2 mRNA levels (10 3 pg/mL, p < .05) 24 h after treatment Figure 5d and Table 3 . There were no effects of IL-6 on BCRP function or ABCG2 mRNA after 4 h of treatment. There were also no effects of TNF-α treatment on P-gp activity or ABCB1 mRNA levels. However, TNF-α treatment decreased BCRP activity (10 3 and 10 4 pg/mL; p < .01) 4 h after treatment, though there were no effects on ABCG2 mRNA levels. IFN-ɣ increased P-gp function (10 3 and 10 4 pg/mL; p < .01), but down-regulated ABCB1 mRNA levels (p < .05) 24 h after treatment Figure 5j and Table 2 . In contrast, IFN-ɣ decreased BCRP function both 4 h (10 3 pg/mL; p < .05) and 24 h (10 3 and 10 4 pg/mL; p < .01) after treatment Figure 5 (k, L), though there were no effects on ABCG2 mRNA levels. The current study has demonstrated, for the first time, that bacterial and viral infection-mimics can impact drug transporter activity in human brain endothelial cells in vitro. hCMEC/D3 cells exposed to TLR ligands and pro-inflammatory cytokines exhibit altered P-gp and BCRP activity and expression. Interestingly, we also showed that these transporters were regulated in opposite directions by all infection mimics, which may indicate a compensatory regulation between P-gp and BCRP function in human BECs. These findings may have considerable implications for biodistribution of drugs in the CNS during bacterial and viral infection or inflammation. LPS, a TLR-4 agonist, caused transporter-specific time-dependant changes in function. LPS induced a long-term (24 h) increase of P-gp activity, which, in vivo, would likely be associated with reduced concentrations of P-gp substrates in the brain, as shown previously using brain capillaries. 20, 21 Conversely, LPS exposure (24 h) decreased BCRP activity. Considering that altered drug transporter function can profoundly impact the accumulation and efficacy of CNS-targeting drugs, findings in the present study may be extremely important in the context of CNS infection-related therapeutics. Chikungunya (CHIKV), Dengue virus (DENV), Human Immunodeficiency virus (HIV), Japanese Encephalitis (JEV), SARS-CoV-2, Zika virus (ZIKV) and West Nile virus (WNV) all belong to the class of ssRNA viruses. 22, 23 In the present study, we have shown that the viral-mimic ssRNA inhibits P-gp function both in the short (4 h) and longerterm (24 h) . This may indicate potential implications in drug transfer across the BBB in vivo, in infections induced by ssRNA viruses. To our surprise, BCRP function was increased by ssRNA exposure at 24 h. Further studies are clearly required to determine the impact of ssRNA exposure on drug and xenobiotics accumulation in animal and human brain. Cytomegalovirus (CMV), DENV and ZIKV activate the host' immune system through TLR-3 stimulation, 24-26 therefore we used the TLR-3 agonist PolyI:C as an alternative viral mimic. We demonstrated that drug transporter function is regulated in a TLR type-specific manner, since ssRNA and PolyI:C induced opposite effects on P-gp and BCRP function. Human endometrial endothelial cells also present distinct responses to TLR-3 and TLR-7 agonists, based on the distinct inflammatory and antiviral immune pathways that they activate. 27 Nevertheless, infections mimicked by PolyI:C may profoundly influence the accumulation of potentially toxic compounds by reducing BCRP activity. Taken together, our results provide evidence that there may be viral-specific effects on human brain protection through modification of P-gp and BCRP function in the brain vascular endothelium. All infection-mimics had opposite effects on P-gp and BCRP function. LPS and PolyI:C stimulated P-gp function, but inhibited BCRP activity. In contrast, ssRNA decreased P-gp function whilst increasing BCRP activity. P-gp and BCRP can modulate the transfer of cytokines and other inflammatory mediators from the brain parenchyma to the blood, suggesting that infection itself might alter the accumulation of these compounds in the brain. While P-gp and BCRP share common inhibitors and substrates, their functional relationship is not well understood. In this regard, Abcb1a −/mutant mice exhibit threefold higher levels of Abcg2 mRNA in brain capillaries compared to wild type animals, 28 suggesting a BCRP compensation for the loss of P-gp. Results from the present study, provide the first evidence of a potential compensatory relationship between P-gp Another interesting finding of this study is that modification of transporter function in BECs by bacterial and viral mimics seems to occur independent of total protein or mRNA abundance. Studies have reported several posttranslational mechanisms regulating P-gp and BCRP activity in BECs, with no changes in gene or total protein expression. They include the insertion/removal of P-gp into/from the cell surface by Caveolin-1, 29,30 ERM (ezrin, radixin, and moesin) proteins, 31 or small GTPases. 32 In addition, phosphorylation 33, 34 or glycosylation 35 can also promote changes in drug transporter function, independent of changes in absolute transporter levels. There were no observable changes in plasma membrane P-gp associated with altered P-gp function in human BECs after infection. In this case, other regulatory mechanisms such as changes in the lipid microenvironment or protein-membrane composition 36, 37 may be contributing to the observed changes in P-gp function. Furthermore, indirect regulation of drug transporter activity mediated by the actin filament-associated protein (AFAP)-1 has been recently shown to inhibit P-gp function without changing total cellular or plasma membrane levels of P-gp protein in BECs. 38 Further studies are required to elucidate which signaling pathways are involved in the regulation of P-gp activity following infection. In the case of BCRP, LPS (bacterial infection mimic) decreased transporter activity and plasma membrane BCRP protein at 24 h while total cellular BCRP levels remained unchanged, suggesting a possible change in BCRP intracellular translocation induced by LPS. Similar regulation of BCRP has been previously reported in human embryonic stem cells 39 and vascular endothelial cells. 40 Pro-inflammatory cytokines, including IL-6, TNF-α, and IFN-ɣ, represents a critical component of the response to bacterial and viral infection. 41 We found in the present study that exposure to IL-6 decreased P-gp function and ABCB1 mRNA levels in BECs, consistent with previous findings in BECs derived from guinea pigs at various stages of development. 6 IL-6 (24 h) also down-regulated BCRP function and ABCG2 mRNA levels in human BECs in vitro, which was consistent with a previous study that utilized 72 h exposure in the same cell line (72 h). 42 TNF-α is a critical mediator of viral neurotrophic infections 43 and in the current study, there were no effects of TNF-α on P-gp function or ABCB1 mRNA levels. However, BCRP activity was acutely (4 h) reduced after TNF-α treatment, consistent with previous reports in human, porcine, and guinea pig BECs. 42, 44 IFN-ɣ plays a central role in viral replication and pathogenesis such as the ZIKV, 45 and disrupts the BBB, inducing the migration of inflammatory cells into the brain. 46 Therefore, we hypothesized IFN-ɣ would impact BBB function by downregulating drug transporter activity in human BECs. Indeed, IFN-ɣ exposure resulted in a significant reduction in BCRP function at 4 h and 24 h. In contrast, we found increased P-gp activity 24 h after exposure to IFN-ɣ. IFN-ɣ mediates BBB disruption following JEV infection in mice. 47 Thus, we can speculate that increased P-gp activity after exposure to this cytokine represents an adaptive mechanism in response to increased BBB permeability. Further studies are needed to address this possibility in human BECs. Notwithstanding, this study has identified the potential of IFN-ɣ to modulate both P-gp and BCRP at the human BBB, in vitro. Although the current study focused on drug transporter activity and expression, infection/ inflammation also changes other aspects of the BBB structure. Indeed, tight junctions (TJ), represent important modulators of BBB integrity following exposure to infective/inflammatory stimuli. Studies have demonstrated that the presence of Claudin-5, and its ratio to other claudins, are crucial to normal BBB function. [48] [49] [50] BBB breakdown induced by downregulation/degradation of TJ is a common feature of virus-induced neuropathogenesis. In vivo and in vitro studies have reported increased BBB permeability with concomitant downregulation of TJ proteins following different models of viral encephalitis, as well as exposure to infective agents, infection mimics, and proinflammatory mediators. [51] [52] [53] [54] [55] [56] [57] [58] [59] [60] [61] Clearly, further studies are required to investigate the relationship of TJ and drug transporter function during infection. Although the calcein-AM activity assay is a commonly used method for in vitro assessment of P-gp function and that calcein has been used as P-gp substrate for several years by different research groups, 7,62-69 some studies report that calcein could also be substrate for other ABC transporters, including MRP1 and MRP2. [70] [71] [72] There is no evidence in the literature of ABCC1/MRP1 or ABCC2/MRP2 responsiveness (altered expression or function) following infection in human BBB. However, considering hCMEC/D3 plasma membrane expression levels of MRP1 (1.65 ± 0.23 fmol/µg protein) compared to P-gp (3.87 ± 0.39 fmol/µg protein), 69 it is possible that the response we found may be driven by a combination of P-gp and MRP1 efflux. We recognize this is a potential limitation and highlight the importance of future studies to dissect the specific roles of P-gp and MRP1 in brain protection during infection. In conclusion, we have shown, for the first time, that bacterial and viral mimics influence the expression and function of major drug transporter systems in hCMEC/D3 cells. The current findings are important in the context of bacterial/viral CNS insult, that include Streptococcus pneumoniae, Haemophilus influenzae, Neisseria meningitidis, arboviruses, coronavirus, enterovirus, herpesvirus, poliovirus and rotavirus . 26, [73] [74] [75] These pathogens can induce the onset of meningitis/encephalitis, leading to severe cerebral injury and long-lasting neurological dysfunction. We provide in vitro evidence that stimulation of TLRs is associated with disrupted P-gp and BCRP function in human BECs. Should a similar effect occur in vivo brain protection against xenobiotics would be compromised and there altered biodistribution of drugs in the CNS could result. Further studies with coculture models would be extremely relevant to highlight the possible influence of astrocytes and microglia on brain endothelial drug transporter response to TLRs ligands. Also, given the transporter-specific sensitivity of P-gp and BCRP in BECs to different infection-mimics, polymicrobial infections may result in diverse patterns of P-gp and BCRP dysfunction in the brain endothelium, and this concept should be further investigated. Our results highlight the need to assess the effects of infection-mimics, and of important human pathogens on the exposure of xenobiotics and accumulation of therapeutic drugs at multiple times across the life course, in vivo. Acute effects of viral exposure on p-glycoprotein function in the mouse fetal blood-brain barrier ATP-binding cassette transporters in reproduction: a new frontier Developmental neurotoxicity test guidelines: problems and perspectives Regulation of multidrug resistance p-glycoprotein in the developing blood-brain barrier: interplay between glucocorticoids and cytokines TGF-β1 regulation of multidrug resistance P-glycoprotein in the developing male bloodbrain barrier Pro-inflammatory cytokine regulation of P-glycoprotein in the developing blood-brain barrier Glucocorticoids modify effects of TGF-β1 on multidrug resistance in the fetal bloodbrain barrier Quantitative targeted absolute proteomic analysis of transporters, receptors and junction proteins for validation of human cerebral microvascular endothelial cell line hCMEC/D3 as a human blood-brain barrier model Recent expansions on cellular models to uncover the scientific barriers towards drug development for Alzheimer's Disease Real-time acquisition of transendothelial electrical resistance in an all-human, in vitro, 3-dimensional, blood-brain barrier model exemplifies tight-junction integrity The hCMEC/D3 cell line as a model of the human blood brain barrier Effect of oxygen on multidrug resistance in the first trimester human placenta Dexamethasone stimulates placental system A transport and trophoblast differentiation in term villous explants Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method Astrocytemediated regulation of multidrug resistance p-glycoprotein in fetal and neonatal brain endothelial cells: age-dependent effects Identification and functional characterization of breast cancer resistance protein in human bronchial epithelial cells (Calu-3) Overlapping substrate and inhibitor specificity of human and murine ABCG2 Differential roles of MDA5 and RIG-I helicases in the recognition of RNA viruses IRFs: master regulators of signalling by Toll-like receptors and cytosolic pattern-recognition receptors Alteration in P-glycoprotein at the blood-brain barrier in the early period of MCAO in rats Inhibition of multidrug resistance transporter-1 facilitates neuroprotective therapies after focal cerebral ischemia TLR7 is a key regulator of innate immunity against Japanese encephalitis virus infection COVID-19: a new challenge for human beings Zika virus depletes neural progenitors in human cerebral organoids through activation of the innate immune receptor TLR3 Human TLR3 recognizes dengue virus and modulates viral replication in vitro Blockade of Lyn kinase upregulates both canonical and non-canonical TLR-3 pathways in THP-1 monocytes exposed to human cytomegalovirus Human endometrial endothelial cells generate distinct inflammatory and antiviral responses to the TLR3 agonist, Poly(I:C) and the TLR8 agonist, viral ssRNA Expression, up-regulation, and transport activity of the multidrug-resistance protein Abcg2 at the mouse blood-brain barrier Modulation of p-glycoprotein function by caveolin-1 phosphorylation Rapid, reversible modulation of blood-brain barrier P-glycoprotein transport activity by vascular endothelial growth factor Proteins regulating the intercellular transfer and function of P-glycoprotein in multidrug-resistant cancer The small GTPases Rab5 and RalA regulate intracellular traffic of P-glycoprotein Pim-1 kinase protects P-glycoprotein from degradation and enables its glycosylation and cell surface expression The 44-kDa Pim-1 kinase phosphorylates BCRP/ABCG2 and thereby promotes its multimerization and drug-resistant activity in human prostate cancer cells The ABC of glycosylation Rapid loss of blood-brain barrier P-glycoprotein activity through transporter internalization demonstrated using a novel in situ proteolysis protection assay Localization of the human breast cancer resistance protein (BCRP/ ABCG2) in lipid rafts/caveolae and modulation of its activity by cholesterol in vitro Actin filament-associated protein 1 (AFAP-1) is a key mediator in inflammatory signaling-induced rapid attenuation of intrinsic P-gp function in human brain capillary endothelial cells Dynamic ABCG2 expression in human embryonic stem cells provides the basis for stress response Hyperuricemia enhances intracellular urate accumulation via down-regulation of cell-surface BCRP/ABCG2 expression in vascular endothelial cells Neuroinflammation and infection: molecular mechanisms associated with dysfunction of neurovascular unit Regulation of BCRP (ABCG2) and P-glycoprotein (ABCB1) by cytokines in a model of the human blood-brain barrier Acute and chronic neurological consequences of early-life Zika virus infection in mice Regulation of major efflux transporters under inflammatory conditions at the blood-brain barrier in vitro Selective activation of type II interferon signaling by Zika Virus NS5 protein Interferon (IFN)-beta 1a and IFN-beta 1b block IFN-gamma-induced disintegration of endothelial junction integrity and barrier Viral infection of the central nervous system and neuroinflammation precede blood-brain barrier disruption during japanese encephalitis virus infection Claudin-1 and claudin-5 expression and tight junction morphology are altered in blood vessels of human glioblastoma multiforme Correlation of tight junction morphology with the expression of tight junction proteins in blood-brain barrier endothelial cells Size-selective loosening of the blood-brain barrier in claudin-5-deficient mice Toll-like receptor 3 mediates West Nile virus entry into the brain causing lethal encephalitis Japanese encephalitis virus induces matrix metalloproteinase-9 expression via a ROS/c-Src/ PDGFR/PI3K/Akt/MAPKs-dependent AP-1 pathway in rat brain astrocytes Viral pathogen-associated molecular patterns regulate blood-brain barrier integrity via competing innate cytokine signals Enhancement of blood-brain barrier permeability and reduction of tight junction protein expression are modulated by chemokines/cytokines induced by rabies virus infection Interferonlambda restricts West Nile virus neuroinvasion by tightening the blood-brain barrier HIV-1-induced alterations of claudin-5 expression at the blood-brain barrier level Toll-like receptor-3 mediates HIV-1-induced interleukin-6 expression in the human brain endothelium via TAK1 and JNK pathways: implications for viral neuropathogenesis Rho-mediated regulation of tight junctions during monocyte migration across the blood-brain barrier in HIV-1 encephalitis (HIVE) Adherens junction: molecular architecture and regulation Expression and regulation of toll-like receptors in cerebral endothelial cells Transitory loss of glia and the subsequent modulation in inflammatory cytokines/chemokines regulate paracellular claudin-5 expression in endothelial cells Calcein accumulation as a fluorometric functional assay of the multidrug transporter In vitro P-glycoprotein assays to predict the in vivo interactions of P-glycoprotein with drugs in the central nervous system Phyllanthin and hypophyllanthin inhibit function of P-gp but not MRP2 in Caco-2 cells In vitro and in vivo anti-tumour effects of MPT0B014, a novel derivative aroylquinoline, and in combination with erlotinib in human non-smallcell lung cancer cells The functional implications of common genetic variation in CYP3A5 and ABCB1 in human proximal tubule cells Rational use of in vitro P-glycoprotein assays in drug discovery Rapid assessment of p-glycoprotein-drug interactions at the blood-brain barrier Transport properties of the multidrug resistance-associated protein (MRP) in human tumour cells Inhibitory effect of the reversal agents V-104, GF120918 and Pluronic L61 on MDR1 Pgp-, MRP1-and MRP2-mediated transport Kinetic analysis of calcein and calcein-acetoxymethylester efflux mediated by the multidrug resistance protein and P-glycoprotein Detection of herpesvirus, enterovirus, and arbovirus infection in patients with suspected central nervous system viral infection in the Western Brazilian Amazon Early-onset neonatal sepsis Host-pathogen interactions in bacterial meningitis We are grateful to Alisa Kostaki for her technical support in this research. The authors report no conflict of interest.