WITHDRAWN: Melatonin improves methamphetamine-induced blood brain barrier impairment through NADPH oxidase-2 in primary rat brain microvascular endothelium cells

WITHDRAWN: Melatonin improves methamphetamine-induced blood brain barrier impairment through NADPH oxidase-2 in primary rat brain microvascular endothelium cells

Author’s Accepted Manuscript Melatonin promotes blood-brain barrier integrity in methamphetamine –induced inflammation in primary rat brain microvascu...

1MB Sizes 0 Downloads 51 Views

Author’s Accepted Manuscript Melatonin promotes blood-brain barrier integrity in methamphetamine –induced inflammation in primary rat brain microvascular endothelial cells Pichaya Jumnongprakhon, Piyarat Govitrapong, Chainarong Tocharus, Jiraporn Tocharus www.elsevier.com/locate/brainres

PII: DOI: Reference:

S0006-8993(16)30438-3 http://dx.doi.org/10.1016/j.brainres.2016.06.014 BRES44965

To appear in: Brain Research Received date: 20 March 2016 Revised date: 10 May 2016 Accepted date: 8 June 2016 Cite this article as: Pichaya Jumnongprakhon, Piyarat Govitrapong, Chainarong Tocharus and Jiraporn Tocharus, Melatonin promotes blood-brain barrier integrity in methamphetamine –induced inflammation in primary rat brain microvascular endothelial cells, Brain Research, http://dx.doi.org/10.1016/j.brainres.2016.06.014 This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting galley proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

1

Melatonin promotes blood-brain barrier integrity in methamphetamine –induced inflammation in primary rat brain microvascular endothelial cells Pichaya Jumnongprakhon1, Piyarat Govitrapong2,3, Chainarong Tocharus1, Jiraporn Tocharus4* 1

Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200,

Thailand 2

Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University,

Bangkok, Thailand 3

Center for Neuroscience and Department of Pharmacology, Faculty of Science, Mahidol

University, Bangkok, Thailand 4

Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200,

Thailand *

Correspondence to. Jiraporn Tocharus, Department of Physiology, Faculty of Medicine,

Chiang Mai University, Chiang Mai 50200, Thailand, Tel.: (6653)945362; fax: (6653) 945365. E-mail: [email protected]

Abstract Melatonin is a neurohormone and has high potent of antioxidant that is widely reported to be active against methamphetamine (METH)-induced toxicity to neuron, glial cells, and brain endothelial cells. However, the role of melatonin on the inflammatory responses which are mostly caused by blood–brain barrier (BBB) impairment by METH administration has not been investigated. This study used the primary rat brain microvascular endothelial cells

2

(BMVECs) to determine the protective mechanism of melatonin on METH-induced inflammatory responses in the BBB via nuclear factor-ĸB (NF-κB) and nuclear factor erythroid 2-related factor-2 (Nrf2) signaling. Herein, we demonstrated that melatonin reduced the level of the inflammatory mediators, including intercellular adhesion molecules (ICAM)-1, vascular cell adhesion molecules (VCAM)-1, matrix metallopeptidase (MMP)-9, inducible nitric oxide synthase (iNOS), and nitric oxide (NO) caused by METH. These responses were related to the decrease of the expression and translocation of the NF-κB p65 subunit and the activity of NADPH oxidase (NOX)-2. In addition, melatonin promoted the antioxidant processes, modulated the expression and translocation of Nrf2, and also increased the level of heme oxygenase (HO)-1, NAD (P) H: quinone oxidoreductase (NQO)-1, γ-glutamylcysteine synthase (γ-GCLC), and the activity of superoxide dismutase (SOD) through NOX2 mechanism. In addition, we found that the protective role of melatonin in METH-induced inflammatory responses in the BBB was mediated through melatonin receptors (MT1/2). We concluded that the interaction of melatonin with its receptor prevented METH-induced inflammatory responses by suppressing the NF-κB signaling and promoting the Nrf2 signaling before BBB impairment. Keywords: Blood–brain barrier, Melatonin, Methamphetamine, NF-κB, Nrf2

1. Introduction Methamphetamine (METH) is the highly potent neurotoxin that is known worldwide to cause neurodegeneration by inducing dysfunction of neurons, glial cells, and the blood–brain barrier (BBB) (Taylor et al., 2013; Loftis and Janowsky, 2014; Cheng et al., 2015; Jumnongprakhon et

3

al., 2014, 2015; Parameyong et al., 2015; Northrop and Yamamoto, 2015; Sun et al., 2015). The primary event of neurodegeneration that is caused by METH has been direct impairment of the BBB prior to mediation of the central nervous system (CNS) damage (Bank and Erikson, 2010). BBB impairment by METH has been reviewed in several mechanisms, including the hyperactivity of NADPH oxidase (NOX)-2 which generates excessive amounts of free radicals, such as reactive oxygen species (ROS) and reactive nitrogen species (RNS) (Ramirez et al., 2009; Park et al., 2012); the dysfunction of cytoskeleton and the transmembrane protein of tight junction, which is the controlling of paracellular permeability (Park et al., 2013; Fernandes et al., 2015); the dysfunction of the uptake and the efflux activities (Elali et al., 2012); and the activation of caspase cascade in cell death response or apoptosis (Abdul et al., 2011;Ma et al., 2014; Fisher et al., 2015). Moreover, overexpression of inflammatory mediators such as inducible nitric oxide synthase (iNOS), nitric oxide (NO), interleukin (IL)-1, and tumor necrosis factor (TNF) α, which is an important factor in inflammatory response, has also been reported (Fernandes et al., 2014; Coelho-Santos et al., 2015; Parikh et al., 2015; Zhang et al., 2015; Skaper et al., 2014; Hussain et al., 2015; Kothur et al., 2015).

Previous studies have demonstrated that the release of cytokines causes progressive BBB impairment by the activation of leukocyte infiltration and monocyte adhesion molecules such as intercellular adhesion molecules (ICAM)-1, vascular cell adhesion molecules (VCAM)-1, and matrix metallopeptidase (MMP)-9 leads to tight junction impairment (Urrutia et al., 2013; Fernandes et al., 2014; Wang et al., 2014; Parikh et al., 2015). Decrease in the levels of tight junction proteins such as ZO-1, occludin, and claudin-5 are associated with BBB dysfunction

4

and increased paracellular permeability, leading to decrease transendothelial electric resistance (TEER) values (Mahajan et al., 2008; Rosas-Hernandez et al., 2013). The nuclear factor-ĸB (NF-κB) signaling is the major pathway that regulates the inflammatory cytokine expression and has been widely studied in inflammatory responses (Kawai and Akira, 2007). Previous studies have reported several stimuli such as lipopolysaccharide (LPS) and METHinduced inflammation in both neuronal cells and glial cells by the overactivation of NF-κB signaling (Flora et al., 2003; Shah et al., 2012; Wires et al., 2012; Permpoonpattana et al., 2013; Coelho-Santos et al., 2015; Jumnongprakhon et al., 2015). Therefore, the inhibition of these negative effects may be beneficial in protecting BBB impairment.

Upon inflammation, the cells maintain homeostasis by regulating the defense system. The inflammatory defending system mostly mediates the phase II antioxidant enzymes, including heme oxygenase (HO)-1, NAD (P) H: quinone oxidoreductase (NQO)-1, and γglutamylcysteine synthase (γ-GCLC), superoxide dismutase (SOD), catalase (CAT) which is regulated by the nuclear factor erythroid 2-related factor-2 (Nrf2) mechanism (Huang et al., 2015). The activation of Nrf2 signaling triggers the Nrf2 protein to translocate into the nucleus and bind to the antioxidant response element (ARE) (Chen et al., 2015). Several reports have reviewed that the Nrf2 mechanism is the important target of toxin, including METH, prior to mediating the cellular stress, inflammation, dysfunction, and death (Pacchioni et al., 2007; Granado et al., 2011; Jumnongprakhon et al., 2015; Ramkissoon and Wells, 2013, 2015). Moreover, the Nrf2 could reduce the activity of NF-κB signaling (Li et al., 2008). Thus, the activation or protection of this system may increase BBB integrity.

5

Melatonin (N-acetyl-5-methoxytryptamine) is mainly produced from the pineal gland that plays an important role of strong antioxidant, anti-inflammation, and anti-apoptosis, and it is widely accepted that it has high potential as regards protecting the CNS from toxic exposure via melatonin receptors (MT1/2), including exposure to METH. Moreover, melatonin has demonstrated that it could protect BBB by ischemic and hemorrhagic stroke, and sepsis or inflammatory responses to infection (Chen et al., 2006; Reiter et al., 2010; Tocharus et al., 2010; Chern et al., 2012; Accuna-Castroviejo et al., 2014; Gracia et al., 2014; Wang et al., 2014; Locoste et al., 2015; Ngunen et al., 2015; Manchester et al., 2015; Zhao et al., 2015). According to its exerting effects, melatonin might be beneficial for protecting BBB in METH administration. In the present study, we hypothesized that melatonin might be able to attenuate METH-induced inflammatory responses in the BBB by suppressing the NF-κB signaling and promoting the Nrf2 signaling through NOX2 mechanism.

2. Results 2.1. Melatonin protect against METH-induced NF-κB mechanism via both NF-κB and NOX2 in BMVECs Our present study demonstrated that METH-induced the excessive expression of ICAM-1, VCAM-1, MMP-9, iNOS, and NO, and that it was also closely related with the activation of inflammation through NF-κB mechanism through NOX2 activity which is possible cause of BBB impairment. Pretreatment with melatonin could promote the BBB integrity that was impaired by METH. To elucidate the protective effect of melatonin on

6

METH-induced BBB impairment as to whether it is directly against the NF-κB mechanism or the NOX2 activity, 10 µM of JSH23 (the selective inhibitor of NF-κB translocation) and 100 µM of apocynin (NOX2 inhibitor) were used in this experiment. The treatment with METH alone significantly increased the expression of ICAM-1, VCAM-1, and MMP-9 (Fig. 1A); iNOS (Fig. 1B); and NO (Fig. 1C), and these effects were significantly reversed by melatonin treatment (p<0.001). Especially, JSH23 completely abolished the expression of these proteins, nearly to the basal level. Herein, it is clearly indicated that melatonin protects against METHmediated expression of ICAM1, VCAM1, iNOS, and NO via directly suppressing the NF-κB mechanism and partially protecting via the NOX2 mechanism that might be involved with the increasing of BBB integrity. However, METH+JSH-23 significantly decreased the expression of MMP-9 (p<0.001) when compared to METH+melatonin. This result suggests that the major regulation of MMP-9 expression that caused by METH is NF-κB mechanism and melatonin might partially abolish METH-toxicity through this mechanism. Next, we investigated the role of melatonin on the NF-κB expression and activity via directly inhibiting NF-κB or NOX2. The results showed that melatonin significantly reduced the translocation and expression of the p65 subunit in BMVECs, as shown in Fig. 1D–F (p<0.001). The co-treatment of JSH23 with melatonin showed results similar to those of the control group. In the presence of the NOX2 inhibitor and by co-treatment with melatonin, the translocation of p65 significantly decreased, in comparison to METH treatment; however, the level was still higher than those in JSH23 treated group. These data suggested that NF-κB mechanism is the major pathway of melatonin to protect from METH-induced inflammation in BMVECs and that it is partly mediated by NOX2 activity.

7

2.2. Melatonin attenuates METH-suppressed Nrf2 mechanism via NOX2 in BMVECs

One of the major factors that caused progressive inflammation in BBB by METH is the decreasing activity of the cellular defending system, especially the reduction of antioxidant enzymes such as glutathione peroxidase (GPx), SOD, HO-1, and NQO-1. Next, we determined the protective effect of melatonin on METH-reduced antioxidant enzymes regarding whether it is closely related to NF-κB mechanism or NOX2 mechanism. METH treatment alone significantly decreased the expression of HO-1, NQO-1, GCLC, and SOD activity, respectively, as shown in Fig. 2A–D, and these effects significantly increased in melatonin treatment (p<0.001). As regards the inhibition of NF-κB, the METH treatment showed results similar to those of METH treated alone. In contrast, the apocynin treatment significantly increased the level of these proteins, similar to the control group (p<0.001). Pretreatment with JSH23 and melatonin followed by METH did not significantly restore these effects. However, apocynin-treated cells showed results similar to those of the control group. Herein, these results indicate that melatonin protects against METH-reduced expression of HO-1, NQO-1, GCLC, and SOD activity via directly suppressing NOX2. Then, we investigated the role of melatonin whether it promoted Nrf2 expression and activity via directly inhibiting NF-κB or NOX2. The results showed that melatonin significantly increased the translocation and expression of Nrf2 in BMVECs compared to METH treated group, as shown in Fig. 2E–G (p<0.001). The co-treatment of JSH23 with melatonin did not alter this effect when compared to METH treatment alone. On the other hand, co-treatment with apocynin and melatonin significantly increased this effect when compared to METH-treated

8

cells. These data suggest that melatonin protects against METH-suppressed Nrf2 signaling in BMVECs mainly through NOX2 mechanism.

2.3. Melatonin diminishes nitrative stress, apoptosis, and cell integrity impairment by METH treatment via NF-κB but not oxidative stress in BMVECs

As shown in Fig 3, we next determined the important role of NF-κB mechanism in inducing cell death and impairment of tight junction by promoting oxidative and nitrative stress caused by METH in BMVECs. The results showed that METH-mediated the ROS level (Fig. 3A), RNS level (Fig. 3B), paracellular permeability (Fig. 3D), caspase-3 level (Fig. 3E), and percentage of apoptotic cells (Fig. 3F), and also in the attenuation of the TEER values (Fig. 3C). When pretreatment with melatonin prior treated with METH, it could prevent these negative responses. To investigate the protective role of melatonin on METH-induced BBB impairment whether involved with NF-κB mechanism. The co-treatment of JSH23 and METH resulted in the reduction of the RNS level (Fig. 3B), paracellular permeability (Fig. 3D), caspase-3 level (Fig. 3E), and percentage of apoptotic cells (Fig. 3F), and also in the promotion of the TEER values (Fig. 3C) similar to those of the control group (p<0.001). However, the ROS level did not change in this treatment when compared to the treatment with METH alone. Thus, the impairment of the BBB caused by METH markedly activated the NF-κB mechanism. Moreover, pretreatment with JSH23 and melatonin or JSH23 alone prior to treatment with METH significantly reversed the negative effects when compared to METH treatment alone. This result suggests that melatonin protects against METH-induced BBB impairment by directly suppressing the NF-κB mechanism.

9

2.4. Melatonin directly prevents METH-induced inflammation by interacted with its receptors (MT1/2)

Finally, we determined whether the protective role of melatonin on METH-induced inflammation is mediated via the MT1/2 receptor. The results showed that pretreatment with luzindole, the antagonist of MT1/2 receptors, prior to treated with melatonin, completely ameliorated the effect of melatonin on NF-κB mechanism by significantly increasing ICAM1, VCAM-1, MMP-9, iNOS, NO, nuclear p65, and total p65, as shown in Figure 4 (p<0.001). Moreover, the activation of Nrf2 significantly decreased the expression of HO-1, NQO-1, GCLC, SOD activity, nuclear Nrf2, and total Nrf2 (Fig. 5, p<0.001). These findings suggest that melatonin protects against METH-induced inflammation in BMVECs is mediated via MT1/2 receptors and resulted in suppressing NF-κB and activating.

3. Discussion

The co-culture of purified primary rat BMVECs and primary mixed glial cells was used as the BBB model in this study for investigating the protective role of melatonin on METH-induced inflammation. This finding demonstrated that melatonin protected against METH-induced inflammation by directly inhibiting the NF-κB signaling and promoting the Nrf2 via NOX2 mechanism. Importantly, the protective role of melatonin in METH-induced inflammation is via interaction with its receptor-mediated signaling.

10

Short-term and long-term METH exposure leading to neurodegenerative diseases has been widely reported (Panenka et al., 2013; Radfar and Rawson, 2014; Yu et al., 2015). The toxicity of METH is manifested in the CNS by impairing the BBB which acts as a strong shield of the CNS through its mediation of cell stress, dysfunction, inflammation, and death (O'SHEA et al., 2014; Northrop and yamamoto, 2015). It is well known that NOX2 is the key regulator of several mechanisms in BBB impairment (Cahill-Smith and Li, 2014), but its role in inflammation is still unclear. Our results demonstrated that apocynin, the NOX2 inhibitor, partly down-regulated the expression of inflammatory mediators via NF-κB signaling caused by METH. Based on this finding, we suggested that NOX2 is one of the regulators of NF-κB in METH-induced BBB impairment. NF-κB signaling is the cellular regulation of inflammation by generating several cytokines such as TNF-α, iNOS, and NO, and also the mediation of the expression of monocytes adhesion molecules which interrupt the function of the tight junction in brain endothelium such as ICAM1 and VCAM1. Moreover, this signaling could promote extracellular matrix enzymes such as MMP-2 and MMP-9 for degrading the laminin and collagen type IV in basal lamina (Csiszar et al., 2008; Zhang et al., 2009; Saito et al.,2013; Yuan et al., 2015). The blockage of this signaling could promote BBB integrity by preserving the tight junction. Our study showed that inhibition of the NF-κB or the NOX2 activity, METH did not activate the expressions of ICAM-1, VCAM-1, MMP-9, iNOS, and NO. Similar results were observed in melatonin-treated cells. However, the decreasing of these responses in NOX2 inhibition was not completely inhibited as compared to the NF-κB inhibition group. Thus, these results suggest that melatonin protects from METH-induced inflammation by directly inhibiting the NF-κB pathway and partly inhibiting the NOX2

11

mechanism. As previous studies have reviewed, the inflammation which triggers the NF-κB signaling was mediated via several mechanisms including NOX2, toll-like receptors (TLRs)-4, and α7-nicotinic acetylcholine receptors (α7-nAChR) (Gloireet al., 2006; Maloney et al., 2009; Gray and Jandeleit-Dahm, 2015; Zhanget al., 2015). From the result, the partial inhibition of NOX2 on NF-κB signaling might be co-activated with other mechanisms, especially the α7nAChR which has been previously reported to have a role in inflammation due to BBB impairment that is caused by METH administration (Zhang et al., 2015). Inhibition of the NFκB activity, METH failed to induce BBB impairment by a decrease in the oxidative damage and the apoptosis, thereby increasing the BBB integrity, as observed in the increasing of the TEER value and the decreasing of the paracellular permeability. Based on these data, we suggest that NF-κB signaling is the major target of melatonin to modulate BBB function caused by METH. However, the inhibition of NF-κB did not completely decrease the ROS level, which was mostly generated by NOX2 (Park et al., 2012; Garrido-Urbani et al., 2014; Xiao et al., 2015). Thus, the inhibition of NF-κB was not the only critical factor in the protection of the BBB. The advantage of melatonin, as this study demonstrated, is the protective effect both on NOX2 mechanism and NF-κB prior to reducing METH toxicity; as a result, it was useful for the protection of the BBB. We further evaluated the protective role of melatonin on METH-induced inflammation via NF-κB regarding whether it required its receptors. The results showed that with luzindole (a non-selective antagonist MT1/2 receptor) pretreatment, melatonin failed to inhibit the translocation of the p65 subunit of NF-κB and the expression of ICAM-1, VCAM-1, MMP-9, iNOS, and NO. Based on this finding, we

12

suggested that melatonin required its receptor for abolishing the inflammation via NF-κB signaling.

As regards inflammatory responses, the cell has its defending mechanism of mediating antioxidant enzymes for protecting against cell stress, dysfunction, inflammation, and death that caused by exposure to toxins (Huang et al., 2015). Previous studies have established that METH caused brain damage by interrupting or reducing antioxidant enzymes, including SOD, catalase (CAT), GPx, HO-1, NQO-1, GCLC, etc. Moreover, the depletion of these enzymes through reduced production induced progressive stress and inflammation of cells prior to dysfunction and death (Kensler et al., 2007; Osburn and Kensler, 2008; Sykiotiset al., 2011; Miyazaki et al., 2011; Permpoonpattana et al., 2013; Huang et al., 2015; Jumnogprakhon et al., 2015). In this study, we found that melatonin could promote the activity of SOD and the expression of HO-1, NQO-1, and GCLC in BMVECs. Next, we proceeded to investigate the involvement of NF-κB or NOX2 in the protective effect of melatonin in METH-induced inflammation in the BBB. Inhibition of NF-κB signaling pathway, METH did not promote the activity of SOD and the expression of HO-1, NQO-1, and GCLC; so, we suggested that the reduction of the antioxidant enzyme by METH did not involve NF-κB signaling. On the other hand, inhibition of NOX2 activity, METH caused promotion of the SOD activity and expression of HO-1, NQO-1, and GCLC were upregulated; therefore, we suggest that METH suppressed antioxidant activity via NOX2 mechanism. Subsequently, we investigated the protective role of melatonin in the promotion of antioxidant enzymes and investigated whether it involved with NOX2. Next, the cells were subjected to co-treatment of apocynin with melatonin and METH, which caused the

13

promotion of the SOD activity and the expression of HO-1, NQO-1, and GCLC, which is similar to the co-treatment of apocynin with METH. Based on this finding, we suggest that the protective role of melatonin which promotes the antioxidant enzyme activity and expression is closely related to NOX2 mechanism. As demonstrated in a previous study, it has been established that antioxidant enzymes are mostly regulated by Nrf2 signaling, and they reported that a decrease in Nrf2 activation causes depletion of antioxidant enzymes (Permpoonpattana et al., 2013; Jumnogprakhon et al., 2015). In agreement with those reviews, it was observed that METH exposure in BMVECs decreased the expression and translocation of Nrf2 in relation with the low expression of HO-1, NQO-1, and GCLC and SOD activity. Moreover, we found that a decrease in Nrf2 activity did not implicate NF-κB signaling but it was closely involved with NOX2. Importantly, melatonin could promote the expression and Nrf2 activity, and this promotion was closely involved with NOX2 devoid of NF-κB signaling. In contrast, a few studies have demonstrated that the inhibition of NF-κB signaling could promote Nrf2 activity (Wijayanti et al., 2004; Pinkaew et al., 2015). Thus, we suggest that the activation of Nrf2 in the brain endothelial cells may be closely related with NOX2 signaling rather than NF-κB signaling because the NF-κB signaling may be a parallel cascade during inflammation. We also found that in the presence of luzindole, melatonin failed to promote the Nrf2 expression and activity including the expression of HO-1, NQO-1, GCLC, and SOD activity caused by METH. Thus, these data suggest that the protective role of melatonin in METH-induced suppressing of Nrf2 signaling requires the interaction of melatonin and its receptor.

3.1. Conclusions

14

We concluded that the interaction of melatonin with its receptor (MT1/2) protected against BBB impairment caused by METH by directly inhibiting the NF-κB signaling and modulating the Nrf2 signaling via NOX2 mechanism (Fig. 6). Thus, melatonin might be beneficial in protecting the BBB from inflammation caused by METH or other pathogens.

4. Experimental procedure

4.1. Reagents and chemicals Melatonin, luzindole, JSH23, apocynin, 4 kDa FITC-dextran, and myeloperoxidase (MPO) were purchased from Sigma-Aldrich Chemical Company (St. Louis, MO, USA). Anti-β-actin, anti-iNOS, anti-p65 subunit, anti-lamin B1, Muse®Annexin V & Dead Cell Kit, and Muse® Caspase-3/7 Kit were purchased from Merck Millipore (Millipore, MA, USA). The following antibodies were used for the western blot analysis: anti-Nrf2, anti-HO-1, anti-NQO-1, antiGCLC, anti-ICAM-1, anti-VCAM-1, and anti-MMP-9 (Abcam, Cambridge, UK); and antimouse IgG peroxidase-conjugated secondary antibody and anti-rabbit IgG peroxidaseconjugated secondary antibody (Millipore, MA, USA). The superoxide dismutase assay kit was purchased from Cayman Chemical Company (Cayman, MI, USA). 4.2. Primary rat brain microvascular endothelial cells (BMVECs) isolation and culture The isolation of the BMVECs was performed according to the protocol from Liu et al., 2013. Briefly, three whole brains of 10-day-old neonatal rats without meninges and large blood vessel were removed, and then were minced into small pieces in a pre-chilled phosphate

15

buffer saline (PBS) and centrifuged at 1000 rpm at 4°C for 5 min. Then, the pellets were homogenized in 25% bovine serum albumin (BSA) and centrifuged at 3000 rpm at 4°C for 10 min. The microvessel pellets were digested with 0.1% collagenase type II (Sigma, St. Louis, MO, USA) for 30 min at 37°C. Then, the digested microvessels were resuspended in DMEM/F12 and centrifuged at 1,000 rpm for 5 min. The precipitate layer was collected and resuspended in DMEM/F12 supplemented with 10% fetal bovine serum (FBS), 3 mg/ml glucose, 0.5 mM glutamine, 3 µM puromycin, and penicillin/streptomycin. The cell suspensions were cultured at 37°C in a humidified atmosphere of 5% CO2 for 3 days. After the growth of the BMVECs for 90–100% confluence, the cells were passaged with 0.25% trypsin:EDTA and resuspended in DMEM/F12 supplemented with 10% FBS, 3 mg/ml glucose, 0.5 mM glutamine, and penicillin/streptomycin in 37°C in a humidified atmosphere of 5% CO2. The anti-Von Willebrand factor VIII and the anti-α actin antibody were stained on the cells for the confirming of brain endothelial cells and observed under a fluorescent microscope (Olympus, Tokyo, Japan) (Data not shown). Protocols were performed in compliance with the Guide for the Care and Use of Laboratory Animal (Chiang Mai University) and were approved by Animal Care and Use committee.

4.3. Primary mixed glial cells isolation and culture Primary mixed glial cells were prepared following a protocol based on the McCarthy and de Vellis, 1980. Briefly, whole brains of three neonatal rats that were 0–2 days old were removed. After that, the brains were homogenized and centrifuged at 3,000 rpm for 10 min. The pellets were resuspended in glial cell maintenance media (DMEM/F12 supplemented with 10% FBS,

16

0.5 mM glutamine, and penicillin/streptomycin) and cultured at 37°C in a humidified atmosphere of 5% CO2. The anti-GFAP was stained on the cells for the confirmation of glial cells, and observed under a fluorescent microscope (Data not shown).

4.4. Cultivation of BBB model in vitro The BBB model was cultivated by co-culturing of the purified BMVECs with primary mixed glial cells. Briefly, the primary mixed glial cells were firstly maintained in 24-well plates at 37°C in a humidified atmosphere, at 5% CO2, for 7 days. The purified BMVECs were then cultured on collagen/fibronectin (Sigma, St. Louis, MO, USA) coated-inserted 0.4 µm transwells and maintained at 37°C in a humidified atmosphere, at 5% CO2, until 100% confluence was reached. The suitable representation of the BBB model in vitro also confirmed the transendothelial electric resistance (TEER) values by the EVOM2 voltometer with STX-2 electrodes, TEER >200Ω∙cm2 (Millipore, Inc., MA, USA). Prior to investigate in all the experiments, the primary mixed glial cells were discarded and switched the culturing media to media free serum.

4.5. Western blot analysis

To perform the western blot analysis, the cultured BMVECs were maintained in collagen/fibronectin coated-inserted trans-wells of 24-well plates at a density of 1×105cells/ml at 37°C until TEER >200 Ω∙cm2. The cells were prior treated with apocynin (100 µM) or JSH23 (10 µM) for 1 h. Then, the cells were pretreated with melatonin (1, 10, and 100 nM) for 2 h, and then incubated in the presence or absence of METH (100 µM, the cytotoxicity dose

17

which confirming by MTT assay) (Data not shown) for 24 h. The cells were harvested and lysed for the subcellular fraction of cytosolic and nuclear proteins, as described previously (Jumnongprakhon et al., 2015). Briefly, the cytosolic protein was prepared by a hypotonic lysis buffer containing 10 mM HEPES (pH 7.9), 1.5 mM magnesium chloride, 10 mM potassium chloride, 0.5 mM phenylmethylsulfonyl fluoride, 0.5 mM dithiothreitol, and a cocktail of protease inhibitors. The nuclear protein was prepared by lysing the pellets after cytosolic preparation with the hypertonic extraction buffer containing 10 mM HEPES (pH 7.9), 0.42 M sodium chloride, 1.5 mM magnesium chloride, 10 mM potassium chloride, 0.5 mM phenylmethylsulfonyl fluoride, 1 mM dithiothreitol, and a cocktail of protease inhibitors. The Bradford protein assay (Bio-Rad Laboratories, Hercules, CA, USA) was used to determine the protein concentration. Equal amounts of 50 µg proteins were separated with the 10–15% SDS-PAGE and transferred to a PVDF membrane (Immobilon-P, Millipore, Bedford, MA, USA). The membranes were then probed overnight with anti-iNOS, anti-p65, anti-ICAM1, anti-VCAM-1, anti-MMP-9, anti-Nrf2, anti-HO-1, anti-NQO-1, and anti-GCLC (1:2000). Then, the blotted cells were then incubated with anti-mouse and anti-rabbit IgG peroxidaseconjugated secondary antibodies (1:4000) (Millipore, MA, USA). Finally, the Immobilon Western HRP substrate (Millipore, MA, USA) was used to incubate the blots prior to exposure to an X-ray film. The densitometry was analyzed by using the Image-J®software. The β-actin was used to normalize the cytosolic fraction and lamin B1 was used to normalize the nuclear fraction.

4.6. Griess reaction assay

18

After the cells were exposed to the condition with the phenol red free media, the culture media were collected and the nitrite production determined by Griess reaction assay. An equal volume of the supernatant reacted with an equal volume of a mixture of 1% sulfanilamide in 5% phosphoric acid for 5 min and 0.1% N-(1-napthyl) ethylenediamine hydrochloride for 5 min in the dark at room temperature. The absorbance was measured using a microplate reader at 540 nm. The known concentrations of sodium nitrite (NaNO2) were used as the standard curve.

4.7. Superoxide dismutase activity assay

After the cells were exposed to the condition, the cells were harvested and lysed with ice-cold lysis buffer containing1% NP-40, 1% sodium deoxycholate, 0.1% sodium dodecyl sulfate, 40 mM β-glycerophosphate, 50 mM sodium fluoride, 2 mM sodium orthovanadate, and a cocktail of protease inhibitors. The SOD activity was determined using an SOD assay kit (Cayman Chemical Company, MI, USA), according to the manufacturer’s protocol, which utilized a tetrazolium salt for the detection of the superoxide radicals generated by xanthine oxidase and hypoxanthine.

4.8. ROS and RNS assay The cultured BMVECs were maintained in 96-well microplates at a density of 1×105 cells/ml at 37°C for 24 h. The cells were prior treated with JSH23 (10 µM) for 1 h. Then, the cells were pretreated with melatonin (1, 10, and 100 nM) for 2 h, and then treated in the presence or absence of METH (100 µM) for 24 h. After the cells were exposed to the condition, the incubation solutions of the 20 µM 2’,7’-dichlorofluorescein diacetate (DCFH-DA) and the 10

19

µM 4,5-diaminofluorescein diacetate (DAF-2DA) in PBS for 2 h at 37°C in the dark were used to determine the ROS and the RNS levels, respectively. The fluorescence values were then measured at excitation/emission wavelengths of 485/535 nm by using a Synergy H4 microplate reader (Biotek, VT, USA).

4.9. TEER measurement and paracellular permeability assay

The cultured BMVECs were maintained in collagen/fibronectin coated-inserted trans-wells of 24-well plates at a density of 1×105cells/ml at 37°C until TEER >200 Ω∙cm2. The cells were prior treated with JSH23 (10 µM) for 1 h. Then, the cells were pretreated with melatonin (1, 10, and 100 nM) for 2 h and then treated in the presence or absence of METH (100 µM) for 24 h. The TEER values were determined by the EVOM2 voltometer with STX-2 electrodes for triplication in each group. The paracellular permeability was determined as the fluorescence intensity of FITC-dextran. Briefly, 5 mg/ml 4 kDa FITC-dextran in a reaction buffer containing 122 mM NaCl, 25 mM NaHCO3, 3 mM KCl, 1.2 mM MgSO4, 0.4 mM K2HPO4, 1.4 mM CaCl2, 10 mM HEPES, and 10 mM glucose was added to the upper chamber at 37°C in the dark for 2 h, and the lower chamber media was then used to determine the fluorescence intensity at an excitation/emission wavelength of 488/525 nm by using a Synergy H4 microplate reader.

4.10. Apoptosis and caspase-3 detection assay by flow cytometry

The cultured BMVECs were maintained in collagen/fibronectin coated-inserted trans-wells of 24-well plates at a density of 1×105cells/ml at 37°C until TEER >200 Ω∙cm2. The cells were

20

prior treated with JSH23 (10 µM) for 1 h. Then, the cells were pretreated with melatonin (1, 10, and 100 nM) for 2 h, and then treated in the presence or absence of METH (100 µM) for 24 h. Then, the cells were trypsinized and resuspended in culture media at a concentration of 1×104 cells/100 µl. For the determination of the apoptotic cells, the cells were incubated with the reaction assay of MuseTMAnnexin-V and the dead cell assay kit (EMD Millipore Biosciences) for 20 min in the dark at room temperature. For the determination of the caspase3 levels, the cells were incubated with caspase-3/7 working solutions (EMD Millipore Biosciences) for 10 min in the dark at room temperature. Finally, the percentage of the apoptotic cells and the caspase-3 level were analyzed by using Muse Cell Analyzer (Merck Millipore, MA, USA), according to the manufacturer’s instruction.

4.11. Statistical analysis

The data are expressed as mean ± SEM of three independent experiments. The statistical difference was analyzed using one-way analysis of variance (ANOVA) followed by Post Hoc Dunnett’s test for comparing the significance between the individual groups (p<0.05).

Disclosure of Interest The authors declare no conflict of interest.

Acknowledgments

21

This work was supported by the CMU Mid-Career Research Fellowship Program, Chiang Mai University, Thailand, as well as by a research grant from TRF (DPG 5780001) and Mahidol University to PG.

References Acuña-Castroviejo, D., et al., 2014. Extrapineal melatonin: sources, regulation, and potential functions. Cell Mol. Life Sci. 71, 2997-3025. Abdul-Muneer, P.M., et al., 2011. Impairment of brain endothelial glucose transporter by methamphetamine causes blood-brain barrier dysfunction. Molec. Neurodegen. 6, 17501756. Banks, W.A., Ericson, M.A., 2010. The blood-brain barrier and immune function and dysfunction. Neurobiol. Dis. 37, 26-32. Cahill-Smith, S., Li, J.M., 2014. Oxidative stress, redox signalling and endothelial dysfunction in ageing-related neurodegenerative diseases: a role of NADPH oxidase 2. J. Clin. Pharmacol. 78, 441-453. Chen, H.Y., et al., 2006. Melatonin decreases neurovascular oxidative/nitrosative damage and protects against early increases in the blood–brain barrier permeability after transient focal cerebral ischemia in mice. J. Pineal Res. 41, 175-182. Chen, B., et al., 2015. The role of Nrf2 in oxidative stress-induced endothelial injuries. J. Endocrin. 225, 83-99. Cheng, M.C., Hsu, S.H., Chen, C.H., 2015. Chronic methamphetamine treatment reduces synaptic plasticity genes expression and changes in their DNA methylation status in the mice brain. Brain Res.15, 765-769. Chern, C.M., et al., 2012. Melatonin ameliorates neural function by promoting endogenous neurogenesis through the MT2 melatonin receptor in ischemic-stroke mice. Free Radic Biol Med. 52, 1634-1647.

22

Coelho-Santos, V., et al., 2015. The TNF-alpha/NF-kappa B signaling pathway has a key role in methamphetamine-induced blood-brain barrier dysfunction. J. Cereb. Blood Flow. Metab. 35,1260-1271. Csiszar, A., et al., 2009. Inflammation and endothelial dysfunction during aging: role of NFkappaB. J. App. Physio. 105, 1333-1341. ElAli, A., et al., 2012. Apolipoprotein-E controls adenosine triphosphate-binding cassette transporters ABCB1 and ABCC1 on cerebral microvessels after methamphetamine intoxication. Stroke. 43,1647-1653. Fernandes, S., et al., 2014. Acetyl-L-Carnitine Prevents Methamphetamine-Induced Structural Damage on Endothelial Cells via ILK-Related MMP-9 Activity. Mol. Neurobiol. 25, 465-477 Fernandes, S., Salta, S., Summavielle, T., 2015. Methamphetamine promotes alpha-tubulin deacetylation in endothelial cells: the protective role of acetyl-l-carnitine. Toxicol. Lett. 234, 131-138. Fisehr, D., Gamieldien, K., Mafunda, P. S., 2015. Methamphetamine is not Toxic but Disrupts the Cell Cycle of Blood-Brain Barrier Endothelial Cells. Neurotox. Res. 28, 8-17. Flora, G., et al., 2003. Methamphetamine potentiates HIV-1 Tat protein-mediated activation of redox-sensitive pathways in discrete regions of the brain. Exp. Neurol. 1, 60-70. García, J.J., et al., 2014. Protective effects of melatonin in reducing oxidative stress and in preserving the fluidity of biological membranes: a review. J. Pineal Res. 56, 225-237. Garrido-Urbani, S., Jaquet, V., Imhof, B.A., 2014. [ROS and NADPH oxidase: key regulators of tumor vascularisation]. Med. Sci. (Paris). 30, 415-421. Gloire, G., Legrand-Poels, S., Piette, J., 2006. NF-kappaB activation by reactive oxygen species: fifteen years later. Biochem. Pharmacol. 72,1493-1505. Granado, N., et al., 2011. Nrf2 deficiency potentiates methamphetamine-induced dopaminergic axonal damage and gliosis in the striatum. Glia. 59, 1850-1863. Gray, S.P., Jandeliet-Dahm, K.A., 2015. The role of NADPH Oxidase in Vascular Disease Hypertension, Atherosclerosis & Stroke. Curr. Pharm. Design. 26, 510-521.

23

Huang, Y., et al., 2015. The complexity of the Nrf2 pathway: beyond the antioxidant response. J. Nutr. Biochem. 15, 179-185. Husain, K., et al., 2015.Inflammation, oxidative stress and renin angiotensin system in atherosclerosis. World J. Biol. Chem. 6, 209-217. Jumnongprakhon, P., et al., 2015. Melatonin Protects Methamphetamine-Induced Neuroinflammation Through NF-kappa B and Nrf2 Pathways in Glioma Cell Line. Neurochem. Res. 40, 1448-1456. Jumnongprakhon, P., et al., 2014. Protective effect of melatonin on methamphetamine-induced apoptosis in glioma cell line. Neurotox. Res. 25, 286-294. Kensler, T.W., Wakabayashi, N., Biswal, S., 2007. Cell survival responses to environmental stresses via the Keap1-Nrf2-ARE pathway. Annu. Rev. Pharmacol. Toxicol. 47, 89-116. Kawai, T., Akira, S., 2007. Signaling to NF-kappa B by Toll-like receptors. Trend. Mol. Med.13, 460-469. Kothur, K., et al., 2015.CSF cytokines/chemokines as biomarkers in neuroinflammatory CNS disorders: A systematic review. Cytokine. 15, 72-77. Lacoste, B., et al., 2015. Anatomical and cellular localization of melatonin MT1 and MT2 receptors in the adult rat brain. J. Pineal Res. 58, 397-417. Li, W., et al., 2008. Activation of Nrf2-antioxidant signaling attenuates NF kappa Binflammatory response and elicits apoptosis. Biochem. Pharmacol. 76,1485-1489. Liu, Y., et al., 2013. A simple method for isolating and culturing the rat brain microvascular endothelial cells. Microvasc. Res. 90, 199-205. Loftis, J.M., Janowsky, A., 2014. Neuroimmune basis of methamphetamine toxicity. Int. Rev. Neurobiol. 118, 165-197. Ma, J., et al., 2014. Methamphetamine induces autophagy as a pro-survival response against apoptotic endothelial cell death through the Kappa opioid receptor. Cell Death Dis. 5, e1099.

24

Mahajan, S.D., et al., 2008. Methamphetamine alters blood brain barrier permeability via the modulation of tight junction expression: Implication for HIV-1 neuropathogenesis in the context of drug abuse. Brain Res. 1203, 133-148. Maloney, E., et al., 2009. Activation of NF-kappaB by palmitate in endothelial cells: a key role for NADPH oxidase-derived superoxide in response to TLR4 activation. Arterioscler. Thromb. Vasc. Biol. 29, 1370-1375. Manchester, L.C., et al., 2015. Melatonin: an ancient molecule that makes oxygen metabolically tolerable. J. Pineal Res. 59, 403-419. Mccarthy, K.D., De Vellis, J., 1980. Preparation of separate astroglial and oligodendroglial cell cultures from rat cerebral tissue. J. Cell Biol. 85, 890-902. Miyazaki, I., et al., 2011. Astrocyte-derived metallothionein protects dopaminergic neurons from dopamine quinone toxicity. Glia. 59, 435-451. Nguyen, X.K., et al., 2015. Liposomal melatonin rescues methamphetamineelicited mitochondrial burdens, pro-apoptosis, and dopaminergic degeneration through the inhibition PKCδ gene. J. Pineal Res. 58, 86-106. Northrop, N.A.,Yamamoto, B.K., 2015. Methamphetamine effects on blood-brain barrier structure and function. Front Neurosci. 9, 69-72. Osburn,W.O., Kensler, T.W., 2008. Nrf2 signaling: an adaptive response pathway for protection against environmental toxic insults. Mutat. Res. 659, 31-39. O'Shea, E., et al., 2014. Current preclinical studies on neuroinflammation and changes in blood–brain barrier integrity by MDMA and methamphetamine. Neuropharmacol. 14, 79-83. Pacchioni, A.M., et al., 2007. Nrf2 gene deletion fails to alter psychostimulant-induced behavior or neurotoxicity. Brain Res. 1, 26-35. Panenka, W.J., et al., 2013. Methamphetamine uses: a comprehensive review of molecular, preclinical and clinical finding. Drug Alcohol Depend. 129, 167-179.

25

Parameyong, A., Govitrapong, P., Chetsawang, B., 2015. Melatonin attenuates the mitochondrial translocation of mitochondrial fission proteins and Bax, cytosolic calcium overload and cell death in methamphetamine-induced toxicity in neuroblastoma SH-SY5Y cells. Mitochondrial 24, 1-8. Park, M., Hennig, B., Toborek, M., 2012. Methamphetamine alters occludin expression via NADPH oxidase-induced oxidative insult and intact caveolae. J. Cell Mol. Med. 16, 362-375. Park, M., et al., 2013. Methamphetamine-induced occludin endocytosis is mediated by the Arp2/3 complex-regulated actin rearrangement. J. Biol. Chem. 288, 33324-33334. Parikh, N.U., et al., 2015. Galectin-1 suppresses methamphetamine induced neuroinflammation in Human brain microvascular endothelial cells: Neuroprotective role in maintaining Blood brain barrier integrity. Brain Res. 1624, 175-187. Perpoonpattana, K., Govitrapong, P., 2013. The anti-inflammatory effect of melatonin on methamphetamine-induced proinflammatory mediators in human neuroblastoma dopamine SH-SY5Y cell lines. Neurotox. Res. 23,189-199. Pinkaew, D., et al., 2015.Association of neuroprotective effect of di-O-demethylcurcumin on Abeta-Induced neurotoxicity with suppression of NF-kappaB and activation of Nrf2. Neurotox. Res. 26, 358-365. Radfar, S.R., Rawson, R.A., 2014. Current research on methamphetamine: epidemiology, medical and psychiatric effects, treatment, and harm reduction efforts. Addict. Health. 6,146-154. Ramkissoon, A., Wells, P.G., 2013. Developmental role of nuclear factor E2-related factor 2 in mitigating methamphetamine fetal toxicity and postnatal neurodevelopmental deficits. Free Radic. Biol. Med. 65, 620-631. Ramkissoon, A., Wells, P.G., 2015. Methamphetamine oxidative stress, neurotoxicity, and functional deficits are modulated by nuclear factor-E2-related factor 2. Free Radic. Biol. Med. 89, 358-368.

26

Ramirez, S.H., et al., 2009.Methamphetamine disrupts blood-brain barrier function by induction of oxidative stress in brain endothelial cells. J. Cereb. Blood Flow. Metab. 29,1933-1945. Reiter, R.J., Manchester, L.C., Tan, D.X., 2010. Neurotoxins: free radical mechanisms and melatonin protection. Curr. Neuropharmacol. 8, 194-210. Rosas-Hernandez, H., et al., 2013. Prolactin protects against the methamphetamine-induced cerebral vascular toxicity. Curr. Neurovasc. Res. 10, 346-355. Saito, T., et al., 2013. Importance of endothelial NF-kappaB signalling in vascular remodelling and aortic aneurysm formation. Cardiovasc. Res. 97,106-114. Shah, A., et al., 2012. Involvement of metabotropic glutamate receptor 5, AKT/PI3K signaling and NF-κB pathway in methamphetamine-mediated increase in IL-6 and IL-8 expression in astrocytes. J. Neuroinflammation. 9, 52-60. Skaper, S.D., Facci, L., Giusti, P., 2014. Neuroinflammation, microglia and mast cells in the pathophysiology of neurocognitive disorders: a review. Neuro. Disord. Drug Targets. 13, 1654-1666. Sun, D., et al., 2015. Neuroprotection of resveratrol against neurotoxicity induced by methamphetamine in mouse mesencephalic dopaminergic neurons. Biofactors. 4, 252-260. Sykiotis, G.P., et al.,2011. The role of the antioxidant and longevity-promoting Nrf2 pathway in metabolic regulation. Curr. Opin. Clin. Nutr. Metab. Care. 14, 41-48. Taylor, S.B., Levis, C.R.,Olive, M.F., 2013.The neurocircuitry of illicit psychostimulant addiction: acute and chronic effects in humans. Subst. Abuse Rehabil. 4, 29-43. Tocharus, J., et al., 2010. Melatonin attenuates methamphetamine-induced overexpression of pro-inflammatory cytokines in microglial cell lines. J. Pineal Res. 48, 347-352. Urrutia, A., et al., 2013. A study on the effect of JNK inhibitor, SP600125, on the disruption of blood-brain barrier induced by methamphetamine. Neurobiol. Dis. 50, 49-58.

27

Wang, Q., et al., 2014. Molecular pathology of brain matrix metalloproteases, claudin5, and aquaporins in forensic autopsy cases with special regard to methamphetamine intoxication. Int. J. legal. Med. 128, 469-474. Wang, Z., et al., 2014. Melatonin alleviates secondary brain damage and neurobehavioral.dysfunction after experime ntal subarachnoid hemorrhage: possible involvement of TLR4-mediated inflammatory pathway. J. Pineal. Res. 55, 399-408. Wijayanti, N., et al., 2004. Role of NF-kappaB and p38 MAP kinase signaling pathways in the lipopolysaccharide-dependent activation of heme oxygenase-1 gene expression. Antioxid. Redox. Signal. 6, 802-810. Wires, E.S., et al., 2012. Methamphetamine activates nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and induces human immunodeficiency virus (HIV) transcription in human microglial cells. J. Neuroviral. 5, 400-410. Xiao, X., et al., 2015. Angiotensin-(1-7) counteracts angiotensin II-induced dysfunction in cerebral endothelial cells via modulating Nox2/ROS and PI3K/NO pathways. Exp. Cell Res. 336, 58-65. Yu, S., et al., 2015. Recent advances in methamphetamine neurotoxicity mechanisms and its molecular pathophysiology. Behav. Neuro.15,103-109. Yuan, F., et al., 2015. SIRT2 inhibition exacerbates neuroinflammation and blood-brain barrier disruption in experimental traumatic brain injury by enhancing NF-κB p65 acetylation and activation. J. Neurochem.136, 581-593. Zhao, L., et al., 2015. Melatonin alleviates brain injury in mice subjected to cecal ligation and puncture via attenuating inflammation, apoptosis, and oxidative stress: the role of SIRT1 signaling. J. Pineal Res. 59, 230-239. Zhang, B., et al., 2015. Alpha 7 nicotinic acetylcholine receptor is required for blood-brain barrier injury-related CNS disorders caused by Cryptococcus neoformans and HIV-1 associated comorbidity factors. BMC Infect. Dis. 15,35-42.

28

Zhang, H., et al., 2009. Role of TNF-alpha in vascular dysfunction. Clinic. Sci. 116, 219-230.

Figure Legends Fig. 1 - Melatonin protects against METH-induced inflammation via NF-κB signaling in BMVECs. The cells were prior treated with (10 µM) JSH23 or (100 µM) apocynin for 1 h. Then, the cells were pretreated with melatonin (1, 10, and 100 nM) for 2 h prior to treatment in the presence or absence of METH (100 µM) for 24 h. Equal amounts of 50 µg protein were separated using electrophoresis and analyzed by western blotting for the expression of ICAM1, VCAM-1, MMP-9 (A), iNOS (B), nuclear p65 subunit (D), cytosolic p65 (E), and total p65 subunit (F) and actin to normalize for the cytosolic fraction and lamin B1 to normalize for the nuclear fraction. The level of nitrite production was determined by Griess reaction (C). The values present the mean ± SEM from three independent experiments. ***P˂ 0.001, in comparison with the control group;

###

P ˂ 0.001, in comparison with the METH group. $$$P

˂ 0.001, in comparison between the melatonin with METH treatment group and JSH-23 with METH treatment group.

29

Fig. 2 - Melatonin attenuates METH- suppressed anti-oxidant defense mechanism of Nrf2 signaling in BMVECs. The cells were prior treated with (10 µM) JSH23 or (100 µM) apocynin for 1 h. Then, the cells were pretreated with melatonin (1, 10, and 100 nM) for 2 h prior to treatment in the presence or absence of METH (100 µM) for 24 h. Equal amounts of 50 µg protein were separated using electrophoresis and analyzed by western blotting for the expression of HO-1 (A), NQO-1 (B), GCLC (C), nuclear Nrf2 (E), cytosolic Nrf2 (F), and total Nrf2 (G) and actin to normalize for the cytosolic fraction and lamin B1 to normalize for the nuclear fraction. The SOD activity was determined using the SOD assay kit (C). The values present the mean ± SEM from three independent experiments. ***P ˂ 0.001, in comparison with the control group; ###P ˂ 0.001, in comparison with the METH group. Fig. 3 - Melatonin protects against METH-induced BBB impairment via NF-κB signaling in BMVECs. The cells were prior treated with (10 µM) JSH23 for 1 h. Then, the cells were pretreated with melatonin (1, 10, and 100 nM) for 2 h prior to treatment in the presence or absence of METH (100 µM) for 24 h. The ROS level was investigated by the CM-H2DCDA (A). The RNS level was investigated by the DAF-2DA (B). The formation of the tight junction was investigated using the TEER value by EVOM2 (C). The paracellular permeability was investigated using the FITC-Dextran intensity by permeability assay (D). The apoptosis determined the activity of the caspase-3 level (E) and the apoptotic cells (F) by flow cytometry. The values present the mean ± SEM from three independent experiments. ***P ˂ 0.001, in comparison with the control group; ###P ˂ 0.001, in comparison with the METH group.

30

Fig. 4 - Melatonin interacts with MT1/2 receptors against METH-induced NF-κB signaling activation. The cells were prior treated with luzindole (1 µM) for 1 h. Then, the cells were pretreated with melatonin (1, 10, and 100 nM) for 2 h prior to treatment in the presence or absence of METH (100 µM) for 24 h. The western blotting analysis was used for determining the expression of ICAM-1, VCAM-1, MMP-9 (A), iNOS (B), nuclear p65 subunit (D), cytosolic p65 (E), and total p65 subunit (F) and actin to normalize for the cytosolic fraction and lamin B1 to normalize for the nuclear fraction. The level of nitrite production was determined by Griess reaction (C). The values present the mean ± SEM from three independent experiments. ***P ˂ 0.001, in comparison with the control group;

###

P ˂ 0.001,

in comparison with the METH group. $$$P ˂ 0.001, in comparison with the melatonin and METH treatment group. Fig. 5 - Melatonin interacts using MT1/2 receptor-mediated Nrf2 signaling in METH treatment. The cells were prior treated with (1 µM) luzindole for 1 h. Then, the cells were pretreated with melatonin (1, 10, and 100 nM) for 2 h prior to treatment in the presence or absence of METH (100 µM) for 24 h. The western blotting analysis was used for determining the expression of HO-1 (A), NQO-1 (B), GCLC (C), nuclear Nrf2 (E), cytosolic Nrf2 (F), and total Nrf2 (G) and actin to normalize for the cytosolic fraction and lamin B1 to normalize for the nuclear fraction. The SOD assay kit was utilized to determine the SOD activity level (C). The values present the mean ± SEM from three independent experiments. ***P ˂ 0.001, in comparison with the control group;

###

P ˂ 0.001, in comparison with the METH group. $$$P

˂ 0.001, in comparison with the melatonin and METH treatment group.

31

Highlights   

METH induces blood brain barrier impairment mediated by NOX-2. Melatonin improves blood brain barrier impairment mediated by NOX-2 inhibition Melatonin acts as blood brain barrier protector through melatonin receptor

32

33

34