Next Article in Journal
Therapeutic Potential of Gynostemma pentaphyllum Extract for Hair Health Enhancement: A Randomized, Double-Blind, Placebo-Controlled Clinical Trial
Previous Article in Journal
The Cholesterol Paradox in Long-Livers from a Sardinia Longevity Hot Spot (Blue Zone)
Previous Article in Special Issue
A Combined Extract Derived from Black Sticky Rice and Dill Improves Clinical Symptoms and Ischemic Stroke Biomarkers in Transient Ischemic Attack and Ischemic Stroke Patients
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Much More than Nutrients: The Protective Effects of Nutraceuticals on the Blood–Brain Barrier in Diseases

by
Anna E. Kocsis
1,
Nóra Kucsápszky
1,
Ana Raquel Santa-Maria
2,
Attila Hunyadi
3,4,5,6,
Mária A. Deli
1,*,† and
Fruzsina R. Walter
1,*,†
1
Biological Barriers Research Group, Institute of Biophysics, HUN-REN Biological Research Centre, H-6726 Szeged, Hungary
2
Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
3
Institute of Pharmacognosy, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary
4
Interdisciplinary Centre of Natural Products, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary
5
HUN-REN-SZTE Biologically Active Natural Products Research Group, Eötvös u. 6, H-6720 Szeged, Hungary
6
Graduate Institute of Natural Products, Kaohsiung Medical University, Shih-Chuan 1st Rd. 100, Kaohsiung 807, Taiwan
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Nutrients 2025, 17(5), 766; https://doi.org/10.3390/nu17050766
Submission received: 22 January 2025 / Revised: 14 February 2025 / Accepted: 19 February 2025 / Published: 21 February 2025

Abstract

:
The dysfunction of the blood–brain barrier (BBB) is well described in several diseases, and is considered a pathological factor in many neurological disorders. This review summarizes the most important groups of natural compounds, including alkaloids, flavonoids, anthocyanidines, carotenoids, lipids, and vitamins that were investigated for their potential protective effects on brain endothelium. The brain penetration of these compounds and their interaction with BBB efflux transporters and solute carriers are discussed. The cerebrovascular endothelium is considered a therapeutic target for natural compounds in diseases. In preclinical studies modeling systemic and central nervous system diseases, nutraceuticals exerted beneficial effects on the BBB. In vivo, they decreased BBB permeability, brain edema, astrocyte swelling, and morphological changes in the vessel structure and basal lamina. At the level of brain endothelial cells, nutraceuticals increased cell survival and decreased apoptosis. From the general endothelial functions, decreased angiogenesis and increased levels of vasodilating agents were demonstrated. From the BBB functions, elevated barrier integrity by tightened intercellular junctions, and increased expression and activity of BBB transporters, such as efflux pumps, solute carriers, and metabolic enzymes, were shown. Nutraceuticals enhanced the antioxidative defense and exerted anti-inflammatory effects at the BBB. The most important signaling changes mediating the increased cell survival and BBB stability were the activation of the WNT, PI3K-AKT, and NRF2 pathways, and inhibition of the MAPK, JNK, ERK, and NF-κB pathways. Nutraceuticals represent a valuable source of new potentially therapeutic molecules to treat brain diseases by protecting the BBB.

1. Importance of Nutraceuticals in Health and Disease

The study of nutraceuticals has gained significant attention in recent years, as they offer a promising approach to maintaining and promoting human health. The term nutraceutical is a hybrid expression between nutrition and pharmaceutical, referring to any bioactive compound that can provide both nutritional and medicinal value. The vast majority of such compounds in the human diet are of herbal origin, i.e., they are bioactive constituents of edible plants. These herbal compounds have benefits in a wide range of therapeutic areas in many formulations [1,2]. Numerous studies have highlighted the potential of nutraceuticals in the prevention and management of chronic diseases, such as cardiovascular diseases, cancer, osteoarthritis, diabetes, and neurological disorders [3,4,5,6,7,8,9,10].
The blood–brain barrier (BBB) is one of the major interfaces between the systemic circulation and the brain sustaining fundamental homeostasis for brain functions. For detailed reviews about the main cell types, transporters, basal membranes, glycocalyx, and signal transduction, see [11,12,13,14,15]. Brain endothelial cells comprising the functional basis for the BBB are one of the main cellular targets for dietary natural products [16]. Here, we focus on factors of the special relationship between nutraceuticals and the BBB. BBB penetration of nutraceuticals, and their interaction with influx and efflux transporters and intracellular signaling pathways, will be discussed. The effect of nutraceuticals on BBB protection based on in vitro and in vivo studies draws attention to the versatile use of these mostly plant-derived natural products.

2. Major Properties of Nutraceuticals with Protective Effects on the Blood–Brain Barrier

A wide range of natural products have been reported for their BBB protective activity, and these cover all major classes of secondary plant metabolites, including terpenoids, phenolic compounds, alkaloids, and organosulfur compounds, as well as some primary metabolites such as ⍵-3 fatty acids and several vitamins (Table 1). In line with this chemical diversity, it is not surprising that key BBB-related physicochemical characteristics of these compounds represent a very large variation. Molecular masses vary between 148 (cinnamic acid) and 659 (fucoxanthin), logarithm of the octanol-water partition coefficient (logP) values vary between −1.6 (ascorbic acid) and 15.6 (lycopene), and topological polar surface area (TPSA) values vary between 0 for lycopene and β-carotene, and 266 for rutin (see Supplementary Materials, Table S1). These ranges are far wider than what would generally be considered ideal for a central nervous system (CNS) active drug candidate [17]. While there are some highly polar compounds that reportedly protect the BBB, like the cationic anthocyanidines and their glucosides, most of the compounds are rather lipophilic up to the extreme, e.g., the tetraterpene carotenoids. While both extremes in terms of hydrophilic–lipophilic properties are well known for manifesting in problems with biodistribution and making it difficult for a compound to pass through the BBB, it is worth stressing that BBB protecting agents do not necessarily need to penetrate through the barrier to exert their effect [14]. Nonetheless, carotenoids provide an interesting example for a highly lipophilic group of nutraceuticals whose members do not become stuck in extracellular membranes, even without an influx mechanism assisting their uptake. Due to their length matching the thickness of phospholipid bilayers, they are perfectly capable of passing through the BBB by passive diffusion [18].
As seen from Table 1, all nutraceuticals identified as BBB-protective hits had been linked to antioxidant activities. The vast majority of them are well-known ‘classical’ antioxidants, such as phenolic compounds able to scavenge toxic free radicals and mitigate oxidative stress-related cellular damage. This is not surprising, considering the key role of oxidative stress in the onset and progression of BBB damage in diseases [14].
Table 1. Blood–brain barrier protective nutraceuticals and natural compounds.
Table 1. Blood–brain barrier protective nutraceuticals and natural compounds.
NutraceuticalPropertiesSourceAntioxidantEfflux Pump Interaction Y/NInflux
Transport Interaction
BBB/Brain Penetration
Alkaloids
Caffeine194 Da
A
Coffee, green and black tea, guarana berriesYes
[19]
No
[20]
CNT2/SLC28A2
[21]
OAT1/SLC22A6
[22]
GLUT1/SLC2A1
[23]
Yes, high
[24,25]
Capsaicin305 Da
A
Chili pepperYes
[26]
Yes
[27,28]
NDYes, high
[29]
Theophylline180 Da
A
Cocoa beans, brewed teaYes
[30]
No
[31]
OAT1
[22]
Yes
[32]
Anthocyanidines
Cyanidin/Cyanidin-3-O-beta-glucoside/Procyanidine287 Da
H
Red wine, elderflower, berries, tea, apple, cinnamonYes *
[33]
Yes
[34,35]
GLUT1/SLC2A1
[36]
Yes
[36,37]
Malvidin/Malvidin-3-O-glucoside331 Da
H
Red wine, berriesYes
[33,38]
Yes
[34]
GLUT1/SLC2A1, GLUT3/SLC2A3 [39]Yes
[40]
Carotenoids
Astaxanthin597 Da
L
Seafood, salmon, trout, algaeYes *Yes
[41]
NDYes *
β-Carotene537 Da
L
Carrots, sweet potato, pumpkinYes *Yes
[31,42]
GLUT4/SLC2A4
[43]
No *
Fucoxanthin659 Da
L
Brown algaeYes *
[44]
Yes
[45]
SLC7A11
[44]
ND *
Lutein569 Da
L
Kale, spinach, orange, egg yolk, avocadoYes *Yes
[46]
SR-B1
[47]
Yes *
Lycopene537 Da
L
Tomato, watermelon grapefruit, pomegranateYes *Yes
[48]
NDYes *
Diarylheptanoids
Curcumin 368 Da
L
Turmeric Yes *
[49]
Yes
[50,51]
GLUT1/SLC2A1
[52]
No
[53]
Flavonoids
Apigenin270 Da
L
Parsley, celery, chamomile teaYes
[54,55]
Yes
[56,57]
GLUT1/SLC2A1
[58]
Yes, low
[56,59]
Catechin/
Epicatechin
290 Da
L
Tea, red wine, cocoaYes
[60,61]
Yes
[62,63]
ASBT/SLC10A2 [64]Yes
[36]
Chrysin254 Da
L
Chamomile, honey, propolis, passion fruitYes
[65,66]
Yes
[67,68]
OATP
[69]
ND
Daidzein254 Da
L
Soy, soy productsYes
[70]
Yes
[71,72]
GLUT4/SLC2A4
[73]
Yes, high
[59]
Fisetin286 Da
L
Onions, leeks, broccoliYes
[74]
Yes
[75]
GLUT4/SLC2A4
[76]
Yes, low
[59]
Genistein270 Da
A
Soy, soy productsYes *
[77]
Yes
[72,78]
Noradrenalin, serotonin transporter [79] GLUT1/SLC2A1 [80]Yes, high
[56,59]
Hesperetin302 Da
L
Citrus fruit, herbs, wineYes *
[81,82]
Yes
[67,83]
NDYes
[37]
Hesperidin611 Da
H
Citrus fruit, herbs, wineYes
[84,85]
Yes
[68,86]
NDYes, low
[56]
Kaempferol286 Da
L
Onion, leeks, broccoli, ginkgo bilobaYes
[87,88]
Yes
[67,71]
GLUT4/SLC2A4
[89]
Yes, low [56,90]
Luteolin286 Da
L
Pepper, leafy greens, celery, broccoliYes
[91]
Yes
[92]
SLC7A11
[93]
Yes, low
[59]
Myricetin318 Da
L
Onions, leeks, broccoliYes
[87]
Yes
[94]
PCFT/SLC46A1
[95]
Yes
[90]
Naringenin272 Da
L
Citrus fruits, herbs, wineYes
[96]
Yes
[65]
NDYes
[35]
Naringin581 Da
H
Citrus fruits, herbs, wineYes
[84]
Yes
[68,97]
NDND
Quercetin302 Da
L
Onion, broccoli, ginkgo biloba,
apple
Yes *Yes
[67,98]
SLC7A11
[99]
Yes, low
[40,56,90]
Rutin611 Da
H
Citrus fruits, herbs, wineYes *Yes
[56,100]
OATP2B1/SLCO2B1
[101]
GLUT4/SLC2A4 [102]
Yes, low [56,59]
Silybin/
Silymarin
482 Da
L
Milk thistleYes *
[103]
Yes
[68]
NDND
Tangeretin372 Da
L
Tangerine, citrus peelYes
[104,105]
Yes
[106]
SGLT1/SLC5A1
[107]
Yes
[108]
Monoterpenes
Borneol154 Da
L
Coriander, ginger oil, rosemary, thymeYes
[109]
Yes
[110,111]
NDYes
[112]
Carvacrol 150 Da
L
Oregano, thymeYes
[113]
Yes
[114]
NDND
Omega-3 fatty acids
Docosahexaenoic acid328 Da
L
Oceanic fish oil, seaweedYes
[115]
Yes
[116]
MFSD2A
[117]
Yes
[117]
Eicosapentaenoic acid302 Da
L
Fish oil, seaweedYes
[118,119]
Yes
[116]
VNUT/LC17A9
[120]
Yes
[121]
Organosulfur compounds
α-Lipoic acid206 Da
L
Broccoli, yeast, meat, kidney, heart, liverYes
[122,123]
Yes
[124]
SMVT/SLC5A6 [125]Yes
[126]
Sulforaphane177 Da
L
Broccoli, kale, cauliflowerYes *Yes
[127]
NDYes
[128]
Phenolic acids
Caffeic acid180 Da
L
Berries, kiwi, plum, appleYes
[129]
Yes
[130]
MCT1/SLC16A1, MCT4/SLC16A3 [131]Yes
[132]
Cinnamic acid148 Da
L
Cinnamon, grape, cocoaYes
[133]
Yes
[134]
MCT1/SLC16A1, MCT4/SLC16A3
[131]
ND
p-Coumaric acid164 Da
L
Berries, kiwi, plum, appleYes
[135]
NDMCT1/SLC16A1, MCT4/SLC16A3 [131]
OAT3/SLC22A8 [136]
ND
Ferulic acid194 Da
L
Grains, nuts, fruits, vegetablesYes
[137,138]
Yes
[139]
MCT1/SLC16A1, MCT4/SLC16A3 [130,140]
OAT3/SLC22A8
[136]
Yes, low
[25]
Gallic acid170 Da
L
Berries, kiwi, plum, appleYes
[141]
Yes
[134]
OAT3/SLC22A8
[136]
Yes
[142]
Rosmarinic acid360 Da
L
Berries, kiwi, plum, appleYes
[143,144,145]
Yes
[146]
OAT1/SLC22A6, OAT3/SLC22A8
[147]
Yes, low
[132]
Stilbenes
Piceatannol244 Da
L
Grape, white tea, passion fruitYes
[148]
No
[149]
MCT1/SLC16A1, MCT4/SLC16A3
[150]
ND
Polydatin390 Da
L
Grapes, cocoa, peanutsYes
[151]
Yes
[152]
NDYes, low
[153]
Pterostilbene256 Da
L
Blueberries, grapesYes
[154]
NDMCT1/SLC16A1, MCT4/SLC16A3
[150]
Yes
[155]
Resveratrol228 Da
L
Grapes, wine, peanutsYes *
[33]
Yes
[156]
NDYes, low
[59,157]
Vitamins
Vitamin C/
Ascorbic acid
176 Da
H
Fruits, vegetablesYes *
[158]
No
[159]
SLC19A1, SLC23A2
[11]
Yes
[160]
Vitamin B9/
Folic acid
441 Da
H
Fruits, vegetables, nutsYes
[161]
NDSLC19A1, SLC46A1
[11]
Yes
[162]
Vitamin D3/
Cholecalciferol
385 Da
L
Fish, milk, meatYes
[163]
NDLRPs
[11]
Yes
[164]
Vitamin E/
α-Tocopherol
431 Da
L
Plant oil, nuts, nut oil, spinach, broccoliYes *Yes
[165]
SR-B1, αTTP, PLTP
[162]
Yes
[165]
Abbreviations: A: amphiphilic; ASBT: Ileal apical sodium-dependent bile acid transporter; GLUT: glucose transporter; H: hydrophilic; OAT: organic anion transporter; SMVT: sodium-dependent multivitamin transporter; L: lipophilic; LRP: LDL receptor-related protein; MCT: monocarboxylic acid transporter; MFSD2A: sodium-dependent lysophosphatidylcholine symporter 1; ND: no data; OATP: organic anion-transporting polypeptide; PCFT: proton-coupled folate transporter; PLTP: phospholipid transfer protein; SGLT1: sodium/glucose cotransporter 1; SLC: solute carrier; SR-B1: scavenger receptor class B type 1; αTTP: α-tocopherol (binding) transport protein; *: [10].
This notion has likely been driving related studies and the selection of compounds to be tested as potential BBB protective agents. However, it is now well understood that antioxidant action is much more complex than free radical scavenging, and is based on the modulation of complex signaling pathways to activate endogenous, primarily enzymatic antioxidant defense mechanisms [166]. This is also demonstrated by the appearance of some non-phenolic antioxidants among the BBB protective dietary compounds. Based on the above, we may postulate that there is much more to BBB protection than free radical scavenging by dietary antioxidants, and that a broad chemical space is still waiting to be explored in this regard.
It should also be noted that several of the compounds presented in Table 1 are well known for their low chemical stability and/or bioavailability. This is particularly true for some phenolic compounds, e.g., curcumin and resveratrol. In such cases, the experimental model may fundamentally determine the outcome of a study. Because of this, while BBB protection may have rightfully been connected to the consumption of these compounds, their decomposition products or metabolites may have played a significant role in the effect [166].
Studies focusing on nutraceuticals affecting CNS functions often neglect the amount of the compound that enters the brain, or how the molecule can cross the BBB. Passage of natural compounds across the BBB was reviewed by Ureña-Vacas et al. [167]. We listed 48 compounds in Table 1, and most of them were studied for BBB or brain penetration either in animal or culture models. We did not find permeability data for eight compounds, namely carvacrol, chrysin, cinnamic acid, p-coumaric acid fucoxanthin, naringin, piceatannol, and silymarin (Table 1). Two well-known lipophilic compounds, β-carotene [10] and curcumin [53], are considered to not cross the BBB, which means that their permeability is so low that it is below the detection level. Very low barrier permeability was measured for four molecules, namely fisetin, luteolin, resveratrol, and rutin [59], and from these, only rutin is hydrophilic (Table 1). Low but measurable BBB penetration was described for seven compounds, namely apigenin, ferulic acid, hesperidin, kaempferol, polydatin, quercetin, and rosmarinic acid (Table 1). High permeability was detected for caffeine, which is used as a permeability marker in BBB studies [24], capsaicin when given systematically [29], daidzein, and genistein [59]. For the remaining 23 compounds, BBB crossing was described (Table 1). We found two comparative studies to evaluate the barrier crossing of several herbal compounds: Shimazu et al. tested BBB permeation in rats and in a rat BBB co-culture model [59], while Yang et al. used rat brain endothelial- and intestinal epithelial-based culture models [56]. A clear ranking for penetration can be established only in comparative studies [56,59]. The limitation of most studies is that they investigate only one compound; therefore, the levels of permeability and brain penetration cannot be compared to other datasets due to differences in models, methods, and calculations [167]. Lipophilicity and passive transport are not the only determinants of BBB crossing for the compounds listed in Table 1; stability, bioavailability and, most importantly, brain endothelial transporters can also play a role.
Active transport systems at the level of brain endothelial cells play an important role in the entry of nutrients to the brain [11]. Nutraceuticals and their glucoside derivatives interact with hexose transporters, such as GLUT-1, -2, and -4, which help their transport across biological barriers. Members of the solute carrier family including organic cationic and anionic transporters (OCTs and OATs), sodium-coupled nucleoside transporters (CNTs), organic anion transporters (OATPs), and monocarboxylate transporters (MCTs), are highly expressed at the BBB, as demonstrated by transcriptomic studies on mouse [168,169,170] and human [171,172] isolated brain microvessels, and also on cultured brain endothelial cells [173,174,175,176].
The effect of nutraceuticals and different herbal compounds on efflux pumps is a well-studied area, and has been described in detail [11,177,178,179,180]. As a general conclusion, multiple herbal compounds interact as ligands or inhibitors with major efflux pumps, which are also present at the BBB, such as P-glycoprotein (PGP, MDR1/ABCB1) or breast cancer resistant protein (BCRP/ABCG2). These compounds can interact with the efflux pumps, decreasing their bioavailability. Upon inhibition, other nutrients or drugs otherwise subject to efflux pump activity can reach the target tissue in a different concentrations, leading to higher absorption and changing the dose delivered to the tissue [181].
From the compounds listed in Table 1, those that were tested for protective effects on both in vivo and culture models of the BBB (Table 2 and Table 3) by more than two research groups are shown in Figure 1. Among the selected 11 compounds, 6 are flavonoids, namely apigenin, catechins, genistein, luteolin, kaempferol and quercetin. There could be several reasons for why flavonoids are the most investigated chemical group for their BBB effects. Firstly, they are present in vegetables such as leafy greens, onions, herbs, and beverages, therefore they are regularly consumed in balanced diets. Secondly, these compounds are also major constituents in many traditional medicines, such as Gingko biloba and chamomile tea. Thirdly, they are widely studied for neuroprotection in preclinical models [182], although much less for their effects on the BBB. The main biomedical research field for curcumin, a diarylheptanoid compound in the spice turmeric, and resveratrol, a stilbene found in grapes, wine, and nuts, is cancer [183], although their role is also investigated in cardiovascular and CNS diseases [184]. It was surprising to find only a handful of studies for these two compounds on BBB models. Regarding the carotenoid astaxanthin and the polyunsaturated fatty acid DHA, their primary food source is fish, seafood, and algae (Figure 1), which are well known for their beneficial effects on general health, brain development, and cognition [185]. The monoterpene borneol that can be found mainly in spices and herbs is mainly investigated for its use to help drug delivery across biological barriers [186].

3. Protective Effects of Nutraceuticals on the BBB in CNS Diseases

While the BBB is essential for preserving neural functions, its disruption or dysfunction is implicated in a wide range of CNS pathologies. Conditions such as neurodegenerative diseases (Alzheimer’s disease, Parkinson’s disease), cerebrovascular disorders (subarachnoid hemorrhage, stroke, ischemia), and traumatic brain injury (TBI), along with different disease states such as hyperlipidemia, oxidative stress, or neurodevelopmental conditions like autism, often involve alterations in BBB integrity and function [187]. These changes can lead to an imbalance in transport mechanisms and intercellular junctional molecule expression, elevation of proinflammatory cytokine levels, and disruption of blood flow, all exacerbating disease progression [12]. Nutraceuticals in preclinical studies could partially or fully restore barrier integrity, decrease brain edema, and block the downregulation of junctional molecule expression, along with reducing oxidative stress and inflammation and restoring healthy BBB phenotypes in CNS pathologies, as summarized in Figure 2.

3.1. In Vivo Investigations

The use of animal models is fundamental in biomedical research to advance our understanding of the pathophysiology, progression, and treatment of diseases [188]. The protective effects of nutraceuticals on BBB were mainly studied in rat or mouse disease models (Table 2).
Table 2. The effects of nutraceuticals on the blood–brain barrier model in animal models of diseases.
Table 2. The effects of nutraceuticals on the blood–brain barrier model in animal models of diseases.
CompoundDisease ModelEffects on BBB ParametersReference
Apigeninsubarachnoid hemorrhage, ratinflammation ↓, BBB disruption ↓,
ZO-1, occludin ↑
[189]
cerebral IR,
MCAO, rat
vascularization/tube formation ↑, cerebral infarction ↓[190]
Astaxanthin and derivativessubarachnoid hemorrhage, mouse BBB disruption ↓[191]
subarachnoid hemorrhage, ratbarrier integrity ↑, brain edema ↓,
IL-1β, TNF-α, MMP-9 expression ↓
[192]
Borneol-R123 permeability in hippocampus ↑, PGP and MRP1 ↓, TJ disruption[193]
cerebral IR
-
blood pressure, cerebrovascular resistance ↓, edema ↓,
BBB integrity, eNOS, CLDN-5, ZO-1 ↑,
ET-1, iNOS, MMP-2/9, ICAM1, LFA-1 ↓
[111,194]
Caffeic acid phenethyl ester TBI, rat and mousevascular integrity ↑, CLDN-5 ↑[195]
β-carotenecerebral IR,
MCAO, mouse
barrier integrity ↑, occludin, ZO-1 ↑
peroxynitrite generation ↓
[196]
CarvacrolTBI, ratbarrier integrity ↑, brain edema ↓, occludin, CLDN-5, ZO-1 ↑, MMP-9 ↓[113]
CatechinTBI, ratbarrier integrity ↑, ZO-1, occludin ↑[197]
ChrysinTBI, ratbarrier integrity ↑, brain EB content ↓[198]
p-Coumaric acidhypoxia, mousebarrier integrity ↑, brain edema ↓, occludin expression ↑[199]
Curcumincerebral IR,
MCAO, rat
barrier integrity ↑, brain EB content ↓ [200]
Hypoxia/hypercap-nia, ratbrain edema ↓, apoptosis ↓, AQP4 levels ↓,[201]
Daidzein cerebral IR,
MCAO, rat
barrier integrity ↑, astrocyte swelling ↓, cytoplasmic vacuolation ↓, edema ↓,
vessel lumen ↑
[202]
Docosahexaenoic acidcerebral IR,
MCAO, rat
barrier integrity ↑, brain edema ↓[203]
cerebral IR,
MCAO, rat
barrier integrity ↑[204]
cerebral IR,
left CCAO, rat
barrier integrity ↑, brain edema ↓, occludin, CLDN-5, ZO-1 ↑, MMP-2/9 ↓[205]
uremia + contrast media, mouseCLDN-5, laminin α-4, -5 ↑[206]
Eicosapentaenoic acidcerebral IR,
left CCAO, rat
barrier integrity ↑, brain edema ↓, occludin, CLDN-5, ZO-1 ↑, MMP-2/9 ↓[205]
uremia + contrast media, mouseCLDN-5, laminin α-4, -5 ↑[206]
Epigallo-catechin gallatecerebral IR,
MCAO, rat
barrier integrity ↑, TJ opening ↓
occludin, CLDN-5, ZO-1 expression ↑
[207]
Ferulic acid + tetramethylpyrazinecerebral IR,
MCAO, rat
barrier integrity ↑, brain edema ↓,
JAM-1, occludin ↑, MMP-9 expression ↓
[208]
Fisetinautism, valproic acid-induced, ratbarrier integrity ↑, CLDN-5 expression ↑[209]
FucoxanthinTBI, mousebarrier integrity ↑, brain edema ↓, occludin, CLDN-5, ZO-1, VE cadherin ↑, MMP-9 ↓, apoptosis and ferroptosis ↓,
BMEC mitophagy ↑
[210]
GenisteinTBI, ratbarrier integrity ↑[211]
HesperetinTBI, mousebarrier integrity ↑, brain edema ↓,
ZO-1, occludin, CLDN-5 ↑,
NLRP3 inflammasome ↓
[212]
Hesperidincerebral IR,
MCAO, mouse
barrier integrity ↑, brain edema ↓,
disruption of CLDN-5 and ZO-1 ↓
[85]
Kaempferolneuroinflammation, LPS-induced, mousebarrier integrity ↑,
occludin, connexin-43 expression ↑
[213]
neuroinflammation and BBB dysfunction, LPS-induced BBB structure restored, brain edema ↓, occludin, connexin-43 expression ↑[214]
Kaempferol-glucoside/Juglanincerebral IR,
MCAO, mouse
BBB permeability ↓, VEGF and VEGFR2 ↓ ZO-1, occludin expression ↑[215]
α-Lipoic acidTBI, ratbarrier integrity ↑, brain EB content ↓[216]
Luteinsubarachnoid hemorrhage, ratvasospasm ↓[217]
TBI, ratIL-1, IL-6, TNF-α, CCL2 ↓, ROS ↓,
SOD, GSH ↑, ICAM-1, ET-1 ↓
[218]
LuteolinAD, Aβ25–35-induced, mouseBBB leakage ↓, astrocyte swelling ↓, CBF ↑ ZO-1, occludin, CLDN-5 expression ↑, [219]
diabetes, high-fat diet and streptozotocin- induced, ratZO-1, occludin and GLUT-1 expression ↑[220]
Lycopenesubarachnoid hemorrhage, ratbarrier integrity ↑, brain edema ↓[221]
hyperlipidemia, high fat diet induced, ratVEGF, VCAM-1 ↓, CLDN-5 ↑,
IL-1, IL-6, and TNF-α ↓
[222]
Malvidincerebral IR,
BCCAO, rat
eNOS ↑, MMP-9 ↓[223]
Naringenincerebral IR,
MCAO, mouse
BBB leakage ↓,
ZO-1, occludin, CLDN-5, β-catenin ↑
[224]
Polydatincerebral IR,
MCAO, rat
barrier integrity ↑, brain edema ↓,
CLDN-5 expression ↑
[225]
cerebral IR,
MCAO, rat
barrier integrity ↑, brain edema ↓,
ZO-1, occludin, CLDN-5 ↑,
TNF-α, IL-1β, IL-6, CCL2 levels ↓,
ICAM-1 and VCAM-1 ↓
[226]
Procyanidin B2cerebral IR,
MCAO, rat
barrier integrity ↑, brain edema ↓, ZO-1 expression ↑[227]
Pterostilbenecerebral IR,
MCAO, rat
barrier integrity ↑, CBF ↑, laminin ↑,
ZO-1, occludin, CLDN-5, VE-cadherin ↑
[228]
cerebral IR,
MCAO, rat
barrier integrity ↑, brain edema ↓,
MMP-2/9 expression ↓
[229]
Quercetin AD, Aβ25–35-induced, mousebarrier integrity ↑, CBF ↑[230]
cerebral ischemia, photothrombosis-induced, ratbarrier integrity ↑, MMP-9 activity ↓[231]
cerebral IR,
BCCAO, rat
barrier integrity ↑, brain endothelial cell swelling ↓, vesicles and vacuoles ↓,
CLDN-5, ZO-1, β-catenin ↑, MMP-9 ↓
[232]
oxidative stress,
PCB-induced, rat
occludin, CLDN-5, JAM-3, ZO-1, AF-6 ↑ [233]
cerebral IR,
MCAO, rat
barrier integrity ↑, occludin, CLDN-5, ZO-1 expression ↑[234]
Quercetin +/− hydroxylsafflor yellow Acerebral IR,
MCAO, mouse
barrier integrity ↑[235]
Resveratrolrecurrent ischemic stroke, ratbarrier integrity ↑, brain edema ↓,
no change in CBF
[236]
Rosmarinic acidMCAO + diabetes, STZ-induced, ratbarrier integrity ↑, brain edema ↓[237]
Tangeretincerebral IR,
MCAO, rat
barrier integrity ↑[238]
Vitamin B9sepsis, cecal ligation and perforation, ratbarrier integrity ↑[239]
Vitamin Ccerebral IR,
MCAO, rat
barrier integrity ↑, MMP-2/9 expression ↓, CLDN-1,CLDN-5, ZO-1 ↑ [240]
Vitamin D3TBI, ratbarrier integrity ↑, brain edema ↓,
ZO-1, occludin expression ↑
[241]
Abbreviations: AD: Alzheimer’s disease; AF-6: ALL-1 fusion partner at chromosome-6; BCCAO: bilateral common carotid artery occlusion; BMEC: brain microvascular endothelial cell; CBF: cerebral blood flow; CCL2: chemokine (C-C motif) ligand 2; CLDN: claudin; EB: Evans blue; eNOS: endothelial nitric oxide synthase; ET-1: endothelin-1; GLUT-1: glucose transporter 1; ICAM1: intercellular adhesion molecule 1; IL: interleukin; iNOS: inducible nitric oxide synthase; IR: ischemia/reperfusion; JAM: junction associated molecule; LFA-1: lymphocyte function-associated antigen 1; MMP: matrix metalloproteinase; NLRP3: microglial nodd-like receptor protein 3; PCB: polychlorinated biphenyl; ROS: reactive oxygen stress; STZ: streptozotocin; TBI: traumatic brain injury; MCAO: transient middle cerebral artery occlusion; TNF-α: tumor necrosis factor-α; VCAM-1: vascular cell adhesion protein 1; ZO-1: zonula occludens-1; ↑: increase; ↓: decrease.

3.1.1. Traumatic Brain Injury

Mechanical traumas affecting the head or spine are the major causes of TBI, in which BBB dysfunction leads to secondary neuronal injuries. BBB breakdown in TBI includes the loss of tight junctions (TJs), increased vesicular transport, extravasation of plasma proteins, and brain edema, causing further neuronal damage [242,243]. Decreased edema and increased BBB integrity after TBI were found after treatment with α-lipoic acid [216], chrysin [198], genistein [211], carvacrol [113], and vitamin D3 [241]. Caffeic acid phenethyl ester also increased BBB integrity by elevating claudin-5 levels [195]. Catechin and hesperetin acted similarly by restoring zonula occludens protein 1 (ZO-1) and occludin expression [197,212]. Hesperetin reduced the NLRP3 inflammasome activation after TBI [212]. Carvacrol protected BBB properties after trauma due to its antioxidant property, restored the expression of junctional proteins, and decreased the level of matrix metalloproteinase-9 (MMP-9; [113]). Lutein in a rat TBI model decreased the level of intercellular adhesion molecule-1 (ICAM-1) and endothelin-1, a potent vasoconstrictor, and elevated nuclear factor erythroid 2-related factor 2 (Nrf-2) expression and antioxidant cell response [218]. Fucoxanthin, similarly to previous molecules, rescued the barrier integrity, increased the expression of junctional proteins, attenuated brain endothelial cell apoptosis and ferroptosis, and increased mitophagy [210].

3.1.2. Cerebrovascular Disorders

In the aging population, the most common CNS diseases are those that affect the cerebral vasculature. Ischemic stroke, hemorrhage, or vascular malformations all pose an immediate life-threatening risk due to impaired blood flow and very sudden functional loss [13]. Ischemic stroke-induced damage is the most frequent cerebrovascular pathology. Ischemic and hemorrhagic types of stroke affect all components of the neurovascular unit [244]. Activation of brain endothelial cells and surrounding cell types, such as astroglia, pericytes, and microglia leads to proinflammatory cytokine and chemokine production initiating neuroinflammation. Downstream steps in stroke pathology are BBB opening and immune cell entry to the brain, causing further BBB dysfunction and worsening inflammation and edema [245]. Nutraceuticals improve stroke outcomes and inflammation in multiple animal models of the diseases (Table 2).
Apigenin induced neovascularization and decreased cerebral infarction in a rat middle cerebral artery occlusion (MCAO) model [190]. Tangeretin, quercetin alone and in combination with hydroxysafflor yellow A, curcumin, hesperidin, polydatin, procyanidin B2, pterostilbene, docosahexaenoic acid (DHA), eicosapentaenoic acid (EPA), and ferulic acid decreased brain edema and increased BBB integrity [83,200,203,204,205,208,225,226,227,228,229,231,235,238]. Curcumin in a hypoxia–hypercapnia model counteracted brain edema formation by decreasing the gene and protein expression of aquaporin 4 (AQP4) water channels [201] found in astrocytic endfeet wrapping brain capillaries (Figure 3). In a model of recurrent ischemic attacks, resveratrol decreased brain water content and improved barrier properties, without a change in cerebral blood flow [236]. Rosmarinic acid protected the BBB against MCAO-induced damage in diabetic rats [237]. DHA and EPA, quercetin, hesperidin, polydatin, procyanidin B2, ferulic acid, and vitamin C also elevated the expression of tight junction (TJ) and junctional associated molecules in animal MCAO models [83,205,208,225,226,227,240,241]. Treatment after reperfusion with DHA and EPA, quercetin, ferulic acid, pterostilbene, and vitamin C increased barrier integrity by decreasing the expression of MMP-2 and/or MMP-9 [205,208,229,231,240]. The kaempferol glucoside juglanin showed similar effects after MCAO by the inhibition of the vascular endothelial growth factor (VEGF)/VEGFR pathway [215]. The same observations were made for naringenin and quercetin, which modulated the GSK-3β/β-catenin pathway [224,232], while green tea polyphenols decreased the level of PKCα [207]. Polydatin downregulated the expression of proinflammatory markers and cell surface molecules in a rat brain ischemia–reperfusion injury [226]. In a mouse MCAO model, β-carotene in emulsified or nanocarrier form increased barrier integrity, ZO-1, and occludin levels, and decreased the production of peroxynitrite that lowers mitochondrial membrane potential and enhances cell apoptosis and necrosis [196]. Daidzein restored the ultrastructural changes at the BBB in a rat model of stroke, decreased astrocyte swelling, cytoplasmic vacuolation, edema, and increased blood vessel lumen [202]. The barrier protective effect of malvidin was also mediated by elevation in eNOS and decrease in MMP-9 [223].
The monoterpene borneol (Table 1, Figure 1) has a dual effect on the BBB. On the one hand, borneol and its derivatives are protective in models of cerebral ischemia–reperfusion injury [111,194]. At a wide range of doses (0.5–600 mg/kg body weight), borneol relaxed blood vessels, and decreased blood pressure and cerebrovascular resistance through lowering endothelin-1 and elevating eNOS expression. Borneol restored BBB integrity and decreased edema in the brain by increasing claudin-5 and ZO1 expression, decreasing MMPs and brain endothelial cell surface adhesion molecules intercellular adhesion molecule-1 (ICAM-1) and lymphocyte function-associated antigen-1 (LFA-1) [111,194].
On the other hand, the BBB opening effect of borneol is well documented. Increased entry of the lipophilic efflux pump substrate rhodamine-123 was described in the hippocampus and hypothalamus of healthy rats treated with 100 or 200 mg/kg body weight borneol for one week [193]. This BBB opening effect was explained by the disruption of brain endothelial tight junctions by transmission electron microscopy and the decreased expression of PGP and multidrug resistance protein MRP1 (Table 2). Borneol not only improved the brain entry of a variety of therapeutic molecules, but also enhanced drug delivery across other biological barriers [186]. Several mechanisms can contribute to the drug penetration enhancing effect of borneol. First, due to its lipophilic nature and small molecular weight, it can form micelles in lipid membranes, which will perturb plasma membrane lipid structures and lead to interendothelial tight junction disruption and enhanced cell membrane permeability [186]. Second, it can increase BBB opening by activation of adenosine receptors [246]. Third, it can elevate the entry of lipophilic and amphiphilic drugs and xenobiotics by inhibiting efflux pumps [186,193]. Based on these effects, borneol is investigated as a potential permeability enhancer for better drug delivery [186].
In a general hypoxia model in mice, cerebral edema was reversed by coumaric acid, similarly to dexamethasone. BBB integrity, occludin levels, and brain edema returned to the level of the non-hypoxic animals [199]. Although atherosclerosis affects mainly larger vessels in the brain [13], the BBB is also damaged during hyperlipidemic conditions. Our laboratory described that an ApoB-100 mouse model for hypertriglyceridemia ([247], Bjelik et al., 2006) presented increased BBB permeability, edema and TJ disruption along with gene expression changes [248]. In this mouse model, proinflammatory interleukin-6 (IL-6) levels were also upregulated [249]. In high-fat-diet-induced rat hyperlipidemia, lycopene restored the level of TJ molecule claudin-5, and decreased VEGF, vascular cell adhesion molecule-1 (VCAM-1) and proinflammatory cytokines [222]. In a rat diabetic model induced by high-fat diet and streptozocin, luteolin increased ZO-1, occludin, and GLUT-1 expression [220].
In a subarachnoid hemorrhage model, apigenin decreased inflammation and BBB disruption by elevating ZO-1 and occludin expression [189]. Hemorrhage-induced vasospasm was decreased by lutein [217]. Astaxanthin-derivative adonixanthin and lycopene lowered BBB leakage during subarachnoid hemorrhage [191,221]. Astaxanthin increased barrier integrity and decreased brain edema through the downregulation of MMP-9 expression [192].

3.1.3. Neurodegenerative Diseases

Changes in BBB functions, namely permeability increase and decreased expression of transporters, such as GLUT-1 and P-glycoprotein, already occur during the preclinical, mild cognitive impairment stage of neurodegenerative diseases, such as Alzheimer’s disease. At later stages, severe BBB damage with microbleeds, leukocyte infiltration, and perivascular neurotoxic aggregates are present [12]. In an animal model of memory deficit induced by β-amyloid peptide 25–35 (Aβ25–35), luteolin increased barrier integrity by upregulating the expression of junctional associated molecule ZO-1 and TJ proteins occludin and claudin-5. It also decreased astrocyte swelling, and increased cerebral blood flow [219]. In a similar model, quercetin protected BBB integrity and elevated cerebral blood flow [230].

3.1.4. Neuroinflammation, Oxidative Stress, and Other Neurodevelopmental Diseases

During systemic inflammation barrier integrity, signaling pathways, transport for molecules, and immune cell trafficking can change at the BBB, all altering the normal physiological function [250]. Kaempferol ameliorated the neuroinflammation-inducing effects of LPS in mouse models, resulting in better barrier integrity and higher expression of occludin and connexin-43 [213,214]. In a similar model, kaempferol decreased shrinkage of brain endothelial cells, edema of astrocytes, and the area surrounding the capillaries in the striatum [214]. Folic acid increased BBB integrity in a rat sepsis model [239].
Few investigations focused on environmentally harmful agents, such as polychlorinated biphenyl, which can induce oxidative stress in animals, and also affects BBB permeability. Quercetin restored the level of TJ proteins and TJ-associated proteins at the BBB, and thus protected its barrier function [233].
In uremia, caused by renal damage, toxins are released that also harm the BBB. These patients often need imaging using contrast media that also affect BBB functions. EPA and DHA protected the integrity of the BBB by the upregulation of claudin-5 and laminin [206].
Changes in BBB functions were described in neurodevelopmental diseases, such as autism spectrum disorder [251]. Valproic acid induces symptoms in rats mimicking autism related phenomena. Fisetin elevated barrier integrity and claudin-5 expression in a rat model of autism [209] by possibly modulating the WNT/β-catenin pathway [251].
To sum up, the majority of the in vivo studies on disease models that investigated the effects of nutraceuticals on the BBB focused on barrier integrity (Table 2). BBB permeability changes were measured by brain edema formation and Evan’s blue dye leakage to the brain, which reflects to albumin extravasation. Investigations focused on TJ breakdown and molecular mechanisms leading to the damage of intercellular connections. Since MMPs degrade TJ and basal membrane proteins, their expression was also described in several studies. While upregulation of cell surface adhesion molecules ICAM-1 and VCAM-1 were observed, leukocyte transmigration in these studies was not investigated. Astrocyte function through the regulation of AQP4 levels could be a novel target for nutraceuticals [252]. Another knowledge gap is that changes in the function or morphology of brain pericytes, which have a crucial role in the development and maintenance of the BBB [253], were not explored (Figure 2). Brain endothelial surface glycocalyx is an important element of the barrier functions of the BBB, and is damaged in systemic and brain inflammatory diseases [16]. Since most nutraceuticals listed in this review have antioxidant effects, they might also protect against brain endothelial glycocalyx damage. Investigations on brain pericytes and brain endothelial glycocalyx can be a promising line of research for nutraceuticals in the future. At present, animal models are indispensable tools in CNS research, driving progress toward novel treatments and a deeper understanding of brain health and disease. One should consider, however, the challenges related to animal experiments, including ethical considerations to refine, replace, and reduce in vivo models [254], species-specific differences, and the difficulty of fully recapitulating the human disease phenotypes. Emerging research technologies, especially human organs-on-chip models of diseases, may solve these problems by replacing the current in vivo methods [255].

3.2. In Vitro Investigations

In line with global efforts to reduce, refine, and replace animal experiments, cell culture models are on the rise, providing crucial understandings of the basic molecular mechanisms of CNS diseases [13]. Many in vitro models of the BBB exist, from simple cell lines to complex primary cell based co-culture models, from rodent and human origin [256]. The barrier tightness and transporter expression of these models should be characterized before they can be considered suitable to establish disease models or to test compound permeability across the BBB [174,175,256]. The state-of-the-art BBB models are based on human stem cells [175,176], but such models have not been used for the investigation of nutraceuticals yet. In this review, only culture models made from brain endothelial cells or cell lines were included (Table 3).
Table 3. In vitro studies: effects of nutraceuticals on brain endothelial cells.
Table 3. In vitro studies: effects of nutraceuticals on brain endothelial cells.
CompoundBBB ModelInjuryEffect on Brain Endothelial CellsReference
Apigeninhuman BMECPMAtube formation ↓, MMP-9 ↓[257]
human BMECOGD/Rcell viability ↑,
cell migration and tube formation ↑, caveolin-1 ↑
[190]
Astaxanthin and derivativeshuman HBMEC
cell line
proliferation ↑, tube formation ↑
cell cycle G0/G1 phase ↓, S phase ↑,
[258]
OGDcell viability ↑, LDH release ↓
human BMEChemoglobin,
collagenase
cell viability ↑, ROS ↓, VE-cadherin ↑[191]
mouse bEnd.3 cell lineOGDRcell viability ↑, apoptosis ↓, FD40 permeability ↓, CLDN-5, ZO-1 ↑[259]
porcine BMECAPP, ADAM10 ↑, BACE-1 ↓,
PGP, ABCA1, LRP-1, Aβ1–40 uptake and transport ↑, cholesterol synthesis ↓
[260]
Borneolrat BMEC, AC co-culturePGP ↓, R123 accumulation ↑, digoxin, verapamil transport ↑[110]
mouse bEnd.3 cell linepuerarin, tetramethylpyrazine permeability ↑, ZO-1 ↓[246]
rat BMECOGDcell viability ↑, apoptosis ↓, CAT ↑, VEGF and VEGFR1 ↑[193]
Caffeinemouse BMECTNF-α + IFN-γVCAM-1 ↓, iNOS ↓[261]
Capsaicinmouse cEND
cell line
TEER ↓, CLDN5 ↓, ZO-1 dislocation[262]
human hCMEC/D3 cell lineTNF-αIL-1β, IL-6 ↓[263]
Catechin/Epicatechin and derivativesrat BMEC, AC, PC co-cultureTNF-α + Il-1βCLDN-5, β-catenin staining ↑,
ROS, NO ↓,
leptin transporter LRP2 ↓
[264]
human BMEC1–421–42 fibril formation ↓, ROS ↓[265]
Chrysinmouse bEnd.3 cell lineLPSVCAM-1 ↓, monocyte adhesion ↓[266]
Cinnamic acid derivativeshuman HBMEC-2
cell line
oxidative stresscell damage ↓, cell viability ↑, mitochondrial transmembrane potential ↑[267]
Curcuminbovine BMECoxidative stresscell damage, LDH release ↓[200]
rat BMECOGDLDH release ↓, IL-1β ↓[268]
porcine BMECBCRP protein ↓, efflux transport ↓[68]
Cyanidin metaboliteHBMEChypoxiacell proliferation ↓, cell viability ↓, cyclin D1, CDK2, CDK4 ↓ [269]
Docosahexaenoic acidrat BMEC, PC, AC co-cultureoligomeric Aβ42 cell viability ↑, ROS production ↓ barrier integrity ↑
SF, albumin permeability ↓,
PGP ↑, R123 accumulation ↓
[270]
BMECOGDapoptosis ↓[271]
porcine BMECIL-1βCalcein-AM accumulation ↑[272]
Fisetinhuman BMECPMAtube formation ↓, MMP-9 ↓[257]
Fucoxanthinmouse bEnd.3 cell linemechanical/
stretch
cell viability ↑, apoptosis↓, TEER ↑, γGT ↑, ACSL4 ↓, PINK1, LC3 ↑[210]
Gallic acid rat BMEC, AC, PC co-cultureTNF-α + Il-1βCLDN-5 and β-catenin staining ↑[264]
Genisteinhuman BMECTNF-αTNF-α, IL-1β, CCL-1, IL-8, ICAM-1 ↓, leukocyte tr.migration ↓[273]
mouse bEnd.3 cell line25–35cell viability ↑,
ROS, and nitrotyrosine ↓, GSH ↑
[274]
Kaempferol
and derivatives
rat RBE4
cell line
ecto-ALP ↑, MPP+ uptake ↑[275]
rat RBE4
cell line
ecto-ALP ↑, insulin uptake ↑[276]
human BMECOGD/Rcell viability ↑, FD permeation ↓,
occludin, ZO-1 ↑
[215]
human BMEChypoxia/reoxygenationcell viability ↑, apoptosis ↓, mitochondrial membrane potential ↑, tube formation ↑, ICAM-1, VCAM-1, IL-1β ↓[277]
α-Lipoic acidbEnd.3,
rat BMEC
OGD/RLDH release ↓[278]
Luteolinhuman BMECPMAtube formation ↓, MMP-9 ↓[257]
human BMEC, AC co-cultureAβ1–40cell viability ↑, TEER ↑,
SF and albumin permeability ↓,
TNF-α, IL-1β, IL-6, IL-8 release ↓
[279]
Lycopene mouse bEnd.3 cell linecell viability ↑[280]
Myricetinhuman BMECOGD/RFD70 permeation ↓, TEER ↑,
TNF-α, IL-1β and IL-6 ↓,
NO and eNOS activity ↑
[281]
human BMECoxidative stresscell viability ↑ [90]
Naringeninmouse b.END5
rat RBE4 cell lines
concentration and time-dependent cellular uptake [37]
Naringinporcine BMECBCRP protein ↑, efflux transport ↑[68]
Piceatannolmouse bEnd.3 cell lineLPS ICAM-1 and VCAM-1 ↓,
iNOS, ROS ↓
[148]
Polydatinprimary rat BMECOGDcell viability ↑, TNF-α, IL-6 ↓
CLDN-5, occludin, ZO-1 ↑
[226]
Procyanidinrat BMECPGP activity ↓, efflux transport↓,
R123 accumulation ↑
[35]
Pterostilbenehuman BMECOGDcell viability ↑, MMP-9 ↓,
CLDN-5, ZO-1, VE-cadherin,
occludin ↑, F/G actin ↓
[228]
Quercetin and metabolitesrat RBEC1 cell lineconcentration- and time-dependent cellular accumulation[282]
human BMECAβ1–40cell viability ↑, LDH release ↓
TEER ↑, albumin and SF permeability ↓, ROS ↓, γGT, ALP ↑
[283]
human BMECoxidative stresscell viability ↑[90]
porcine BMECBCRP protein ↑, efflux transport ↑[68]
human BMEChypoxia/reoxygenetionviability ↑, migration, angiogenesis ↑, CLDN-5 and ZO-1 ↑, VCAM-1 ↓, ROS ↓[284]
mouse bEnd.3 cell lineGlaesserella parasuis infectionIl-6, Il-8, Il-18, TNF-α, MMP-9, ANG-2, ET-1 ↓, ZO-1, occludin, CLDN-5 ↑[285]
Quercetin-biapigenin nanoparticleshuman hCMEC/D3 cell lineoxidative stresscell viability ↑, TEER ↓[286]
Quercetin +/− hydroxysafflor yellow A human hCMEC/D3 cell lineOGDcell viability ↑, TEER ↑[235]
Resveratrol rat BMECOGDcell viability ↑[236]
rat BMEC, AC, PC co-cultureTNF-α + Il-1βalbumin permeability ↓, CLDN-5 and β-catenin staining ↑, NO ↓[264]
RutinHBMEChypoxiacell proliferation ↓, cell viability ↓, cyclin D1, CDK2, CDK4 ↑[269]
Silymarinhuman
HBEC-5i
cell line
AGEcell migration ↓, tube formation ↓[287]
Sulforaphanehuman hCMEC/D3
cell line
NRF2 gene silencing by siRNAmitochondrial ABCB10 ↑[288]
mouse BMEC-GLUT1 ↑, HK1, PDK1, GSK, PKM2, ATP production ↑, NQO1, CAT, GSTs, TXN1, GSR ↑, ABCD3, ABCB6 ↑, ferroportin-1 ↑[127]
Tangeretinhuman HBMEC cell lineOGDcell viability ↑, ROS and MDA ↓, SOD activity ↑, NO and iNOS ↓[289]
Theophyllinemouse BMECTNF-α + IFN-γVCAM-1 ↓, iNOS ↓[261]
Vitamin Ehuman
HBEC-5i
cell line
oxidative stresscell viability ↑, apoptosis ↓, mitochondrial membrane potential ↑, ROS ↓, GSH ↑, SOD, GPX, CAT ↑, cytosolic HO-1 and NQO1 ↑[290]
Abbreviations: Aβ: amyloid-β; ABC: ATP binding cassette transporter; AC: astrocyte; ADAM10: A disintegrin and metalloproteinase domain-containing protein 10; AGE: advanced glycation end products; ALP: alkaline phosphatase; APP: amyloid precursor protein; ATP: adenosine triphosphate; BACE-1: β-site of APP cleaving enzyme; beta-secretase; BBB: blood–brain barrier; BCRP: breast cancer resistance protein; BMEC: brain microvascular endothelial cells; calcein-AM: calcein-acetoxymethyl ester; CAT: catalase; CCL: Chemokine (C-C motif) ligand; CDK: cyclin dependent kinase; CLDN-5: claudin-5; ET-1: endothelin-1; FD: FITC-dextran; LDH: lactate dehydrogenase; GLUT1: glucose transporter 1; GSH: glutathione; GSK: glucokinase; glutathione-disulfide reductase; GPX: glutathione peroxidase; GST: glutathione S-transferase; γGT: γ-glutamyl transpeptidase; HK1: hexokinase type 1; HO-1: heme oxygenase 1; IFN-γ: interferon-γ; Il-1β: interleukin-1β; iNOS: inducible nitric oxide synthase; LPS: lipopolysaccharide; MDA: malondialdehyde; MMP: matrix metalloproteinase; MPP+: 1-methyl-4-phenylpyridinium; NO: nitric oxide; NQO1: NAD(P)H dehydrogenase (quinone) 1; NRF2: nuclear factor erythroid 2-related factor 2; OGD: oxygen-glucose deprivation; OGD/R: oxygen-glucose deprivation/reoxygenation; PC: pericyte; PDK1: pyruvate dehydrogenase kinase 1; PKM2: pyruvate kinase isozyme type 2; PGP: P-glycoprotein; PMA: phorbol 12-myristate 13-acetate; R123: rhodamine 123; ROS: reactive oxygen species; SF: sodium fluorescein; SOD: superoxide dismutase; TEER: transendothelial electrical resistance; TNF-α: tumor necrosis factor-α; TXN1: thioredoxin 1; VCAM-1: vascular cell adhesion molecule 1; VEGF: vascular endothelial growth factor; VEGFR: vascular endothelial growth factor receptor; ZO-1: zonula occludens-1; ↑: increase; ↓: decrease.

3.2.1. Cell Viability

The effects of nutraceutical compounds on the viability of brain endothelial was studied the most in culture BBB models, but rarely in uninjured, control conditions (Table 3). Nutraceutical compounds at low concentrations are non-toxic, but high, suprapharmacological concentrations of the flavonoids apigenin, genistein, hesperidin, kaempferol, rutin, and quercetin decreased the viability of rat brain endothelial cells measured by the metabolic assay MTT [56]. In contrast, lycopene increased cell viability in mouse bEnd.3 cell line compared to control group [280].
In all of the models, using different modalities to induce cell injury, nutraceuticals exerted protective effects (Table 3). Among the oxidative stress models, cinnamic acid derivatives inhibited H2O2-induced cell damage, suppressed the decrease in mitochondrial transmembrane potential, and increased cell viability in HBMEC-2 cell line [267]. Myricetin and quercetin, the main components of blackberry polyphenols, were also protective against H2O2-induced cell toxicity [90]. Quercetin and apigenin nanoparticle formulation counteracted the metabolic activity decrease caused by the oxidant tert-butyl hydroperoxide [286]. Cell injury measured by LDH release and induced by oxidative stress was inhibited by curcumin [200,268].
Fucoxanthin increased cell viability and inhibited apoptosis and ferroptosis after mechanical stretch injury, an in vitro TBI model, in mouse bEnd.3 cell line by modulating ACSL4, PINK1, and LC3genes regulating autophagy [210]. Astaxanthin and its derivatives, adonixanthin and adonirubin, inhibited cell death in a brain hemorrhage human brain endothelial cell model [191].
Luteolin and quercetin elevated brain endothelial cell viability after treatment with Aβ1–40 [279,283]. DHA showed similar protection in all the neurovascular cell types treated with an oligomeric Aβ1–42 [270]. Genistein also counteracted cell death induced by Aβ25–35 in bEnd.3 cells [274].
Stroke is modeled in vitro by oxygen and glucose deprivation (OGD) that can be followed by a reoxygenation period (OGD/R). This was the most investigated pathological BBB culture model for nutraceuticals (Table 3). Astaxanthin, kaempferol and its derivative juglanin, pterostilbene, and quercetin, with or without hydroxysafflor yellow A, resveratrol, apigenin, borneol, DHA, α-lipoic acid, and tangeretin, promoted cell viability after hypoxia or OGD/R in cultured brain endothelial cells [84,190,193,215,228,235,236,258,259,271,278,289]. Polydatin rescued brain endothelial cell viability in an OGD model [226]. Kaempferol and other agents like resveratrol, quercetin, and polydatin in a plant extract increased cell survival and mitochondrial membrane potential in a hypoxia/regeneration model [277]. Vitamin E protected human brain endothelial cells against oxidative-stress-induced cell death and apoptosis [290].

3.2.2. Cell Proliferation, Migration, and Tube Formation

VEGF together with angiopoietins are the most important mediators of angiogenesis in the brain [253]. VEGF, as its name suggests, elevates brain endothelial cell proliferation, but it also enhances BBB permeability; therefore, its levels are reduced after BBB reaches its full maturity, and the barrier tightness is increased. VEGF levels are increased in CNS pathologies, especially after stroke, when angiogenesis can help neuronal survival in the penumbra region [14].
Under healthy conditions, astaxanthin increased brain endothelial cell proliferation and migration in a scratch assay, and also increased tube formation. The resting G0/G1 phase of the cell cycle was decreased, while the more active S phase was increased by astaxanthin [258]. Apigenin, luteolin, and fisetin decreased the tube forming capacity of human brain endothelial cells [257]. Elderberry and elderflower extracts rich in cyanidin metabolite and rutin inhibited cell proliferation by the downregulation of cyclin D1, CDK2, and CDK4 cell cycle promoters in human brain endothelial cells [269].
Advanced glycation end products promote cell migration and tube formation of endothelial cells. Sylimarin decreased this effect through the GSK-3β-dependent inhibition of VEGF release [287]. In contrast, borneol enhanced the level of VEGF, and decreased VEGFR1 expression in rat brain endothelial cells after OGD pushing cell signaling towards angiogenesis [193]. Quercetin enhanced cell migration in a scratch assay, and increased tube formation in a hypoxia/reoxygenation model [284]. In an OGD/R model, apigenin increased caveolin-1 expression, cell migration, and tube formation in human brain endothelial cells via the VEGF pathway [203]. Kaempferol, resveratrol, quercetin, and polydatin, the main components of a plant extract, promoted tube formation and cell layer regeneration in a hypoxia/reoxygenation BBB model [277].

3.2.3. Barrier Integrity

The intercellular connections of brain endothelial cells are composed molecularly of tight and adherens junction proteins linked with junction associated molecules. In CNS and systemic diseases, brain endothelial TJs weaken, causing BBB leakage that worsens disease pathology [12]. On the other hand, opening the TJs between brain endothelial cells can enable paracellular drug delivery [291,292]. Therefore, the question of whether the TJs need to be reinforced or weakened depends on the goal of the study.
Using in vitro BBB models, barrier opening was induced by borneol, which downregulated ZO-1 expression in bEnd.3 cells and enhanced the permeability of other natural compounds [246]. Capsaicin treatment decreased claudin-5 expression and promoted intracellular redistribution of ZO-1 molecules in cEND cells [262]. A quercetin-biapigenin nanoparticle formulation caused a mild, but significant, decrease in the transendothelial electrical resistance of the hCMEC/D3 model [286].
One of the main hallmarks of Alzheimer’s disease pathology is the perivascular accumulation of Aβ peptide aggregates [12]. This pathology can be mimicked by treatment with fibrillary Aβ1–40 that damages basic barrier functions, decreases the electrical resistance, and increases the permeability of the in vitro BBB models. These changes were counteracted by luteolin treatment in a human co-culture BBB model [279]. Quercetin restored BBB integrity in a similar system [283]. In a rat co-culture BBB model, impaired barrier tightness measured by decreased resistance and increased permeability induced by oligomeric Aβ42 was blocked by the addition of DHA [270].
Myricetin increased barrier integrity measured by resistance elevation and transcellular permeability decrease [281]. Astaxanthin increased barrier integrity and upregulated claudin-5 and ZO-1 expression in b.End3 cells in an OGDR model [259]. Juglanin (kaempferol-3-O-α-L-arabinofuranoside) decreased paracellular permeability and restored occludin and ZO-1 expression in human brain endothelial cells [215]. Interestingly, juglanin significantly increased the expression of these junctional molecules compared to the untreated control group, suggesting a BBB tightening effect of the compound [215]. Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) is a long, non-coding RNA that protects the BBB after stroke. Polydatin increased the expression of junctional molecules claudin-5, ZO-1 and occludin in an OGD model via MALAT1 upregulation [226]. After 3 and 12 h of OGD injury, the actin structure of human brain endothelial cells was changed, which was reversed concentration-dependently by pterostilbene [228]. Pterostilbene inhibited the redistribution of occludin, claudin-5, and VE-cadherin from the junctions to the cytoplasm. It also increased the expression of occludin, claudin-5, ZO-1, VE-cadherin, and basal membrane component laminin, while MMP-9 was downregulated [228]. Quercetin in combination with hydroxysafflor yellow A increased the resistance of hCMEC/D3 cells in an OGD model [235], while quercetin alone increased claudin-5 and ZO-1 levels in human brain endothelial cells after hypoxia/reoxygenation [284].
Few other in vitro injury models were studied for BBB integrity. Grape phenolic compounds epicatechin, gallic acid, and resveratrol upregulated claudin-5 and β-catenin immunostaining in a triple co-culture model of the BBB treated with proinflammatory cytokines to model neuroinflammation [264]. The astaxanthin derivative adonixanthin increased VE-cadherin levels in human brain endothelial cells treated with hemoglobin and collagenase, pathological factors in brain hemorrhage [191]. To model TBI, b.End3 cells were injured by mechanical stretch. Fucoxanthin elevated resistance and thus barrier integrity in the model [210]. Glasserella parasuis bacterial infection causes meningitis in pigs. An infection model with this microbe decreased the junctional expression of ZO-1, occludin, and claudin-5 in bEnd.3 cells, which was reversed by quercetin [285].

3.2.4. Antioxidative and Anti-Inflammatory Effects

The antioxidative effects of nutraceuticals (Table 1) is one of the central elements of their biological actions and protective effects on the CNS and the BBB [182,293]. This can be a direct chemical scavenging of reactive oxygen species (ROS), or an indirect effect by the elevation of the cellular antioxidative defense through regulatory factors [6,14,293]. The latter includes the upregulation of key antioxidant enzymes such as superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPX), glutathione-disulfide reductase (GSR), and downregulation of enzymes producing excess nitric oxide (NO) like the inducible nitric oxide synthase (iNOS/NOS2). While ROSs contribute to inflammation, the main regulators are cytokines and chemokines, which are produced in all cells of the neurovascular unit and are induced by pathological stimuli, such as bacterial lipopolysaccharides (LPS) [294]. As we discuss below, nutraceuticals downregulate the level of proinflammatory cytokines that also contribute to their protective actions on brain endothelial cells (Figure 3).
Figure 3. The molecular basis of the protective effects of nutraceuticals on cells of the blood–brain barrier. Elements of brain endothelial cell and astrocyte functions that are involved in BBB damage and rescued by nutraceuticals are shown. ABCD3: ATP-binding cassette sub-family D member 3; ALP: alkaline phosphatase; ANG-2: angiopoietin 2; AQP4: aquaporin-4; ATP: adenosine triphosphate; Aβ1–40: amyloid-β1–40; BCRP: breast cancer resistance protein; CAT: catalase; CAV-1: caveolin-1; CLDDN-5: claudin-5; COX-2: cyclooxygenase-2; CX43: connexin-43; eNOS: endothelial NOS; ET-1: endothelin 1; GLUT1: glucose transporter 1; GPX: glutathione peroxidase; GSH: glutathione; GSR: glutathione-disulfide reductase; GSTs: glutathione S-transferases; HK1: hexokinase Type I; HO-1: heme oxygenase-1; IL-1β; -6; -8; -18: interleukin-1β; -6; -8; -18; iNOS: inducible nitric oxide synthase; JAM-1; -3: junction associated molecule-1; -3; LRP1; -2: low density lipoprotein receptor-related protein 1; MDA: malondialdehyde; MMP-2; -9: matrix metalloproteinase-2; -9; NF-κB: nuclear factor kappa-light-chain-enhancer of activated B; NO: nitric oxide; NQO1: NAD(P)H quinone oxidoreductase 1 and -2; OCLN: occludin; PDK1; -2: pyruvate dehydrogenase kinase 1; -2; P-gp: P-glycoprotein; PKM2: pyruvate kinase isozyme type 2; ROS: reactive oxygen species; SOD: uperoxide dismutase; TNF-α: tumor necrosis factor-α; TXN1: thioredoxin 1; VCAM-1: vascular cell adhesion molecule-1; VEGF: vascular endothelial growth factor; VEGFR2: vascular endothelial growth factor receptor 2; ZO-1: zonula occludens-1; γGT: gamma-glutamyl transpeptidase; ↑: increase; ↓: decrease. Created in BioRender. Kucsápszky N.; Santa Maria; A. (2025) https://BioRender.com/g20w064 (accessed on 10 February 2025).
Figure 3. The molecular basis of the protective effects of nutraceuticals on cells of the blood–brain barrier. Elements of brain endothelial cell and astrocyte functions that are involved in BBB damage and rescued by nutraceuticals are shown. ABCD3: ATP-binding cassette sub-family D member 3; ALP: alkaline phosphatase; ANG-2: angiopoietin 2; AQP4: aquaporin-4; ATP: adenosine triphosphate; Aβ1–40: amyloid-β1–40; BCRP: breast cancer resistance protein; CAT: catalase; CAV-1: caveolin-1; CLDDN-5: claudin-5; COX-2: cyclooxygenase-2; CX43: connexin-43; eNOS: endothelial NOS; ET-1: endothelin 1; GLUT1: glucose transporter 1; GPX: glutathione peroxidase; GSH: glutathione; GSR: glutathione-disulfide reductase; GSTs: glutathione S-transferases; HK1: hexokinase Type I; HO-1: heme oxygenase-1; IL-1β; -6; -8; -18: interleukin-1β; -6; -8; -18; iNOS: inducible nitric oxide synthase; JAM-1; -3: junction associated molecule-1; -3; LRP1; -2: low density lipoprotein receptor-related protein 1; MDA: malondialdehyde; MMP-2; -9: matrix metalloproteinase-2; -9; NF-κB: nuclear factor kappa-light-chain-enhancer of activated B; NO: nitric oxide; NQO1: NAD(P)H quinone oxidoreductase 1 and -2; OCLN: occludin; PDK1; -2: pyruvate dehydrogenase kinase 1; -2; P-gp: P-glycoprotein; PKM2: pyruvate kinase isozyme type 2; ROS: reactive oxygen species; SOD: uperoxide dismutase; TNF-α: tumor necrosis factor-α; TXN1: thioredoxin 1; VCAM-1: vascular cell adhesion molecule-1; VEGF: vascular endothelial growth factor; VEGFR2: vascular endothelial growth factor receptor 2; ZO-1: zonula occludens-1; γGT: gamma-glutamyl transpeptidase; ↑: increase; ↓: decrease. Created in BioRender. Kucsápszky N.; Santa Maria; A. (2025) https://BioRender.com/g20w064 (accessed on 10 February 2025).
Nutrients 17 00766 g003
An example is sulforaphane, which increased the anti-oxidative stress responses and redox signaling in mouse brain endothelial cells by upregulating key enzymes and proteins such as CAT, GSR, NAD(P)H dehydrogenase quinone 1 (NQO1) and thioredoxin 1 (TXN1) [127].
When ischemia damages brain tissue, the mitochondria in brain endothelial cells enhance oxidative stress by decreasing SOD, CAT, and GPX activity, and increasing ROS and malondialdehyde (MDA) levels. Treatment with tangeretin decreased ROS and MDA levels, elevated SOD activity, and decreased iNOS levels [289]. Borneol partly restored the imbalance caused by OGD in brain endothelial cells by increasing CAT expression [193]. ROS levels were decreased, and VCAM-1 upregulation was blocked by quercetin in a hypoxia/reoxygenation model [284]. In a similar setting kaempferol and other agents like resveratrol, quercetin, and polydatin in a plant extract had the same effect, but they also downregulated ICAM-1 expression [277]. Myricetin, polydatin and curcumin decreased proinflammatory cytokine levels in an in vitro OGD/R model [226,268,281].
Proinflammatory cytokine or LPS treatments are often used to model neuroinflammation in an in vitro setting. Caffeine and theophylline inhibited iNOS production and VCAM-1 expression in mouse brain endothelial cells treated with tumor necrosis factor-α (TNF-α) and interferon-γ (IFN-γ) cytokines [261]. Epicatechin counteracted the increase of ROS release in a similar model in a triple co-culture BBB system [264]. In another study, a capsaicin derivative, nitro-capsaicin, inhibited the release of interleukin-1β (IL-1β) and IL-6 and prevented TNF-α-induced inflammation in hCMEC/D3 cells [263]. Cytokine-mediated up-regulation of TNF-α, IL-1β, IL-8, chemokine (C-C motif) ligand 2 (CCL2/MCP-1) and ICAM-1 was reversed by the treatment of brain endothelial cells by genistein. This isoflavone also inhibited leukocyte transmigration across the cultured brain endothelial monolayer [273]. In an LPS-induced injury model, chrysin decreased the expression of VCAM-1, and subsequently inhibited monocyte adhesion to mouse bEnd.3 cells [266]. In a similar model, piceatannol also decreased the presence of VCAM-1 and ICAM-1 on the surface of brain endothelial cells and lowered iNOS levels and ROS [148].
Oxidative stress was also induced in several types of injury models. ROS release was decreased by astaxanthin after treatment with hemoglobin in a human brain endothelial cells modeling hemorrhage in vitro [191]. Vitamin E reduced ROSs and increased the level of glutathione and antioxidant enzymes SOD, GPX, CAT, NQO1, and heme oxygenase 1 (HO-1), as well as the mitochondrial membrane potential in human brain endothelial cells injured by oxidative stress [290].
Fibrillary Aβ1–40 increased proinflammatory cytokine levels in a human BBB co-culture model, which could be reversed by luteolin [279]. ROS levels in brain endothelial cells are elevated by fibrillary Aβ1–40 which was inhibited by co-treatment with quercetin through SOD activation [283]. DHA also lowered ROS production in neurovascular cell types treated with Aβ oligomers [270]. ROS levels were decreased, while the antioxidant glutathione concentration was increased in brain endothelial cells after treatment with genistein in a Aβ25–35 model [274].

3.2.5. Vasoactive Agents

In addition to their specific BBB characteristics, brain endothelial cells, similarly to peripheral vascular endothelium, produce vasoactive agents, like NO, endothelin-1 (ET-1), prostaglandins, and adrenomedullin. This is a neglected area of in vitro BBB research especially in stem cell-derived BBB models, in which endothelial characteristics should be unequivocally demonstrated [295]. NO is a key vasodilator molecule synthesized in brain endothelial cells by the endothelial NOS enzyme (eNOS/NOS3). NO helps maintain BBB functions, and is an important inter- and intracellular signaling molecule [296]. As discussed above, excess NO produced by iNOS contributes to oxidative stress; therefore, NO levels at physiological and in inflammatory conditions are well regulated.
BBB integrity was rescued by myricetin in an in vitro ischemia/reoxygenation model by activating the eNOS/NO pathway in human brain endothelial cells [281]. The elevated NO production in a neuroinflammation model of the BBB in vitro was decreased by grape phenolic compounds epicatechin and resveratrol [264]. In an OGD model, increased levels of NO and iNOS were reduced by tangeretin [289].
Endothelin-1, a vasoconstrictor was upregulated in mouse bEnd.3 cells infected with G. parasuis, an in vitro meningitis model, which was reduced by quercetin [285].
Prostaglandins are vasoactive lipid regulators of the cerebral blood flow produced by cyclooxygenase (COX) enzymes. Apigenin, fisetin, and luteolin inhibited increased gene and protein expression of COX-2 induced by phorbol 12-myristate 13-acetate (PMA) in human brain endothelial cells [257]. In OGD models both DHA [271] and polydatin [226] reduced COX-2 levels in brain endothelial cells.

3.2.6. Effects on BBB Efflux and Influx Transport

Efflux transporters provide chemical defense at the level of BBB by actively blocking the entry of neurotoxic compound to the brain [11]. The dysfunction and downregulation of the two most studied efflux transporters at the BBB, PGP, and BCRP is linked to pathological changes in neurodegenerative diseases, like Alzheimer’s and Parkinson’s diseases [12]. Upregulation of efflux transport at the BBB contributes to the barrier functions, while inhibition of efflux pump activity and downregulation of efflux transporter weaken the chemical defense of the brain and increase the brain entry of efflux pump substrate drugs [181]. As we listed in Table 1, many nutraceuticals interact with influx transporters that are crucial to provide nutrients for brain cells [11]. The most important influx transporter group at the BBB is the solute carriers that are indispensable to carry hydrophilic nutrients and nutraceuticals across the BBB.
Citrus flavonoids hesperetin, naringenin, and their metabolites are taken up by mouse and rat brain endothelial lines in a concentration and time-dependent manner [37]. Quercetin also showed elevated accumulation with time and concentration in rat brain endothelial cells [282]. Sulforaphan upregulated GLUT1, the most important influx transporter of glucose at the BBB, in mouse brain endothelial cells [127]. In healthy cultured porcine brain endothelial cells, astaxanthin upregulated the levels of PGP efflux pump, low density lipoprotein receptor-related protein 1 (LRP1), the receptor of apolipoprotein E, and apolipoprotein A1. Astaxanthin also enhanced Aβ1–40 uptake and transport [260]. Kaempferol increased the uptake of the hormone insulin and the monoaminergic neurotoxin 1-methyl-4-phenylpyridinium (MPP+) in rat RBE4 cells [275,276].
In contrast to astaxanthin, other compounds decreased efflux pump expression and activity (Table 3). Borneol downregulated the expression of PGP and increased the accumulation of rhodamine 123, an efflux pump ligand, in brain endothelial cells and the transport of digoxin and verapamil across the BBB model [110]. Borneol also elevated the permeability of two CNS drugs, puerarin and tetramethylpyrazine via activation of A1A and A2A adenosine receptors, and enhanced BBB permeability in bEnd.3 cells [246]. In rat brain endothelial cells, procyanidin also lowered PGP pump activity leading to decreased efflux transport of rhodamine 123 [35]. BCRP protein level and the efflux transport activity was decreased by curcumin, quercetin, and naringin in brain endothelial cells [68].
In models of injury, protective effects were seen. DHA (30 µM) increased PGP activity and decreased rhodamine 123 accumulation in primary rat brain endothelial cells treated with Aβ oligomers [270]. In porcine brain endothelial cells, DHA (5 µM) increased the accumulation of calcein-acetoxymethyl ester (calcein-AM), a ligand of efflux pumps PGP and MRP1, but did not change plasma membrane fluidity or the expression of PGP, indicating that it decreased the efflux pump activity by other mechanisms [272]. DHA treatment in the same model inhibited the effect of the proinflammatory cytokine IL-1β, which increased efflux pump activity measured by decreased cellular accumulation of calcein-AM (Table 3). Epicatechin, a phenolic compound, counteracted the increased expression of leptin transporter LRP2 in primary rat brain endothelial cells treated with cytokines to induce inflammation [264].

3.2.7. BBB Enzymes and Metabolism

Brain endothelial cells express many enzymes, including phase 1 and 2 drug metabolizing enzymes that act as local protection and detoxification systems and work in tandem with efflux pumps at the BBB [14,174]. From these enzymes, γ-glutamyl transpeptidase (γ-GT) and tissue alkaline phosphatase (ALP/TNAP) are considered as BBB markers, since they are highly enriched in brain endothelial cells. In line with this, high levels of γ-GT can reflect good brain endothelial cell functions and protects against BBB permeability increase in stroke [297].
In human brain endothelial cells injured by Aβ1–40 treatment, quercetin increased the level of BBB marker enzymes γGT and ALP [283]. Fucoxanthin elevated γ-GT levels in the mouse bEnd.3 cell line, helping to restore BBB integrity in a TBI model [210]. Kaempferol increased ecto-ALP activity in rat brain endothelial cells, which was linked to increased insulin and MPP+ uptake [275,276], while sulforaphane upregulated glutathione S-transferases (GSTs) that participate in phase 2 drug metabolism at the BBB [127].
Astaxanthin treatment reduced cholesterol synthesis, decreased the activity of β-secretase (BACE1), and upregulated the non-amyloidogenic ADAM10 metalloproteinase in porcine brain endothelial cells [260]. Astaxanthin thus shifted the metabolism of amyloid precursor protein, and more soluble, and less aggregating peptides were generated.
One of the hallmark of brain endothelial cells is the large number of mitochondria that produce ATP for energy-demanding efflux pumps, enzymes, and ion pumps at the BBB. In pathologies, energy demand increases while ATP levels drop. Sulforaphane increased ATP production in brain endothelial cells by upregulating many important metabolic enzymes and mitochondrial transporters including hexokinase type 1 (HK1), pyruvate dehydrogenase kinase 1 (PDK1), glucokinase (GSK), pyruvate kinase isozyme type 2 (PKM2), ABCD3, ABCB6, and ferroportin-1/SLC40A1 [127].

4. BBB Signaling Pathways Regulated by Nutraceuticals

The literature on signaling pathways regulated by nutraceuticals is vast, and includes different models and diseases such as tumors or tumor cells, cardiovascular, metabolic, or CNS diseases [298]. From this large knowledge base, we only selected data that prove the regulation of signaling pathways in established BBB preclinical models both in vivo and in vitro (Table 4). As summarized in Figure 4, the most important signaling pathways that mediate the protective effects of nutraceuticals on the BBB converge on increased brain endothelial cell survival and BBB stability.
The VEGF pathway is crucial in brain angiogenesis and BBB development [299]. Brain endothelial VEGF signaling is polarized, and luminal VEGF activates AKT and leads to cytoprotection, while abluminal VEGF increases BBB permeability via p38 [300]. The activation of the VEGF pathway at the BBB was demonstrated by apigenin [190], borneol [193], DHA [271], kaempferol [215,277], and silymarin [287]. VEGF-induced cell survival is mediated by the upregulation of antiapoptotic BCL2 protein, and downregulation of apoptotic BAX protein and caspase-3 and -9 enzymes (Table 4, Figure 4).
The phosphatidylinositol 3-kinase (PI3K)-AKT signaling pathway was the second-most investigated (Table 4) one that leads to the protection of both brain endothelial cells and BBB functions (Figure 4). AKT in brain endothelial cells was activated by cyanidin [301], α-lipoic acid [278], lycopene [280], myricetin [281], quercetin [232] and sulforaphane [127,288]. The inhibition of mitogen-activated protein kinase (MAPK), c-Jun N-terminal kinase (JNK), extracellular signal-regulated kinases (ERK), that crosstalk with the PI3K-AKT pathway, also results in increased brain endothelial cell and BBB protection (Figure 4). The downstream effectors of both PI3K-AKT activation and MAPK-JNK-ERK inhibition are the downregulation/inhibition of glycogen synthase kinase-3β (GSK3β) and the activation of nuclear factor erythroid 2-related factor 2 (NRF2). The translocation of NRF2 to cell nuclei results in the transcription of phase II antioxidant enzymes (Figure 3) that increase the level of antioxidant molecules such as glutathione and decrease the level of ROS [293]. NRF2 activation was observed in BBB models by genistein [274], myricetin [281], quercetin [284], and sulforaphane [127,288]. NRF2 upregulation is also central to the neuroprotective effects of chrysin, naringenin, quercetin and sulforaphane [293], further supporting the link between neuroprotection and BBB protection. Indeed, nutraceuticals that target the NRF2 antioxidant pathway upregulate redox resilience genes, modulate the neurosteroid homeostasis, enhance brain resilience and neuronal adaptive responses in CNS diseases [302].
Despite the importance of the canonical WNT signaling via β-catenin in the development and maintenance of the BBB [253,299], few studies investigated this pathway for natural compounds. WNT activation was observed in BBB models by astaxanthin [191], pterostilbene [228], and quercetin [230] that led to increased BBB stability (Figure 4).
Neuroinflammation is a common feature for CNS diseases and the transcription factor NF-κB mediates the release of proinflammatory cytokines and chemokines in the cells of the neurovascular unit. NF-κB, as a central hub in the inflammatory pathways (Figure 4), was the most investigated signaling molecule in the preclinical BBB models (Table 4). Except for borneol, all of the compounds, namely apigenin [189], capsaicin [263], carvacrol [113], chrysin [266], curcumin [268], gallic acid, resveratrol [264], luteolin [279], piceatannol [148], and quercetin [230] decreased the expression, nuclear translocation, or activity of NF-κB, resulting in BBB protection. Activation of NF-κB also contributes to cell death; therefore, its downregulation/inhibition may also lead to cell survival, but this pathway is not shown in Figure 4.
Table 4. BBB signaling pathways regulated by nutraceuticals.
Table 4. BBB signaling pathways regulated by nutraceuticals.
NutraceuticalBBB Signaling Pathway Interactions Reference
ApigeninCOX-2 ↓
TLR4, IκB, NF-κB ↓
BECN1 ↓, VEGF, mTOR ↑
[257]
[189]
[190]
Astaxanthincaspase-3 ↓, pGSK3β ↓
WNT7A, β-catenin, CCND1 ↑,
ERK activation ↓
p75NTR ↓
PPAR-α activation ↑
[258]
[191]
[259]
[260]
BorneolNF-κB activation ↑
A1AR, A2AR ↑
BCL-2 ↑, BAX ↓, Ca2+ ↓, VEGF ↑, VEGFR1 ↓
[110]
[246]
[193]
CapsaicinTRPV1 activity and Ca2+
NF-κB activity, nuclear translocation ↓
[303]
[263]
β-CaroteneAKT, FKHR, and ERK1/2 phosphorylation ↓ [196]
CarvacrolTRPM7 activation ↓
caspase-3 ↓, BAX ↓, BCL-2 ↑, NF-KB ↓
[304]
[113]
Catechin/Epicatechin/Epigallocatechin gallatePKCα ↓[207]
[197]
Chrysinp38 MAPK and JNK activation ↓,
NF-κB p65 translocation ↓
[266]
Curcuminp38 MAPK and NFκB activation ↓[268]
CyanidinAKT ↑, caspase-3 ↓, ERK1/2 ↓[301]
Docosahexaenoic acidANG2 ↓,VEGF ↑
PGE2, PGI2, COX-2 ↓
[271]
FisetinCOX-2 ↓[257]
Fucoxanthincaspase-3 ↓[210]
Gallic acidNF-κB nuclear translocation ↓[264]
GenisteinNRF2, PI3K ↑[274]
HesperidinFOXO3a nuclear translocation ↓[85]
Kaempferol and derivativesVEGF and VEGFR2 ↓
VEGF ↑
[215]
[277]
α-Lipoic acidAKT and mTOR phosphorylation ↑[278]
LuteolinCOX-2 ↓
NFκ-B activation ↓
[257]
[279]
LycopeneAKT activation ↑, LXR-β ↑[280]
MyricetinAKT and NRF2 activation ↑[281]
Naringeninp-GSK-3β ↓[224]
PiceatannolNF-κB, MAPK, p38, JNK ↓
p-IKKα/β, p-IκBα, p-p65 ↓
[148]
PolydatinCREB/PGC-1α/PPARγ ↑
COX-2 ↓
[226]
Pterostilbenec-Met, c-Jun and c-Myc proteins ↑
WNT pathway activation ↑
[228]
QuercetinKEAP1/NRF2 activation ↑, ATF6/GRP78 ↓
VEGF ↓, PI3K/AKT/ERK activation ↑
WNT ↑, GSK-3β expression ↓
NF-kB p65, RAGE ↓
[284]
[285]
[232]
[230]
ResveratrolNF-κB nuclear translocation ↓[264]
SilymarinVEGF release ↓[287]
SulforaphanNRF2 ↑, AKT phosphorylation ↑
NRF2 ↑
[127,288]
Tangeretincaspase-3 ↓, JNK activation ↓[289]
Vitamin E/α-TocopherolBAX, caspase-9/caspase-3 ↓, BCL-2 ↑,
NRF2 ↑
[290]
Abbreviations: A1AR: adenosine A1 receptor; A2AR: adenosine A2 receptor; ANG2: angiopoietin 2; ATF6: activating transcription factor 6; BAX: BCL-2-like protein 4; BCL-2: B-cell lymphoma 2 protein; BECN1: Beclin-1; CCND1: Cyclin D1; COX-2: cyclooxygenase-2; CREB: cAMP response element-binding protein; ERK: extracellular signal-regulated kinases; FKHR: forkhead transcription factor; FOXO3a: Forkhead box O3; GRP78: 78 kDa glucose-regulated protein; GSK-3β: Glycogen synthase kinase-3β; JNK: c-Jun N-terminal kinase; KEAP1: Kelch-like ECH-associated protein 1; LXR-β: Liver X receptor-β; MAPK: mitogen-activated protein kinase; mTOR: mammalian target of rapamycin; NF-κB: nuclear factor κ-light-chain-enhancer of activated B; NRF2: nuclear factor erythroid 2-related factor 2; PGC-1α: Peroxisome proliferator-activated receptor gamma co-activator 1α; PGE2: prostaglandin E2; PGI2: prostaglandin I2; PI3K: phosphoinositol 3-kinase; PKCα: Protein kinase Cα; PPARγ: Peroxisome proliferative activated receptor γ; RAGE: receptor for advanced glycation end products; TLR4: Toll-like receptor 4; TRPV1: transient receptor potential cation channel subfamily V member 1; VEGF: vascular endothelial growth factor; VEGFR: vascular endothelial growth factor receptor; WNT7A: Protein Wnt-7a; ↑: increase; ↓: decrease.
COX enzymes produce a complex array of proinflammatory and anti-inflammatory prostanoid and endocannabinoid lipid mediators that are important regulators of neuroinflammation. COX-1 and -2 are important anti-inflammatory pharmaceutical targets for disease modification, and COX inhibitors alone or in combination are examined in clinical trials of neurological diseases [305]. BBB protection by COX-2 inhibition (Figure 4) was observed for apigenin [257], DHA [271], fisetin [257], luteolin [257], and polydatin [226].
Some nutraceuticals can activate or inhibit ion channels at the BBB. The alkaloid capsaicin, an activator of TRPV1, increased intracellular Ca2+ levels in cultured brain endothelial cells [303], although no further studies were performed to reveal the functional consequence of the channel activation. The monoterpene carvacrol, a TRPM7 inhibitor, reduced TRMP7 expression in spinal cord vessels, decreased the inflammatory response and stabilized the blood–spinal cord barrier in an injury model [304].
Several nutraceuticals listed in Table 4 belong to the group of phytoestrogens, including curcumin, genistein, kaempferol, resveratrol, and quercetin. These compounds may also exert beneficial effects on the BBB via estrogen receptors, the activation of protective signaling pathways like PI3K-AKT, and inhibition of JNK, ERK, and p38 [202]. BBB protection by phytoestrogens can be mediated by endothelial nitric oxide synthase (eNOS) and the VEGF pathway.
Borneol has a dual, concentration-dependent effect on the BBB. At high concentrations, it increased BBB permeability by activation of NF-κB and adenosin A1A and A2A receptors [110,246]. However, at low concentrations, it also increased BBB stability by elevating BCL-2 and VEGF and reducing BAX and intracellular Ca2+ levels [193].
As a conclusion, the most important signaling changes mediating the increased cell survival and BBB stability were the activation of the WNT, PI3K-AKT, and NRF2 pathways, and inhibition of the MAPK, JNK, ERK, and NF-κB pathways (Figure 4). It should be noted that there is crosstalk between many elements of these pathways, and signaling networks at the BBB are very complex and still not fully understood.

5. Toxicity and Drug Interactions

This review focused on the protective effects of nutraceuticals, but toxic effects resulting in adverse events and drug interactions were also described for certain nutraceutical supplements [184]. These were reported mainly for dietary supplements containing herbal extracts. Green tea and ginseng extracts were related to acute liver injury, while gingko extracts caused decreased blood coagulation [184]. While most nutraceuticals are considered generally safe, toxicity depends on dose and concentration, and also on individual pharmacogenetics. Caffeine is an example, which, if overdosed, can trigger cardiovascular problems in some individuals [306]. As we described in the cell viability section, the listed compounds were not toxic for brain endothelial cells in the studies, except for very high concentrations of six tested flavonoids [56]. For most of the compounds, toxicity evaluations on different cell types or organ models are missing.
Another potential safety issue is that nutraceuticals by their interaction with drug efflux and influx transporters and drug metabolizing enzymes can cause drug interactions [184]. Most of the listed compounds interact with efflux transporters (Table 1) expressed at the BBB, in the liver, intestines, and kidneys that highly influence the absorption and biodistribution of many prescription drugs such as cyclosporin A, verapamil, digoxin, and many others [11]. The interactions of nutraceuticals with drug efflux pumps may result in higher intestinal absorption, lower excretion, and higher organ penetration of other efflux pump ligand medicines, resulting in their altered pharmacokinetics and pharmacodynamics. These potential detrimental effects of nutraceuticals should be carefully considered in addition to their beneficial or protective properties.

6. Conclusions and Future Perspectives

Although the research field of nutraceuticals is promising and provides an almost endless number of molecules to test, the clinical application of nutraceuticals to protect the BBB requires further research. There are several challenges ahead. We should better understand the mechanisms of action of nutraceutical compounds, and optimize their bioavailability and dosing. On the experimental pharmacology level, more critical and systematic reviews would be needed. The preclinical studies reviewed here used single compounds in single disease models. Systematic testing of compound combinations or combinations of nutraceuticals with non-nutraceutical molecules or medicines for synergistic effects and investigations of the same compound on multiple or complex models are missing. Human BBB-on-chip models may provide platforms [295] to solve some of these problems. On the clinical pharmacology level, rigorous and reliable information on the safety and efficacy of nutraceuticals can be obtained from randomized controlled trials [184], which are missing related to the protection of the BBB. This is due to several reasons. The details of BBB dysfunction in diseases, especially clinically, is still not fully understood. Measurements of BBB leakage by dynamic contrast-enhanced MRI or blood biomarkers and BBB dysfunction, like transporter activity or neuroinflammation by PET in patients, are already available [307], but only in few centers. We can expect progress in the adaptation and use of these clinical methods that would accelerate clinical studies on BBB protection in a wide range of diseases. Finally, we cannot expect that nutraceuticals will solve all of the therapeutic problems related to diseases with BBB dysfunction, but they may provide novel active agents or support existing pharmacotherapy. We are optimistic about the future of nutraceuticals as adjunct therapies in combating CNS pathologies and preserving BBB integrity.

Supplementary Materials

The following information can be downloaded at https://www.mdpi.com/article/10.3390/nu17050766/s1, Table S1: LogP and topological polar surface area (TPSA) values for natural products reported as BBB protective agents. Refs. [308,309,310,311].

Author Contributions

Conceptualization: M.A.D. and F.R.W.; analysis and data collection: A.E.K., N.K., A.H., M.A.D. and F.R.W., figures: N.K., A.R.S.-M. and F.R.W.; funding acquisition: A.H. and M.A.D.; project administration: A.H., M.A.D. and F.R.W.; resources: A.H. and M.A.D.; supervision: A.H., M.A.D. and F.R.W.; writing—original draft: A.E.K., N.K., A.R.S.-M., A.H., M.A.D. and F.R.W.; writing—review and editing: A.E.K., A.H., M.A.D. and F.R.W. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by grant 2022-1.2.6-TÉT-IPARI-TR-2022-00024 (D.M.A. and A.H.) from the National Research, Development and Innovation Office of Hungary. F.R.W. was funded by the Hungarian Research Network grant number SA-111/2021.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Atanasov, A.G.; Zotchev, S.B.; Dirsch, V.M.; International Natural Product Sciences Taskforce; Supuran, C.T. Natural products in drug discovery: Advances and opportunities. Nat. Rev. Drug Discov. 2021, 20, 200–216. [Google Scholar] [CrossRef] [PubMed]
  2. Puri, V.; Nagpal, M.; Singh, I.; Singh, M.; Dhingra, G.A.; Huanbutta, K.; Dheer, D.; Sharma, A.; Sangnim, T. A Comprehensive Review on Nutraceuticals: Therapy Support and Formulation Challenges. Nutrients 2022, 14, 4637. [Google Scholar] [CrossRef] [PubMed]
  3. Iriti, M.; Vitalini, S.; Fico, G.; Faoro, F. Neuroprotective herbs and foods from different traditional medicines and diets. Molecules 2010, 15, 3517–3555. [Google Scholar] [CrossRef] [PubMed]
  4. Castrogiovanni, P.; Trovato, F.M.; Loreto, C.; Nsir, H.; Szychlinska, M.A.; Musumeci, G. Nutraceutical Supplements in the Management and Prevention of Osteoarthritis. Int. J. Mol. Sci. 2016, 17, 2042. [Google Scholar] [CrossRef] [PubMed]
  5. Golla, U. Emergence of nutraceuticals as the alternative medications for pharmaceuticals. Int. J. Complement. Altern. Med. 2018, 11, 155–158. [Google Scholar] [CrossRef]
  6. Silva, R.F.M.; Pogačnik, L. Polyphenols from Food and Natural Products: Neuroprotection and Safety. Antioxidants 2020, 9, 61. [Google Scholar] [CrossRef]
  7. Zhu, T.; Wang, L.; Wang, L.P.; Wan, Q. Therapeutic targets of neuroprotection and neurorestoration in ischemic stroke: Applications for natural compounds from medicinal herbs. Biomed. Pharmacother. 2022, 148, 112719. [Google Scholar] [CrossRef]
  8. Banji, O.J.F.; Banji, D.; Makeen, H.A.; Alqahtani, S.S.; Alshahrani, S. Neuroinflammation: The Role of Anthocyanins as Neuroprotectants. Curr. Neuropharmacol. 2022, 20, 2156–2174. [Google Scholar] [CrossRef]
  9. Marino, P.; Pepe, G.; Basilicata, M.G.; Vestuto, V.; Marzocco, S.; Autore, G.; Procino, A.; Gomez-Monterrey, I.M.; Manfra, M.; Campiglia, P. Potential Role of Natural Antioxidant Products in Oncological Diseases. Antioxidants 2023, 12, 704. [Google Scholar] [CrossRef]
  10. Feng, J.; Zheng, Y.; Guo, M.; Ares, I.; Martínez, M.; Lopez-Torres, B.; Martínez-Larrañaga, M.R.; Wang, X.; Anadón, A.; Martínez, M.A. Oxidative stress, the blood-brain barrier and neurodegenerative diseases: The critical beneficial role of dietary antioxidants. Acta Pharm. Sin. B. 2023, 13, 3988–4024. [Google Scholar] [CrossRef]
  11. Campos-Bedolla, P.; Walter, F.R.; Veszelka, S.; Deli, M.A. Role of the blood-brain barrier in the nutrition of the central nervous system. Arch. Med. Res. 2014, 45, 610–638. [Google Scholar] [CrossRef] [PubMed]
  12. Sweeney, M.D.; Zhao, Z.; Montagne, A.; Nelson, A.R.; Zlokovic, B.V. Blood-Brain Barrier: From Physiology to Disease and Back. Physiol. Rev. 2019, 99, 21–78. [Google Scholar] [CrossRef] [PubMed]
  13. Andjelkovic, A.V.; Stamatovic, S.M.; Phillips, C.M.; Martinez-Revollar, G.; Keep, R.F. Modeling blood-brain barrier pathology in cerebrovascular disease in vitro: Current and future paradigms. Fluids Barriers CNS 2020, 17, 44. [Google Scholar] [CrossRef] [PubMed]
  14. Archie, S.R.; Al Shoyaib, A.; Cucullo, L. Blood-Brain Barrier Dysfunction in CNS Disorders and Putative Therapeutic Targets: An Overview. Pharmaceutics 2021, 13, 1779. [Google Scholar] [CrossRef]
  15. Walter, F.R.; Santa-Maria, A.R.; Mészáros, M.; Veszelka, S.; Dér, A.; Deli, M.A. Surface charge, glycocalyx, and blood-brain barrier function. Tissue Barriers 2021, 9, 1904773. [Google Scholar] [CrossRef]
  16. de Rus Jacquet, A.; Layé, S.; Calon, F. How nutrients and natural products act on the brain: Beyond pharmacology. Cell Rep. Med. 2023, 4, 101243. [Google Scholar] [CrossRef]
  17. Wager, T.T.; Hou, X.; Verhoest, P.R.; Villalobos, A. Central Nervous System Multiparameter Optimization Desirability: Application in Drug Discovery. ACS Chem. Neurosci. 2016, 7, 767–775. [Google Scholar] [CrossRef]
  18. Lakey-Beitia, J.; Kumar, D.J.; Hegde, M.L.; Rao, K.S. Carotenoids as Novel Therapeutic Molecules Against Neurodegenerative Disorders: Chemistry and Molecular Docking Analysis. Int. J. Mol. Sci. 2019, 20, 5553. [Google Scholar] [CrossRef]
  19. Gao, L.; Sun, W.; Zhang, L.; Liang, C.; Zhang, D. Caffeine upregulates SIRT3 expression to ameliorate astrocytes-mediated HIV-1 Tat neurotoxicity via suppression of EGR1 signaling pathway. J. Neurovirol. 2024, 30, 286–302. [Google Scholar] [CrossRef]
  20. Netsch, M.I.; Gutmann, H.; Luescher, S.; Brill, S.; Schmidlin, C.B.; Kreuter, M.H.; Drewe, J. Inhibitory activity of a green tea extract and some of its constituents on multidrug resistance-associated protein 2 functionality. Planta Med. 2005, 71, 135–141. [Google Scholar] [CrossRef]
  21. McCall, A.L.; Millington, W.R.; Wurtman, R.J. Blood-brain barrier transport of caffeine: Dose-related restriction of adenine transport. Life Sci. 1982, 31, 2709–2715. [Google Scholar] [CrossRef] [PubMed]
  22. Sugawara, M.; Mochizuki, T.; Takekuma, Y.; Miyazaki, K. Structure-affinity relationship in the interactions of human organic anion transporter 1 with caffeine, theophylline, theobromine and their metabolites. Biochim. Biophys. Acta 2005, 1714, 85–92. [Google Scholar] [CrossRef] [PubMed]
  23. Gunnink, L.K.; Busscher, B.M.; Wodarek, J.A.; Rosette, K.A.; Strohbehn, L.E.; Looyenga, B.D.; Louters, L.L. Caffeine inhibition of GLUT1 is dependent on the activation state of the transporter. Biochimie 2017, 137, 99–105. [Google Scholar] [CrossRef] [PubMed]
  24. Nakagawa, S.; Deli, M.A.; Kawaguchi, H.; Shimizudani, T.; Shimono, T.; Kittel, A.; Tanaka, K.; Niwa, M. A new blood-brain barrier model using primary rat brain endothelial cells, pericytes and astrocytes. Neurochem. Int. 2009, 54, 253–263. [Google Scholar] [CrossRef] [PubMed]
  25. Lardeau, A.; Poquet, L. Phenolic acid metabolites derived from coffee consumption are unlikely to cross the blood-brain barrier. J. Pharm. Biomed. Anal. 2013, 76, 134–138. [Google Scholar] [CrossRef]
  26. Guo, S.Y.; Yang, G.P.; Jiang, D.J.; Wang, F.; Song, T.; Tan, X.H.; Sun, Z.Q. Protection of capsaicin against hypoxia-reoxygenation-induced apoptosis of rat hippocampal neurons. Can. J. Physiol. Pharmacol. 2008, 86, 785–792. [Google Scholar] [CrossRef]
  27. Nabekura, T.; Kamiyama, S.; Kitagawa, S. Effects of dietary chemopreventive phytochemicals on P-glycoprotein function. Biochem. Biophys. Res. Commun. 2005, 327, 866–870. [Google Scholar] [CrossRef]
  28. Okura, T.; Ibe, M.; Umegaki, K.; Shinozuka, K.; Yamada, S. Effects of dietary ingredients on function and expression of P-glycoprotein in human intestinal epithelial cells. Biol. Pharm. Bull. 2010, 33, 255–259. [Google Scholar] [CrossRef]
  29. Rollyson, W.D.; Stover, C.A.; Brown, K.C.; Perry, H.E.; Stevenson, C.D.; McNees, C.A.; Ball, J.G.; Valentovic, M.A.; Dasgupta, P. Bioavailability of capsaicin and its implications for drug delivery. J. Control. Release 2014, 196, 96–105. [Google Scholar] [CrossRef]
  30. Wu, F.; Liu, R.; Shen, X.; Xu, H.; Sheng, L. Study on the interaction and antioxidant activity of theophylline and theobromine with SOD by spectra and calculation. Spectrochim. Acta A Mol. Biomol. Spectrosc. 2019, 215, 354–362. [Google Scholar] [CrossRef]
  31. Kupsáková, I.; Rybár, A.; Docolomanský, P.; Drobná, Z.; Stein, U.; Walther, W.; Barancík, M.; Breier, A. Reversal of P-glycoprotein mediated vincristine resistance of L1210/VCR cells by analogues of pentoxifylline. A QSAR study. Eur. J. Pharm. Sci. 2004, 21, 283–293. [Google Scholar] [CrossRef] [PubMed]
  32. Tsai, T.H.; Liu, M.C. Determination of unbound theophylline in rat blood and brain by microdialysis and liquid chromatography. J. Chromatogr. A 2004, 1032, 97–101. [Google Scholar] [CrossRef] [PubMed]
  33. Dani, C.; Pasquali, M.A.; Oliveira, M.R.; Umezu, F.M.; Salvador, M.; Henriques, J.A.; Moreira, J.C. Protective effects of purple grape juice on carbon tetrachloride-induced oxidative stress in brains of adult Wistar rats. J. Med. Food. 2008, 11, 55–61. [Google Scholar] [CrossRef] [PubMed]
  34. Dreiseitel, A.; Oosterhuis, B.; Vukman, K.V.; Schreier, P.; Oehme, A.; Locher, S.; Hajak, G.; Sand, P.G. Berry anthocyanins and anthocyanidins exhibit distinct affinities for the efflux transporters BCRP and MDR1. Br. J. Pharmacol. 2009, 158, 1942–1950. [Google Scholar] [CrossRef]
  35. He, L.; Zhao, C.; Yan, M.; Zhang, L.Y.; Xia, Y.Z. Inhibition of P-glycoprotein function by procyanidine on blood-brain barrier. Phytother. Res. 2009, 23, 933–937. [Google Scholar] [CrossRef]
  36. Faria, A.; Pestana, D.; Teixeira, D.; Azevedo, J.; De Freitas, V.; Mateus, N.; Calhau, C. Flavonoid transport across RBE4 cells: A blood-brain barrier model. Cell Mol. Biol. Lett. 2010, 15, 234–241. [Google Scholar] [CrossRef]
  37. Youdim, K.A.; Dobbie, M.S.; Kuhnle, G.; Proteggente, A.R.; Abbott, N.J.; Rice-Evans, C. Interaction between flavonoids and the blood-brain barrier: In vitro studies. J. Neurochem. 2003, 85, 180–192. [Google Scholar] [CrossRef]
  38. Xu, Y.; Ke, H.; Li, Y.; Xie, L.; Su, H.; Xie, J.; Mo, J.; Chen, W. Malvidin-3-O-Glucoside from Blueberry Ameliorates Nonalcoholic Fatty Liver Disease by Regulating Transcription Factor EB-Mediated Lysosomal Function and Activating the Nrf2/ARE Signaling Pathway. J. Agric. Food Chem. 2021, 69, 4663–4673. [Google Scholar] [CrossRef]
  39. Oliveira, H.; Roma-Rodrigues, C.; Santos, A.; Veigas, B.; Brás, N.; Faria, A.; Calhau, C.; de Freitas, V.; Baptista, P.V.; Mateus, N.; et al. GLUT1 and GLUT3 involvement in anthocyanin gastric transport- Nanobased targeted approach. Sci. Rep. 2019, 9, 789. [Google Scholar] [CrossRef]
  40. Faria, A.; Meireles, M.; Fernandes, I.; Santos-Buelga, C.; Gonzalez-Manzano, S.; Dueñas, M.; de Freitas, V.; Mateus, N.; Calhau, C. Flavonoid metabolites transport across a human BBB model. Food Chem. 2014, 149, 190–196. [Google Scholar] [CrossRef]
  41. Iizuka, M.; Ayaori, M.; Uto-Kondo, H.; Yakushiji, E.; Takiguchi, S.; Nakaya, K.; Hisada, T.; Sasaki, M.; Komatsu, T.; Yogo, M.; et al. Astaxanthin enhances ATP-binding cassette transporter A1/G1 expressions and cholesterol efflux from macrophages. J. Nutr. Sci. Vitaminol. 2012, 58, 96–104. [Google Scholar] [CrossRef] [PubMed]
  42. Teng, Y.N.; Sheu, M.J.; Hsieh, Y.W.; Wang, R.Y.; Chiang, Y.C.; Hung, C.C. β-carotene reverses multidrug resistant cancer cells by selectively modulating human P-glycoprotein function. Phytomedicine 2016, 23, 316–323. [Google Scholar] [CrossRef] [PubMed]
  43. Liu, X.; Wang, N.; Gao, Z. β-Carotene regulates glucose transport and insulin resistance in gestational diabetes mellitus by increasing the expression of SHBG. Clin. Exp. Pharmacol. Physiol. 2022, 49, 1307–1318. [Google Scholar] [CrossRef] [PubMed]
  44. Du, H.F.; Wu, J.W.; Zhu, Y.S.; Hua, Z.H.; Jin, S.Z.; Ji, J.C.; Wang, C.S.; Qian, G.Y.; Jin, X.D.; Ding, H.M. Fucoxanthin Induces Ferroptosis in Cancer Cells via Downregulation of the Nrf2/HO-1/GPX4 Pathway. Molecules 2024, 29, 2832. [Google Scholar] [CrossRef] [PubMed]
  45. Chandra, F.; Tania, T.F.; Nurcahyanti, A.D.R. Bixin and Fuxoxanthin Alone and in Combination with Cisplatin Regulate ABCC1 and ABCC2 Transcription in A549 Lung Cancer Cells. J. Pharm. Bioallied Sci. 2023, 15, 15–20. [Google Scholar] [CrossRef]
  46. Molnár, J.; Gyémánt, N.; Mucsi, I.; Molnár, A.; Szabó, M.; Körtvélyesi, T.; Varga, A.; Molnár, P.; Tóth, G. Modulation of multidrug resistance and apoptosis of cancer cells by selected carotenoids. In Vivo 2004, 18, 237–244. [Google Scholar]
  47. Sato, Y.; Kondo, Y.; Sumi, M.; Takekuma, Y.; Sugawara, M. Intracellular uptake mechanism of lutein in retinal pigment epithelial cells. J. Pharm. Pharm. Sci. 2013, 16, 494–501. [Google Scholar] [CrossRef]
  48. Palozza, P.; Simone, R.; Catalano, A.; Parrone, N.; Monego, G.; Ranelletti, F.O. Lycopene regulation of cholesterol synthesis and efflux in human macrophages. J. Nutr. Biochem. 2011, 22, 971–978. [Google Scholar] [CrossRef]
  49. Banji, D.; Banji, O.J.; Dasaroju, S.; Annamalai, A.R. Piperine and curcumin exhibit synergism in attenuating D-galactose induced senescence in rats. Eur. J. Pharmacol. 2013, 703, 91–99. [Google Scholar] [CrossRef]
  50. Chearwae, W.; Shukla, S.; Limtrakul, P.; Ambudkar, S.V. Modulation of the function of the multidrug resistance-linked ATP-binding cassette transporter ABCG2 by the cancer chemopreventive agent curcumin. Mol. Cancer Ther. 2006, 5, 1995–2006. [Google Scholar] [CrossRef]
  51. Shukla, S.; Zaher, H.; Hartz, A.; Bauer, B.; Ware, J.A.; Ambudkar, S.V. Curcumin inhibits the activity of ABCG2/BCRP1, a multidrug resistance-linked ABC drug transporter in mice. Pharm. Res. 2009, 26, 480–487. [Google Scholar] [CrossRef] [PubMed]
  52. Gunnink, L.K.; Alabi, O.D.; Kuiper, B.D.; Gunnink, S.M.; Schuiteman, S.J.; Strohbehn, L.E.; Hamilton, K.E.; Wrobel, K.E.; Louters, L.L. Curcumin directly inhibits the transport activity of GLUT1. Biochimie 2016, 125, 179–185. [Google Scholar] [CrossRef] [PubMed]
  53. Askarizadeh, A.; Barreto, G.E.; Henney, N.C.; Majeed, M.; Sahebkar, A. Neuroprotection by curcumin: A review on brain delivery strategies. Int. J. Pharm. 2020, 585, 119476. [Google Scholar] [CrossRef]
  54. Han, J.Y.; Ahn, S.Y.; Kim, C.S.; Yoo, S.K.; Kim, S.K.; Kim, H.C.; Hong, J.T.; Oh, K.W. Protection of apigenin against kainate-induced excitotoxicity by anti-oxidative effects. Biol. Pharm. Bull. 2012, 35, 1440–1446. [Google Scholar] [CrossRef]
  55. Zhao, L.; Wang, J.L.; Wang, Y.R.; Fa, X.Z. Apigenin attenuates copper-mediated β-amyloid neurotoxicity through antioxidation, mitochondrion protection and MAPK signal inactivation in an AD cell model. Brain Res. 2013, 1492, 33–45. [Google Scholar] [CrossRef]
  56. Yang, Y.; Bai, L.; Li, X.; Xiong, J.; Xu, P.; Guo, C.; Xue, M. Transport of active flavonoids, based on cytotoxicity and lipophilicity: An evaluation using the blood-brain barrier cell and Caco-2 cell models. Toxicol. In Vitro 2014, 28, 388–396. [Google Scholar] [CrossRef]
  57. Ren, K.; Jiang, T.; Zhou, H.F.; Liang, Y.; Zhao, G.J. Apigenin Retards Atherogenesis by Promoting ABCA1-Mediated Cholesterol Efflux and Suppressing Inflammation. Cell Physiol. Biochem. 2018, 47, 2170–2184. [Google Scholar] [CrossRef]
  58. Melstrom, L.G.; Salabat, M.R.; Ding, X.Z.; Milam, B.M.; Strouch, M.; Pelling, J.C.; Bentrem, D.J. Apigenin inhibits the GLUT-1 glucose transporter and the phosphoinositide 3-kinase/Akt pathway in human pancreatic cancer cells. Pancreas 2008, 37, 426–431. [Google Scholar] [CrossRef]
  59. Shimazu, R.; Anada, M.; Miyaguchi, A.; Nomi, Y.; Matsumoto, H. Evaluation of Blood-Brain Barrier Permeability of Polyphenols, Anthocyanins, and Their Metabolites. J. Agric. Food Chem. 2021, 69, 11676–11686. [Google Scholar] [CrossRef]
  60. Werba, J.P.; Misaka, S.; Giroli, M.G.; Shimomura, K.; Amato, M.; Simonelli, N.; Vigo, L.; Tremoli, E. Update of green tea interactions with cardiovascular drugs and putative mechanisms. J. Food Drug Anal. 2018, 26, S72–S77. [Google Scholar] [CrossRef]
  61. Almajano, M.P.; Vila, I.; Gines, S. Neuroprotective effects of white tea against oxidative stress-induced toxicity in striatal cells. Neurotox. Res. 2011, 20, 372–378. [Google Scholar] [CrossRef] [PubMed]
  62. Vaidyanathan, J.B.; Walle, T. Transport and metabolism of the tea flavonoid (-)-epicatechin by the human intestinal cell line Caco-2. Pharm. Res. 2001, 18, 1420–1425. [Google Scholar] [CrossRef] [PubMed]
  63. Kikuchi, T.; Hayashi, A.; Ikeda, N.; Morita, O.; Tasaki, J. Multidrug resistance-associated protein 2 (MRP2) is an efflux transporter of EGCG and its metabolites in the human small intestine. J. Nutr. Biochem. 2022, 107, 109071. [Google Scholar] [CrossRef] [PubMed]
  64. Annaba, F.; Kumar, P.; Dudeja, A.K.; Saksena, S.; Gill, R.K.; Alrefai, W.A. Green tea catechin EGCG inhibits ileal apical sodium bile acid transporter ASBT. Am. J. Physiol. Gastrointest. Liver Physiol. 2010, 298, G467–G473. [Google Scholar] [CrossRef] [PubMed]
  65. He, X.L.; Wang, Y.H.; Bi, M.G.; Du, G.H. Chrysin improves cognitive deficits and brain damage induced by chronic cerebral hypoperfusion in rats. Eur. J. Pharmacol. 2012, 680, 41–48. [Google Scholar] [CrossRef] [PubMed]
  66. Li, X.; Li, X.; Deng, L. Chrysin reduces inflammation and oxidative stress and improves ovarian function in D-gal-induced premature ovarian failure. Bioengineered 2022, 13, 8291–8301. [Google Scholar] [CrossRef]
  67. Mitsunaga, Y.; Takanaga, H.; Matsuo, H.; Naito, M.; Tsuruo, T.; Ohtani, H.; Sawada, Y. Effect of bioflavonoids on vincristine transport across blood-brain barrier. Eur. J. Pharmacol. 2000, 395, 193–201. [Google Scholar] [CrossRef]
  68. Kaur, M.; Badhan, R.K. Phytochemical mediated-modulation of the expression and transporter function of breast cancer resistance protein at the blood-brain barrier: An in-vitro study. Brain Res. 2017, 1654 Pt A, 9–23. [Google Scholar] [CrossRef]
  69. Mohos, V.; Fliszár-Nyúl, E.; Ungvári, O.; Bakos, É.; Kuffa, K.; Bencsik, T.; Zsidó, B.Z.; Hetényi, C.; Telbisz, Á.; Özvegy-Laczka, C.; et al. Effects of Chrysin and Its Major Conjugated Metabolites Chrysin-7-Sulfate and Chrysin-7-Glucuronide on Cytochrome P450 Enzymes and on OATP, P-gp, BCRP, and MRP2 Transporters. Drug Metab. Dispos. 2020, 48, 1064–1073. [Google Scholar] [CrossRef]
  70. Yu, Z.; Yang, L.; Deng, S.; Liang, M. Daidzein ameliorates LPS-induced hepatocyte injury by inhibiting inflammation and oxidative stress. Eur. J. Pharmacol. 2020, 885, 173399. [Google Scholar] [CrossRef]
  71. Limtrakul, P.; Khantamat, O.; Pintha, K. Inhibition of P-glycoprotein function and expression by kaempferol and quercetin. J. Chemother. 2005, 17, 86–95. [Google Scholar] [CrossRef] [PubMed]
  72. Merino, G.; Perez, M.; Real, R.; Egido, E.; Prieto, J.G.; Alvarez, A.I. In vivo inhibition of BCRP/ABCG2 mediated transport of nitrofurantoin by the isoflavones genistein and daidzein: A comparative study in Bcrp1 (−/−) mice. Pharm. Res. 2010, 27, 2098–2105. [Google Scholar] [CrossRef] [PubMed]
  73. Cheong, S.H.; Furuhashi, K.; Ito, K.; Nagaoka, M.; Yonezawa, T.; Miura, Y.; Yagasaki, K. Daidzein promotes glucose uptake through glucose transporter 4 translocation to plasma membrane in L6 myocytes and improves glucose homeostasis in Type 2 diabetic model mice. J. Nutr. Biochem. 2014, 25, 136–143. [Google Scholar] [CrossRef] [PubMed]
  74. Li, D.; Liu, X.; Pi, W.; Zhang, Y.; Yu, L.; Xu, C.; Sun, Z.; Jiang, J. Fisetin Attenuates Doxorubicin-Induced Cardiomyopathy In Vivo and In Vitro by Inhibiting Ferroptosis Through SIRT1/Nrf2 Signaling Pathway Activation. Front. Pharmacol. 2022, 12, 808480. [Google Scholar] [CrossRef] [PubMed]
  75. Yoshimura, S.; Kawano, K.; Matsumura, R.; Sugihara, N.; Furuno, K. Inhibitory effect of flavonoids on the efflux of N-acetyl 5-aminosalicylic acid intracellularly formed in Caco-2 cells. J. Biomed. Biotechnol. 2009, 2009, 467489. [Google Scholar] [CrossRef]
  76. Watanabe, M.; Hisatake, M.; Fujimori, K. Fisetin Suppresses Lipid Accumulation in Mouse Adipocytic 3T3-L1 Cells by Repressing GLUT4-Mediated Glucose Uptake through Inhibition of mTOR-C/EBPα Signaling. J. Agric. Food Chem. 2015, 63, 4979–4987. [Google Scholar] [CrossRef]
  77. El-Far, Y.M.; Khodir, A.E.; Emarah, Z.A.; Ebrahim, M.A.; Al-Gayyar, M.M.H. Chemopreventive and hepatoprotective effects of genistein via inhibition of oxidative stress and the versican/PDGF/PKC signaling pathway in experimentally induced hepatocellular carcinoma in rats by thioacetamide. Redox Rep. 2022, 27, 9–20. [Google Scholar] [CrossRef]
  78. Kodaira, H.; Kusuhara, H.; Fujita, T.; Ushiki, J.; Fuse, E.; Sugiyama, Y. Quantitative evaluation of the impact of active efflux by p-glycoprotein and breast cancer resistance protein at the blood-brain barrier on the predictability of the unbound concentrations of drugs in the brain using cerebrospinal fluid concentration as a surrogate. J. Pharmacol. Exp. Ther. 2011, 339, 935–944. [Google Scholar] [CrossRef]
  79. Toyohira, Y.; Ueno, S.; Tsutsui, M.; Itoh, H.; Sakai, N.; Saito, N.; Takahashi, K.; Yanagihara, N. Stimulatory effects of the soy phytoestrogen genistein on noradrenaline transporter and serotonin transporter activity. Mol. Nutr. Food Res. 2010, 54, 516–524. [Google Scholar] [CrossRef]
  80. Vera, J.C.; Reyes, A.M.; Cárcamo, J.G.; Velásquez, F.V.; Rivas, C.I.; Zhang, R.H.; Strobel, P.; Iribarren, R.; Scher, H.I.; Slebe, J.C.; et al. Genistein is a natural inhibitor of hexose and dehydroascorbic acid transport through the glucose transporter, GLUT1. J. Biol. Chem. 1996, 271, 8719–8724. [Google Scholar] [CrossRef]
  81. Choi, E.J.; Ahn, W.S. Neuroprotective effects of chronic hesperetin administration in mice. Arch. Pharm. Res. 2008, 31, 1457–1462. [Google Scholar] [CrossRef] [PubMed]
  82. Li, J.; Wang, T.; Liu, P.; Yang, F.; Wang, X.; Zheng, W.; Sun, W. Hesperetin ameliorates hepatic oxidative stress and inflammation via the PI3K/AKT-Nrf2-ARE pathway in oleic acid-induced HepG2 cells and a rat model of high-fat diet-induced NAFLD. Food Funct. 2021, 12, 3898–3918. [Google Scholar] [CrossRef] [PubMed]
  83. Morris, M.E.; Zhang, S. Flavonoid-drug interactions: Effects of flavonoids on ABC transporters. Life Sci. 2006, 78, 2116–2130. [Google Scholar] [CrossRef]
  84. Ikemura, M.; Sasaki, Y.; Giddings, J.C.; Yamamoto, J. Preventive effects of hesperidin, glucosyl hesperidin and naringin on hypertension and cerebral thrombosis in stroke-prone spontaneously hypertensive rats. Phytother. Res. 2012, 26, 1272–1277. [Google Scholar] [CrossRef]
  85. Lee, B.K.; Hyun, S.W.; Jung, Y.S. Yuzu and Hesperidin Ameliorate Blood-Brain Barrier Disruption during Hypoxia via Antioxidant Activity. Antioxidants 2020, 9, 843. [Google Scholar] [CrossRef]
  86. El-Readi, M.Z.; Hamdan, D.; Farrag, N.; El-Shazly, A.; Wink, M. Inhibition of P-glycoprotein activity by limonin and other secondary metabolites from Citrus species in human colon and leukaemia cell lines. Eur. J. Pharmacol. 2010, 626, 139–145. [Google Scholar] [CrossRef]
  87. Lei, Y.; Chen, J.; Zhang, W.; Fu, W.; Wu, G.; Wei, H.; Wang, Q.; Ruan, J. In vivo investigation on the potential of galangin, kaempferol and myricetin for protection of D-galactose-induced cognitive impairment. Food Chem. 2012, 135, 2702–2707. [Google Scholar] [CrossRef]
  88. Yuan, Y.; Zhai, Y.; Chen, J.; Xu, X.; Wang, H. Kaempferol Ameliorates Oxygen-Glucose Deprivation/Reoxygenation-Induced Neuronal Ferroptosis by Activating Nrf2/SLC7A11/GPX4 Axis. Biomolecules 2021, 11, 923. [Google Scholar] [CrossRef]
  89. Kitakaze, T.; Jiang, H.; Nomura, T.; Hironao, K.Y.; Yamashita, Y.; Ashida, H. Kaempferol Promotes Glucose Uptake in Myotubes through a JAK2-Dependent Pathway. J. Agric. Food Chem. 2020, 68, 13720–13729. [Google Scholar] [CrossRef]
  90. Figueira, I.; Garcia, G.; Pimpão, R.C.; Terrasso, A.P.; Costa, I.; Almeida, A.F.; Tavares, L.; Pais, T.F.; Pinto, P.; Ventura, M.R.; et al. Polyphenols journey through blood-brain barrier towards neuronal protection. Sci. Rep. 2017, 7, 11456. [Google Scholar] [CrossRef]
  91. Sharma, V.; Mishra, M.; Ghosh, S.; Tewari, R.; Basu, A.; Seth, P.; Sen, E. Modulation of interleukin-1beta mediated inflammatory response in human astrocytes by flavonoids: Implications in neuroprotection. Brain Res. Bull. 2007, 73, 55–63. [Google Scholar] [CrossRef] [PubMed]
  92. Park, H.S.; Lee, K.; Kim, S.H.; Hong, M.J.; Jeong, N.J.; Kim, M.S. Luteolin improves hypercholesterolemia and glucose intolerance through LXRα-dependent pathway in diet-induced obese mice. J. Food Biochem. 2020, 44, e13358. [Google Scholar] [CrossRef] [PubMed]
  93. Han, Z.; Batudeligen Chen, H.; Narisu Anda Xu, Y.; Xue, L. Luteolin attenuates CCl4-induced hepatic injury by inhibiting ferroptosis via SLC7A11. BMC Complement. Med. Ther. 2024, 24, 193. [Google Scholar] [CrossRef] [PubMed]
  94. Tan, K.W.; Li, Y.; Paxton, J.W.; Birch, N.P.; Scheepens, A. Identification of novel dietary phytochemicals inhibiting the efflux transporter breast cancer resistance protein (BCRP/ABCG2). Food Chem. 2013, 138, 2267–2274. [Google Scholar] [CrossRef] [PubMed]
  95. Furumiya, M.; Inoue, K.; Nishijima, C.; Yamashiro, T.; Inaoka, E.; Ohta, K.; Hayashi, Y.; Yuasa, H. Noncompetitive inhibition of proton-coupled folate transporter by myricetin. Drug Metab. Pharmacokinet. 2014, 29, 312–316. [Google Scholar] [CrossRef]
  96. Li, Y.; He, B.; Zhang, C.; He, Y.; Xia, T.; Zeng, C. Naringenin Attenuates Isoprenaline-Induced Cardiac Hypertrophy by Suppressing Oxidative Stress through the AMPK/NOX2/MAPK Signaling Pathway. Nutrients 2023, 15, 1340. [Google Scholar] [CrossRef]
  97. Ali, M.M.; Agha, F.G.; El-Sammad, N.M.; Hassan, S.K. Modulation of anticancer drug-induced P-glycoprotein expression by naringin. Z. Naturforsch. C J. Biosci. 2009, 64, 109–116. [Google Scholar] [CrossRef]
  98. Ott, M.; Huls, M.; Cornelius, M.G.; Fricker, G. St. John’s Wort constituents modulate P-glycoprotein transport activity at the blood-brain barrier. Pharm. Res. 2010, 27, 811–822. [Google Scholar] [CrossRef]
  99. Lin, X.; Zhao, X.; Chen, Q.; Wang, X.; Wu, Y.; Zhao, H. Quercetin ameliorates ferroptosis of rat cardiomyocytes via activation of the SIRT1/p53/SLC7A11 signaling pathway to alleviate sepsis-induced cardiomyopathy. Int. J. Mol. Med. 2023, 52, 116. [Google Scholar] [CrossRef]
  100. Kumar, K.K.; Priyanka, L.; Gnananath, K.; Babu, P.R.; Sujatha, S. Pharmacokinetic drug interactions between apigenin, rutin and paclitaxel mediated by P-glycoprotein in rats. Eur. J. Drug Metab. Pharmacokinet. 2015, 40, 267–276. [Google Scholar] [CrossRef]
  101. Ogura, J.; Koizumi, T.; Segawa, M.; Yabe, K.; Kuwayama, K.; Sasaki, S.; Kaneko, C.; Tsujimoto, T.; Kobayashi, M.; Yamaguchi, H.; et al. Quercetin-3-rhamnoglucoside (rutin) stimulates transport of organic anion compounds mediated by organic anion transporting polypeptide 2B1. Biopharm. Drug Dispos. 2014, 35, 173–182. [Google Scholar] [CrossRef] [PubMed]
  102. Hsu, C.Y.; Shih, H.Y.; Chia, Y.C.; Lee, C.H.; Ashida, H.; Lai, Y.K.; Weng, C.F. Rutin potentiates insulin receptor kinase to enhance insulin-dependent glucose transporter 4 translocation. Mol. Nutr. Food Res. 2014, 58, 1168–1176. [Google Scholar] [CrossRef] [PubMed]
  103. Tsai, M.J.; Liao, J.F.; Lin, D.Y.; Huang, M.C.; Liou, D.Y.; Yang, H.C.; Lee, H.J.; Chen, Y.T.; Chi, C.W.; Huang, W.C.; et al. Silymarin protects spinal cord and cortical cells against oxidative stress and lipopolysaccharide stimulation. Neurochem. Int. 2010, 57, 867–875. [Google Scholar] [CrossRef] [PubMed]
  104. Sun, P.; Huang, R.; Qin, Z.; Liu, F. Influence of Tangeretin on the Exponential Regression of Inflammation and Oxidative Stress in Streptozotocin-Induced Diabetic Nephropathy. Appl. Biochem. Biotechnol. 2022, 194, 3914–3929. [Google Scholar] [CrossRef] [PubMed]
  105. Peng, B.; Hu, J.; Sun, Y.; Huang, Y.; Peng, Q.; Zhao, W.; Xu, W.; Zhu, L. Tangeretin alleviates inflammation and oxidative response induced by spinal cord injury by activating the Sesn2/Keap1/Nrf2 pathway. Phytother. Res. 2024, 38, 4555–4569. [Google Scholar] [CrossRef]
  106. Honda, Y.; Ushigome, F.; Koyabu, N.; Morimoto, S.; Shoyama, Y.; Uchiumi, T.; Kuwano, M.; Ohtani, H.; Sawada, Y. Effects of grapefruit juice and orange juice components on P-glycoprotein- and MRP2-mediated drug efflux. Br. J. Pharmacol. 2004, 143, 856–864. [Google Scholar] [CrossRef]
  107. Satsu, H.; Shibata, R.; Suzuki, H.; Kimura, S.; Shimizu, M. Inhibitory Effect of Tangeretin and Cardamonin on Human Intestinal SGLT1 Activity In Vitro and Blood Glucose Levels in Mice In Vivo. Nutrients 2021, 13, 3382. [Google Scholar] [CrossRef]
  108. Datla, K.P.; Christidou, M.; Widmer, W.W.; Rooprai, H.K.; Dexter, D.T. Tissue distribution and neuroprotective effects of citrus flavonoid tangeretin in a rat model of Parkinson’s disease. Neuroreport 2001, 12, 3871–3875. [Google Scholar] [CrossRef]
  109. Bansod, S.; Chilvery, S.; Saifi, M.A.; Das, T.J.; Tag, H.; Godugu, C. Borneol protects against cerulein-induced oxidative stress and inflammation in acute pancreatitis mice model. Environ. Toxicol. 2021, 36, 530–539. [Google Scholar] [CrossRef]
  110. Fan, X.; Chai, L.; Zhang, H.; Wang, Y.; Zhang, B.; Gao, X. Borneol Depresses P-Glycoprotein Function by a NF-κB Signaling Mediated Mechanism in a Blood Brain Barrier in Vitro Model. Int. J. Mol. Sci. 2015, 16, 27576–27588. [Google Scholar] [CrossRef]
  111. Chen, N.; Wen, J.; Wang, Z.; Wang, J. Multiple regulation and targeting effects of borneol in the neurovascular unit in neurodegenerative diseases. Basic Clin. Pharmacol. Toxicol. 2022, 130, 5–19. [Google Scholar] [CrossRef] [PubMed]
  112. Li, W.R.; Chen, R.Y.; Yang, L.; Huang, T.L.; Xu, Q.W.; Mi, S.Q.; Wang, N.S. Pharmacokinetics of natural borneol after oral administration in mice brain and its effect on excitation ratio. Eur. J. Drug Metab. Pharmacokinet. 2012, 37, 39–44. [Google Scholar] [CrossRef] [PubMed]
  113. Abbasloo, E.; Khaksari, M.; Sanjari, M.; Kobeissy, F.; Thomas, T.C. Carvacrol decreases blood-brain barrier permeability post-diffuse traumatic brain injury in rats. Sci. Rep. 2023, 13, 14546. [Google Scholar] [CrossRef] [PubMed]
  114. Dos Santos Barbosa, C.R.; Scherf, J.R.; de Freitas, T.S.; de Menezes, I.R.A.; Pereira, R.L.S.; Dos Santos, J.F.S.; de Jesus, S.S.P.; Lopes, T.P.; de Sousa Silveira, Z.; de Morais Oliveira-Tintino, C.D.; et al. Effect of Carvacrol and Thymol on NorA efflux pump inhibition in multidrug-resistant (MDR) Staphylococcus aureus strains. J. Bioenerg. Biomembr. 2021, 53, 489–498. [Google Scholar] [CrossRef]
  115. Li, G.; Li, Y.; Xiao, B.; Cui, D.; Lin, Y.; Zeng, J.; Li, J.; Cao, M.J.; Liu, J. Antioxidant Activity of Docosahexaenoic Acid (DHA) and Its Regulatory Roles in Mitochondria. J. Agric. Food Chem. 2021, 69, 1647–1655. [Google Scholar] [CrossRef]
  116. Kuan, C.Y.; Walker, T.H.; Luo, P.G.; Chen, C.F. Long-chain polyunsaturated fatty acids promote paclitaxel cytotoxicity via inhibition of the MDR1 gene in the human colon cancer Caco-2 cell line. J. Am. Coll. Nutr. 2011, 30, 265–273. [Google Scholar] [CrossRef]
  117. Nguyen, L.N.; Ma, D.; Shui, G.; Wong, P.; Cazenave-Gassiot, A.; Zhang, X.; Wenk, M.R.; Goh, E.L.; Silver, D.L. Mfsd2a is a transporter for the essential omega-3 fatty acid docosahexaenoic acid. Nature 2014, 509, 503–506. [Google Scholar] [CrossRef]
  118. El Mahdy, R.N.; Nader, M.A.; Helal, M.G.; Abu-Risha, S.E.; Abdelmageed, M.E. Eicosapentaenoic acid mitigates ulcerative colitis-induced by acetic acid through modulation of NF-κB and TGF-β/ EGFR signaling pathways. Life Sci. 2023, 327, 121820. [Google Scholar] [CrossRef]
  119. Xiao, B.; Li, Y.; Lin, Y.; Lin, J.; Zhang, L.; Wu, D.; Zeng, J.; Li, J.; Liu, J.W.; Li, G. Eicosapentaenoic acid (EPA) exhibits antioxidant activity via mitochondrial modulation. Food Chem. 2022, 373 Pt A, 131389. [Google Scholar] [CrossRef]
  120. Kato, Y.; Ohsugi, K.; Fukuno, Y.; Iwatsuki, K.; Harada, Y.; Miyaji, T. Vesicular nucleotide transporter is a molecular target of eicosapentaenoic acid for neuropathic and inflammatory pain treatment. Proc. Natl. Acad. Sci. USA 2022, 119, e2122158119. [Google Scholar] [CrossRef]
  121. Edmond, J. Essential polyunsaturated fatty acids and the barrier to the brain: The components of a model for transport. J. Mol. Neurosci. 2001, 16, 181–193; discussion 215–221. [Google Scholar] [CrossRef] [PubMed]
  122. Qi, B.; Zheng, Y.; Gao, W.; Qi, Z.; Gong, Y.; Liu, Y.; Wang, Y.; Cheng, X.; Ning, M.; Lang, Y.; et al. Alpha-lipoic acid impedes myocardial ischemia-reperfusion injury, myocardial apoptosis, and oxidative stress by regulating HMGB1 expression. Eur. J. Pharmacol. 2022, 933, 175295. [Google Scholar] [CrossRef] [PubMed]
  123. Skibska, B.; Kochan, E.; Stanczak, A.; Lipert, A.; Skibska, A. Antioxidant and Anti-inflammatory Effects of α-Lipoic Acid on Lipopolysaccharide-induced Oxidative Stress in Rat Kidney. Arch. Immunol. Ther. Exp. 2023, 71, 16. [Google Scholar] [CrossRef] [PubMed]
  124. Cheng, L.C.; Su, K.H.; Kou, Y.R.; Shyue, S.K.; Ching, L.C.; Yu, Y.B.; Wu, Y.L.; Pan, C.C.; Lee, T.S. α-Lipoic acid ameliorates foam cell formation via liver X receptor α-dependent upregulation of ATP-binding cassette transporters A1 and G1. Free Radic. Biol. Med. 2011, 50, 47–54. [Google Scholar] [CrossRef] [PubMed]
  125. Zehnpfennig, B.; Wiriyasermkul, P.; Carlson, D.A.; Quick, M. Interaction of α-Lipoic Acid with the Human Na+/Multivitamin Transporter (hSMVT). J. Biol. Chem. 2015, 290, 16372–16382. [Google Scholar] [CrossRef]
  126. Spector, R. Fatty acid transport through the blood-brain barrier. J. Neurochem. 1988, 50, 639–643. [Google Scholar] [CrossRef]
  127. Sajja, R.K.; Kaisar, M.A.; Vijay, V.; Desai, V.G.; Prasad, S.; Cucullo, L. In Vitro Modulation of Redox and Metabolism Interplay at the Brain Vascular Endothelium: Genomic and Proteomic Profiles of Sulforaphane Activity. Sci. Rep. 2018, 8, 12708. [Google Scholar] [CrossRef]
  128. Jazwa, A.; Rojo, A.I.; Innamorato, N.G.; Hesse, M.; Fernández-Ruiz, J.; Cuadrado, A. Pharmacological targeting of the transcription factor Nrf2 at the basal ganglia provides disease modifying therapy for experimental parkinsonism. Antioxid. Redox Signal. 2011, 14, 2347–2360. [Google Scholar] [CrossRef]
  129. He, J.; He, Z.; Wang, H.; Zhang, C.; Pei, T.; Yan, S.; Yan, Y.; Wang, F.; Chen, Y.; Yuan, N.; et al. Caffeic acid alleviates skeletal muscle atrophy in 5/6 nephrectomy rats through the TLR4/MYD88/NF-kB pathway. Biomed. Pharmacother. 2024, 174, 116556. [Google Scholar] [CrossRef]
  130. Wortelboer, H.M.; Usta, M.; van Zanden, J.J.; van Bladeren, P.J.; Rietjens, I.M.; Cnubben, N.H. Inhibition of multidrug resistance proteins MRP1 and MRP2 by a series of alpha,beta-unsaturated carbonyl compounds. Biochem. Pharmacol. 2005, 69, 1879–1890. [Google Scholar] [CrossRef]
  131. Konishi, Y.; Hagiwara, K.; Shimizu, M. Transepithelial transport of fluorescein in Caco-2 cell monolayers and use of such transport in in vitro evaluation of phenolic acid availability. Biosci. Biotechnol. Biochem. 2002, 66, 2449–2457. [Google Scholar] [CrossRef] [PubMed]
  132. Hase, T.; Shishido, S.; Yamamoto, S.; Yamashita, R.; Nukima, H.; Taira, S.; Toyoda, T.; Abe, K.; Hamaguchi, T.; Ono, K.; et al. Rosmarinic acid suppresses Alzheimer’s disease development by reducing amyloid β aggregation by increasing monoamine secretion. Sci. Rep. 2019, 9, 8711. [Google Scholar] [CrossRef] [PubMed]
  133. Kose, S.B.E.; Kocasari, F.S. Protective effect of cinnamic acid on orthophenylphenol-induced oxidative stress in rats. Vet. Res. Forum. 2022, 13, 187–192. [Google Scholar] [CrossRef] [PubMed]
  134. Basu, S.; Jana, S.; Patel, V.B.; Patel, H. Effects of piperine, cinnamic acid and gallic acid on rosuvastatin pharmacokinetics in rats. Phytother. Res. 2013, 27, 1548–1556. [Google Scholar] [CrossRef] [PubMed]
  135. Mani, A.; Kushwaha, K.; Khurana, N.; Gupta, J. p-Coumaric acid attenuates high-fat diet-induced oxidative stress and nephropathy in diabetic rats. J. Anim. Physiol. Anim. Nutr. 2022, 106, 872–880. [Google Scholar] [CrossRef]
  136. Wang, L.; Sweet, D.H. Potential for food-drug interactions by dietary phenolic acids on human organic anion transporters 1 (SLC22A6), 3 (SLC22A8), and 4 (SLC22A11). Biochem. Pharmacol. 2012, 84, 1088–1095. [Google Scholar] [CrossRef]
  137. Sun, S.; Ruan, Y.; Yan, M.; Xu, K.; Yang, Y.; Shen, T.; Jin, Z. Ferulic Acid Alleviates Oxidative Stress-Induced Cardiomyocyte Injury by the Regulation of miR-499-5p/p21 Signal Cascade. Evid. Based Complement. Altern. Med. 2021, 2021, 1921457. [Google Scholar] [CrossRef]
  138. Ermis, A.; Aritici Colak, G.; Acikel-Elmas, M.; Arbak, S.; Kolgazi, M. Ferulic Acid Treats Gastric Ulcer via Suppressing Oxidative Stress and Inflammation. Life 2023, 13, 388. [Google Scholar] [CrossRef]
  139. Muthusamy, G.; Balupillai, A.; Ramasamy, K.; Shanmugam, M.; Gunaseelan, S.; Mary, B.; Prasad, N.R. Ferulic acid reverses ABCB1-mediated paclitaxel resistance in MDR cell lines. Eur. J. Pharmacol. 2016, 786, 194–203. [Google Scholar] [CrossRef]
  140. Poquet, L.; Clifford, M.N.; Williamson, G. Transport and metabolism of ferulic acid through the colonic epithelium. Drug Metab. Dispos. 2008, 36, 190–197. [Google Scholar] [CrossRef]
  141. Lin, Y.; Luo, T.; Weng, A.; Huang, X.; Yao, Y.; Fu, Z.; Li, Y.; Liu, A.; Li, X.; Chen, D.; et al. Gallic Acid Alleviates Gouty Arthritis by Inhibiting NLRP3 Inflammasome Activation and Pyroptosis Through Enhancing Nrf2 Signaling. Front. Immunol. 2020, 11, 580593. [Google Scholar] [CrossRef] [PubMed]
  142. Pervin, M.; Unno, K.; Nakagawa, A.; Takahashi, Y.; Iguchi, K.; Yamamoto, H.; Hoshino, M.; Hara, A.; Takagaki, A.; Nanjo, F.; et al. Blood brain barrier permeability of (-)-epigallocatechin gallate, its proliferation-enhancing activity of human neuroblastoma SH-SY5Y cells, and its preventive effect on age-related cognitive dysfunction in mice. Biochem. Biophys. Rep. 2017, 9, 180–186. [Google Scholar] [CrossRef] [PubMed]
  143. Joardar, S.; Dewanjee, S.; Bhowmick, S.; Dua, T.K.; Das, S.; Saha, A.; De Feo, V. Rosmarinic Acid Attenuates Cadmium-Induced Nephrotoxicity via Inhibition of Oxidative Stress, Apoptosis, Inflammation and Fibrosis. Int. J. Mol. Sci. 2019, 20, 2027. [Google Scholar] [CrossRef] [PubMed]
  144. Yao, Y.; Li, R.; Liu, D.; Long, L.; He, N. Rosmarinic acid alleviates acetaminophen-induced hepatotoxicity by targeting Nrf2 and NEK7-NLRP3 signaling pathway. Ecotoxicol. Environ. Saf. 2022, 241, 113773. [Google Scholar] [CrossRef] [PubMed]
  145. Luo, W.; Tao, Y.; Chen, S.; Luo, H.; Li, X.; Qu, S.; Chen, K.; Zeng, C. Rosmarinic Acid Ameliorates Pulmonary Ischemia/Reperfusion Injury by Activating the PI3K/Akt Signaling Pathway. Front. Pharmacol. 2022, 13, 860944. [Google Scholar] [CrossRef]
  146. Nyandwi, J.B.; Ko, Y.S.; Jin, H.; Yun, S.P.; Park, S.W.; Kim, H.J. Rosmarinic Acid Increases Macrophage Cholesterol Efflux through Regulation of ABCA1 and ABCG1 in Different Mechanisms. Int. J. Mol. Sci. 2021, 22, 8791. [Google Scholar] [CrossRef]
  147. Wang, L.; Sweet, D.H. Competitive inhibition of human organic anion transporters 1 (SLC22A6), 3 (SLC22A8) and 4 (SLC22A11) by major components of the medicinal herb Salvia miltiorrhiza (Danshen). Drug Metab. Pharmacokinet. 2013, 28, 220–228. [Google Scholar] [CrossRef]
  148. Zhou, Y.; Khan, H.; Hoi, M.P.M.; Cheang, W.S. Piceatannol Protects Brain Endothelial Cell Line (bEnd.3) against Lipopolysaccharide-Induced Inflammation and Oxidative Stress. Molecules 2022, 27, 1206. [Google Scholar] [CrossRef]
  149. Rubic, T.; Lorenz, R.L. Downregulated CD36 and oxLDL uptake and stimulated ABCA1/G1 and cholesterol efflux as anti-atherosclerotic mechanisms of interleukin-10. Cardiovasc. Res. 2006, 69, 527–535. [Google Scholar] [CrossRef]
  150. Ibacache-Chía, A.P.; Sierralta, J.A.; Schüller, A. The Inhibitory Effects of the Natural Stilbene Piceatannol on Lactate Transport In Vitro Mediated by Monocarboxylate Transporters. Mol. Nutr. Food Res. 2024, 68, e2400414. [Google Scholar] [CrossRef]
  151. Shah, F.A.; Kury, L.A.; Li, T.; Zeb, A.; Koh, P.O.; Liu, F.; Zhou, Q.; Hussain, I.; Khan, A.U.; Jiang, Y.; et al. Polydatin Attenuates Neuronal Loss via Reducing Neuroinflammation and Oxidative Stress in Rat MCAO Models. Front. Pharmacol. 2019, 10, 663. [Google Scholar] [CrossRef] [PubMed]
  152. Peng, Y.; Xu, J.; Zeng, Y.; Chen, L.; Xu, X.L. Polydatin attenuates atherosclerosis in apolipoprotein E-deficient mice: Role of reverse cholesterol transport. Phytomedicine 2019, 62, 152935. [Google Scholar] [CrossRef] [PubMed]
  153. Yang, J.; Wang, Y.; Cai, X.; Qu, B.; Zhang, Y.; Sun, Z.; Yan, J. Comparative pharmacokinetics and tissue distribution of polydatin, resveratrol, and emodin after oral administration of Huzhang and Huzhang-Guizhi herb-pair extracts to rats. J. Ethnopharmacol. 2024, 318 Pt B, 117010. [Google Scholar] [CrossRef]
  154. Dos Santos Lacerda, D.; Türck, P.; Gazzi de Lima-Seolin, B.; Colombo, R.; Duarte Ortiz, V.; Poletto Bonetto, J.H.; Campos-Carraro, C.; Bianchi, S.E.; Belló-Klein, A.; Linck Bassani, V.; et al. Pterostilbene reduces oxidative stress, prevents hypertrophy and preserves systolic function of right ventricle in cor pulmonale model. Br. J. Pharmacol. 2017, 174, 3302–3314. [Google Scholar] [CrossRef] [PubMed]
  155. Wang, Y.; Su, Y.; Yang, Y.; Jin, H.; Wu, M.; Wang, Q.; Sun, P.; Zhang, J.; Yang, X.; Shu, X. Increased brain uptake of pterostilbene loaded folate modified micellar delivery system. Drug Deliv. 2022, 29, 3071–3086. [Google Scholar] [CrossRef] [PubMed]
  156. Planas, J.M.; Alfaras, I.; Colom, H.; Juan, M.E. The bioavailability and distribution of trans-resveratrol are constrained by ABC transporters. Arch. Biochem. Biophys. 2012, 527, 67–73. [Google Scholar] [CrossRef]
  157. Juan, M.E.; Maijó, M.; Planas, J.M. Quantification of trans-resveratrol and its metabolites in rat plasma and tissues by HPLC. J. Pharm. Biomed. Anal. 2010, 51, 391–398. [Google Scholar] [CrossRef]
  158. Yen, G.C.; Duh, P.D.; Tsai, H.L. Antioxidant and pro-oxidant properties of ascorbic acid and gallic acid. Food Chem. 2002, 79, 307–313. [Google Scholar] [CrossRef]
  159. Wartenberg, M.; Hoffmann, E.; Schwindt, H.; Grünheck, F.; Petros, J.; Arnold, J.R.; Hescheler, J.; Sauer, H. Reactive oxygen species-linked regulation of the multidrug resistance transporter P-glycoprotein in Nox-1 overexpressing prostate tumor spheroids. FEBS Lett. 2005, 579, 4541–4549. [Google Scholar] [CrossRef]
  160. May, J.M. Vitamin C transport and its role in the central nervous system. Subcell. Biochem. 2012, 56, 85–103. [Google Scholar] [CrossRef]
  161. Xu, Y.; Wang, H.; Wang, Y.; Zheng, Y.; Sun, G. Effects of folate on arsenic toxicity in Chang human hepatocytes: Involvement of folate antioxidant properties. Toxicol. Lett. 2010, 195, 44–50. [Google Scholar] [CrossRef] [PubMed]
  162. Spector, R.; Johanson, C.E. Vitamin transport and homeostasis in mammalian brain: Focus on Vitamins B and E. J. Neurochem. 2007, 103, 425–438. [Google Scholar] [CrossRef] [PubMed]
  163. Mokhtari, Z.; Hekmatdoost, Z.; Nourian, M. Antioxidant efficacy of vitamin D. J. Parathyr. Dis. 2017, 5, 11–16. [Google Scholar]
  164. Pardridge, W.M.; Sakiyama, R.; Coty, W.A. Restricted transport of vitamin D and A derivatives through the rat blood-brain barrier. J. Neurochem. 1985, 44, 1138–1141. [Google Scholar] [CrossRef]
  165. Tang, J.; Fu, Q.; Wang, Y.; Racette, K.; Wang, D.; Liu, F. Vitamin E reverses multidrug resistance in vitro and in vivo. Cancer Lett. 2013, 336, 149–157. [Google Scholar] [CrossRef]
  166. Hunyadi, A. The mechanism(s) of action of antioxidants: From scavenging reactive oxygen/nitrogen species to redox signaling and the generation of bioactive secondary metabolites. Med. Res. Rev. 2019, 39, 2505–2533. [Google Scholar] [CrossRef]
  167. Ureña-Vacas, I.; Aznar de la Riera, M.B.; Serrano Dolores, R.; González-Burgos, E. A new frontier in neuropharmacology: Recent progress in natural products research for blood–brain barrier crossing. Curr. Res. Biotechnol. 2024, 8, 100235. [Google Scholar] [CrossRef]
  168. Munji, R.N.; Soung, A.L.; Weiner, G.A.; Sohet, F.; Semple, B.D.; Trivedi, A.; Gimlin, K.; Kotoda, M.; Korai, M.; Aydin, S.; et al. Profiling the mouse brain endothelial transcriptome in health and disease models reveals a core blood-brain barrier dysfunction module. Nat. Neurosci. 2019, 22, 1892–1902. [Google Scholar] [CrossRef]
  169. Sabbagh, M.F.; Heng, J.S.; Luo, C.; Castanon, R.G.; Nery, J.R.; Rattner, A.; Goff, L.A.; Ecker, J.R.; Nathans, J. Transcriptional and epigenomic landscapes of CNS and non-CNS vascular endothelial cells. eLife 2018, 7, e36187. [Google Scholar] [CrossRef]
  170. Vanlandewijck, M.; He, L.; Mäe, M.A.; Andrae, J.; Ando, K.; Del Gaudio, F.; Nahar, K.; Lebouvier, T.; Laviña, B.; Gouveia, L.; et al. A molecular atlas of cell types and zonation in the brain vasculature. Nature 2018, 554, 475–480. [Google Scholar] [CrossRef]
  171. Garcia, F.J.; Sun, N.; Lee, H.; Godlewski, B.; Mathys, H.; Galani, K.; Zhou, B.; Jiang, X.; Ng, A.P.; Mantero, J.; et al. Single-cell dissection of the human brain vasculature. Nature 2022, 603, 893–899. [Google Scholar] [CrossRef] [PubMed]
  172. Yang, A.C.; Vest, R.T.; Kern, F.; Lee, D.P.; Agam, M.; Maat, C.A.; Losada, P.M.; Chen, M.B.; Schaum, N.; Khoury, N.; et al. A human brain vascular atlas reveals diverse mediators of Alzheimer’s risk. Nature 2022, 603, 885–892. [Google Scholar] [CrossRef] [PubMed]
  173. Kalari, K.R.; Thompson, K.J.; Nair, A.A.; Tang, X.; Bockol, M.A.; Jhawar, N.; Swaminathan, S.K.; Lowe, V.J.; Kandimalla, K.K. BBBomics-Human Blood Brain Barrier Transcriptomics Hub. Front. Neurosci. 2016, 10, 71. [Google Scholar] [CrossRef] [PubMed]
  174. Veszelka, S.; Tóth, A.; Walter, F.R.; Tóth, A.E.; Gróf, I.; Mészáros, M.; Bocsik, A.; Hellinger, É.; Vastag, M.; Rákhely, G.; et al. Comparison of a Rat Primary Cell-Based Blood-Brain Barrier Model With Epithelial and Brain Endothelial Cell Lines: Gene Expression and Drug Transport. Front. Mol. Neurosci. 2018, 11, 166. [Google Scholar] [CrossRef] [PubMed]
  175. Santa-Maria, A.R.; Heymans, M.; Walter, F.R.; Culot, M.; Gosselet, F.; Deli, M.A.; Neuhaus, W. Transport Studies Using Blood-Brain Barrier In Vitro Models: A Critical Review and Guidelines. Handb. Exp. Pharmacol. 2022, 273, 187–204. [Google Scholar] [CrossRef]
  176. Porkoláb, G.; Mészáros, M.; Szecskó, A.; Vigh, J.P.; Walter, F.R.; Figueiredo, R.; Kálomista, I.; Hoyk, Z.; Vizsnyiczai, G.; Gróf, I.; et al. Synergistic induction of blood-brain barrier properties. Proc. Natl. Acad. Sci. USA 2024, 121, e2316006121. [Google Scholar] [CrossRef]
  177. Zhou, S.; Lim, L.Y.; Chowbay, B. Herbal modulation of P-glycoprotein. Drug Metab. Rev. 2004, 36, 57–104. [Google Scholar] [CrossRef]
  178. Youdim, K.A.; Shukitt-Hale, B.; Joseph, J.A. Flavonoids and the brain: Interactions at the blood-brain barrier and their physiological effects on the central nervous system. Free Radic. Biol. Med. 2004, 37, 1683–1693. [Google Scholar] [CrossRef]
  179. Li, Y.; Revalde, J.; Paxton, J.W. The effects of dietary and herbal phytochemicals on drug transporters. Adv. Drug Deliv. Rev. 2017, 116, 45–62. [Google Scholar] [CrossRef]
  180. Li, J.; Wang, S.; Tian, F.; Zhang, S.Q.; Jin, H. Advances in Pharmacokinetic Mechanisms of Transporter-Mediated Herb-Drug Interactions. Pharmaceuticals 2022, 15, 1126. [Google Scholar] [CrossRef]
  181. Fricker, G. Drug interactions with natural products at the blood brain barrier. Curr. Drug Metab. 2008, 9, 1019–1026. [Google Scholar] [CrossRef] [PubMed]
  182. Pekdemir, B.; Raposo, A.; Saraiva, A.; Lima, M.J.; Alsharari, Z.D.; BinMowyna, M.N.; Karav, S. Mechanisms and Potential Benefits of Neuroprotective Agents in Neurological Health. Nutrients 2024, 16, 4368. [Google Scholar] [CrossRef] [PubMed]
  183. Auti, A.; Tathode, M.; Marino, M.M.; Vitiello, A.; Ballini, A.; Miele, F.; Mazzone, V.; Ambrosino, A.; Boccellino, M. Nature’s weapons: Bioactive compounds as anti-cancer agents. AIMS Public Health 2024, 11, 747–772. [Google Scholar] [CrossRef] [PubMed]
  184. Andrew, R.; Izzo, A.A. Principles of pharmacological research of nutraceuticals. Br. J. Pharmacol. 2017, 174, 1177–1194. [Google Scholar] [CrossRef]
  185. Lund, E.K. Health benefits of seafood; is it just the fatty acids? Food Chem. 2013, 140, 413–420. [Google Scholar] [CrossRef]
  186. Kulkarni, M.; Sawant, N.; Kolapkar, A.; Huprikar, A.; Desai, N. Borneol: A Promising Monoterpenoid in Enhancing Drug Delivery Across Various Physiological Barriers. AAPS PharmSciTech. 2021, 22, 145. [Google Scholar] [CrossRef]
  187. Profaci, C.P.; Munji, R.N.; Pulido, R.S.; Daneman, R. The blood-brain barrier in health and disease: Important unanswered questions. J. Exp. Med. 2020, 217, e20190062. [Google Scholar] [CrossRef]
  188. Mukherjee, P.; Roy, S.; Ghosh, D.; Nandi, S.K. Role of animal models in biomedical research: A review. Lab. Anim. Res. 2022, 38, 18. [Google Scholar] [CrossRef]
  189. Zhang, T.; Su, J.; Guo, B.; Wang, K.; Li, X.; Liang, G. Apigenin protects blood-brain barrier and ameliorates early brain injury by inhibiting TLR4-mediated inflammatory pathway in subarachnoid hemorrhage rats. Int. Immunopharmacol. 2015, 28, 79–87. [Google Scholar] [CrossRef]
  190. Pang, Q.; Zhao, Y.; Chen, X.; Zhao, K.; Zhai, Q.; Tu, F. Apigenin Protects the Brain against Ischemia/Reperfusion Injury via Caveolin-1/VEGF In Vitro and In Vivo. Oxid. Med. Cell Longev. 2018, 2018, 7017204. [Google Scholar] [CrossRef]
  191. Iwata, S.; Imai, T.; Shimazawa, M.; Ishibashi, T.; Hayashi, M.; Hara, H.; Nakamura, S. Protective effects of the astaxanthin derivative, adonixanthin, on brain hemorrhagic injury. Brain Res. 2018, 1698, 130–138. [Google Scholar] [CrossRef] [PubMed]
  192. Zhang, X.S.; Zhang, X.; Zhang, Q.R.; Wu, Q.; Li, W.; Jiang, T.W.; Hang, C.H. Corrigendum to “Astaxanthin reduces matrix metalloproteinase-9 expression and activity in the brain after experimental subarachnoid hemorrhage in rats” [Brain Res. 1624 (2015) 113–124]. Brain Res. 2024, 1850, 149390. [Google Scholar] [CrossRef] [PubMed]
  193. Yu, B.; Zhong, F.M.; Yao, Y.; Deng, S.Q.; Xu, H.Q.; Lu, J.F.; Ruan, M.; Shen, X.C. Synergistic protection of tetramethylpyrazine phosphate and borneol on brain microvascular endothelium cells injured by hypoxia. Am. J. Transl. Res. 2019, 11, 2168–2180. [Google Scholar]
  194. Li, Y.; Ren, M.; Wang, J.; Ma, R.; Chen, H.; Xie, Q.; Li, H.; Li, J.; Wang, J. Progress in Borneol Intervention for Ischemic Stroke: A Systematic Review. Front. Pharmacol. 2021, 12, 606682. [Google Scholar] [CrossRef]
  195. Zhao, J.; Pati, S.; Redell, J.B.; Zhang, M.; Moore, A.N.; Dash, P.K. Caffeic Acid phenethyl ester protects blood-brain barrier integrity and reduces contusion volume in rodent models of traumatic brain injury. J. Neurotrauma 2012, 29, 1209–1218. [Google Scholar] [CrossRef]
  196. Mahmood, Q.; Lu, N.; Wang, X.; Du, Y.; Ghori, M.U.; Tian, B.; Yang, H.; Han, F.; Jiang, G.; Lu, Y. Targeted delivery of β-carotene potentially prevents blood-brain barrier breakdown after stroke in mice. Phytomed. Plus 2023, 3, 100426. [Google Scholar] [CrossRef]
  197. Jiang, Z.; Zhang, J.; Cai, Y.; Huang, J.; You, L. Catechin attenuates traumatic brain injury-induced blood-brain barrier damage and improves longer-term neurological outcomes in rats. Exp. Physiol. 2017, 102, 1269–1277. [Google Scholar] [CrossRef]
  198. Rashno, M.; Sarkaki, A.; Farbood, Y.; Rashno, M.; Khorsandi, L.; Naseri, M.K.G.; Dianat, M. Possible mechanisms involved in the neuroprotective effects of chrysin against mild traumatic brain injury-induced spatial cognitive decline: An in vivo study in a rat model. Brain Res. Bull. 2023, 204, 110779. [Google Scholar] [CrossRef]
  199. Li, Y.; Han, J.; Zhang, Y.; Chen, Y.; Zhang, Y. Prophylactic effect and mechanism of p-coumaric acid against hypoxic cerebral edema in mice. Respir. Physiol. Neurobiol. 2019, 260, 95–104. [Google Scholar] [CrossRef]
  200. Jiang, J.; Wang, W.; Sun, Y.J.; Hu, M.; Li, F.; Zhu, D.Y. Neuroprotective effect of curcumin on focal cerebral ischemic rats by preventing blood-brain barrier damage. Eur. J. Pharmacol. 2007, 561, 54–62. [Google Scholar] [CrossRef]
  201. Yu, L.S.; Fan, Y.Y.; Ye, G.; Li, J.; Feng, X.P.; Lin, K.; Dong, M.; Wang, Z. Curcumin alleviates brain edema by lowering AQP4 expression levels in a rat model of hypoxia-hypercapnia-induced brain damage. Exp. Ther. Med. 2016, 11, 709–716. [Google Scholar] [CrossRef] [PubMed]
  202. Liu, R.; Zhong, X.; Zeng, J.; Huang, Z.; Li, X.; Xiao, H.; Chen, Q.; Li, D. 3′-Daidzein sulfonate sodium inhibits neuronal apoptosis induced by cerebral ischemia-reperfusion. Int. J. Mol. Med. 2017, 39, 1021–1028. [Google Scholar] [CrossRef] [PubMed]
  203. Pan, H.C.; Kao, T.K.; Ou, Y.C.; Yang, D.Y.; Yen, Y.J.; Wang, C.C.; Chuang, Y.H.; Liao, S.L.; Raung, S.L.; Wu, C.W.; et al. Protective effect of docosahexaenoic acid against brain injury in ischemic rats. J. Nutr. Biochem. 2009, 20, 715–725. [Google Scholar] [CrossRef] [PubMed]
  204. Hong, S.H.; Khoutorova, L.; Bazan, N.G.; Belayev, L. Docosahexaenoic acid improves behavior and attenuates blood-brain barrier injury induced by focal cerebral ischemia in rats. Exp. Transl. Stroke Med. 2015, 7, 3. [Google Scholar] [CrossRef]
  205. Zhang, W.; Zhang, H.; Mu, H.; Zhu, W.; Jiang, X.; Hu, X.; Shi, Y.; Leak, R.K.; Dong, Q.; Chen, J.; et al. Omega-3 polyunsaturated fatty acids mitigate blood-brain barrier disruption after hypoxic-ischemic brain injury. Neurobiol. Dis. 2016, 91, 37–46. [Google Scholar] [CrossRef]
  206. Shin, J.A.; Park, H.; Choi, H.; Chang, Y.K.; Kim, J.J.; Ham, Y.R.; Na, K.R.; Lee, K.W.; Choi, D.E. ω-3 Polyunsaturated Fatty Acids Improve the Blood-Brain-Barrier Integrity in Contrast-Induced Blood-Brain-Barrier Injury in Uremic Mice. Int. J. Mol. Sci. 2023, 24, 12168. [Google Scholar] [CrossRef]
  207. Liu, X.; Wang, Z.; Wang, P.; Yu, B.; Liu, Y.; Xue, Y. Green tea polyphenols alleviate early BBB damage during experimental focal cerebral ischemia through regulating tight junctions and PKCalpha signaling. BMC Complement. Altern. Med. 2013, 13, 187. [Google Scholar] [CrossRef]
  208. Xu, S.H.; Yin, M.S.; Liu, B.; Chen, M.L.; He, G.W.; Zhou, P.P.; Cui, Y.J.; Yang, D.; Wu, Y.L. Tetramethylpyrazine-2′-O-sodium ferulate attenuates blood-brain barrier disruption and brain oedema after cerebral ischemia/reperfusion. Hum. Exp. Toxicol. 2017, 36, 670–680. [Google Scholar] [CrossRef]
  209. Mehra, S.; Ahsan, A.U.; Sharma, M.; Budhwar, M.; Chopra, M. Gestational Fisetin Exerts Neuroprotection by Regulating Mitochondria-Directed Canonical Wnt Signaling, BBB Integrity, and Apoptosis in Prenatal VPA-Induced Rodent Model of Autism. Mol. Neurobiol. 2024, 61, 4001–4020. [Google Scholar] [CrossRef]
  210. Zhang, L.; Hu, Z.; Bai, W.; Peng, Y.; Lin, Y.; Cong, Z. Fucoxanthin ameliorates traumatic brain injury by suppressing the blood-brain barrier disruption. iScience 2023, 26, 108270. [Google Scholar] [CrossRef]
  211. Soltani, Z.; Khaksari, M.; Jafari, E.; Iranpour, M.; Shahrokhi, N. Is genistein neuroprotective in traumatic brain injury? Physiol. Behav. 2015, 152 Pt A, 26–31. [Google Scholar] [CrossRef]
  212. Song, H.; Ding, Z.; Chen, J.; Chen, T.; Wang, T.; Huang, J. The AMPK-SIRT1-FoxO1-NF-κB signaling pathway participates in hesperetin-mediated neuroprotective effects against traumatic brain injury via the NLRP3 inflammasome. Immunopharmacol. Immunotoxicol. 2022, 44, 970–983. [Google Scholar] [CrossRef] [PubMed]
  213. Cheng, X.; Yang, Y.L.; Yang, H.; Wang, Y.H.; Du, G.H. Kaempferol alleviates LPS-induced neuroinflammation and BBB dysfunction in mice via inhibiting HMGB1 release and down-regulating TLR4/MyD88 pathway. Int. Immunopharmacol. 2018, 56, 29–35. [Google Scholar] [CrossRef] [PubMed]
  214. Yang, Y.L.; Cheng, X.; Li, W.H.; Liu, M.; Wang, Y.H.; Du, G.H. Kaempferol Attenuates LPS-Induced Striatum Injury in Mice Involving Anti-Neuroinflammation, Maintaining BBB Integrity, and Down-Regulating the HMGB1/TLR4 Pathway. Int. J. Mol. Sci. 2019, 20, 491. [Google Scholar] [CrossRef] [PubMed]
  215. Liu, J.; Chen, L.; Zhang, X.; Pan, L.; Jiang, L. The Protective Effects of Juglanin in Cerebral Ischemia Reduce Blood-Brain Barrier Permeability via Inhibition of VEGF/VEGFR2 Signaling. Drug Des. Dev. Ther. 2020, 14, 3165–3175. [Google Scholar] [CrossRef] [PubMed]
  216. Toklu, H.Z.; Hakan, T.; Biber, N.; Solakoğlu, S.; Oğünç, A.V.; Sener, G. The protective effect of alpha lipoic acid against traumatic brain injury in rats. Free Radic. Res. 2009, 43, 658–667. [Google Scholar] [CrossRef] [PubMed]
  217. Turk, C.; Camlar, M.; Diniz, G.; Arslan, F.D.; Oren, M.M.; Ozer, F. Effects of Lutein on Brain Damage and Vasospasm in an Experimental Subarachnoid Hemorrhage Model. World Neurosurg. 2020, 143, e450–e455. [Google Scholar] [CrossRef]
  218. Tan, D.; Yu, X.; Chen, M.; Chen, J.; Xu, J. Lutein protects against severe traumatic brain injury through anti-inflammation and antioxidative effects via ICAM-1/Nrf-2. Mol. Med. Rep. 2017, 16, 4235–4240. [Google Scholar] [CrossRef]
  219. Liu, R.; Gao, M.; Qiang, G.F.; Zhang, T.T.; Lan, X.; Ying, J.; Du, G.H. The anti-amnesic effects of luteolin against amyloid beta(25-35) peptide-induced toxicity in mice involve the protection of neurovascular unit. Neuroscience 2009, 162, 1232–1243. [Google Scholar] [CrossRef]
  220. Moustafa, E.M.; Moawed, F.S.M.; Elmaghraby, D.F. Luteolin/ZnO nanoparticles attenuate neuroinflammation associated with diabetes via regulating MicroRNA-124 by targeting C/EBPA. Environ. Toxicol. 2023, 38, 2691–2704. [Google Scholar] [CrossRef]
  221. Wu, A.; Liu, R.; Dai, W.; Jie, Y.; Yu, G.; Fan, X.; Huang, Q. Lycopene attenuates early brain injury and inflammation following subarachnoid hemorrhage in rats. Int. J. Clin. Exp. Med. 2015, 8, 14316–14322. [Google Scholar] [PubMed]
  222. Wang, W.; Yang, W.; Shen, Z.; Wen, S.; Hu, M. The Dose-Response Effect of Lycopene on Cerebral Vessel and Neuron Impairment Induced by Hyperlipidemia. J. Agric. Food Chem. 2018, 66, 13173–13182. [Google Scholar] [CrossRef] [PubMed]
  223. Lapi, D.; Chiurazzi, M.; Di Maro, M.; Mastantuono, T.; Battiloro, L.; Sabatino, L.; Ricci, S.; Di Carlo, A.; Starita, N.; Guida, B.; et al. Malvidin’s Effects on Rat Pial Microvascular Permeability Changes Due to Hypoperfusion and Reperfusion Injury. Front. Cell Neurosci. 2016, 10, 153. [Google Scholar] [CrossRef] [PubMed]
  224. Yang, Y.; Li, L.; Yu, L.; Xia, Y.; Fang, Z.; Wang, S. Naringenin Protected Against Blood Brain Barrier Breakdown after Ischemic Stroke through GSK-3β/β-Catenin Pathway. Neurochem. Res. 2024, 50, 17. [Google Scholar] [CrossRef] [PubMed]
  225. Ji, H.; Zhang, X.; Du, Y.; Liu, H.; Li, S.; Li, L. Polydatin modulates inflammation by decreasing NF-κB activation and oxidative stress by increasing Gli1, Ptch1, SOD1 expression and ameliorates blood-brain barrier permeability for its neuroprotective effect in pMCAO rat brain. Brain Res. Bull. 2012, 87, 50–59. [Google Scholar] [CrossRef]
  226. Ruan, W.; Li, J.; Xu, Y.; Wang, Y.; Zhao, F.; Yang, X.; Jiang, H.; Zhang, L.; Saavedra, J.M.; Shi, L.; et al. MALAT1 Up-Regulator Polydatin Protects Brain Microvascular Integrity and Ameliorates Stroke Through C/EBPβ/MALAT1/CREB/PGC-1α/PPARγ Pathway. Cell Mol. Neurobiol. 2019, 39, 265–286. [Google Scholar] [CrossRef]
  227. Wu, S.; Yue, Y.; Li, J.; Li, Z.; Li, X.; Niu, Y.; Xiang, J.; Ding, H. Procyanidin B2 attenuates neurological deficits and blood-brain barrier disruption in a rat model of cerebral ischemia. Mol. Nutr. Food Res. 2015, 59, 1930–1941. [Google Scholar] [CrossRef]
  228. Yang, Z.H.; Liu, Y.J.; Ban, W.K.; Liu, H.B.; Lv, L.J.; Zhang, B.Y.; Liu, A.L.; Hou, Z.Y.; Lu, J.; Chen, X.; et al. Pterostilbene alleviated cerebral ischemia/reperfusion-induced blood-brain barrier dysfunction via inhibiting early endothelial cytoskeleton reorganization and late basement membrane degradation. Food Funct. 2023, 14, 8291–8308. [Google Scholar] [CrossRef]
  229. Yan, W.; Ren, D.; Feng, X.; Huang, J.; Wang, D.; Li, T.; Zhang, D. Neuroprotective and Anti-Inflammatory Effect of Pterostilbene Against Cerebral Ischemia/Reperfusion Injury via Suppression of COX-2. Front. Pharmacol. 2021, 12, 770329. [Google Scholar] [CrossRef]
  230. Liu, R.; Zhang, T.T.; Zhou, D.; Bai, X.Y.; Zhou, W.L.; Huang, C.; Song, J.K.; Meng, F.R.; Wu, C.X.; Li, L.; et al. Quercetin protects against the Aβ(25-35)-induced amnesic injury: Involvement of inactivation of rage-mediated pathway and conservation of the NVU. Neuropharmacology 2013, 67, 419–431. [Google Scholar] [CrossRef]
  231. Lee, J.K.; Kwak, H.J.; Piao, M.S.; Jang, J.W.; Kim, S.H.; Kim, H.S. Quercetin reduces the elevated matrix metalloproteinases-9 level and improves functional outcome after cerebral focal ischemia in rats. Acta Neurochir. 2011, 153, 1321–1329. [Google Scholar] [CrossRef] [PubMed]
  232. Jin, Z.; Ke, J.; Guo, P.; Wang, Y.; Wu, H. Quercetin improves blood-brain barrier dysfunction in rats with cerebral ischemia reperfusion via Wnt signaling pathway. Am. J. Transl. Res. 2019, 11, 4683–4695. [Google Scholar] [PubMed]
  233. Selvakumar, K.; Prabha, R.L.; Saranya, K.; Bavithra, S.; Krishnamoorthy, G.; Arunakaran, J. Polychlorinated biphenyls impair blood-brain barrier integrity via disruption of tight junction proteins in cerebrum, cerebellum and hippocampus of female Wistar rats: Neuropotential role of quercetin. Hum. Exp. Toxicol. 2013, 32, 706–720. [Google Scholar] [CrossRef]
  234. Yang, R.; Shen, Y.J.; Chen, M.; Zhao, J.Y.; Chen, S.H.; Zhang, W.; Song, J.K.; Li, L.; Du, G.H. Quercetin attenuates ischemia reperfusion injury by protecting the blood-brain barrier through Sirt1 in MCAO rats. J. Asian Nat. Prod. Res. 2022, 24, 278–289. [Google Scholar] [CrossRef] [PubMed]
  235. Li, X.; Yang, Y.; Feng, P.; Wang, H.; Zheng, M.; Zhu, Y.; Zhong, K.; Hu, J.; Ye, Y.; Lu, L.; et al. Quercetin improves the protection of hydroxysafflor yellow a against cerebral ischemic injury by modulating of blood-brain barrier and src-p-gp-mmp-9 signalling. Heliyon 2024, 10, e31002. [Google Scholar] [CrossRef] [PubMed]
  236. Clark, D.; Tuor, U.I.; Thompson, R.; Institoris, A.; Kulynych, A.; Zhang, X.; Kinniburgh, D.W.; Bari, F.; Busija, D.W.; Barber, P.A. Protection against recurrent stroke with resveratrol: Endothelial protection. PLoS ONE 2012, 7, e47792. [Google Scholar] [CrossRef]
  237. Luan, H.; Kan, Z.; Xu, Y.; Lv, C.; Jiang, W. Rosmarinic acid protects against experimental diabetes with cerebral ischemia: Relation to inflammation response. J. Neuroinflamm. 2013, 10, 28. [Google Scholar] [CrossRef]
  238. Yang, T.; Feng, C.; Wang, D.; Qu, Y.; Yang, Y.; Wang, Y.; Sun, Z. Neuroprotective and Anti-inflammatory Effect of Tangeretin Against Cerebral Ischemia-Reperfusion Injury in Rats. Inflammation 2020, 43, 2332–2343. [Google Scholar] [CrossRef]
  239. Novochadlo, M.; Goldim, M.P.; Bonfante, S.; Joaquim, L.; Mathias, K.; Metzker, K.; Machado, R.S.; Lanzzarin, E.; Bernades, G.; Bagio, E.; et al. Folic acid alleviates the blood brain barrier permeability and oxidative stress and prevents cognitive decline in sepsis-surviving rats. Microvasc. Res. 2021, 137, 104193. [Google Scholar] [CrossRef]
  240. Chang, C.Y.; Chen, J.Y.; Wu, M.H.; Hu, M.L. Therapeutic treatment with vitamin C reduces focal cerebral ischemia-induced brain infarction in rats by attenuating disruptions of blood brain barrier and cerebral neuronal apoptosis. Free Radic. Biol. Med. 2020, 155, 29–36. [Google Scholar] [CrossRef]
  241. Yang, J.; Wang, K.; Hu, T.; Wang, G.; Wang, W.; Zhang, J. Vitamin D3 Supplement Attenuates Blood-Brain Barrier Disruption and Cognitive Impairments in a Rat Model of Traumatic Brain Injury. Neuromol. Med. 2021, 23, 491–499. [Google Scholar] [CrossRef] [PubMed]
  242. Shlosberg, D.; Benifla, M.; Kaufer, D.; Friedman, A. Blood-brain barrier breakdown as a therapeutic target in traumatic brain injury. Nat. Rev. Neurol. 2010, 6, 393–403. [Google Scholar] [CrossRef] [PubMed]
  243. Cash, A.; Theus, M.H. Mechanisms of Blood-Brain Barrier Dysfunction in Traumatic Brain Injury. Int. J. Mol. Sci. 2020, 21, 3344. [Google Scholar] [CrossRef] [PubMed]
  244. Keep, R.F.; Zhou, N.; Xiang, J.; Andjelkovic, A.V.; Hua, Y.; Xi, G. Vascular disruption and blood-brain barrier dysfunction in intracerebral hemorrhage. Fluids Barriers CNS 2014, 11, 18. [Google Scholar] [CrossRef]
  245. Bernardo-Castro, S.; Sousa, J.A.; Brás, A.; Cecília, C.; Rodrigues, B.; Almendra, L.; Machado, C.; Santo, G.; Silva, F.; Ferreira, L.; et al. Pathophysiology of Blood-Brain Barrier Permeability Throughout the Different Stages of Ischemic Stroke and Its Implication on Hemorrhagic Transformation and Recovery. Front. Neurol. 2020, 11, 594672. [Google Scholar] [CrossRef]
  246. Wu, J.Y.; Li, Y.J.; Yang, L.; Hu, Y.Y.; Hu, X.B.; Tang, T.T.; Wang, J.M.; Liu, X.Y.; Xiang, D.X. Borneol and A-asarone as adjuvant agents for improving blood-brain barrier permeability of puerarin and tetramethylpyrazine by activating adenosine receptors. Drug Deliv. 2018, 25, 1858–1864. [Google Scholar] [CrossRef]
  247. Bjelik, A.; Bereczki, E.; Gonda, S.; Juhász, A.; Rimanóczy, A.; Zana, M.; Csont, T.; Pákáski, M.; Boda, K.; Ferdinandy, P.; et al. Human apoB overexpression and a high-cholesterol diet differently modify the brain APP metabolism in the transgenic mouse model of atherosclerosis. Neurochem. Int. 2006, 49, 393–400. [Google Scholar] [CrossRef]
  248. Hoyk, Z.; Tóth, M.E.; Lénárt, N.; Nagy, D.; Dukay, B.; Csefová, A.; Zvara, Á.; Seprényi, G.; Kincses, A.; Walter, F.R.; et al. Cerebrovascular Pathology in Hypertriglyceridemic APOB-100 Transgenic Mice. Front. Cell Neurosci. 2018, 12, 380. [Google Scholar] [CrossRef]
  249. Barabási, B.; Barna, L.; Santa-Maria, A.R.; Harazin, A.; Molnár, R.; Kincses, A.; Vigh, J.P.; Dukay, B.; Sántha, M.; Tóth, M.E.; et al. Role of interleukin-6 and interleukin-10 in morphological and functional changes of the blood-brain barrier in hypertriglyceridemia. Fluids Barriers CNS 2023, 20, 15. [Google Scholar] [CrossRef]
  250. Galea, I. The blood-brain barrier in systemic infection and inflammation. Cell Mol. Immunol. 2021, 18, 2489–2501. [Google Scholar] [CrossRef]
  251. Gozal, E.; Jagadapillai, R.; Cai, J.; Barnes, G.N. Potential crosstalk between sonic hedgehog-WNT signaling and neurovascular molecules: Implications for blood-brain barrier integrity in autism spectrum disorder. J. Neurochem. 2021, 159, 15–28. [Google Scholar] [CrossRef] [PubMed]
  252. Tesse, A.; Grossini, E.; Tamma, G.; Brenner, C.; Portincasa, P.; Marinelli, R.A.; Calamita, G. Aquaporins as Targets of Dietary Bioactive Phytocompounds. Front. Mol. Biosci. 2018, 5, 30. [Google Scholar] [CrossRef] [PubMed]
  253. Langen, U.H.; Ayloo, S.; Gu, C. Development and Cell Biology of the Blood-Brain Barrier. Annu. Rev. Cell Dev. Biol. 2019, 35, 591–613. [Google Scholar] [CrossRef] [PubMed]
  254. Stresser, D.M.; Kopec, A.K.; Hewitt, P.; Hardwick, R.N.; Van Vleet, T.R.; Mahalingaiah, P.K.S.; O’Connell, D.; Jenkins, G.J.; David, R.; Graham, J.; et al. Towards in vitro models for reducing or replacing the use of animals in drug testing. Nat. Biomed. Eng. 2024, 8, 930–935. [Google Scholar] [CrossRef]
  255. Ingber, D.E. Human organs-on-chips for disease modelling, drug development and personalized medicine. Nat. Rev. Genet. 2022, 23, 467–491. [Google Scholar] [CrossRef]
  256. Helms, H.C.; Abbott, N.J.; Burek, M.; Cecchelli, R.; Couraud, P.O.; Deli, M.A.; Förster, C.; Galla, H.J.; Romero, I.A.; Shusta, E.V.; et al. In vitro models of the blood-brain barrier: An overview of commonly used brain endothelial cell culture models and guidelines for their use. J. Cereb. Blood Flow. Metab. 2016, 36, 862–890. [Google Scholar] [CrossRef]
  257. Tahanian, E.; Sanchez, L.A.; Shiao, T.C.; Roy, R.; Annabi, B. Flavonoids targeting of IκB phosphorylation abrogates carcinogen-induced MMP-9 and COX-2 expression in human brain endothelial cells. Drug Des. Dev. Ther. 2011, 5, 299–309. [Google Scholar] [CrossRef]
  258. Xu, Y.; Zhang, J.; Jiang, W.; Zhang, S. Astaxanthin induces angiogenesis through Wnt/β-catenin signaling pathway. Phytomedicine 2015, 22, 744–751. [Google Scholar] [CrossRef]
  259. Kuo, M.H.; Lee, H.F.; Tu, Y.F.; Lin, L.H.; Cheng, Y.Y.; Lee, H.T. Astaxanthin Ameliorates Ischemic-Hypoxic-Induced Neurotrophin Receptor p75 Upregulation in the Endothelial Cells of Neonatal Mouse Brains. Int. J. Mol. Sci. 2019, 20, 6168. [Google Scholar] [CrossRef]
  260. Fanaee-Danesh, E.; Gali, C.C.; Tadic, J.; Zandl-Lang, M.; Carmen Kober, A.; Agujetas, V.R.; de Dios, C.; Tam-Amersdorfer, C.; Stracke, A.; Albrecher, N.M.; et al. Astaxanthin exerts protective effects similar to bexarotene in Alzheimer’s disease by modulating amyloid-beta and cholesterol homeostasis in blood-brain barrier endothelial cells. Biochim. Biophys. Acta Mol. Basis Dis. 2019, 1865, 2224–2245. [Google Scholar] [CrossRef]
  261. Bereta, M.; Bereta, J.; Georgoff, I.; Coffman, F.D.; Cohen, S.; Cohen, M.C. Methylxanthines and calcium-mobilizing agents inhibit the expression of cytokine-inducible nitric oxide synthase and vascular cell adhesion molecule-1 in murine microvascular endothelial cells. Exp. Cell Res. 1994, 212, 230–242. [Google Scholar] [CrossRef] [PubMed]
  262. Kaiser, M.; Burek, M.; Britz, S.; Lankamp, F.; Ketelhut, S.; Kemper, B.; Förster, C.; Gorzelanny, C.; Goycoolea, F.M. The Influence of Capsaicin on the Integrity of Microvascular Endothelial Cell Monolayers. Int. J. Mol. Sci. 2018, 20, 122. [Google Scholar] [CrossRef] [PubMed]
  263. Jamornwan, S.; Chokpanuwat, T.; Uppakara, K.; Laorob, T.; Wichai, U.; Ketsawatsomkron, P.; Saengsawang, W. Nitro Capsaicin Suppressed Microglial Activation and TNF-α-Induced Brain Microvascular Endothelial Cell Damage. Biomedicines 2022, 10, 2680. [Google Scholar] [CrossRef] [PubMed]
  264. Ardid-Ruiz, A.; Harazin, A.; Barna, L.; Walter, F.R.; Bladé, C.; Suárez, M.; Deli, M.A.; Aragonès, G. The effects of Vitis vinifera L. phenolic compounds on a blood-brain barrier culture model: Expression of leptin receptors and protection against cytokine-induced damage. J. Ethnopharmacol. 2020, 247, 112253. [Google Scholar] [CrossRef]
  265. Lee, S.B.; Choi, E.H.; Jeong, K.H.; Kim, K.S.; Shim, S.M.; Kim, G.H. Effect of catechins and high-temperature-processed green tea extract on scavenging reactive oxygen species and preventing Aβ1-42 fibrils’ formation in brain microvascular endothelium. Nutr. Neurosci. 2020, 23, 363–373. [Google Scholar] [CrossRef]
  266. Lee, B.K.; Lee, W.J.; Jung, Y.S. Chrysin Attenuates VCAM-1 Expression and Monocyte Adhesion in Lipopolysaccharide-Stimulated Brain Endothelial Cells by Preventing NF-κB Signaling. Int. J. Mol. Sci. 2017, 18, 1424. [Google Scholar] [CrossRef]
  267. Zhang, W.X.; Wang, H.; Cui, H.R.; Guo, W.B.; Zhou, F.; Cai, D.S.; Xu, B.; Jia, X.H.; Huang, X.M.; Yang, Y.Q.; et al. Design, synthesis and biological evaluation of cinnamic acid derivatives with synergetic neuroprotection and angiogenesis effect. Eur. J. Med. Chem. 2019, 183, 111695. [Google Scholar] [CrossRef]
  268. Dong, H.J.; Shang, C.Z.; Peng, D.W.; Xu, J.; Xu, P.X.; Zhan, L.; Wang, P. Curcumin attenuates ischemia-like injury induced IL-1β elevation in brain microvascular endothelial cells via inhibiting MAPK pathways and nuclear factor-κB activation. Neurol. Sci. 2014, 35, 1387–1392. [Google Scholar] [CrossRef]
  269. Lamy, S.; Muhire, É.; Annabi, B. Antiproliferative efficacy of elderberries and elderflowers (Sambucus canadensis) on glioma and brain endothelial cells under normoxic and hypoxic conditions. J. Funct. Foods 2018, 40, 164–179. [Google Scholar] [CrossRef]
  270. Veszelka, S.; Tóth, A.E.; Walter, F.R.; Datki, Z.; Mózes, E.; Fülöp, L.; Bozsó, Z.; Hellinger, E.; Vastag, M.; Orsolits, B.; et al. Docosahexaenoic acid reduces amyloid-β induced toxicity in cells of the neurovascular unit. J. Alzheimer’s Dis. 2013, 36, 487–501. [Google Scholar] [CrossRef]
  271. Chen, X.; Wang, Q.; Zhan, L.; Shu, A. Effects and mechanisms of docosahexaenoic acid on the generation of angiopoietin-2 by rat brain microvascular endothelial cells under an oxygen- and glucose-deprivation environment. SpringerPlus 2016, 5, 1518. [Google Scholar] [CrossRef] [PubMed]
  272. Torres-Vergara, P.; Penny, J. Pro-inflammatory and anti-inflammatory compounds exert similar effects on P-glycoprotein in blood-brain barrier endothelial cells. J. Pharm. Pharmacol. 2018, 70, 713–722. [Google Scholar] [CrossRef] [PubMed]
  273. Lee, Y.W.; Lee, W.H. Protective effects of genistein on proinflammatory pathways in human brain microvascular endothelial cells. J. Nutr. Biochem. 2008, 19, 819–825. [Google Scholar] [CrossRef] [PubMed]
  274. Xi, Y.D.; Yu, H.L.; Ding, J.; Ma, W.W.; Yuan, L.H.; Feng, J.F.; Xiao, Y.X.; Xiao, R. Flavonoids protect cerebrovascular endothelial cells through Nrf2 and PI3K from β-amyloid peptide-induced oxidative damage. Curr. Neurovasc. Res. 2012, 9, 32–41. [Google Scholar] [CrossRef]
  275. Calhau, C.; Martel, F.; Soares-da-Silva, P.; Hipólito-Reis, C.; Azevedo, I. Regulation of [(3)H]MPP(+) transport by phosphorylation/dephosphorylation pathways in RBE4 cells: Role of ecto-alkaline phosphatase. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2002, 365, 349–356. [Google Scholar] [CrossRef]
  276. Calhau, C.; Martel, F.; Pinheiro-Silva, S.; Pinheiro, H.; Soares-da-Silva, P.; Hipólito-Reis, C.; Azevedo, I. Modulation of insulin transport in rat brain microvessel endothelial cells by an ecto-phosphatase activity. J. Cell Biochem. 2002, 84, 389–400. [Google Scholar] [CrossRef]
  277. Li, M.T.; Ke, J.; Guo, S.F.; Shan, L.L.; Gong, J.H.; Qiao, T.C.; Tian, H.Y.; Wu, Y.; Peng, Z.Y.; Zeng, X.Q.; et al. Huzhangqingmaiyin protected vascular endothelial cells against cerebral small vessel disease through inhibiting inflammation. J. Ethnopharmacol. 2024, 318 Pt A, 116905. [Google Scholar] [CrossRef]
  278. Xie, R.; Li, X.; Ling, Y.; Shen, C.; Wu, X.; Xu, W.; Gao, X. Alpha-lipoic acid pre- and post-treatments provide protection against in vitro ischemia-reperfusion injury in cerebral endothelial cells via Akt/mTOR signaling. Brain Res. 2012, 1482, 81–90. [Google Scholar] [CrossRef]
  279. Zhang, J.X.; Xing, J.G.; Wang, L.L.; Jiang, H.L.; Guo, S.L.; Liu, R. Luteolin Inhibits Fibrillary β-Amyloid1-40-Induced Inflammation in a Human Blood-Brain Barrier Model by Suppressing the p38 MAPK-Mediated NF-κB Signaling Pathways. Molecules 2017, 22, 334. [Google Scholar] [CrossRef]
  280. Xu, X.D.; Teng, Y.; Zou, J.Y.; Ye, Y.; Song, H.; Wang, Z.Y. Effects of lycopene on vascular remodeling through the LXR-PI3K-AKT signaling pathway in APP/PS1 mice. Biochem. Biophys. Res. Commun. 2020, 526, 699–705. [Google Scholar] [CrossRef]
  281. Zhang, S.; Hu, X.; Guo, S.; Shi, L.; He, Q.; Zhang, P.; Yu, S.; Zhao, R. Myricetin ameliorated ischemia/reperfusion-induced brain endothelial permeability by improvement of eNOS uncoupling and activation eNOS/NO. J. Pharmacol. Sci. 2019, 140, 62–72. [Google Scholar] [CrossRef] [PubMed]
  282. Ishisaka, A.; Ichikawa, S.; Sakakibara, H.; Piskula, M.K.; Nakamura, T.; Kato, Y.; Ito, M.; Miyamoto, K.; Tsuji, A.; Kawai, Y.; et al. Accumulation of orally administered quercetin in brain tissue and its antioxidative effects in rats. Free Radic. Biol. Med. 2011, 51, 1329–1336. [Google Scholar] [CrossRef] [PubMed]
  283. Li, Y.; Zhou, S.; Li, J.; Sun, Y.; Hasimu, H.; Liu, R.; Zhang, T. Quercetin protects human brain microvascular endothelial cells from fibrillar β-amyloid1-40-induced toxicity. Acta Pharm. Sin. B 2015, 5, 47–54. [Google Scholar] [CrossRef] [PubMed]
  284. Li, M.T.; Ke, J.; Guo, S.F.; Wu, Y.; Bian, Y.F.; Shan, L.L.; Liu, Q.Y.; Huo, Y.J.; Guo, C.; Liu, M.Y.; et al. The Protective Effect of Quercetin on Endothelial Cells Injured by Hypoxia and Reoxygenation. Front. Pharmacol. 2021, 12, 732874. [Google Scholar] [CrossRef]
  285. Sun, P.; Yang, Y.; Yang, L.; Qian, Y.; Liang, M.; Chen, H.; Zhang, J.; Qiu, Y.; Guo, L.; Fu, S. Quercetin Protects Blood-Brain Barrier Integrity via the PI3K/Akt/Erk Signaling Pathway in a Mouse Model of Meningitis Induced by Glaesserella parasuis. Biomolecules 2024, 14, 696. [Google Scholar] [CrossRef]
  286. Oliveira, A.I.; Pinho, C.; Sarmento, B.; Dias, A.C.P. Quercetin-biapigenin nanoparticles are effective to penetrate the blood-brain barrier. Drug Deliv. Transl. Res. 2022, 12, 267–281. [Google Scholar] [CrossRef]
  287. Alhusban, A.; Alkhazaleh, E.; El-Elimat, T. Silymarin Ameliorates Diabetes-Induced Proangiogenic Response in Brain Endothelial Cells through a GSK-3β Inhibition-Induced Reduction of VEGF Release. J. Diabetes Res. 2017, 2017, 2537216. [Google Scholar] [CrossRef]
  288. Sajja, R.K.; Prasad, S.; Tang, S.; Kaisar, M.A.; Cucullo, L. Blood-brain barrier disruption in diabetic mice is linked to Nrf2 signaling deficits: Role of ABCB10? Neurosci. Lett. 2017, 653, 152–158. [Google Scholar] [CrossRef]
  289. Wu, C.; Zhao, J.; Chen, Y.; Li, T.; Zhu, R.; Zhu, B.; Zhang, Y. Tangeretin protects human brain microvascular endothelial cells against oxygen-glucose deprivation-induced injury. J. Cell Biochem. 2019, 120, 4883–4891. [Google Scholar] [CrossRef]
  290. Shieh, P.; Hsu, S.S.; Liang, W.Z. Mechanisms underlying protective effects of vitamin E against mycotoxin deoxynivalenol-induced oxidative stress and its related cytotoxicity in primary human brain endothelial cells. Environ. Toxicol. 2021, 36, 1375–1388. [Google Scholar] [CrossRef]
  291. Deli, M.A. Potential use of tight junction modulators to reversibly open membranous barriers and improve drug delivery. Biochim. Biophys. Acta 2009, 1788, 892–910. [Google Scholar] [CrossRef] [PubMed]
  292. Greene, C.; Campbell, M. Tight junction modulation of the blood brain barrier: CNS delivery of small molecules. Tissue Barriers 2016, 4, e1138017. [Google Scholar] [CrossRef] [PubMed]
  293. Tavakkoli, A.; Iranshahi, M.; Hasheminezhad, S.H.; Hayes, A.W.; Karimi, G. The neuroprotective activities of natural products through the Nrf2 upregulation. Phytother. Res. 2019, 33, 2256–2273. [Google Scholar] [CrossRef] [PubMed]
  294. Banks, W.A.; Kovac, A.; Morofuji, Y. Neurovascular unit crosstalk: Pericytes and astrocytes modify cytokine secretion patterns of brain endothelial cells. J. Cereb. Blood Flow. Metab. 2018, 38, 1104–1118. [Google Scholar] [CrossRef] [PubMed]
  295. Deli, M.A.; Porkoláb, G.; Kincses, A.; Mészáros, M.; Szecskó, A.; Kocsis, A.E.; Vigh, J.P.; Valkai, S.; Veszelka, S.; Walter, F.R.; et al. Lab-on-a-chip models of the blood-brain barrier: Evolution, problems, perspectives. Lab Chip 2024, 24, 1030–1063. [Google Scholar] [CrossRef] [PubMed]
  296. Scarpellino, G.; Brunetti, V.; Berra-Romani, R.; De Sarro, G.; Guerra, G.; Soda, T.; Moccia, F. The Unexpected Role of the Endothelial Nitric Oxide Synthase at the Neurovascular Unit: Beyond the Regulation of Cerebral Blood Flow. Int. J. Mol. Sci. 2024, 25, 9071. [Google Scholar] [CrossRef] [PubMed]
  297. Black, K.L.; Baba, T.; Pardridge, W.M. Enzymatic barrier protects brain capillaries from leukotriene C4. J. Neurosurg. 1994, 81, 745–751. [Google Scholar] [CrossRef]
  298. Gosslau, A.; Chen, K.Y. Nutraceuticals, apoptosis, and disease prevention. Nutrition 2004, 20, 95–102. [Google Scholar] [CrossRef]
  299. Rattner, A.; Wang, Y.; Nathans, J. Signaling Pathways in Neurovascular Development. Annu. Rev. Neurosci. 2022, 45, 87–108. [Google Scholar] [CrossRef]
  300. Dragoni, S.; Turowski, P. Polarised VEGFA signalling at vascular blood–neural barriers. Int. J. Mol. Sci. 2018, 19, 1378. [Google Scholar] [CrossRef]
  301. Xu, J.W.; Ikeda, K.; Yamori, Y. Inhibitory effect of polyphenol cyanidin on TNF-alpha-induced apoptosis through multiple signaling pathways in endothelial cells. Atherosclerosis 2007, 193, 299–308. [Google Scholar] [CrossRef] [PubMed]
  302. Scuto, M.; Majzúnová, M.; Torcitto, G.; Antonuzzo, S.; Rampulla, F.; Di Fatta, E.; Trovato Salinaro, A. Functional Food Nutrients, Redox Resilience Signaling and Neurosteroids for Brain Health. Int. J. Mol. Sci. 2024, 25, 12155. [Google Scholar] [CrossRef] [PubMed]
  303. Golech, S.A.; McCarron, R.M.; Chen, Y.; Bembry, J.; Lenz, F.; Mechoulam, R.; Shohami, E.; Spatz, M. Human brain endothelium: Coexpression and function of vanilloid and endocannabinoid receptors. Brain Res. Mol. Brain Res. 2004, 132, 87–92. [Google Scholar] [CrossRef] [PubMed]
  304. Park, C.S.; Lee, J.Y.; Choi, H.Y.; Yune, T.Y. Suppression of Transient Receptor Potential Melastatin 7 by Carvacrol Protects against Injured Spinal Cord by Inhibiting Blood-Spinal Cord Barrier Disruption. J. Neurotrauma 2022, 39, 735–749. [Google Scholar] [CrossRef] [PubMed]
  305. Vezzani, A.; Balosso, S.; Varvel, N.H.; Dingledine, R. Anti-inflammatory strategies for disease modification: Focus on therapies close to clinical translation. In Jasper’s Basic Mechanisms of the Epilepsies, 5th ed.; Noebels, J.L., Avoli, M., Rogawski, M.A., Vezzani, A., Delgado-Escueta, A.V., Eds.; Oxford University Press: New York, NY, USA, 2024; Chapter 74. [Google Scholar]
  306. Rodak, K.; Kokot, I.; Kratz, E.M. Caffeine as a Factor. Influencing the Functioning of the Human. Body-Friend or Foe? Nutrients 2021, 13, 3088. [Google Scholar] [CrossRef]
  307. van Hameren, G.; Aboghazleh, R.; Parker, E.; Dreier, J.P.; Kaufer, D.; Friedman, A. From spreading depolarization to blood-brain barrier dysfunction: Navigating traumatic brain injury for novel diagnosis and therapy. Nat. Rev. Neurol. 2024, 20, 408–425. [Google Scholar] [CrossRef]
  308. Li, Y.; Luo, Z.; Li, G.; Belwal, T.; Li, L.; Xu, Y.; Su, B.; Lin, X. Interference-free Detection of Caffeine in Complex Matrices Using a Nanochannel Electrode Modified with Binary Hydrophilic-Hydrophobic PDMS. ACS Sens. 2021, 6, 1604–1612. [Google Scholar] [CrossRef]
  309. Sharma, N.; Phan, H.T.T.; Yoda, T.; Shimokawa, N.; Vestergaard, M.C.; Takagi, M. Effects of Capsaicin on Biomimetic Membranes. Biomimetics 2019, 4, 17. [Google Scholar] [CrossRef]
  310. Konovalova, I.S.; Shishkina, S.V.; Wyshusek, M.; Patzer, M.; Reiss, G.J. Supramolecular architecture of theophylline polymorphs, monohydrate and co-crystals with iodine: Study from the energetic viewpoint. RSC Adv. 2024, 14, 29774–29788. [Google Scholar] [CrossRef]
  311. Whaley, W.L.; Rummel, J.D.; Kastrapeli, N. Interactions of genistein and related isoflavones with lipid micelles. Langmuir 2006, 22, 7175–7184. [Google Scholar] [CrossRef]
Figure 1. The most tested nutraceuticals for protective effects on the blood–brain barrier both in vivo and in vitro. The chemical composition and some of the food sources of the selected 11 compounds are shown. Created in BioRender. Kucsápszky N., Santa Maria, A. (2025) https://BioRender.com/j91e333 (accessed on 10 February 2025).
Figure 1. The most tested nutraceuticals for protective effects on the blood–brain barrier both in vivo and in vitro. The chemical composition and some of the food sources of the selected 11 compounds are shown. Created in BioRender. Kucsápszky N., Santa Maria, A. (2025) https://BioRender.com/j91e333 (accessed on 10 February 2025).
Nutrients 17 00766 g001
Figure 2. The structure of the blood–brain barrier in physiological and pathological conditions. The schematic drawing shows the protective effects of nutraceuticals on major components of BBB dysfunction. Created in BioRender. Kucsápszky N., Santa Maria, A. (2025) https://BioRender.com/p74u637 (accessed on 10 February 2025).
Figure 2. The structure of the blood–brain barrier in physiological and pathological conditions. The schematic drawing shows the protective effects of nutraceuticals on major components of BBB dysfunction. Created in BioRender. Kucsápszky N., Santa Maria, A. (2025) https://BioRender.com/p74u637 (accessed on 10 February 2025).
Nutrients 17 00766 g002
Figure 4. Signaling pathways regulated by nutraceuticals in BBB dysfunction. The simplified drawing shows the most important signaling pathways contributing to the protective effects of nutraceuticals on the BBB. The pathways converge on increased brain endothelial cell survival and BBB stability. BAX: BCL-2-like protein 4; BCL2: B-cell lymphoma 2 protein; CK: cytokines; COX-2: cyclooxygenase-2; ERK: extracellular signal-regulated kinases; GSK-3β: Glycogen synthase kinase-3β; JNK: c-Jun N-terminal kinase; MAPK: mitogen-activated protein kinase; NF-κB: nuclear factor kappa-light-chain-enhancer of activated B; NRF2: nuclear factor erythroid 2-related factor 2; PGs: prostaglandins; PI3K: phosphoinositol 3-kinase; TLR4: toll-like receptor 4; VEGF: vascular endothelial growth factor; ↑: increase; ↓: decrease. Created in BioRender. Kucsápszky N., Santa-Maria, AR. (2025) https://BioRender.com/d25n102 (accessed on 10 February 2025).
Figure 4. Signaling pathways regulated by nutraceuticals in BBB dysfunction. The simplified drawing shows the most important signaling pathways contributing to the protective effects of nutraceuticals on the BBB. The pathways converge on increased brain endothelial cell survival and BBB stability. BAX: BCL-2-like protein 4; BCL2: B-cell lymphoma 2 protein; CK: cytokines; COX-2: cyclooxygenase-2; ERK: extracellular signal-regulated kinases; GSK-3β: Glycogen synthase kinase-3β; JNK: c-Jun N-terminal kinase; MAPK: mitogen-activated protein kinase; NF-κB: nuclear factor kappa-light-chain-enhancer of activated B; NRF2: nuclear factor erythroid 2-related factor 2; PGs: prostaglandins; PI3K: phosphoinositol 3-kinase; TLR4: toll-like receptor 4; VEGF: vascular endothelial growth factor; ↑: increase; ↓: decrease. Created in BioRender. Kucsápszky N., Santa-Maria, AR. (2025) https://BioRender.com/d25n102 (accessed on 10 February 2025).
Nutrients 17 00766 g004
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kocsis, A.E.; Kucsápszky, N.; Santa-Maria, A.R.; Hunyadi, A.; Deli, M.A.; Walter, F.R. Much More than Nutrients: The Protective Effects of Nutraceuticals on the Blood–Brain Barrier in Diseases. Nutrients 2025, 17, 766. https://doi.org/10.3390/nu17050766

AMA Style

Kocsis AE, Kucsápszky N, Santa-Maria AR, Hunyadi A, Deli MA, Walter FR. Much More than Nutrients: The Protective Effects of Nutraceuticals on the Blood–Brain Barrier in Diseases. Nutrients. 2025; 17(5):766. https://doi.org/10.3390/nu17050766

Chicago/Turabian Style

Kocsis, Anna E., Nóra Kucsápszky, Ana Raquel Santa-Maria, Attila Hunyadi, Mária A. Deli, and Fruzsina R. Walter. 2025. "Much More than Nutrients: The Protective Effects of Nutraceuticals on the Blood–Brain Barrier in Diseases" Nutrients 17, no. 5: 766. https://doi.org/10.3390/nu17050766

APA Style

Kocsis, A. E., Kucsápszky, N., Santa-Maria, A. R., Hunyadi, A., Deli, M. A., & Walter, F. R. (2025). Much More than Nutrients: The Protective Effects of Nutraceuticals on the Blood–Brain Barrier in Diseases. Nutrients, 17(5), 766. https://doi.org/10.3390/nu17050766

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

Article Metrics

Back to TopTop