Next Article in Journal
The Suppression of Ubiquitin C-Terminal Hydrolase L1 Promotes the Transdifferentiation of Auditory Supporting Cells into Hair Cells by Regulating the mTOR Pathway
Previous Article in Journal
4-Oxo-2-Nonenal- and Agitation-Induced Aggregates of α-Synuclein and Phosphorylated α-Synuclein with Distinct Biophysical Properties and Biomedical Applications
Previous Article in Special Issue
Human Glial Cells as Innovative Targets for the Therapy of Central Nervous System Pathologies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Emerging Role of ABC Transporters in Glia Cells in Health and Diseases of the Central Nervous System

by
Maria Villa
1,†,
Jingyun Wu
1,†,
Stefanie Hansen
1 and
Jens Pahnke
1,2,3,4,*
1
Translational Neurodegeneration Research and Neuropathology Lab, Department of Clinical Medicine (KlinMed), Medical Faculty, University of Oslo (UiO) and Section of Neuropathology Research, Department of Pathology (PAT), Clinics for Laboratory Medicine (KLM), Oslo University Hospital (OUS), Sognsvannsveien 20, NO-0372 Oslo, Norway
2
Institute of Nutritional Medicine (INUM)/Lübeck Institute of Dermatology (LIED), University of Lübeck (UzL) and University Medical Center Schleswig-Holstein (UKSH), Ratzeburger Allee 160, D-23538 Lübeck, Germany
3
Department of Pharmacology, Faculty of Medicine, University of Latvia (LU), Jelgavas iela 3, LV-1004 Rīga, Latvia
4
School of Neurobiology, Biochemistry and Biophysics, The Georg S. Wise Faculty of Life Sciences, Tel Aviv University (TAU), Tel Aviv IL-6997801, Israel
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Cells 2024, 13(9), 740; https://doi.org/10.3390/cells13090740
Submission received: 28 February 2024 / Revised: 15 April 2024 / Accepted: 20 April 2024 / Published: 24 April 2024
(This article belongs to the Special Issue Emerging Roles of Glial Cells in Human Health and Disease)

Abstract

:
ATP-binding cassette (ABC) transporters play a crucial role for the efflux of a wide range of substrates across different cellular membranes. In the central nervous system (CNS), ABC transporters have recently gathered significant attention due to their pivotal involvement in brain physiology and neurodegenerative disorders, such as Alzheimer’s disease (AD). Glial cells are fundamental for normal CNS function and engage with several ABC transporters in different ways. Here, we specifically highlight ABC transporters involved in the maintenance of brain homeostasis and their implications in its metabolic regulation. We also show new aspects related to ABC transporter function found in less recognized diseases, such as Huntington’s disease (HD) and experimental autoimmune encephalomyelitis (EAE), as a model for multiple sclerosis (MS). Understanding both their impact on the physiological regulation of the CNS and their roles in brain diseases holds promise for uncovering new therapeutic options. Further investigations and preclinical studies are warranted to elucidate the complex interplay between glial ABC transporters and physiological brain functions, potentially leading to effective therapeutic interventions also for rare CNS disorders.

Table of Contents

1. Introduction…………………………………………………………………………….2
 1.1. ABC Transporters in Physiological Brain Function………………………………..3
  1.1.1. ABC Transporters and Neuronal Lipid Metabolism
  1.1.2. ABC Transporters and Neuronal Drug Efflux
 1.2. Oligodendrocytes………………………………………………………………….4
  1.2.1. ABC Transporters in Oligodendrocyte Differentiation and Myelination
  1.2.2. ABC Transporters in Oligodendrocyte Survival and Protection
  1.2.3. ABC Transporters in Oligodendrocyte-Mediated Ion Homeostasis
 1.3. Astrocytes…………………………………………………………………………5
  1.3.1. ABC Transporters in Astrocyte–Neuron Communication
  1.3.2. ABC Transporters in Astrocyte-Mediated Blood–Brain Barrier Function
 1.4. ABC Transporters in Blood–Brain Barrier Integrity…………………………….6
  1.4.1. ABC Transporters in Nutrient Transport across the BBB
  1.4.2. ABC Transporters in Neurovascular Coupling
 1.5. ABC Transporter Function in Immune Cells of the Brain…………………………8
  1.5.1. Lymphocytes/Monocytes
  1.5.2. Microglia
  1.5.3. Macrophages
2. Function of ABC Transporters in Brain Diseases……………………………………13
 2.1. ABC Transporter Dysfunction in Neurodegenerative Disorders…………………13
  2.1.1. ABC Transporters as Risk Factor for Alzheimer’s Disease
  2.1.2. ABC Transporters in Movement Disorders
 2.2. Other, Non-Inflammatory Brain Diseases………………………………………..17
  2.2.1. White Matter Disorders—Adrenoleukodystrophy ABCD1
  2.2.2. ABC Transporters in Epilepsy
 2.3. ABC Transporter Function in Neuroinflammatory Diseases………………………18
  2.3.1. Multiple Sclerosis
  2.3.2. Detecting Clinical Effects of ABC Transporter-deficiency in EAE
3. General Physiological Effects of ABC Transporters on Modifying Diseases ….....21
 3.1. Export of Toxic Metabolites/Peptides………………………………………………21
 3.2. Examples of Molecular Mechanisms Influenced by ABC Transporters……………21
  3.2.1. Modulation of Steroid Hormone Signaling
4. Summary………………………………………………………………………………….23
References………………………………………………………………………………...24

1. Introduction

Neurological diseases encompass a wide range of disorders that affect various functions of the central nervous system (CNS), both grossly or subtly, and often cell-specifically, leading to impairment in motor control and co-ordination, and memory and cognition deficiencies, as well as other neurological symptoms [1,2]. Amongst various molecular mechanisms implicated in the pathogenesis of neurological diseases, the role of ATP-binding cassette (ABC) transporters has gained increasing attention during the past two decades [3,4,5]. ABC transporters are a superfamily of transmembrane proteins that play essential roles in the transportation of a diverse range of molecules across cellular membranes. The expression and activity of ABC transporters in the CNS are critical for normal brain function. Specifically, these transporters are expressed at the blood–brain (BBB) and blood–choroid plexus barriers (BCPB), where they control the influx and efflux of endogenous compounds, xenobiotics, and therapeutic drugs [6,7]. They are involved in maintaining the brain’s homeostasis, regulating the entry and efflux of substrates, and protecting cells against toxic compounds [4,8]. ABC transporters are differentially expressed in different brain regions [9,10] and cell types, including neurons and glia cells such as astrocytes and oligodendrocytes, in which they play essential roles, e.g., for maintaining cellular lipid homeostasis, regulating neurotransmitter levels, and modulating inflammatory responses in the CNS [6,7].
Emerging evidence suggests that dysfunction of ABC transporters contributes to the development and progression of various neurological and neurodegenerative diseases, including Alzheimer’s disease (AD) [11,12,13,14], Parkinson’s disease (PD) [15,16,17,18,19], Huntington’s disease (HD) [20,21], and drug-resistant epilepsy [22,23,24,25,26]. For instance, in AD, alterations of ABC transporters such as ABCA1, ABCA7, ABCB1, ABCC1, and ABCG2 have been associated with the impaired clearance of amyloid-β (Aβ) peptides, leading to their accumulation in the brain and vessel walls and the formation of neurotoxic plaques or cerebral amyloid angiopathy (CAA) [11,12,13,14,27,28,29,30].
Understanding the precise roles of ABC transporters in neurological and neurodegenerative diseases is of great importance for several reasons. Firstly, elucidating the mechanisms underlying their dysregulation can provide valuable insights into disease pathogenesis and identify potential therapeutic targets. Secondly, ABC transporters have the potential to influence the pharmacokinetics and efficacy of therapeutic drugs used for the treatment of neurological diseases [7]. Thirdly, studying the interplay between ABC transporters and other tissue and cellular processes, such as inflammation and oxidative stress, can help to unravel complex disease mechanisms [5].
In this review, we will summarize the current knowledge about the involvement of ABC transporters in neurological and neurodegenerative diseases with a special emphasis on glial cells. We will discuss their expression patterns, physiological functions, and emerging evidence implicating their dysfunction in disease pathogenesis, and highlight their significance for the development of effective therapeutic interventions.

1.1. ABC Transporters in Physiological Brain Function

Neurons are highly specialized cells responsible for maintaining brain homeostasis and generating and/or transmitting electrical signals in the brain [31,32]. Besides basic metabolic mechanisms, the proper functioning of neurons relies on the maintenance of precise intra- and extracellular ion concentrations [33,34], and the synthesis and reuptake of neurotransmitters [35] in different cellular compartments and in other cells, such as astrocytes [36], as well as on the lipid composition of cellular and intracellular membranes [37,38].
In this regard, ABC transporters have been recognized as key players for maintaining neuronal homeostasis by regulating the transport of various substrates across the neuronal cell membrane (Figure 1). These transporters are located in the cell membranes of various glial cells, such as astrocytes, microglia, and oligodendrocytes, and are crucial for maintaining homeostasis of the brain microenvironment. For example, ABC transporters participate in the regulation of neurotransmitter levels by controlling their reuptake and recycling [39,40]. In addition, ABC transporters are involved in the transport of lipids and lipoproteins. Thereby, they contribute to sustaining the structural integrity and dynamic composition of cellular membranes [41,42,43]. Moreover, ABC transporters are crucial in transporting various metabolites, such as sugars, amino acids, lipids, and other small molecules involved in energy production and cellular homeostasis, ensuring the right balance for metabolic processes within neurons [44,45,46,47,48].

1.1.1. ABC Transporters and Neuronal Lipid Metabolism

The lipid metabolism plays a fundamental role in neuronal structure, function, and signaling [49]. ABC transporters play a vital role in this metabolism as they constitute a significant family of proteins that contribute to neuronal lipid homeostasis by regulating the translocation of lipids across neuronal membranes [50,51]. ABCA1, ABCA2, ABCA7, and ABCG1 are integral components in the intricate regulation of phospholipid and cholesterol homeostasis, playing crucial roles in neuronal cholesterol efflux [52,53,54]. For instance, by regulating cholesterol efflux, they help maintain the cellular cholesterol balance and prevent the accumulation of cholesterol in neurons [21,55]. The association between cholesterol levels and Aβ peptide generation is a critical aspect of AD research. The involvement of ABCA1, ABCA7, and ABCG1 in modulating APP processing adds supportive evidence to these transporters constituting potential therapeutic targets for mitigating Aβ production [52,56]. ABCG1 and ABCG4 transporters have also been identified as key regulators of cellular lipid transport in neurons, affecting the composition of lipid rafts and the processing of amyloid-β precursor protein (APP) [57,58,59]. Moreover, ABCD1, ABCD2, and ABCD3 have been implicated in the transport of fatty acids into peroxisomes, underscoring their role in lipid catabolism and the etiology of peroxisomal disorders [60]. Recent research suggested that the expression of the ABCC4 transporter influences the accumulation of bioactive lipids, such as eicosanoids, which are associated with inflammation and neuronal signaling [61,62]. Overall, the intricate interplay between ABC transporters and neuronal lipid metabolism highlights their crucial role in maintaining neuronal membrane integrity, synaptic function, and overall brain health.

1.1.2. ABC Transporters and Neuronal Drug Efflux

Neurons are often exposed to various exogenous compounds, including therapeutic drugs, environmental toxins, and xenobiotics. ABC transporters, particularly ABCB1, also known as P-glycoprotein (P-gp), and ABCG2, also known as breast cancer resistance protein (BCRP), are vital components of the neuronal drug efflux mechanism. They hereby play a crucial role in protecting neurons from the potential toxicity of these compounds by modulating the pharmacokinetic and pharmacodynamic profiles of therapeutic agents and actively transporting them out of the cells [26,63,64].
ABCB1 is highly expressed in neurons and has been extensively studied for its efflux functions [65]. This transporter can limit the entry of drugs into neurons and contribute to the development of drug resistance in neurological disorders such as Amyotrophic Lateral Sclerosis (ALS). In ALS, an induced pharmaco-resistance has been observed, suggesting a selective upregulation of these transporters in disease-affected CNS regions, both at the mRNA and protein levels [64,66,67,68]. In epilepsy, the overexpression of ABCB1 is proven to be an important factor for the limited efficacy of antiepileptic drugs, as these transporters reduce the effective drug concentration at the site of lesions [69,70]. Moreover, the expression of other ABC transporters, such as ABCG2 and ABCC1, has been detected in neurons and implicated in the efflux of drugs and toxic metabolites [63]. Thus, understanding the mechanisms of neuronal drug efflux mediated by ABC transporters is essential for optimizing drug delivery to the brain and developing strategies to overcome drug resistance in the treatment of neurological conditions [71].

1.2. Oligodendrocytes

1.2.1. ABC Transporters in Oligodendrocyte Differentiation and Myelination

Oligodendrocytes are glial cells in the CNS that play a crucial role for myelination, the process of forming the myelin sheath around neuronal axons and dendrites [72,73]. Myelin provides insulation and facilitates efficient electrical signal transmission [73]. Recent studies have proven that ABC transporters are implicated in the complex processes of oligodendrocyte differentiation and myelination. For instance, ABCA2 has been identified as a key transporter involved in lipid trafficking and synthesis during myelination, with its expression predominantly occurring in oligodendrocytes during the maturation of the developing rat brain [3,74]. This evidence implies that ABCA2 plays a crucial role in the transport of cholesterol and other lipids to the developing myelin sheaths [74,75]. Furthermore, ABC transporters such as ABCG1 and ABCG4 have been shown to regulate the efflux of lipids from oligodendrocytes, which, in turn, impacts myelin integrity and maintenance [76,77].

1.2.2. ABC Transporters in Oligodendrocyte Survival and Protection

ABCC1, also known as multidrug resistance-associated protein 1 (MRP1), is highly expressed in oligodendrocytes and plays a crucial role in the export of potentially toxic metabolites and xenobiotics [78]. Dysfunction of ABCC1 has been associated with impaired oligodendrocyte survival and susceptibility to oxidative stress [79,80]. Additionally, another key transporter, ABCG2, has been suggested to contribute to cellular survival by protecting against apoptosis induced by various stressors, including cytokines. While this research is primarily focused on placental cells, the protective mechanisms of ABCG2 may be extrapolated to oligodendrocytes, where similar stress responses are crucial for cell survival [81]. Notably, ABC transporters such as ABCB1 and ABCG2 contribute to the efflux of neurotoxic compounds from oligodendrocytes, thereby protecting them from damage [28]. Nevertheless, it is important to consider that the precise mechanisms of ABC transporters in oligodendrocyte survival and protection are still being elucidated.

1.2.3. ABC Transporters in Oligodendrocyte-Mediated Ion Homeostasis

Oligodendrocytes also play a critical role in maintaining ion homeostasis in the CNS, which is essential for proper neuronal function [73]. ABC transporters expressed in oligodendrocytes contribute to the regulation of ion concentrations in the extracellular environment [82]. For instance, the ATP-dependent ion transporter sulfonylurea receptor 1 (SUR1)/ABCC8 has been shown to be involved in potassium channel regulation in oligodendrocytes [83,84], preventing excessive potassium accumulation which could be detrimental to neuronal function. Furthermore, ABC transporters have been identified as crucial for maintaining metal ion homeostasis in microorganisms [85,86,87,88], which may provide insights into similar functions in oligodendrocytes. For example, the identified metal-specific ABC transport systems, while studied in bacteria, suggest a conserved mechanism that could be relevant for oligodendrocyte function, and, here particularly, for iron ion uptake, which is important for myelin production [89]. While little research specifically focusing on ABC transporters in oligodendrocyte-mediated ion homeostasis is available, it is worth emphasizing that the dysregulation of ion transporters in oligodendrocytes can lead to imbalances in ion concentrations for myelination processes, compromising neuronal excitability and neurotransmission [84].
In conclusion, ABC transporters expressed in oligodendrocytes play crucial roles in oligodendrocyte differentiation, myelination, survival, protection, and ion homeostasis (Figure 1). By regulating lipid transport, the efflux of toxic compounds, and ion balance, ABC transporters actively contribute to the physiologic function of oligodendrocytes and the maintenance of myelin integrity. Dysregulation or dysfunction of oligodendrocytic ABC transporters can have profound implications for myelin formation and function.

1.3. Astrocytes

1.3.1. ABC Transporters in Astrocyte–Neuron Communication

Astrocytes are the most abundant glial cells in the brain, supporting neuronal function and maintaining brain homeostasis. One key aspect of astrocyte function is their participation in astrocyte–neuron communication, whereby they regulate the extracellular environment and provide essential metabolic support to neurons [90]. Details on ABC transporters in astrocyte–neuron communication are limited. However, several studies highlight the importance of glial cells and their interactions with neurons through various mechanisms, indirectly indicating the potential involvement of ABC transporters. For example, ABCC1 is involved in the efflux of glutathione, glutathione-conjugates, and other organic anions from astrocytes, which can modulate synaptic transmission and neuronal excitability [91,92]. Additionally, the SUR1/ABCC8 is highly expressed in astrocytes and regulates potassium fluxes, which influence neuronal excitability and synaptic activity [93,94]. Furthermore, ABCA1 and ABCG1 play pivotal roles in cholesterol efflux from astrocytes to neurons, contributing to the regulation of cholesterol homeostasis in the brain [48,95,96].

1.3.2. ABC Transporters in Astrocyte-Mediated Blood–Brain Barrier Function

Astrocytes play a vital role in maintaining the integrity and function of the BBB, a highly selective barrier that serves as an interface between the CNS and the peripheral circulation and regulates the passage of molecules between the blood and the brain [90]. ABC transporters expressed in astrocyte processes contribute to the efflux of xenobiotics and drugs across the BBB, limiting their entry into the brain (Figure 1) [97]. The activity of these astrocytic ABC transporters can influence drug efficacy, brain drug concentrations, and susceptibility to neurotoxic compounds. For instance, the drug resistance transporters ABCB1 and ABCG2 are highly expressed in astrocyte end-feet where they actively transport a wide range of substances back into the bloodstream, including therapeutic drugs and toxins [98,99]. Interestingly, chronic intermittent hypoxia and sustained hypoxia can both lead to BBB dysfunction characterized by the altered expression and activity of ABCB1 and ABCG2 [100]. This finding highlights the impact of environmental factors on BBB integrity and its associated transport mechanisms. In terms of astrocyte-mediated functions, the astrocyte-derived apolipoprotein E4 (APOE4) was proven to impair BBB integrity, leading to increased leakage and decreased astrocyte end-feet coverage of blood vessels, suggesting a role for astrocyte-derived factors in BBB function and transporter expression [101].
Overall, ABC transporters expressed in astrocytes are integral to the dynamic communication between astrocytes and neurons (Figure 1). They play a pivotal role in the function of the BBB, where they control the transit of a variety of molecules, including neurotransmitters, ions, and xenobiotics. This function is essential not only for normal cerebral operations but also for generally preserving CNS equilibrium. Their involvement in drug resistance, particularly in relation to CNS pharmacotherapy, underscores their potential as therapeutic targets to enhance drug delivery to the brain and address CNS pathologies [100,102]. Therefore, astrocytic ABC transporters are not only essential for maintaining a homeostatic environment conducive to optimal neuronal function, but also offer a promising avenue for therapeutic intervention.

1.4. ABC Transporters in Blood–Brain Barrier Integrity

The BBB is a highly selective and dynamic structure composed of endothelial cells, pericytes, and astrocyte end-feet that regulates the exchange of substances between the blood and the brain [103]. ABC transporters expressed in endothelial cells and pericytes play a crucial role in maintaining BBB integrity (Figure 1) [63]. Among other ABC transporters, ABCB1 and ABCG2 are highly expressed in endothelial cells of the BBB, where they actively pump out a wide range of substances from the brain, including toxic compounds and therapeutic drugs [104,105]. This protective function is vital, not only for normal brain physiology but also for minimizing the impact of age-related neurodegenerative diseases on BBB integrity [106]. However, the expression of ABC transporters can be modulated by various factors, including oxidative stress, diet and environmental pollutants. With regard to BBB integrity, this modulation has implications for drug delivery to the brain. It can be beneficial in some cases, but also presents challenges for the treatment of CNS disorders. Innovative approaches are being explored to target signaling events that could reduce the activity of ABC transporters, thereby enhancing drug delivery to the brain [107]. Additionally, pericytes express various ABC transporters, such as ABCC1, which contributes to the efflux of xenobiotics and metabolites across the BBB [10]. The co-ordinated action of ABC transporters in endothelial cells and pericytes is essential for maintaining the selective permeability of the BBB and preventing the entry of neurotoxic substances into the brain [108].

1.4.1. ABC Transporters in Nutrient Transport across the BBB

Endothelial cells of the BBB are responsible for regulating the transport of essential nutrients, such as glucose, amino acids, and vitamins from the bloodstream into the brain [109,110]. This selective transport is essential for maintaining cerebral homeostasis and providing the brain with the necessary substrates for proper function. ABC transporters expressed in endothelial cells play a significant role in the process of facilitating the bidirectional movement of various nutrients across the BBB (Figure 1), such as the efflux of glutathione, an essential antioxidant, or glutathione-coupled metabolites by ABCC1 [91,111]. Moreover, ABCC1 and ABCC4 are also expressed in brain endothelial cells and contribute to nutrient transport across the BBB. ABCC4 on the other hand, is involved in the efflux of cyclic nucleotides and prostaglandins, which may have implications on brain signaling and metabolism [112]. Similarly, ABCB1, ABCG2, and ABCC1 are known to play a crucial role in maintaining the integrity of the BBB by restricting the permeation of neurotoxic chemicals. They allow the passage of necessary metabolic substrates and limit brain uptake of nutrients, ensuring proper brain function. Their selective transport mechanism is critical for nutrient delivery, as well as for preventing the accumulation of potentially neurotoxic substances in the brain [113].

1.4.2. ABC Transporters in Neurovascular Coupling

Neurovascular coupling is the process by which changes in neuronal activity are coupled with changes in cerebral blood flow, ensuring that the brain receives an adequate blood supply to meet its metabolic demands [114]. Both endothelial cells and pericytes contribute to neurovascular coupling through the regulation of vascular tone and blood flow [115]. While the specific details on ABC transporters in neurovascular coupling have not been elucidated, it is understood that these transporters can influence this coupling indirectly through their control over the brain microenvironment. For example, ABC transporters expressed in endothelial cells modulate the production and release of vasoactive substances [109]. Specifically, ABCB1 and ABCG2 transporters have been shown to influence the release of endothelium-derived relaxing factors, such as nitric oxide, which regulates vascular tone [116]. Additionally, pericytes, which are closely associated with endothelial cells, express various ABC transporters involved in the regulation of pericyte contractility and vascular tone [117]. Moreover, indirect evidence for ABC transporter involvement in neurovascular coupling exists, such as for ABC transporters in AD pathogenesis, which is characterized by altered BBB permeability and neurovascular dysfunction. This alteration in transporter expression and function can contribute to the vascular Aβ pathology of AD by affecting nutrient transport and waste removal across the BBB [118]. It appears highly plausible that any dysfunction of these transporters could lead to altered neurovascular coupling and contribute to various neurological disorders [22].
In conclusion, ABC transporters expressed in endothelial cells and pericytes play critical roles in BBB integrity, nutrient transport across the BBB, and neurovascular coupling [63,119,120]. By regulating the efflux of toxic compounds, nutrient transport, and vascular tone, these transporters actively contribute to the proper functioning of the brain’s vasculature and maintenance of brain homeostasis. Dysregulation or dysfunction of endothelial and pericyte ABC transporters can have significant implications for BBB integrity, nutrient supply to the brain, and cerebral blood flow regulation in general, which may contribute to the pathogenesis of several neurological disorders [121]. Further research is needed to fully elucidate the specific mechanisms and therapeutic potential of these transporters in endothelial cells and pericytes.
Figure 1. Graphical overview of the role of ABC transporters in different brain cells, such as glia, neurons, and vascular cells (endothelia and pericytes). The different molecules fluxed out of the cells are represented by different symbols, which are explained in the legend (at the bottom of the figure). The transporters implicated in the efflux of these substances are represented in different colors and specified for each cell. The black arrows originate at the cells in which the effects take place and defined actions or effects of certain transporters are specified at the arrowheads.
Figure 1. Graphical overview of the role of ABC transporters in different brain cells, such as glia, neurons, and vascular cells (endothelia and pericytes). The different molecules fluxed out of the cells are represented by different symbols, which are explained in the legend (at the bottom of the figure). The transporters implicated in the efflux of these substances are represented in different colors and specified for each cell. The black arrows originate at the cells in which the effects take place and defined actions or effects of certain transporters are specified at the arrowheads.
Cells 13 00740 g001

1.5. ABC Transporter Function in Immune Cells of the Brain

It has been widely accepted that the disruption of the BBB is a common feature in different neurological disorders, such as Multiple sclerosis (MS), AD, and PD [122,123,124], and leads to the infiltration of immune cells into the brain [125]. ABC transporters are differentially expressed on and have varying important functions in the various cells of the brain’s immune system (Figure 2), modulating the immune response and neuroinflammation caused by the invasion of peripheral immune cells and resident microglia.

1.5.1. Lymphocytes/Monocytes

ABC transporters have been described to be involved in immune responses in several ways (Figure 2). They play a role in regulating lymphocytes and monocytes [126,127]; are implicated in modulating the immune response through cell maturation [128], activation [129], adhesion, and migration [126]; and perform their functions through the release of mediators, such as C-C motif chemokine ligand 2 (CCL2), platelet-activating factor (PAF), or prostaglandin E2 [127,130,131], or by general mechanisms such as cholesterol regulation [132,133,134,135]. The specific roles of the different ABC transporters in the immune response are further described:
  • Role of ABC Transporters Related to T-cell Functions
    ABCB1 is implicated in modulating the T-cell response, including cell infiltration, proliferation, and cytokine secretion. ABCB1 exerts these functions by interfering with dendritic cell function and maturation through the release of CCL2 [128]. Furthermore, ABCB1 plays a part in CD4+ and CD8+ T-cell migration and adhesion [126]. In addition, ABCB1 has an effect on CD8+ T-cell activation, expression, and generation of memory cell pools [129,136]. Inflamed endothelial cells, in which we find extensive ABCB1 expression, also secrete CCL2, affecting mainly CD8+ T-cell adhesion [126]. ABCG1 impacts inflammation mainly by regulating cholesterol. In the absence of ABCG1, cholesterol accumulates and, thereby, improves CD4+ T-cell receptor signaling. Furthermore, ABCG1 deficiency was shown to increase the absolute number of CD4+ T-cells, likely by the amplification of extracellular signal–regulated kinase (ERK) signaling. ABCG1 deficiency also enhances Treg (regulatory T)-cell development through their cholesterol content [135,136]. Inversely, ABCG1 upregulation is related to reduced T-cell proliferation [132,133,134,136].
  • Role of ABC Transporters Related to NKT-cell Functions
    ABCA7 and ABCG1 are implicated in influencing NKT (natural killer T)-cell functions in immune responses. ABCA7 deficiency impedes the binding of NKT-cells and antigen-presenting cells (APCs), by CD1d accumulation in the late endosomal compartment, thereby altering NKT-cell development and activation [132,136]. In contrast to previous statements, there is evidence supporting the involvement of ABCG1 in the correct development of NKT-cells, as ABCG1 deficiency in NKT-cells resulted in a reduced number of these cells [132,133,134,136].
  • Role of ABC Transporters in Monocyte Migration
    Monocyte migration is influenced by the ABCB1 transporter regulating the CCL2 release from reactive astrocytes [127]. Similarly, the ABCC1 transporter controls monocyte migration across the BBB due to its mediation of CCL2 release by reactive astrocytes [22,127].
  • Role of ABC Transporters in the Release of Inflammatory Mediators
    ABCB1, ABCC1, ABCC4, and ABCC8 have been implicated in the regulation of inflammatory mediators. Specifically, ABCC1 and ABCC4 are able to transport inflammatory mediators such as prostaglandin E2 [22,131]. Additionally, ABCC1 is involved in the leukotriene C4 (LTC4) release in all cells producing this mediator [22,92,137]. ABCC8 is implicated in altering inflammatory responses: the silencing of Abcc8 alters SUR1-TRPM4 function in astrocytes, causing a reduced inflammatory response in EAE with a reduction in inflammatory infiltrates and inflammatory cytokines [138]. ABCB1 mediates the secretion of PAF by reactive astrocytes, thereby contributing to the modulation of the immune response [130].
Figure 2. Graphical overview of the role of ABC transporters in maintaining brain immune functions by regulating lymphocyte, monocyte, and dendritic cell functions, also in collaboration with astrocytes. The different molecules fluxed out of the cells are represented by different symbols, which are explained in the legend (at the bottom of the figure). The ABC transporters implicated in the efflux of these substances are represented in different colors and specified for each cell. The black arrows originate at the cells in which the effects take place, and defined actions or effects of certain transporters are specified at the arrowheads. Discontinued lines represent interaction between cells.
Figure 2. Graphical overview of the role of ABC transporters in maintaining brain immune functions by regulating lymphocyte, monocyte, and dendritic cell functions, also in collaboration with astrocytes. The different molecules fluxed out of the cells are represented by different symbols, which are explained in the legend (at the bottom of the figure). The ABC transporters implicated in the efflux of these substances are represented in different colors and specified for each cell. The black arrows originate at the cells in which the effects take place, and defined actions or effects of certain transporters are specified at the arrowheads. Discontinued lines represent interaction between cells.
Cells 13 00740 g002

1.5.2. Microglia

ABC transporter expression has been described in microglia and macrophages in which they play essential roles in facilitating immune responses (Figure 3). Their function in microglia ranges from specifically modulating membrane cholesterol efflux [95,139,140,141] to the general efflux of diverse substrates [142,143]. The depletion of ABCA1 and ABCG1 in microglia has been shown to cause allodynia through the disruption of the formation of cholesterol-enriched rafts to which TLR4 co-localizes [144].
  • ABC Transporters in Microglia Cholesterol Efflux
    ABCA1 expression, at both the mRNA and protein level, has been found in microglia. In ABCA1-deficient mice, a consequent decrease in cholesterol efflux from microglia is seen, including reduced APOE lipidation, thereby increasing the risk of AD [82,95,139,140]. In addition, an upregulation in ABCG4 mRNA and ABCG4 protein levels in microglia located closely to Aβ plaques in human brains has been detected. As this transporter mediates cholesterol efflux, its upregulation improves APOE lipidation and could be considered a microglial protective response [82,141].
  • ABC Transporters in Aβ Clearance
    ABCA7 is highly expressed in microglia in which it is thought to play a role in modulating Aβ clearance. Deficiency of this transporter resulted in a reduced clearance of Aβ peptides [145,146].
  • ABC Transporters in General Substance Efflux and Drug Resistance
    ABCB1 expression was found in cell cultures of rat microglia, where it was localized at the plasma and nuclear membranes [82,147]. Its most probable role is thought to be the mediation of xenobiotics and toxic or general substances efflux [142,143]. This function has, for example, been documented by demonstrating that ABCB1 prevented the entry of digoxin in rat microglia cell cultures [142].
    In other studies, Abcc1 mRNA and ABCC1 protein expression and function have been found in rat microglia cultures and the MLS-9 cell line [143,148,149]. Here, ABCC1 transporters played a role in the resistance to drugs, such as the chemotherapeutic vincristine [148]. The ABCC1 transporter is localized mainly in the plasma membrane, but also in caveolae and clathrin-coated vesicles [82,147,150].
    Abcc4 and Abcc5 mRNA, as well as ABCC4 and ABCC5 proteins, have been detected in rat microglia cultures [82,143,147,149,151]. Functional effects of both transporters in rat and mouse microglia cell lines have been implicated in the transport of antiretroviral 9-(2-phosphonylmethoxyethyl)adenine which is a substance used against the human immunodeficiency virus (HIV) [147,151].
  • ABCD1, a Transporter Directly Related to a Disease
    The ABCD1 gene codes for the ABCD1 protein, also known as the adrenoleukodystrophy (ALD) protein for its role in this specific disease [152]. Mutations affecting the ABCD1 transporter lead to ALD due to the dysfunctional transport of very-long-chain fatty acids into peroxisomes [82,152]. Abcd1 mRNA and ABCD1 protein have been found in rodent CNS, and ABCD1 protein expression has been measured post mortem in human brains [82,152,153]. Specifically, ABCD1 is highly expressed in microglial cells [152]. Therefore, these cells could play a prominent role in the pathogenesis of the disease.
  • Other ABC Transporters
    Abcc3 mRNA is expressed in rat microglia [82,147,149]. Nevertheless, no information about its functionality in these cells is available.
    Abcc6 mRNA expression has been reported in newborn rat glial cell (including, but not exclusive to, microglia) cultures [82,154]. However, in another study, no Abcc6 expression was found in adult rat cell cultures of microglia [149]. Therefore, a study focusing specifically on microglia cultures from newborn rats could help to elucidate if Abcc6 mRNA is indeed present on and/or in these cells.
    ABCG2 protein expression has been found in rat microglia primary cultures. Abcg2 mRNA and ABCG2 protein expression were also seen in a mouse microglia cell line (BV-2 cells), in which ABCG2 expression and function are downregulated by LPS-induced inflammation [82,147,155,156]. A reduction in ABCG2 function could lead to the accumulation of toxic substances in the brain [156]. The specific function of this transporter, however, was not demonstrated, probably due to the low expression in the BV2 cells or a lack of its localization to the plasma membrane [155].
In summary, the functions that various ABC transporters have in microglia have been mainly ascribed to their role in drug efflux and not to a direct impact on immune function.

1.5.3. Macrophages

In macrophages, different ABC transporters have been described. As in microglia, these transporters can modulate cholesterol efflux [157,158,159,160,161,162,163,164], but are also implicated in phagocytosis [165] and Aβ clearance [145,166] (Figure 3).
  • Cholesterol Efflux and Lipid Homeostasis
    ABCA1, ABCA5, ABCA6, ABCA9, ABCB4, ABCG1, and ABCG4 have been detected in macrophages, where they are involved in cholesterol efflux and lipid homeostasis [161,162,163,164,167,168,169,170]. It has been described and widely accepted that dysfunction of these transporters leads to an exacerbated immune response due to the dysregulation of these cells [157,159,160,169,170,171,172].
  • Phagocytosis and Aβ Clearance
    ABCA7 has been implicated in the phagocytosis of apoptotic cells by macrophages. The presence of this transporter on the cell surface of macrophages where it co-localizes with low-density lipoprotein receptor-related protein 1 (LRP1) is essential for ERK signaling [165]. Additionally, as stated for microglia, this transporter has also been proposed to play a direct role in Aβ clearance in macrophages [145,166].
Figure 3. Graphical overview of the role of ABC transporters in microglia and macrophages. The different molecules fluxed out of the cells are represented by different symbols which are explained in the legend (at the bottom of the figure). The transporters implicated in the efflux of these substances are represented in different colors and specified for each cell. The black arrows originate at the cells in which the effects take place and defined actions of effects of certain transporters are specified at the arrowheads.
Figure 3. Graphical overview of the role of ABC transporters in microglia and macrophages. The different molecules fluxed out of the cells are represented by different symbols which are explained in the legend (at the bottom of the figure). The transporters implicated in the efflux of these substances are represented in different colors and specified for each cell. The black arrows originate at the cells in which the effects take place and defined actions of effects of certain transporters are specified at the arrowheads.
Cells 13 00740 g003

2. Function of ABC Transporters in Brain Diseases

2.1. ABC Transporter Dysfunction in Neurodegenerative Disorders

The important physiological role of ABC transporters for the correct functioning of the brain is undeniable. ABC transporters have a crucial function for transporting various substrates, as well as removing toxins and harmful molecules. Functional dysregulation of these transporters is associated with various diseases in several functional systems of our body, such as neurodegenerative, cardio-metabolic, and liver disorders [173,174,175,176,177,178,179]. Among these, neurodegenerative diseases are becoming an increasingly important and studied category. Counteracting the dysregulation of specific ABC transporters is emerging as a new therapeutic approach for some neurodegenerative diseases, such as AD, PD, and even extremely rare diseases such as frontotemporal dementia (FTD) in which ABCA2, ABCA3, ABCA4, ABCA7, ABCA9, ABCA10, and ABCA13 expression was found to be altered in patients with FTD with TDP-43 [180].

2.1.1. ABC Transporters as Risk Factors for Alzheimer’s Disease

In AD, the accumulation of Aβ peptides in the brain is a key pathological hallmark of the disease. Cholesterol plays a role in Aβ metabolism. High levels of cholesterol were related to Aβ production, whereas cholesterol depletion was related to decreased Aβ levels [181,182]. ABC transporters are implicated in facilitating Aβ aggregation in different ways, specifically through:
  • their participation in the lipid metabolism [183,184,185];
  • their involvement in Aβ phagocytosis;
  • Aβ trafficking into lysosomes [145,166,186].
ABC transporters such as ABCA1, ABCA7, ABCB1, ABCC1, ABCG1, and ABCG2 have been shown to play a critical role in the clearance of Aβ from the AD brain [27,28,187]. Mutations related to ABCA1 have been associated with AD. However, ABCA1 has also been implicated in AD resistance [188]. Furthermore, it has been described that ABCA1 overexpression or stimulation leads to a better prognosis in mice with AD [183,185,189]. ABCA2 has been implicated in AD pathogenesis, since its upregulation is related to an increase in APP synthesis [190]. In addition, ABCA2 depletion is associated with decreased Aβ production [184]. ABCA5 could play a protective role in AD as an increase in its expression is related to a decreased Aβ load in vitro [189,191].
As mentioned above, ABCA7 participates in Aβ clearance and production [56,145,166,175,187]. Coding sequence mutations and promoter and intron nucleotide polymorphisms related to the human ABCA7 locus have been associated with an increased risk for AD, promoting and highlighting ABCA7 as a new significant risk marker besides APOE4 [188,189,192,193,194,195].
ABCB1, ABCC1, and ABCG2 also play a role in AD progression and Aβ clearance. Therefore, targeting these transporters could be a potential therapeutic strategy [196,197].
In general, the dysfunction of specific ABC transporters can lead to the accumulation of Aβ in the brain, which can contribute to the development and progression of AD. Modulating ABC transporters has been previously suggested as a possibility for AD therapeutics in mice [12,198]. However, it has to be strongly emphasized that export via the BBB or BCPB facilitated through ABC transporters, e.g., mouse Abcc1, requires additional transporters, such as Oat3 and Abcc4/Mrp4 [199]. The latter are expressed very differently between mice and human, some even fully lacking expression in humans. These important species differences will play a confounding and possibly also detrimental role, hampering the development of drugs and PET tracers using ABC transporters.
  • ABCA1-APOE Axis
    An impairment of the ABCA1- and SR-BI-mediated cholesterol efflux pathways and HDL anti-inflammatory activity has already been reported in AD [200].
    Different ABCA1 genetic variants related to the risk of developing AD have been described. For example, the SNPs rs2230806 and rs2230808 have been associated with the risk of AD in Caribbean Hispanics using APOE-conditional models [201,202]. Furthermore, the SNPs rs2230805 and rs2230806 have been characterized as important markers for different forms of dementia, including AD, in a Swedish cohort [203]. Moreover, some polymorphisms in the coding region (R219K, I883M, and R1587K) or in the promoter region (C-14T) of ABCA1 were related to a higher risk of developing AD in a Spanish cohort [204]. In this cohort, it was also found that the ABCA1-14T allele increased the risk of AD synergistically when acting together with the APOE4 allele, probably by increasingly inefficient cholesterol efflux from glial cells [204]. Furthermore, an association between a rare loss-of-function variant in ABCA1 (N1800H) and increased risk of AD was found [205]. Some microRNAs which regulate ABCA1 have been implicated in Aβ accumulation (miR-106b) [206] or cholesterol metabolism (miR-758 and miR-33) [207,208,209,210].
    Modulating ABCA1 function showed its functional impact in AD models. It has been described that the ABCA1 agonist mifepriston improved aggregation pathologies driven by APOE4 [211]. Earlier findings by Koldamova et al. revealed that the absence of ABCA1 significantly reduces the levels of APOE in the brain [212]. With lower levels of APOE, there is a notable increase in Aβ deposition in the brains of APP23 mice. This suggests that ABCA1 is essential for maintaining normal APOE levels in the brain, which, in turn, affects the clearance of Aβ [212]. Wahrle et al. investigated the potential therapeutic role of targeting ABCA1 in AD [183]. Their study, based on the PDAPP mouse model, found that enhancing the function of ABCA1 through overexpression (mPrP-mAbca1) led to a significant reduction in Aβ accumulation in the brain. The study also demonstrated that enhancing ABCA1 function led to the increased lipidation of APOE-containing vesicles and clearance of Aβ from the brain [183].
  • ABCA7
    There are several ABCA7 gene variants associated with an increased risk for AD. Most of these variations influence the risk for AD depending on ethnic background. In general, ABCA7 loss-of-function variants lead to increased AD risk [192,193,213,214,215]. A putative protective coding variant (p. G215S) was identified in people of British and North American ancestry [216]. Another genomic study found that increased ABCA7 expression due to the rs3764650T allele reduces the risk of AD, suggesting that dysfunction of the ABCA7 transporter may contribute to the accumulation of Aβ in the brain [217].
  • ABCB1
    It has been demonstrated that Aβ is transported by ABCB1 in vitro [218], and Aβ clearance deficiency is associated with a low expression of the transporter in vivo [11,29]. Thus, the ABCB1 transporter seems to be directly implicated in AD pathogenesis. At the same time, Aβ reduces the expression of ABCB1, leading to increased Aβ deposition and exacerbated disease [11,219]. A study investigating the potential therapeutic role of targeting ABCB1 ubiquitination in Tg2576 mice demonstrated that preventing ubiquitination enhanced ABCB1 function and led to the increased clearance of Aβ, resulting in decreased Aβ brain levels [220]. Another study, a post mortem neuropathological study suggested that dysfunction or the decreased expression of ABCB1 could lead to the build-up of Aβ in the walls of cerebral blood vessels, contributing to the pathogenesis of CAA [11]. In summary, many findings to date emphasize the role of ABCB1 in regulating Aβ levels in the brain and, thereby, its potential relevance for understanding the early mechanisms of AD development.
  • ABCC1
    ABCC1 has been implicated as an important early modulator of AD onset, already before the deposition of Aβ starts. Krohn et al. [197] studied different mouse models lacking various ABC transporters (ABCB1 or ABCC1) and/or the protease neprilysin (NEP), a major Aβ-degrading enzyme. NEP deficiency itself induces higher endogenous mouse Aβ levels. Mice lacking both ABCC1 and NEP showed a marked early astrocyte reaction upon increased mouse Aβ, specifically in the dentate gyrus and amygdala regions, the locations where AD pathology is first seen in patients. Thus, this NEP × ABCC1 double-knockout model represents the first non-transgenic, preclinical AD mouse model [197].
    It has also been described that ABCC1 deficiency leads to increased Aβ levels in APPtg mice (APPPS1-21 model), while its activation, using thiethylperazine, reduces Aβ load [12]. These results suggest that ABCC1 transporters may play a role during AD development and could be a target for the modulation of disease onset, as well as a diagnostic tool for functional PET imaging [221,222].
  • ABCG2
    ABCG2 was shown to directly interact with Aβ in AD. For example, a study revealed that this transporter controls the entry of peripheral Aβ into the CNS [196]. It was shown that ABCG2 is upregulated in human cerebral vessels and in the AD mouse brain, and that its dysfunction may contribute to the accumulation of TAU in the brain [223]. The ABCG2 C421A polymorphism has been associated with an increased susceptibility to AD, particularly when combined with the APOE4 allele [224]. ABCG2 has been suggested to reduce toxicity and inflammation in the brain, which are key components in the pathology of AD [225]. Moreover, studies have found that the brain distribution of dual ABCB1/ABCG2 substrates is unchanged in an AD mouse model (APPPS1-21), indicating that the brain distribution of clinically used ABCB1/ABCG2 substrate drugs may not differ between AD patients and healthy individuals [226].
In summary, recent research has highlighted the importance of ABC transporters in the clearance of Aβ and possibly also TAU, two proteins that are implicated in the pathogenesis of AD [227,228]. Targeting these transporters may offer a promising therapeutic strategy for the treatment of AD. To assess potential therapeutic implications of ABC transporters, further research is needed. This will also lead to a better understanding of the precise roles of these transporters in the clearance of Aβ and TAU.

2.1.2. ABC Transporters in Movement Disorders

  • α-Synucleinopathies
    ABCB1 plays a role in α-synucleinopathies, such as PD. Mutations related to ABCB1 have been associated with the risk of developing PD. For example, in an ethnic Chinese population, genetic ABCB1 variations [SNPs—e12/1236(C/T), e21/2677(G/T/A), and e26/3435(C/T)] were shown to increase the risk of PD [229]. However, this genotype did not reach statistical significance in European populations [230,231]. In another study, also in an ethnic Chinese population, genetic variation regarding ABCB1 (SNPs e21/2677T and e26/3435T) was associated with protection against the risk of developing PD [232].
    On the other hand, exposition to pesticides is a risk factor for developing this pathology. This risk becomes especially noticeable in people carrying the 3435T allele of the ABCB1 gene, which is associated with a decreased pump function [233].
    Using PET (11C-Verapamil tracer), Bartels and colleagues demonstrated the decreased BBB function of ABCB1 in disease-specific brain regions at later disease stages of PD, as well as multiple system atrophy (MSA) and Lewy body dementia (LBD) [19,234]. It was proposed that the accumulation of toxic substances, such as pesticides but also aggregated proteins, due to the loss of ABCB1 function associated with aging, is a key incident in the development of PD and other neurodegenerative diseases [18,235].
  • Huntington’s Disease
    Studies have indicated that cholesterol homeostasis may be disrupted in individuals with HD, potentially influencing disease progression. These abnormalities in cholesterol metabolism could impact neuronal function and, thus, contribute to the neurodegenerative processes seen in HD [20,236]. Given the abundant amount of evidence delineating the regulation of cholesterol by ABC transporters [20,237,238,239], the respective transporters involved in cholesterol efflux may be targets for HD modulation and treatment. For example, silencing astrocytic ABCA1 resulted in poor neurite outgrowth in HD neurons [21]. Furthermore, ABCA1 and ABCG4 are reduced in primary astrocytes from R6/2 and YAC128 mice, two mouse models mimicking many features of HD [20]. A study using the R6/2 mouse model found that ABCB10 is also implicated in modulating HD pathology, participating in the UPRmt pathway. ABCB10 is downregulated in HD cells, and its depletion is related to cell death and the production of reactive oxygen species [173]. On the other hand, ABCB10 overexpression has shown the opposite effect, presenting itself as a therapeutic opportunity [173]. Moreover, ABCD1 might be involved in HD due to its role in reacting to oxidative stress, which is a key factor in HD pathology [240].
    A transcriptome-wide association study (TWAS) identified several genomic modifiers of HD onset [241]. The findings of this study indirectly highlight the significance of exploring ABCA7 in HD, given its prominent association to AD [189] and its close genetic location 5′ of the detected HMHA1 locus. This connection implies the general potential relevance of ABCA7 for the modulation of neurodegenerative mechanisms. At the same time, the genomic region encompassing HMHA1 and ABCA7 emerges as a critical area for further research into the molecular underpinnings of HD [241].
    The exact mechanisms and specific roles of ABC transporters in HD are still under investigation by us. Utilizing ABC transporter ko mice to examine modulatory effects of these transporters addresses a critical knowledge gap in the understanding of the role of ABC transporters in HD onset and progression. Very recently, the longitudinal characterization of ABCA7-deficient HD mice showed delaying effects on HD onset and progression in a sex-dependent manner. An understanding of the molecular mechanisms could have implications for new therapeutic strategies. Modulating the ABC transporter activity may offer novel approaches for addressing abnormal or dysfunctional lipid metabolism, protein transport, and neurodegenerative processes associated with HD. While some studies suggest altered cholesterol levels or metabolism in HD patients [242], the mechanisms involved are still the subject of investigation, especially to differentiate whether these changes are a cause or consequence of the disease. Further research is needed to uncover the precise mechanisms and therapeutic potential of targeting ABC transporters in HD.

2.2. Other, Non-Inflammatory Brain Diseases

2.2.1. White Matter Disorders—Adrenoleukodystrophy ABCD1

As stated before, mutations in ABCD1 are directly implicated in X-linked adrenoleukodystrophy. Mutations affecting ABCD1 cause an accumulation of very long-chain fatty acids in the brain, the spinal cord, and the adrenal gland cortex, leading to neurological and endocrine dysfunctions. More than 800 mutations have been described for this disease [243,244,245].

2.2.2. ABC Transporters in Epilepsy

The atypical expression and/or function of ABC transporters are assumed to contribute to refractory epilepsy [246], likely due to the role of inflammation on ABC transporter modulation [247,248]. Furthermore, the expression and activity of efflux transporters are increased, both in epilepsy patients and animal models of epilepsy, thereby counteracting the increased CNS permeability typical of this pathology [23,249,250,251,252,253,254]. This increase plays an important role for epilepsy treatment strategies. Since these transporters are implicated in drug efflux, anti-epileptic treatment becomes less effective [249,255,256,257,258,259,260,261]. Additionally, ABC transporters may also play a direct role in the development of pathologies related to epilepsy, such as hippocampal sclerosis (HS) and cortical dysplasia (CD) [24,251,262].

Hippocampal Sclerosis of Epilepsy and Aging

Hippocampal sclerosis is a common pathology encountered in mesial temporal lobe epilepsy and other epilepsy syndromes, and is defined by neuronal loss and reactive astrocyte proliferation [263]. In drug-resistant epilepsy patients, the overexpression of ABCB1 and ABCC1 was found on reactive astrocytes and the overexpression of ABCB1-3 and ABCC4 was detected on endothelial cells [249,264,265,266].
Hippocampal sclerosis of aging (HS-aging) is related to a genetic risk locus, which is an ABCC9 SNP pair (rs704178 and rs704180). Loss of function of the ABCC9 transporter, which is located on pericyte membranes, contributes to the disruption of the neurovascular unit followed by the development of cognitive impairment [267,268].
  • Cortical Dysplasia
    Cortical dysplasia [269] arises from abnormal stem/progenitor cell function, in which ABC transporters are involved [270,271], and could account for seizure relapse after epilepsy surgery [272]. ABCB1 and ABCC1 have been detected in human tissue samples of focal cortical dysplasia and benign tumor lesions in therapy-refractory epilepsy patients [23,262,273].
  • Resistance to Treatment
    ABC transporters are considered the most clinically important drug efflux pumps in drug-resistant epilepsy [249]. ABCB1 plays a role in oxcarbazepine resistance, as evidenced by the finding that brain levels of this transporter in treated epilepsy patients are inversely proportional to the levels of oxcarbazepine active metabolites [71,274]. Further corroborating this relationship, it has been shown that the co-administration of an ABCB1 inhibitor together with oxcarbazepine leads to a decrease in pilocarpine-induced limbic motor seizures and seizure severity [261]. In animal models, ABCB1 has also been proven to mediate the access of other antiepileptic drugs to the brain [275,276,277,278,279]. In humans, ABCB1 has been shown to be overexpressed in brain tissue from medically intractable epilepsy patients, and the transporter is assumed to mediate the export of the antiepileptic drug phenytoin from the brain, leading to inadequate antiepileptic drug levels [280]. ABCB1, but also ABCC1, ABCC2, and ABCC5, expression is increased in brains from medically intractable epilepsy patients, hinting at their role in the resistance to treatments [23,24,67,249,262,280]. In fact, the co-administration of antiepileptic treatments and ABCC1 inhibitors has been shown to improve the efficacy of antiepileptic drugs such as oxcarbazepine [261], as well as increasing the levels of phenytoin [281] and carbamazepine [276]. The same has been evidenced regarding ABCC2 inhibition, which significantly improved the anticonvulsant effect of the antiepileptic drug phenytoin [282]. Of note, the excessive use of some antiepileptic drugs may induce an increased expression of ABC transporters, actually contributing to the development of drug-resistance [249,283,284,285].

2.3. ABC Transporter Function in Neuroinflammatory Diseases

2.3.1. Multiple Sclerosis

MS is a chronic autoimmune neuroinflammatory disorder characterized mainly by CNS white matter damage and secondary neurodegeneration affecting the grey matter, as reviewed in [286]. It is the most common chronic inflammatory demyelinating disease of the CNS [287,288] and one of the most common causes of neurological disabilities in young adults [289]. MS is considered an autoimmune disease in which symptoms are caused by the damage to whiter matter tracts and, thus, are related to the affected regions. The symptoms have been widely reviewed and usually include sensory, motor, and visual deficits, but also include autonomic problems such as bladder dysfunction and cognitive and affective disorders [290,291,292]. MS can be categorized into different forms, largely depending on the onset and evolution of symptoms: relapsing–remitting (RRMS), primary progressive (PPMS), or secondary progressive (SPMS) [288,290,293]. Its pathology is mainly caused by inflammation and the resulting demyelination. MS is characterized by an inflammatory process triggering a cascade of further pathological events. Briefly, activated T-cells infiltrate into the CNS and differentiate, activating and attracting more inflammatory cells. B-cells, monocytes, and macrophages also migrate through the BBB, participating in the inflammatory process. This inflammation leads to demyelination by directly damaging oligodendrocytes, which, then, is followed by secondary axonal loss. During this process, accompanying astro- and microgliosis occurs, thus astrocytes and microglia are involved in this pathology [288,294,295]. It is thought that relapses are triggered by or are related to inflammatory events which cause inflammatory cell infiltration waves into the CNS. Long-term disability due to MS is caused by the ultimate irreversible axonal damage and neuronal loss [288,294].

Alterations of ABC Transporters Modulating MS

To study the role of ABC transporters and their alterations in the progression and treatment of MS, researchers have used tissue from patients [126,127,296,297]. The course of human MS can be recapitulated to some extent in a rodent model of the disease experimental autoimmune encephalomyelitis (EAE). Reproducing this pathology in rodents appears to be a good and replicable option [126,128,296,298]. In this regard, it is important to emphasize the diverse functions that ABC transporters can facilitate from modulating cholesterol efflux, acting as drug transporters and participating in immune response modulation (see Section 1.5). The roles that different ABC transporters might play in MS are further described below.
Stearoyl-CoA desaturase-1 (SCD1) is an essential enzyme for the conversion of saturated into mono-unsaturated fatty acids. In MS, the sustained accumulation of myelin in phagocytes promotes a shift in their reparative phenotype through SCD1. It has been reported that an increase in SCD1 leads to a reduction of ABCA1 cell surface expression, affecting cholesterol efflux and promoting lipid accumulation, thus resulting in an inflammatory phagocyte phenotype [299]. In addition, SCD1 inhibition or deficiency prevented ABCA1 reduction and correlated with an improvement in re-myelination [299].
Regarding ABCB1, the drug-induced functional downregulation in endothelial cells (at the BBB) in active and inactive lesions of EAE revealed beneficial effects on nerve loss [300]. The loss of ABCB1, as seen in astrocytes, might play a beneficial role for treatment, improving CNS access for some medications. On the other hand, this reduction may also be implicated in increased neurotoxicity [127].
ABCC1 is increased in foamy macrophages and reactive astrocytes, where it plays a role for their CCL2-secretion during the course of MS, which has a detrimental role [127]. Targeting astrocytic SUR1-TRPM4/ABCC8 could be a promising therapeutic approach due to its inhibition during EAE [138,301].
On the other hand, it has been described that peroxisomal membrane protein 70 (ABCD3, PMP70) is reduced in the grey matter of MS patients. This alteration could be related to direct peroxisomal changes affecting MS onset and development [297].
Notably, RRMS patients have a diminished cholesterol efflux via ABCG1, both at serum and cellular levels. Furthermore, these patients also present with decreased monocyte mRNA levels of ABCG1. A reduction in cholesterol efflux due to the dysregulation of ABCG1 might result in a disproportionate immune response and an exacerbated progression of the disease [302] since altered cholesterol homeostasis is implicated in inflammation and cellular inflammatory phenotypes.
On the other hand, ABCG2 expression in foamy macrophages in MS was found to be increased [127]. This transporter plays a role in the teriflunomide treatment of EAE in mice. Here, it has been shown that, in ABCG2-deficient mice, therapeutic effects of the treatment were observed after using a lower dose of the drug compared to mice with a functional ABCG2 transporter [303]. Furthermore, the ABCG2 transporter is also implicated in mitoxantrone treatment, which is used as an escalation therapy in highly active MS [304]. Therefore, the modulation of ABCG2 could help to improve the treatment effectiveness.
In summary, ABC transporters appear to be crucially involved in various aspects of MS pathology, and, thus, their regulation and modulation could present a promising strategy to better study and understand, as well as more effectively treat, this disease.

2.3.2. Detecting Clinical Effects of ABC Transporter Deficiency in EAE, a Murine Model of MS

To examine the effects and roles of different ABC transporters in neuroinflammatory diseases, we employed the murine model for human MS, EAE. Active EAE was induced in ABCB1a/b, ABCC1, and ABCA7 knockout mice, respectively, using age- and gender-matched C57BL/6J mice as controls.
ABCB1a/b, ABCC1, and ABCA7 deficiency all lead to a significantly reduced disease burden and this improved the clinical EAE course in the respective ko lines as compared to C57BL/6 controls, starting in and lasting for all or much of the early effector phase (Figure 4, Figure 5 and Figure 6, [305]). Regarding ABCB1 deficiency, our results, thus, recapitulate and corroborate previous data by Kooji et al. who also demonstrated an improved clinical EAE course in the absence of the ABCB1 transporter [128].
In summary, our results confirm the involvement of these three ABC transporters in the modulation of induced neuroinflammation. However, the precise mechanisms underlying this modulation need to be elucidated. One can assume that these are related to the known roles of ABC transporters in the immune response as described in Section 1.5 of this article. Our results also point to ABCB1, ABCC1, and ABCA7 as potentially useful targets for modulating the MS disease course in patients.

3. General Physiological Effects of ABC Transporters on Modifying Diseases

3.1. Export of Toxic Metabolites/Peptides

ABC transporters play a crucial role as defensive elements by transporting toxic metabolites from the space surrounding the brain vessels, e.g., the glymphatic space, back into the blood flow. Their role in the clearance of toxic metabolites is important in a number of neurodegenerative disorders, such as AD and PD, and has been described in detail elsewhere [19,235,306,307,308].
Other less recognized findings include the following:
  • The amount of the ABC transporter ABCB1 was found to be reduced in Creutzfeldt–Jakob Disease (CJD) and could be involved in removing misfolded prion protein [309].
  • The development of alimentary Progressive Supranuclear Palsy (PSP) from Annonaceae fruit intake has direct neurotoxic effects, but also leads to the accumulation of harmful molecules through ABCB1 inhibition [310,311].

3.2. Examples of Molecular Mechanisms Influenced by ABC Transporters

ABC transporters are crucial in several cellular compartments, from plasma membrane, lysosomes, and peroxisomes, to mitochondria, where the disruption of their function can influence various molecular pathways/mechanisms and induce diseases.
  • As already discussed, dysfunction of ABCD1 leads to ALD due to a defective transport of very-long-chain fatty acids into peroxisomes, hindering their degradation [312].
  • Mutations in ABCD4 alter vitamin B12 metabolism due to a failure of its release from lysosomes [313].
  • ABCB9 was implicated in the release of peptides from lysosomes [314].
  • ABC B subfamily transporters are implicated in mitochondrial mechanisms:
    ABCB6 is implicated in porphyrin transport into mitochondria and heme biosynthesis [315,316]. Its deficiency is associated with increased mortality in mice under extreme demand in porphyrin conditions [317].
    The ABCB7 transporter is essential for iron metabolism. It is needed for hematopoiesis, and its mutation causes X-linked sideroblastic anemia and ataxia [318,319,320,321].
    ABCB8 is also implicated in iron mechanisms, particularly in iron export from mitochondria [322].
    ABCB10 is found in mitochondria handling ROS [323].
  • ABC transporters of the ABC C subfamily have been linked to the functionality and modulation of ion channels. For instance, ABCC7, also known as cystic fibrosis transmembrane conductance regulator, is a chloride channel itself [324]. In addition, ABCC8 and ABCC9, also known as SUR1 and SUR2, respectively, are modulators of potassium channels [325,326,327].
  • The role of several ABC transporters, mainly of the ABC A subfamily, is widely recognized for the lipid transport of various lipid classes [328,329]. Involvement in lipid transport provides an immediate influence on all pathways of the cell’s signaling systems through direct interaction with signaling molecules, e.g., fatty acids or steroid hormones, or through indirect interaction via the fluidity of membranes, e.g., cholesterol content and the movement/clustering of transmembrane receptors. ABCA1 has been described to transport cholesterol and phospholipids, essential membrane lipids [330,331]. ABCA2 has been suggested to play a role in sterol transport [332]. ABCA3 is implicated in lipid metabolism and phospholipid homeostasis for surfactants [333,334,335]. ABCA4 has been described to import N-retinylidene-phosphatidylethanolamine and phosphatidylethanolamine [43].
The link between general molecular processes involving lipids, especially membrane lipids, and sterol-related mechanisms is important to address and will be further described in the next section.

3.2.1. Modulation of Steroid Hormone Signaling by ABC Transporters

Steroid hormone regulation has been widely reviewed to play a role in the development of neurodegenerative diseases [336,337]. Sex hormones control neuron proliferation and differentiation (anabolic effects), and changes in hormone balances during andropause and menopause are linked to neurodegenerative diseases [338,339,340]. For example, changes in estradiol levels during the female lifespan are thought to be related to the increased risk of AD in women as compared to men [341].
Some ABC transporters are able to modulate the traffic of steroid hormones and their metabolites [342,343]. ABCB1 has been shown to transport cortisol and aldosterone [344]. The ABC C subfamily is implicated in steroid hormone regulation. ABCC1 plays a role in regulating steroid hormone homeostasis, and the lack of ABCC1 causes a reduction in concentrations of testicular steroids [345]. Moreover, ABCC1 transports estrone 3-sulfate and dehydroepiandrosterone 3-sulphate (DHEAS) in the presence of glutathione [346,347]. ABCC3, but also ABCC2, has been shown to transport some metabolites of testosterone and androgen glucuronides [348,349]. For instance, ABCC3 transports estrogen, β-estradiol 17-(β-D-glucuronide) (E217βG), and dehydroepiandrosterone (DHEA), which is the precursor molecule of different sex hormones, including testosterone and estradiol [350,351]. It has been found that ABCC4 also transports DHEA and conjugated steroids [347]. Furthermore, ABCC8 also transports steroid sulfates such as DHEA and E217βG [352]. ABCG2, however, has been demonstrated to transport sulfate-conjugates of estrogens, but not free estrone nor 17β-estradiol [353].
It is important to consider the relevance of cholesterol in the metabolism of steroid hormones, since it can serve as a precursor for the synthesis of steroid hormones or neurosteroids [354]. The intriguing findings of disease-delaying effects in male and even disease-preventing effects in female HD mice (model zQ175dn [239]) upon the knockout of ABCA7 function demand a more intense exploration of the multifaceted roles of ABC transporters, particularly those belonging to the ABC A subfamily. Beyond their established functions in lipid transport and lipid homeostasis [42], emerging evidence suggests potential links between ABC transporters and the steroid hormone signaling modulation of the body’s anabolic vs. catabolic balance. Future investigations exploring the specific mechanisms by which ABC A subfamily transporters impact steroid hormone pathways may yield valuable information for developing targeted interventions in the context of HD and other illnesses.

4. Summary

In conclusion, we here reviewed the current knowledge about the expression and function of different ABC transporters in glial and various other cells in the CNS. We, furthermore, delineated the importance of ABC transporters for several neurodegenerative and also neuroinflammatory diseases. Finally, we illustrated the promising roles that modulating the expression and function of these transporters could have on the treatment and development of neurodegenerative and neuroinflammatory CNS diseases.

Author Contributions

Conceptualization: J.P.; methodology: M.V., J.W. and S.H.; validation: S.H.; formal analysis: S.H.; investigation: S.H.; resources: J.P.; data curation: S.H. and J.P.; writing—original draft: M.V., J.W. and J.P.; writing—review and editing: all; visualization: M.V., S.H. and J.P.; supervision: J.P.; project administration: J.P.; funding acquisition: J.P. All authors have read and agreed to the published version of the manuscript.

Funding

J.P. received funding from the Deutsche Forschungsgemeinschaft/Germany (DFG, #263024513); Nasjonalforeningen (#16154), HelseSØ/Norway (#2019054, #2019055, #2022046); Barnekreftforeningen (#19008); EEA grant/Norges grants (TAČR, TARIMAD #TO01000078); and Norges forskningsrådet/Norway (#295910 NAPI, #327571 PETABC). PETABC is an EU Joint Programme—Neurodegenerative Disease Research (JPND) project. PETABC is supported through the following funding organizations under the aegis of JPND—www.jpnd.eu (NFR #327571—Norway, FFG #882717—Austria, BMBF #01ED2106—Germany, MSMT #8F21002–Czech Republic, #ES RTD/2020/26—Latvia, ANR #20-JPW2-0002-04—France, SRC #2020-02905—Sweden). The Norwegian Advance Proteomics Infrastructure (NAPI, www.napi.uio.no, accessed on 20 April 2024) is a National Infrastructure Initiative.

Institutional Review Board Statement

The animal study protocol was approved by the Forsøksdyrutvalget and Mattilsynet (FOTS #10678, 21 January 2017) for studies involving animals.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data files and figures can be downloaded from the ‘Pahnke Lab-open access’ project at https://dx.doi.org/10.17605/OSF.IO/VWQ58.

Acknowledgments

We thank Ivan Eiriz for the support with animal caretaking and breeding. We thank Anna Maria Górska and Irene Santos-García for the comments on the manuscript.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

Aβ—amyloid-β, ABC—ATP-binding cassette, AD—Alzheimer’s disease, ALD—adrenoleukodystrophy, APP—amyloid-β precursor protein, APOE—apolipoprotein E, ASYN—α-synuclein, BBB—blood–brain barrier, BCPB—blood–choroid plexus barrier, CAA—cerebral amyloid angiopathy, CCL2—C-C motif chemokine ligand 2, CD—cluster of differentiation/cortical dysplasia, CNS—central nervous system, DC—dendritic cell, EAE—experimental autoimmune encephalomyelitis, ERK—extracellular signal-regulated kinase, FTD—frontotemporal dementia, GFAP—glial fibrillary acid protein, HD—Huntington’s disease, HS—hippocampal sclerosis, HTT—Huntingtin, ko—knockout, LBD—Lewy body disease, MDR—multi-drug resistance, MDR1—multi-drug resistance 1 (ABCB1, P-gp), MRP—multi-drug resistance-related, MRP1—multi-drug resistance-related 1 (ABCC1), MS—multiple sclerosis, NKT—natural killer T (-cell), P-gp—P-glycoprotein (ABCB1, MDR1), PAF—platelet-activating factor, PD—Parkinson’s disease, PPMS—primary progressive multiple sclerosis, PSP—progressive supranuclear palsy, RRMS—remittent–relapsing multiple sclerosis, SCD1—stearoyl-CoA desaturase-1, SNP—single nucleotide polymorphism, SPMS—secondary progressive multiple sclerosis, Treg—regulatory T-cell.

References

  1. Thakur, K.T.; Albanese, E.; Giannakopoulos, P.; Jette, N.; Linde, M.; Prince, M.J.; Steiner, T.J.; Dua, T. Neurological Disorders. In Mental, Neurological, and Substance Use Disorders: Disease Control Priorities, 3rd ed.; Patel, V., Chisholm, D., Dua, T., Laxminarayan, R., Medina-Mora, M.E., Eds.; International Bank for Reconstruction and Development/The World Bank: Washington, DC, USA, 2016; Volume 4. [Google Scholar]
  2. Feigin, V.L.; Nichols, E.; Alam, T.; Bannick, M.S.; Beghi, E.; Blake, N.; Culpepper, W.J.; Dorsey, E.R.; Elbaz, A.; Ellenbogen, R.G.; et al. Global, regional, and national burden of neurological disorders, 1990–2016: A systematic analysis for the Global Burden of Disease Study 2016. Lancet Neurol. 2019, 18, 459–480. [Google Scholar] [CrossRef]
  3. Kim, W.S.; Weickert, C.S.; Garner, B. Role of ATP-binding cassette transporters in brain lipid transport and neurological disease. J. Neurochem. 2008, 104, 1145–1166. [Google Scholar] [CrossRef]
  4. Katzeff, J.S.; Kim, W.S. ATP-binding cassette transporters and neurodegenerative diseases. Essays Biochem. 2021, 65, 1013–1024. [Google Scholar]
  5. Pahnke, J.; Wolkenhauer, O.; Krohn, M.; Walker, L.C. Clinico-pathologic function of cerebral ABC transporters—Implications for the pathogenesis of Alzheimer’s disease. Curr. Alzheimer Res. 2008, 5, 396–405. [Google Scholar] [CrossRef]
  6. Miller, D.S. Regulation of ABC transporters at the blood-brain barrier. Clin. Pharmacol. Ther. 2015, 97, 395–403. [Google Scholar] [CrossRef]
  7. Moitra, K.; Dean, M. Evolution of ABC transporters by gene duplication and their role in human disease. Biol. Chem. 2011, 392, 29–37. [Google Scholar] [CrossRef]
  8. Locher, K.P. Review. Structure and mechanism of ATP-binding cassette transporters. Philos. Trans. R. Soc. Lond. B Biol. Sci. 2009, 364, 239–245. [Google Scholar] [CrossRef]
  9. Bernstein, H.G.; Hildebrandt, J.; Dobrowolny, H.; Steiner, J.; Bogerts, B.; Pahnke, J. Morphometric analysis of the cerebral expression of ATP-binding cassette transporter protein ABCB1 in chronic schizophrenia: Circumscribed deficits in the habenula. Schizophr. Res. 2016, 177, 52–58. [Google Scholar] [CrossRef]
  10. Bernstein, H.G.; Holzl, G.; Dobrowolny, H.; Hildebrandt, J.; Trubner, K.; Krohn, M.; Bogerts, B.; Pahnke, J. Vascular and extravascular distribution of the ATP-binding cassette transporters ABCB1 and ABCC1 in aged human brain and pituitary. Mech. Ageing Dev. 2014, 141–142, 12–21. [Google Scholar] [CrossRef]
  11. Vogelgesang, S.; Warzok, R.W.; Cascorbi, I.; Kunert-Keil, C.; Schroeder, E.; Kroemer, H.K.; Siegmund, W.; Walker, L.C.; Pahnke, J. The role of P-glycoprotein in cerebral amyloid angiopathy; implications for the early pathogenesis of Alzheimer’s disease. Curr. Alzheimer Res. 2004, 1, 121–125. [Google Scholar] [CrossRef]
  12. Krohn, M.; Lange, C.; Hofrichter, J.; Scheffler, K.; Stenzel, J.; Steffen, J.; Schumacher, T.; Bruning, T.; Plath, A.S.; Alfen, F.; et al. Cerebral amyloid-beta proteostasis is regulated by the membrane transport protein ABCC1 in mice. J. Clin. Investig. 2011, 121, 3924–3931. [Google Scholar] [CrossRef]
  13. Pahnke, J.; Langer, O.; Krohn, M. Alzheimer’s and ABC transporters--new opportunities for diagnostics and treatment. Neurobiol. Dis. 2014, 72 Pt A, 54–60. [Google Scholar] [CrossRef]
  14. Dib, S.; Pahnke, J.; Gosselet, F. Role of ABCA7 in Human Health and in Alzheimer’s Disease. Int. J. Mol. Sci. 2021, 22, 4603. [Google Scholar] [CrossRef]
  15. Vautier, S.; Fernandez, C. ABCB1: The role in Parkinson’s disease and pharmacokinetics of antiparkinsonian drugs. Expert Opin. Drug Metab. Toxicol. 2009, 5, 1349–1358. [Google Scholar] [CrossRef]
  16. Narayan, S.; Sinsheimer, J.S.; Paul, K.C.; Liew, Z.; Cockburn, M.; Bronstein, J.M.; Ritz, B. Genetic variability in ABCB1, occupational pesticide exposure, and Parkinson’s disease. Environ. Res. 2015, 143 Pt A, 98–106. [Google Scholar] [CrossRef]
  17. Haussermann, P.; Kuhn, W.; Przuntek, H.; Muller, T. Integrity of the blood-cerebrospinal fluid barrier in early Parkinson’s disease. Neurosci. Lett. 2001, 300, 182–184. [Google Scholar] [CrossRef]
  18. Bartels, A.L.; Kortekaas, R.; Bart, J.; Willemsen, A.T.; de Klerk, O.L.; de Vries, J.J.; van Oostrom, J.C.; Leenders, K.L. Blood-brain barrier P-glycoprotein function decreases in specific brain regions with aging: A possible role in progressive neurodegeneration. Neurobiol. Aging 2009, 30, 1818–1824. [Google Scholar] [CrossRef]
  19. Bartels, A.L.; Willemsen, A.T.; Kortekaas, R.; de Jong, B.M.; de Vries, R.; de Klerk, O.; van Oostrom, J.C.; Portman, A.; Leenders, K.L. Decreased blood-brain barrier P-glycoprotein function in the progression of Parkinson’s disease, PSP and MSA. J. Neural Transm. 2008, 115, 1001–1009. [Google Scholar] [CrossRef]
  20. Leoni, V.; Caccia, C. The impairment of cholesterol metabolism in Huntington disease. Biochim. Biophys. Acta 2015, 1851, 1095–1105. [Google Scholar] [CrossRef]
  21. Valenza, M.; Marullo, M.; Di Paolo, E.; Cesana, E.; Zuccato, C.; Biella, G.; Cattaneo, E. Disruption of astrocyte-neuron cholesterol cross talk affects neuronal function in Huntington’s disease. Cell Death Differ. 2015, 22, 690–702. [Google Scholar] [CrossRef]
  22. Gil-Martins, E.; Barbosa, D.J.; Silva, V.; Remiao, F.; Silva, R. Dysfunction of ABC transporters at the blood-brain barrier: Role in neurological disorders. Pharmacol. Ther. 2020, 213, 107554. [Google Scholar] [CrossRef]
  23. Dombrowski, S.M.; Desai, S.Y.; Marroni, M.; Cucullo, L.; Goodrich, K.; Bingaman, W.; Mayberg, M.R.; Bengez, L.; Janigro, D. Overexpression of multiple drug resistance genes in endothelial cells from patients with refractory epilepsy. Epilepsia 2001, 42, 1501–1506. [Google Scholar] [CrossRef]
  24. Aronica, E.; Gorter, J.A.; Ramkema, M.; Redeker, S.; Ozbas-Gerceker, F.; van Vliet, E.A.; Scheffer, G.L.; Scheper, R.J.; van der Valk, P.; Baayen, J.C.; et al. Expression and cellular distribution of multidrug resistance-related proteins in the hippocampus of patients with mesial temporal lobe epilepsy. Epilepsia 2004, 45, 441–451. [Google Scholar] [CrossRef]
  25. Qosa, H.; Mohamed, L.A.; Alqahtani, S.; Abuasal, B.S.; Hill, R.A.; Kaddoumi, A. Transporters as Drug Targets in Neurological Diseases. Clin. Pharmacol. Ther. 2016, 100, 441–453. [Google Scholar] [CrossRef]
  26. Moore, J.M.; Bell, E.L.; Hughes, R.O.; Garfield, A.S. ABC transporters: Human disease and pharmacotherapeutic potential. Trends Mol. Med. 2023, 29, 152–172. [Google Scholar] [CrossRef]
  27. Elali, A.; Rivest, S. The role of ABCB1 and ABCA1 in beta-amyloid clearance at the neurovascular unit in Alzheimer’s disease. Front. Physiol. 2013, 4, 45. [Google Scholar] [CrossRef]
  28. Aykac, A.; Sehirli, A.O. The Function and Expression of ATP-Binding Cassette Transporters Proteins in the Alzheimer’s Disease. Glob. Med. Genet. 2021, 8, 149–155. [Google Scholar] [CrossRef]
  29. Vogelgesang, S.; Cascorbi, I.; Schroeder, E.; Pahnke, J.; Kroemer, H.K.; Siegmund, W.; Kunert-Keil, C.; Walker, L.C.; Warzok, R.W. Deposition of Alzheimer’s beta-amyloid is inversely correlated with P-glycoprotein expression in the brains of elderly non-demented humans. Pharmacogenetics 2002, 12, 535–541. [Google Scholar] [CrossRef]
  30. Kuhn, J.; Sharman, T. Cerebral Amyloid Angiopathy; StatPearls: Treasure Island, FL, USA, 2024. [Google Scholar]
  31. Bigbee, J.W. Cells of the Central Nervous System: An Overview of Their Structure and Function. Adv. Neurobiol. 2023, 29, 41–64. [Google Scholar]
  32. Ludwig, P.E.; Reddy, V.; Varacallo, M. Neuroanatomy, Neurons; StatPearls: Treasure Island, FL, USA, 2024. [Google Scholar]
  33. Erecinska, M.; Silver, I.A. Ions and energy in mammalian brain. Prog. Neurobiol. 1994, 43, 37–71. [Google Scholar] [CrossRef]
  34. Gleichmann, M.; Mattson, M.P. Neuronal calcium homeostasis and dysregulation. Antioxid. Redox Signal 2011, 14, 1261–1273. [Google Scholar] [CrossRef] [PubMed]
  35. Teleanu, R.I.; Niculescu, A.G.; Roza, E.; Vladacenco, O.; Grumezescu, A.M.; Teleanu, D.M. Neurotransmitters-Key Factors in Neurological and Neurodegenerative Disorders of the Central Nervous System. Int. J. Mol. Sci. 2022, 23, 5954. [Google Scholar] [CrossRef] [PubMed]
  36. Verkhratsky, A.; Nedergaard, M.; Hertz, L. Why are astrocytes important? Neurochem. Res. 2015, 40, 389–401. [Google Scholar] [CrossRef] [PubMed]
  37. Egawa, J.; Pearn, M.L.; Lemkuil, B.P.; Patel, P.M.; Head, B.P. Membrane lipid rafts and neurobiology: Age-related changes in membrane lipids and loss of neuronal function. J. Physiol. 2016, 594, 4565–4579. [Google Scholar] [CrossRef] [PubMed]
  38. Hussain, G.; Wang, J.; Rasul, A.; Anwar, H.; Imran, A.; Qasim, M.; Zafar, S.; Kamran, S.K.S.; Razzaq, A.; Aziz, N.; et al. Role of cholesterol and sphingolipids in brain development and neurological diseases. Lipids Health Dis. 2019, 18, 26. [Google Scholar] [CrossRef] [PubMed]
  39. Bauer, B.; Hartz, A.M.; Pekcec, A.; Toellner, K.; Miller, D.S.; Potschka, H. Seizure-induced up-regulation of P-glycoprotein at the blood-brain barrier through glutamate and cyclooxygenase-2 signaling. Mol. Pharmacol. 2008, 73, 1444–1453. [Google Scholar] [CrossRef] [PubMed]
  40. Bortfeld, M.; Rius, M.; Konig, J.; Herold-Mende, C.; Nies, A.T.; Keppler, D. Human multidrug resistance protein 8 (MRP8/ABCC11), an apical efflux pump for steroid sulfates, is an axonal protein of the CNS and peripheral nervous system. Neuroscience 2006, 137, 1247–1257. [Google Scholar] [CrossRef]
  41. Coleman, J.A.; Quazi, F.; Molday, R.S. Mammalian P4-ATPases and ABC transporters and their role in phospholipid transport. Biochim. Biophys. Acta 2013, 1831, 555–574. [Google Scholar] [CrossRef]
  42. Quazi, F.; Molday, R.S. Lipid transport by mammalian ABC proteins. Essays Biochem. 2011, 50, 265–290. [Google Scholar]
  43. Quazi, F.; Lenevich, S.; Molday, R.S. ABCA4 is an N-retinylidene-phosphatidylethanolamine and phosphatidylethanolamine importer. Nat. Commun. 2012, 3, 925. [Google Scholar] [CrossRef]
  44. Harris, M.T.; Hussain, S.S.; Inouye, C.M.; Castle, A.M.; Castle, J.D. Reinterpretation of the localization of the ATP binding cassette transporter ABCG1 in insulin-secreting cells and insights regarding its trafficking and function. PLoS ONE 2018, 13, e0198383. [Google Scholar] [CrossRef] [PubMed]
  45. Liu, H.; Cheng, M.; Zhao, S.; Lin, C.; Song, J.; Yang, Q. ATP-Binding Cassette Transporter Regulates N,N’-diacetylchitobiose Transportation and Chitinase Production in Trichoderma asperellum T4. Int. J. Mol. Sci. 2019, 20, 2412. [Google Scholar] [CrossRef] [PubMed]
  46. Hsieh, V.; Kim, M.J.; Gelissen, I.C.; Brown, A.J.; Sandoval, C.; Hallab, J.C.; Kockx, M.; Traini, M.; Jessup, W.; Kritharides, L. Cellular cholesterol regulates ubiquitination and degradation of the cholesterol export proteins ABCA1 and ABCG1. J. Biol. Chem. 2014, 289, 7524–7536. [Google Scholar] [CrossRef] [PubMed]
  47. Abe-Dohmae, S.; Ikeda, Y.; Matsuo, M.; Hayashi, M.; Okuhira, K.; Ueda, K.; Yokoyama, S. Human ABCA7 supports apolipoprotein-mediated release of cellular cholesterol and phospholipid to generate high density lipoprotein. J. Biol. Chem. 2004, 279, 604–611. [Google Scholar] [CrossRef] [PubMed]
  48. Chen, J.; Zhang, X.; Kusumo, H.; Costa, L.G.; Guizzetti, M. Cholesterol efflux is differentially regulated in neurons and astrocytes: Implications for brain cholesterol homeostasis. Biochim. Biophys. Acta 2013, 1831, 263–275. [Google Scholar] [CrossRef] [PubMed]
  49. Tracey, T.J.; Steyn, F.J.; Wolvetang, E.J.; Ngo, S.T. Neuronal Lipid Metabolism: Multiple Pathways Driving Functional Outcomes in Health and Disease. Front. Mol. Neurosci. 2018, 11, 10. [Google Scholar] [CrossRef] [PubMed]
  50. Tarling, E.J.; de Aguiar Vallim, T.Q.; Edwards, P.A. Role of ABC transporters in lipid transport and human disease. Trends Endocrinol. Metab. TEM 2013, 24, 342–350. [Google Scholar] [CrossRef] [PubMed]
  51. Takahashi, K.; Kimura, Y.; Nagata, K.; Yamamoto, A.; Matsuo, M.; Ueda, K. ABC proteins: Key molecules for lipid homeostasis. Med. Mol. Morphol. 2005, 38, 2–12. [Google Scholar] [CrossRef] [PubMed]
  52. Kim, W.S.; Rahmanto, A.S.; Kamili, A.; Rye, K.A.; Guillemin, G.J.; Gelissen, I.C.; Jessup, W.; Hill, A.F.; Garner, B. Role of ABCG1 and ABCA1 in regulation of neuronal cholesterol efflux to apolipoprotein E discs and suppression of amyloid-beta peptide generation. J. Biol. Chem. 2007, 282, 2851–2861. [Google Scholar] [CrossRef]
  53. Davis, W., Jr. The ATP-binding cassette transporter-2 (ABCA2) regulates esterification of plasma membrane cholesterol by modulation of sphingolipid metabolism. Biochim. Biophys. Acta 2014, 1841, 168–179. [Google Scholar] [CrossRef]
  54. Le, L.T.M.; Thompson, J.R.; Dehghani-Ghahnaviyeh, S.; Pant, S.; Dang, P.X.; French, J.B.; Kanikeyo, T.; Tajkhorshid, E.; Alam, A. Cryo-EM structures of human ABCA7 provide insights into its phospholipid translocation mechanisms. EMBO J. 2023, 42, e111065. [Google Scholar] [CrossRef]
  55. Picataggi, A.; Rodrigues, A.; Cromley, D.A.; Wang, H.; Wiener, J.P.; Garliyev, V.; Billheimer, J.T.; Grabiner, B.C.; Hurt, J.A.; Chen, A.C.; et al. Specificity of ABCA7-mediated cell lipid efflux. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 2022, 1867, 159157. [Google Scholar] [CrossRef] [PubMed]
  56. Sakae, N.; Liu, C.C.; Shinohara, M.; Frisch-Daiello, J.; Ma, L.; Yamazaki, Y.; Tachibana, M.; Younkin, L.; Kurti, A.; Carrasquillo, M.M.; et al. ABCA7 Deficiency Accelerates Amyloid-beta Generation and Alzheimer’s Neuronal Pathology. J. Neurosci. 2016, 36, 3848–3859. [Google Scholar] [CrossRef] [PubMed]
  57. Sano, O.; Ito, S.; Kato, R.; Shimizu, Y.; Kobayashi, A.; Kimura, Y.; Kioka, N.; Hanada, K.; Ueda, K.; Matsuo, M. ABCA1, ABCG1, and ABCG4 are distributed to distinct membrane meso-domains and disturb detergent-resistant domains on the plasma membrane. PLoS ONE 2014, 9, e109886. [Google Scholar] [CrossRef] [PubMed]
  58. Sano, O.; Tsujita, M.; Shimizu, Y.; Kato, R.; Kobayashi, A.; Kioka, N.; Remaley, A.T.; Michikawa, M.; Ueda, K.; Matsuo, M. ABCG1 and ABCG4 Suppress gamma-Secretase Activity and Amyloid beta Production. PLoS ONE 2016, 11, e0155400. [Google Scholar] [CrossRef]
  59. Ehehalt, R.; Keller, P.; Haass, C.; Thiele, C.; Simons, K. Amyloidogenic processing of the Alzheimer beta-amyloid precursor protein depends on lipid rafts. J. Cell Biol. 2003, 160, 113–123. [Google Scholar] [CrossRef] [PubMed]
  60. Tawbeh, A.; Gondcaille, C.; Trompier, D.; Savary, S. Peroxisomal ABC Transporters: An Update. Int. J. Mol. Sci. 2021, 22, 6093. [Google Scholar] [CrossRef] [PubMed]
  61. Nakamura, Y.; Nakanishi, T.; Tamai, I. Membrane Transporters Contributing to PGE(2) Distribution in Central Nervous System. Biol. Pharm. Bull. 2018, 41, 1337–1347. [Google Scholar] [CrossRef] [PubMed]
  62. Reid, G.; Wielinga, P.; Zelcer, N.; van der Heijden, I.; Kuil, A.; de Haas, M.; Wijnholds, J.; Borst, P. The human multidrug resistance protein MRP4 functions as a prostaglandin efflux transporter and is inhibited by nonsteroidal antiinflammatory drugs. Proc. Natl. Acad. Sci. USA 2003, 100, 9244–9249. [Google Scholar] [CrossRef]
  63. Shen, S.; Zhang, W. ABC transporters and drug efflux at the blood-brain barrier. Rev. Neurosci. 2010, 21, 29–53. [Google Scholar] [CrossRef]
  64. Jablonski, M.R.; Jacob, D.A.; Campos, C.; Miller, D.S.; Maragakis, N.J.; Pasinelli, P.; Trotti, D. Selective increase of two ABC drug efflux transporters at the blood-spinal cord barrier suggests induced pharmacoresistance in ALS. Neurobiol. Dis. 2012, 47, 194–200. [Google Scholar] [CrossRef]
  65. Davis, T.P.; Sanchez-Covarubias, L.; Tome, M.E. P-glycoprotein trafficking as a therapeutic target to optimize CNS drug delivery. Adv. Pharmacol. 2014, 71, 25–44. [Google Scholar]
  66. Cox, B.; Nicolai, J.; Williamson, B. The role of the efflux transporter, P-glycoprotein, at the blood-brain barrier in drug discovery. Biopharm. Drug Dispos. 2023, 44, 113–126. [Google Scholar] [CrossRef]
  67. Loscher, W.; Potschka, H. Drug resistance in brain diseases and the role of drug efflux transporters. Nat. Rev. Neurosci. 2005, 6, 591–602. [Google Scholar] [CrossRef]
  68. Loscher, W.; Potschka, H. Role of drug efflux transporters in the brain for drug disposition and treatment of brain diseases. Prog. Neurobiol. 2005, 76, 22–76. [Google Scholar] [CrossRef] [PubMed]
  69. Feldmann, M.; Koepp, M. ABC Transporters and Drug Resistance in Patients with Epilepsy. Curr. Pharm. Des. 2016, 22, 5793–5807. [Google Scholar] [CrossRef] [PubMed]
  70. Xiong, J.; Mao, D.A.; Liu, L.Q. Research Progress on the Role of ABC Transporters in the Drug Resistance Mechanism of Intractable Epilepsy. BioMed Res. Int. 2015, 2015, 194541. [Google Scholar] [CrossRef] [PubMed]
  71. Loscher, W.; Potschka, H.; Sisodiya, S.M.; Vezzani, A. Drug Resistance in Epilepsy: Clinical Impact, Potential Mechanisms, and New Innovative Treatment Options. Pharmacol. Rev. 2020, 72, 606–638. [Google Scholar] [CrossRef] [PubMed]
  72. Bradl, M.; Lassmann, H. Oligodendrocytes: Biology and pathology. Acta Neuropathol. 2010, 119, 37–53. [Google Scholar] [CrossRef]
  73. Simons, M.; Nave, K.A. Oligodendrocytes: Myelination and Axonal Support. Cold Spring Harb. Perspect. Biol. 2015, 8, a020479. [Google Scholar] [CrossRef]
  74. Mack, J.T.; Townsend, D.M.; Beljanski, V.; Tew, K.D. The ABCA2 transporter: Intracellular roles in trafficking and metabolism of LDL-derived cholesterol and sterol-related compounds. Curr. Drug Metab. 2007, 8, 47–57. [Google Scholar] [CrossRef] [PubMed]
  75. Mack, J.T.; Beljanski, V.; Soulika, A.M.; Townsend, D.M.; Brown, C.B.; Davis, W.; Tew, K.D. “Skittish” Abca2 knockout mice display tremor, hyperactivity, and abnormal myelin ultrastructure in the central nervous system. Mol. Cell Biol. 2007, 27, 44–53. [Google Scholar] [CrossRef]
  76. Marangon, D.; Boccazzi, M.; Lecca, D.; Fumagalli, M. Regulation of Oligodendrocyte Functions: Targeting Lipid Metabolism and Extracellular Matrix for Myelin Repair. J. Clin. Med. 2020, 9, 470. [Google Scholar] [CrossRef] [PubMed]
  77. Hegyi, Z.; Homolya, L. Functional Cooperativity between ABCG4 and ABCG1 Isoforms. PLoS ONE 2016, 11, e0156516. [Google Scholar] [CrossRef] [PubMed]
  78. Wu, C.; Chakrabarty, S.; Jin, M.; Liu, K.; Xiao, Y. Insect ATP-Binding Cassette (ABC) Transporters: Roles in Xenobiotic Detoxification and Bt Insecticidal Activity. Int. J. Mol. Sci. 2019, 20, 2829. [Google Scholar] [CrossRef] [PubMed]
  79. Spaas, J.; van Veggel, L.; Schepers, M.; Tiane, A.; van Horssen, J.; Wilson, D.M., 3rd; Moya, P.R.; Piccart, E.; Hellings, N.; Eijnde, B.O.; et al. Oxidative stress and impaired oligodendrocyte precursor cell differentiation in neurological disorders. Cell Mol. Life Sci. 2021, 78, 4615–4637. [Google Scholar] [CrossRef] [PubMed]
  80. Rose, J.; Brian, C.; Woods, J.; Pappa, A.; Panayiotidis, M.I.; Powers, R.; Franco, R. Mitochondrial dysfunction in glial cells: Implications for neuronal homeostasis and survival. Toxicology 2017, 391, 109–115. [Google Scholar] [CrossRef] [PubMed]
  81. Evseenko, D.A.; Murthi, P.; Paxton, J.W.; Reid, G.; Emerald, B.S.; Mohankumar, K.M.; Lobie, P.E.; Brennecke, S.P.; Kalionis, B.; Keelan, J.A. The ABC transporter BCRP/ABCG2 is a placental survival factor, and its expression is reduced in idiopathic human fetal growth restriction. FASEB J. 2007, 21, 3592–3605. [Google Scholar] [CrossRef] [PubMed]
  82. Hartz, A.M.; Bauer, B. ABC transporters in the CNS—An inventory. Curr. Pharm. Biotechnol. 2011, 12, 656–673. [Google Scholar] [CrossRef]
  83. Fogal, B.; McClaskey, C.; Yan, S.; Yan, H.; Rivkees, S.A. Diazoxide promotes oligodendrocyte precursor cell proliferation and myelination. PLoS ONE 2010, 5, e10906. [Google Scholar] [CrossRef]
  84. Jha, R.M.; Rani, A.; Desai, S.M.; Raikwar, S.; Mihaljevic, S.; Munoz-Casabella, A.; Kochanek, P.M.; Catapano, J.; Winkler, E.; Citerio, G.; et al. Sulfonylurea Receptor 1 in Central Nervous System Injury: An Updated Review. Int. J. Mol. Sci. 2021, 22, 11899. [Google Scholar] [CrossRef] [PubMed]
  85. Mandal, S.K.; Nayak, S.G.; Kanaujia, S.P. Identification and characterization of metal uptake ABC transporters in Mycobacterium tuberculosis unveil their ligand specificity. Int. J. Biol. Macromol. 2021, 185, 324–337. [Google Scholar] [CrossRef] [PubMed]
  86. Neville, S.L.; Sjohamn, J.; Watts, J.A.; MacDermott-Opeskin, H.; Fairweather, S.J.; Ganio, K.; Carey Hulyer, A.; McGrath, A.P.; Hayes, A.J.; Malcolm, T.R.; et al. The structural basis of bacterial manganese import. Sci. Adv. 2021, 7, eabg3980. [Google Scholar] [CrossRef] [PubMed]
  87. Porcheron, G.; Garenaux, A.; Proulx, J.; Sabri, M.; Dozois, C.M. Iron, copper, zinc, and manganese transport and regulation in pathogenic Enterobacteria: Correlations between strains, site of infection and the relative importance of the different metal transport systems for virulence. Front. Cell Infect. Microbiol. 2013, 3, 90. [Google Scholar] [CrossRef] [PubMed]
  88. Lewis, V.G.; Ween, M.P.; McDevitt, C.A. The role of ATP-binding cassette transporters in bacterial pathogenicity. Protoplasma 2012, 249, 919–942. [Google Scholar] [CrossRef] [PubMed]
  89. Mandal, S.K.; Adhikari, R.; Sharma, A.; Chandravanshi, M.; Gogoi, P.; Kanaujia, S.P. Designating ligand specificities to metal uptake ABC transporters in Thermus thermophilus HB8. Metallomics 2019, 11, 597–612. [Google Scholar] [CrossRef] [PubMed]
  90. Miller, S.J. Astrocyte Heterogeneity in the Adult Central Nervous System. Front. Cell. Neurosci. 2018, 12, 401. [Google Scholar] [CrossRef] [PubMed]
  91. Cole, S.P. Multidrug resistance protein 1 (MRP1, ABCC1), a “multitasking” ATP-binding cassette (ABC) transporter. J. Biol. Chem. 2014, 289, 30880–30888. [Google Scholar] [CrossRef] [PubMed]
  92. Bakos, E.; Homolya, L. Portrait of multifaceted transporter, the multidrug resistance-associated protein 1 (MRP1/ABCC1). Pflugers Arch. 2007, 453, 621–641. [Google Scholar] [CrossRef]
  93. Martinez-Valverde, T.; Vidal-Jorge, M.; Martinez-Saez, E.; Castro, L.; Arikan, F.; Cordero, E.; Radoi, A.; Poca, M.A.; Simard, J.M.; Sahuquillo, J. Sulfonylurea Receptor 1 in Humans with Post-Traumatic Brain Contusions. J. Neurotrauma 2015, 32, 1478–1487. [Google Scholar] [CrossRef]
  94. Tsymbalyuk, O.; Gerzanich, V.; Mumtaz, A.; Andhavarapu, S.; Ivanova, S.; Makar, T.K.; Sansur, C.A.; Keller, A.; Nakamura, Y.; Bryan, J.; et al. SUR1, newly expressed in astrocytes, mediates neuropathic pain in a mouse model of peripheral nerve injury. Mol. Pain. 2021, 17, 17448069211006603. [Google Scholar] [CrossRef] [PubMed]
  95. Hirsch-Reinshagen, V.; Zhou, S.; Burgess, B.L.; Bernier, L.; McIsaac, S.A.; Chan, J.Y.; Tansley, G.H.; Cohn, J.S.; Hayden, M.R.; Wellington, C.L. Deficiency of ABCA1 impairs apolipoprotein E metabolism in brain. J. Biol. Chem. 2004, 279, 41197–41207. [Google Scholar] [CrossRef] [PubMed]
  96. Behl, T.; Kaur, I.; Sehgal, A.; Kumar, A.; Uddin, M.S.; Bungau, S. The Interplay of ABC Transporters in Abeta Translocation and Cholesterol Metabolism: Implicating Their Roles in Alzheimer’s Disease. Mol. Neurobiol. 2021, 58, 1564–1582. [Google Scholar] [CrossRef] [PubMed]
  97. Loscher, W.; Potschka, H. Blood-brain barrier active efflux transporters: ATP-binding cassette gene family. NeuroRx 2005, 2, 86–98. [Google Scholar] [CrossRef] [PubMed]
  98. Lye, P.; Bloise, E.; Imperio, G.E.; Chitayat, D.; Matthews, S.G. Functional Expression of Multidrug-Resistance (MDR) Transporters in Developing Human Fetal Brain Endothelial Cells. Cells 2022, 11, 2259. [Google Scholar] [CrossRef] [PubMed]
  99. Baello, S.; Iqbal, M.; Bloise, E.; Javam, M.; Gibb, W.; Matthews, S.G. TGF-beta1 regulation of multidrug resistance P-glycoprotein in the developing male blood-brain barrier. Endocrinology 2014, 155, 475–484. [Google Scholar] [CrossRef]
  100. Zolotoff, C.; Voirin, A.C.; Puech, C.; Roche, F.; Perek, N. Intermittent Hypoxia and Its Impact on Nrf2/HIF-1alpha Expression and ABC Transporters: An in Vitro Human Blood-Brain Barrier Model Study. Cell Physiol. Biochem. 2020, 54, 1231–1248. [Google Scholar]
  101. Jackson, R.J.; Meltzer, J.C.; Nguyen, H.; Commins, C.; Bennett, R.E.; Hudry, E.; Hyman, B.T. APOE4 derived from astrocytes leads to blood-brain barrier impairment. Brain 2022, 145, 3582–3593. [Google Scholar] [CrossRef]
  102. Al-Majdoub, Z.M.; Al Feteisi, H.; Achour, B.; Warwood, S.; Neuhoff, S.; Rostami-Hodjegan, A.; Barber, J. Proteomic Quantification of Human Blood-Brain Barrier SLC and ABC Transporters in Healthy Individuals and Dementia Patients. Mol. Pharm. 2019, 16, 1220–1233. [Google Scholar] [CrossRef]
  103. Kadry, H.; Noorani, B.; Cucullo, L. A blood-brain barrier overview on structure, function, impairment, and biomarkers of integrity. Fluids Barriers CNS 2020, 17, 69. [Google Scholar] [CrossRef]
  104. Poller, B.; Drewe, J.; Krahenbuhl, S.; Huwyler, J.; Gutmann, H. Regulation of BCRP (ABCG2) and P-glycoprotein (ABCB1) by cytokines in a model of the human blood-brain barrier. Cell Mol. Neurobiol. 2010, 30, 63–70. [Google Scholar] [CrossRef]
  105. Traxl, A.; Mairinger, S.; Filip, T.; Sauberer, M.; Stanek, J.; Poschner, S.; Jager, W.; Zoufal, V.; Novarino, G.; Tournier, N.; et al. Inhibition of ABCB1 and ABCG2 at the Mouse Blood-Brain Barrier with Marketed Drugs To Improve Brain Delivery of the Model ABCB1/ABCG2 Substrate [(11)C]erlotinib. Mol. Pharm. 2019, 16, 1282–1293. [Google Scholar] [CrossRef]
  106. ElAli, A.; Hermann, D.M. ATP-binding cassette transporters and their roles in protecting the brain. Neuroscientist 2011, 17, 423–436. [Google Scholar] [CrossRef]
  107. Miller, D.S. Regulation of ABC transporters blood-brain barrier: The good, the bad, and the ugly. Adv. Cancer Res. 2015, 125, 43–70. [Google Scholar] [PubMed]
  108. Miller, D.S. Regulation of P-glycoprotein and other ABC drug transporters at the blood-brain barrier. Trends Pharmacol. Sci. 2010, 31, 246–254. [Google Scholar] [CrossRef] [PubMed]
  109. Alahmari, A. Blood-Brain Barrier Overview: Structural and Functional Correlation. Neural Plast. 2021, 2021, 6564585. [Google Scholar] [CrossRef]
  110. Rand, D.; Ravid, O.; Atrakchi, D.; Israelov, H.; Bresler, Y.; Shemesh, C.; Omesi, L.; Liraz-Zaltsman, S.; Gosselet, F.; Maskrey, T.S.; et al. Endothelial Iron Homeostasis Regulates Blood-Brain Barrier Integrity via the HIF2alpha-Ve-Cadherin Pathway. Pharmaceutics 2021, 13, 311. [Google Scholar] [CrossRef]
  111. Cole, S.P.; Deeley, R.G. Transport of glutathione and glutathione conjugates by MRP1. Trends Pharmacol. Sci. 2006, 27, 438–446. [Google Scholar] [CrossRef] [PubMed]
  112. Jedlitschky, G.; Tirschmann, K.; Lubenow, L.E.; Nieuwenhuis, H.K.; Akkerman, J.W.; Greinacher, A.; Kroemer, H.K. The nucleotide transporter MRP4 (ABCC4) is highly expressed in human platelets and present in dense granules, indicating a role in mediator storage. Blood 2004, 104, 3603–3610. [Google Scholar] [CrossRef]
  113. Morris, M.E.; Rodriguez-Cruz, V.; Felmlee, M.A. SLC and ABC Transporters: Expression, Localization, and Species Differences at the Blood-Brain and the Blood-Cerebrospinal Fluid Barriers. AAPS J. 2017, 19, 1317–1331. [Google Scholar] [CrossRef]
  114. Stackhouse, T.L.; Mishra, A. Neurovascular Coupling in Development and Disease: Focus on Astrocytes. Front. Cell Dev. Biol. 2021, 9, 702832. [Google Scholar] [CrossRef]
  115. Brown, L.S.; Foster, C.G.; Courtney, J.M.; King, N.E.; Howells, D.W.; Sutherland, B.A. Pericytes and Neurovascular Function in the Healthy and Diseased Brain. Front. Cell. Neurosci. 2019, 13, 282. [Google Scholar] [CrossRef]
  116. Sissung, T.M.; Baum, C.E.; Kirkland, C.T.; Gao, R.; Gardner, E.R.; Figg, W.D. Pharmacogenetics of membrane transporters: An update on current approaches. Mol. Biotechnol. 2010, 44, 152–167. [Google Scholar] [CrossRef]
  117. ElAli, A.; Theriault, P.; Rivest, S. The role of pericytes in neurovascular unit remodeling in brain disorders. Int. J. Mol. Sci. 2014, 15, 6453–6474. [Google Scholar] [CrossRef] [PubMed]
  118. Jia, Y.; Wang, N.; Zhang, Y.; Xue, D.; Lou, H.; Liu, X. Alteration in the Function and Expression of SLC and ABC Transporters in the Neurovascular Unit in Alzheimer’s Disease and the Clinical Significance. Aging Dis. 2020, 11, 390–404. [Google Scholar] [CrossRef] [PubMed]
  119. Abbott, N.J. Blood-brain barrier structure and function and the challenges for CNS drug delivery. J. Inherit. Metab. Dis. 2013, 36, 437–449. [Google Scholar] [CrossRef] [PubMed]
  120. Gomez-Zepeda, D.; Taghi, M.; Scherrmann, J.M.; Decleves, X.; Menet, M.C. ABC Transporters at the Blood-Brain Interfaces, Their Study Models, and Drug Delivery Implications in Gliomas. Pharmaceutics 2019, 12, 20. [Google Scholar] [CrossRef]
  121. Simoes Da Gama, C.; Morin-Brureau, M. Study of BBB Dysregulation in Neuropathogenicity Using Integrative Human Model of Blood-Brain Barrier. Front. Cell. Neurosci. 2022, 16, 863836. [Google Scholar] [CrossRef]
  122. Larochelle, C.; Alvarez, J.I.; Prat, A. How do immune cells overcome the blood-brain barrier in multiple sclerosis? FEBS Lett. 2011, 585, 3770–3780. [Google Scholar] [CrossRef]
  123. Rossi, B.; Santos-Lima, B.; Terrabuio, E.; Zenaro, E.; Constantin, G. Common Peripheral Immunity Mechanisms in Multiple Sclerosis and Alzheimer’s Disease. Front. Immunol. 2021, 12, 639369. [Google Scholar] [CrossRef]
  124. Tan, E.K.; Chao, Y.X.; West, A.; Chan, L.L.; Poewe, W.; Jankovic, J. Parkinson disease and the immune system—Associations, mechanisms and therapeutics. Nat. Rev. 2020, 16, 303–318. [Google Scholar] [CrossRef] [PubMed]
  125. Lecuyer, M.A.; Kebir, H.; Prat, A. Glial influences on BBB functions and molecular players in immune cell trafficking. Biochim. Biophys. Acta 2016, 1862, 472–482. [Google Scholar] [CrossRef] [PubMed]
  126. Kooij, G.; Kroon, J.; Paul, D.; Reijerkerk, A.; Geerts, D.; van der Pol, S.M.; van Het Hof, B.; Drexhage, J.A.; van Vliet, S.J.; Hekking, L.H.; et al. P-glycoprotein regulates trafficking of CD8(+) T cells to the brain parenchyma. Acta Neuropathol. 2014, 127, 699–711. [Google Scholar] [CrossRef] [PubMed]
  127. Kooij, G.; Mizee, M.R.; van Horssen, J.; Reijerkerk, A.; Witte, M.E.; Drexhage, J.A.; van der Pol, S.M.; van Het Hof, B.; Scheffer, G.; Scheper, R.; et al. Adenosine triphosphate-binding cassette transporters mediate chemokine (C-C motif) ligand 2 secretion from reactive astrocytes: Relevance to multiple sclerosis pathogenesis. Brain 2011, 134 Pt 2, 555–570. [Google Scholar] [CrossRef] [PubMed]
  128. Kooij, G.; Backer, R.; Koning, J.J.; Reijerkerk, A.; van Horssen, J.; van der Pol, S.M.; Drexhage, J.; Schinkel, A.; Dijkstra, C.D.; den Haan, J.M.; et al. P-glycoprotein acts as an immunomodulator during neuroinflammation. PLoS ONE 2009, 4, e8212. [Google Scholar] [CrossRef] [PubMed]
  129. Chen, M.L.; Sun, A.; Cao, W.; Eliason, A.; Mendez, K.M.; Getzler, A.J.; Tsuda, S.; Diao, H.; Mukori, C.; Bruno, N.E.; et al. Physiological expression and function of the MDR1 transporter in cytotoxic T lymphocytes. J. Exp. Med. 2020, 217, e20191388. [Google Scholar] [CrossRef] [PubMed]
  130. Kooij, G.; van Horssen, J.; Bandaru, V.V.; Haughey, N.J.; de Vries, H.E. The Role of ATP-Binding Cassette Transporters in Neuro-Inflammation: Relevance for Bioactive Lipids. Front. Pharmacol. 2012, 3, 74. [Google Scholar] [CrossRef]
  131. Csandl, M.A.; Conseil, G.; Cole, S.P. Cysteinyl Leukotriene Receptor 1/2 Antagonists Nonselectively Modulate Organic Anion Transport by Multidrug Resistance Proteins (MRP1-4). Drug Metab. Dispos. 2016, 44, 857–866. [Google Scholar] [CrossRef]
  132. Nowyhed, H.N.; Chandra, S.; Kiosses, W.; Marcovecchio, P.; Andary, F.; Zhao, M.; Fitzgerald, M.L.; Kronenberg, M.; Hedrick, C.C. ATP Binding Cassette Transporter ABCA7 Regulates NKT Cell Development and Function by Controlling CD1d Expression and Lipid Raft Content. Sci. Rep. 2017, 7, 40273. [Google Scholar] [CrossRef]
  133. Armstrong, A.J.; Gebre, A.K.; Parks, J.S.; Hedrick, C.C. ATP-binding cassette transporter G1 negatively regulates thymocyte and peripheral lymphocyte proliferation. J. Immunol. 2010, 184, 173–183. [Google Scholar] [CrossRef]
  134. Sag, D.; Wingender, G.; Nowyhed, H.; Wu, R.; Gebre, A.K.; Parks, J.S.; Kronenberg, M.; Hedrick, C.C. ATP-binding cassette transporter G1 intrinsically regulates invariant NKT cell development. J. Immunol. 2012, 189, 5129–5138. [Google Scholar] [CrossRef] [PubMed]
  135. Cheng, H.Y.; Gaddis, D.E.; Wu, R.; McSkimming, C.; Haynes, L.D.; Taylor, A.M.; McNamara, C.A.; Sorci-Thomas, M.; Hedrick, C.C. Loss of ABCG1 influences regulatory T cell differentiation and atherosclerosis. J. Clin. Investig. 2016, 126, 3236–3246. [Google Scholar] [CrossRef] [PubMed]
  136. Thurm, C.; Schraven, B.; Kahlfuss, S. ABC Transporters in T Cell-Mediated Physiological and Pathological Immune Responses. Int. J. Mol. Sci. 2021, 22, 9186. [Google Scholar] [CrossRef] [PubMed]
  137. Leier, I.; Jedlitschky, G.; Buchholz, U.; Cole, S.P.; Deeley, R.G.; Keppler, D. The MRP gene encodes an ATP-dependent export pump for leukotriene C4 and structurally related conjugates. J. Biol. Chem. 1994, 269, 27807–27810. [Google Scholar] [CrossRef] [PubMed]
  138. Makar, T.K.; Gerzanich, V.; Nimmagadda, V.K.; Jain, R.; Lam, K.; Mubariz, F.; Trisler, D.; Ivanova, S.; Woo, S.K.; Kwon, M.S.; et al. Silencing of Abcc8 or inhibition of newly upregulated Sur1-Trpm4 reduce inflammation and disease progression in experimental autoimmune encephalomyelitis. J. Neuroinflamm. 2015, 12, 210. [Google Scholar] [CrossRef] [PubMed]
  139. Koldamova, R.P.; Lefterov, I.M.; Ikonomovic, M.D.; Skoko, J.; Lefterov, P.I.; Isanski, B.A.; DeKosky, S.T.; Lazo, J.S. 22R-hydroxycholesterol and 9-cis-retinoic acid induce ATP-binding cassette transporter A1 expression and cholesterol efflux in brain cells and decrease amyloid beta secretion. J. Biol. Chem. 2003, 278, 13244–13256. [Google Scholar] [CrossRef]
  140. Fukumoto, H.; Deng, A.; Irizarry, M.C.; Fitzgerald, M.L.; Rebeck, G.W. Induction of the cholesterol transporter ABCA1 in central nervous system cells by liver X receptor agonists increases secreted Abeta levels. J. Biol. Chem. 2002, 277, 48508–48513. [Google Scholar] [CrossRef] [PubMed]
  141. Uehara, Y.; Yamada, T.; Baba, Y.; Miura, S.; Abe, S.; Kitajima, K.; Higuchi, M.A.; Iwamoto, T.; Saku, K. ATP-binding cassette transporter G4 is highly expressed in microglia in Alzheimer’s brain. Brain Res. 2008, 1217, 239–246. [Google Scholar] [CrossRef]
  142. Lee, G.; Schlichter, L.; Bendayan, M.; Bendayan, R. Functional expression of P-glycoprotein in rat brain microglia. J. Pharmacol. Exp. Ther. 2001, 299, 204–212. [Google Scholar]
  143. Ballerini, P.; Di Iorio, P.; Ciccarelli, R.; Caciagli, F.; Poli, A.; Beraudi, A.; Buccella, S.; D’Alimonte, I.; D’Auro, M.; Nargi, E.; et al. P2Y1 and cysteinyl leukotriene receptors mediate purine and cysteinyl leukotriene co-release in primary cultures of rat microglia. Int. J. Immunopathol. Pharmacol. 2005, 18, 255–268. [Google Scholar] [CrossRef]
  144. Navia-Pelaez, J.M.; Choi, S.H.; Dos Santos Aggum Capettini, L.; Xia, Y.; Gonen, A.; Agatisa-Boyle, C.; Delay, L.; Goncalves Dos Santos, G.; Catroli, G.F.; Kim, J.; et al. Normalization of cholesterol metabolism in spinal microglia alleviates neuropathic pain. J. Exp. Med. 2021, 218, e20202059. [Google Scholar] [CrossRef] [PubMed]
  145. Fu, Y.; Hsiao, J.H.; Paxinos, G.; Halliday, G.M.; Kim, W.S. ABCA7 Mediates Phagocytic Clearance of Amyloid-beta in the Brain. J. Alzheimers Dis. 2016, 54, 569–584. [Google Scholar] [CrossRef] [PubMed]
  146. Li, H.; Karl, T.; Garner, B. Understanding the function of ABCA7 in Alzheimer’s disease. Biochem. Soc. Trans. 2015, 43, 920–923. [Google Scholar] [CrossRef] [PubMed]
  147. Ashraf, T.; Kao, A.; Bendayan, R. Functional expression of drug transporters in glial cells: Potential role on drug delivery to the CNS. Adv. Pharmacol. 2014, 71, 45–111. [Google Scholar] [PubMed]
  148. Dallas, S.; Zhu, X.; Baruchel, S.; Schlichter, L.; Bendayan, R. Functional expression of the multidrug resistance protein 1 in microglia. J. Pharmacol. Exp. Ther. 2003, 307, 282–290. [Google Scholar] [CrossRef] [PubMed]
  149. Hirrlinger, J.; Konig, J.; Dringen, R. Expression of mRNAs of multidrug resistance proteins (Mrps) in cultured rat astrocytes, oligodendrocytes, microglial cells and neurones. J. Neurochem. 2002, 82, 716–719. [Google Scholar] [CrossRef]
  150. Dallas, S.; Ronaldson, P.T.; Bendayan, M.; Bendayan, R. Multidrug resistance protein 1-mediated transport of saquinavir by microglia. Neuroreport 2004, 15, 1183–1186. [Google Scholar] [CrossRef]
  151. Dallas, S.; Schlichter, L.; Bendayan, R. Multidrug resistance protein (MRP) 4- and MRP 5-mediated efflux of 9-(2-phosphonylmethoxyethyl)adenine by microglia. J. Pharmacol. Exp. Ther. 2004, 309, 1221–1229. [Google Scholar] [CrossRef] [PubMed]
  152. Fouquet, F.; Zhou, J.M.; Ralston, E.; Murray, K.; Troalen, F.; Magal, E.; Robain, O.; Dubois-Dalcq, M.; Aubourg, P. Expression of the adrenoleukodystrophy protein in the human and mouse central nervous system. Neurobiol. Dis. 1997, 3, 271–285. [Google Scholar] [CrossRef]
  153. Pollard, H.; Moreau, J.; Aubourg, P. Localization of mRNAs for adrenoleukodystrophy and the 70 kDa peroxisomal (PMP70) proteins in the rat brain during post-natal development. J. Neurosci. Res. 1995, 42, 433–437. [Google Scholar] [CrossRef]
  154. Berezowski, V.; Landry, C.; Dehouck, M.P.; Cecchelli, R.; Fenart, L. Contribution of glial cells and pericytes to the mRNA profiles of P-glycoprotein and multidrug resistance-associated proteins in an in vitro model of the blood-brain barrier. Brain Res. 2004, 1018, 1–9. [Google Scholar] [CrossRef] [PubMed]
  155. Lee, G.; Babakhanian, K.; Ramaswamy, M.; Prat, A.; Wosik, K.; Bendayan, R. Expression of the ATP-binding cassette membrane transporter, ABCG2, in human and rodent brain microvessel endothelial and glial cell culture systems. Pharm. Res. 2007, 24, 1262–1274. [Google Scholar] [CrossRef] [PubMed]
  156. Gibson, C.J.; Hossain, M.M.; Richardson, J.R.; Aleksunes, L.M. Inflammatory regulation of ATP binding cassette efflux transporter expression and function in microglia. J. Pharmacol. Exp. Ther. 2012, 343, 650–660. [Google Scholar] [CrossRef] [PubMed]
  157. Oram, J.F.; Lawn, R.M.; Garvin, M.R.; Wade, D.P. ABCA1 is the cAMP-inducible apolipoprotein receptor that mediates cholesterol secretion from macrophages. J. Biol. Chem. 2000, 275, 34508–34511. [Google Scholar] [CrossRef]
  158. Savary, S.; Denizot, F.; Luciani, M.; Mattei, M.; Chimini, G. Molecular cloning of a mammalian ABC transporter homologous to Drosophila white gene. Mamm. Genome 1996, 7, 673–676. [Google Scholar] [CrossRef] [PubMed]
  159. Klucken, J.; Buchler, C.; Orso, E.; Kaminski, W.E.; Porsch-Ozcurumez, M.; Liebisch, G.; Kapinsky, M.; Diederich, W.; Drobnik, W.; Dean, M.; et al. ABCG1 (ABC8), the human homolog of the Drosophila white gene, is a regulator of macrophage cholesterol and phospholipid transport. Proc. Natl. Acad. Sci. USA 2000, 97, 817–822. [Google Scholar] [CrossRef] [PubMed]
  160. Kennedy, M.A.; Barrera, G.C.; Nakamura, K.; Baldan, A.; Tarr, P.; Fishbein, M.C.; Frank, J.; Francone, O.L.; Edwards, P.A. ABCG1 has a critical role in mediating cholesterol efflux to HDL and preventing cellular lipid accumulation. Cell Metab. 2005, 1, 121–131. [Google Scholar] [CrossRef] [PubMed]
  161. Ye, D.; Meurs, I.; Ohigashi, M.; Calpe-Berdiel, L.; Habets, K.L.; Zhao, Y.; Kubo, Y.; Yamaguchi, A.; Van Berkel, T.J.; Nishi, T.; et al. Macrophage ABCA5 deficiency influences cellular cholesterol efflux and increases susceptibility to atherosclerosis in female LDLr knockout mice. Biochem. Biophys. Res. Commun. 2010, 395, 387–394. [Google Scholar] [CrossRef] [PubMed]
  162. Piehler, A.; Kaminski, W.E.; Wenzel, J.J.; Langmann, T.; Schmitz, G. Molecular structure of a novel cholesterol-responsive A subclass ABC transporter, ABCA9. Biochem. Biophys. Res. Commun. 2002, 295, 408–416. [Google Scholar] [CrossRef]
  163. Pennings, M.; Hildebrand, R.B.; Ye, D.; Kunne, C.; Van Berkel, T.J.; Groen, A.K.; Van Eck, M. Bone marrow-derived multidrug resistance protein ABCB4 protects against atherosclerotic lesion development in LDL receptor knockout mice. Cardiovasc. Res. 2007, 76, 175–183. [Google Scholar] [CrossRef]
  164. Engel, T.; Lorkowski, S.; Lueken, A.; Rust, S.; Schluter, B.; Berger, G.; Cullen, P.; Assmann, G. The human ABCG4 gene is regulated by oxysterols and retinoids in monocyte-derived macrophages. Biochem. Biophys. Res. Commun. 2001, 288, 483–488. [Google Scholar] [CrossRef] [PubMed]
  165. Jehle, A.W.; Gardai, S.J.; Li, S.; Linsel-Nitschke, P.; Morimoto, K.; Janssen, W.J.; Vandivier, R.W.; Wang, N.; Greenberg, S.; Dale, B.M.; et al. ATP-binding cassette transporter A7 enhances phagocytosis of apoptotic cells and associated ERK signaling in macrophages. J. Cell Biol. 2006, 174, 547–556. [Google Scholar] [CrossRef] [PubMed]
  166. Kim, W.S.; Li, H.; Ruberu, K.; Chan, S.; Elliott, D.A.; Low, J.K.; Cheng, D.; Karl, T.; Garner, B. Deletion of Abca7 increases cerebral amyloid-beta accumulation in the J20 mouse model of Alzheimer’s disease. J. Neurosci. 2013, 33, 4387–4394. [Google Scholar] [CrossRef]
  167. Kaminski, W.E.; Wenzel, J.J.; Piehler, A.; Langmann, T.; Schmitz, G. ABCA6, a novel a subclass ABC transporter. Biochem. Biophys. Res. Commun. 2001, 285, 1295–1301. [Google Scholar] [CrossRef] [PubMed]
  168. Schumacher, T.; Benndorf, R.A. ABC Transport Proteins in Cardiovascular Disease-A Brief Summary. Molecules 2017, 22, 589. [Google Scholar] [CrossRef] [PubMed]
  169. Vasiliou, V.; Vasiliou, K.; Nebert, D.W. Human ATP-binding cassette (ABC) transporter family. Hum. Genom. 2009, 3, 281–290. [Google Scholar] [CrossRef] [PubMed]
  170. Woodward, O.M.; Kottgen, A.; Kottgen, M. ABCG transporters and disease. FEBS J. 2011, 278, 3215–3225. [Google Scholar] [CrossRef]
  171. Wang, N.; Westerterp, M. ABC Transporters, Cholesterol Efflux, and Implications for Cardiovascular Diseases. Adv. Exp. Med. Biol. 2020, 1276, 67–83. [Google Scholar]
  172. Kerr, I.D.; Hutchison, E.; Gerard, L.; Aleidi, S.M.; Gelissen, I.C. Mammalian ABCG-transporters, sterols and lipids: To bind perchance to transport? Biochim. Biophys. Acta Mol. Cell Biol. Lipids 2021, 1866, 158860. [Google Scholar] [CrossRef]
  173. Fu, Z.; Liu, F.; Liu, C.; Jin, B.; Jiang, Y.; Tang, M.; Qi, X.; Guo, X. Mutant huntingtin inhibits the mitochondrial unfolded protein response by impairing ABCB10 mRNA stability. Biochim. Biophys. Acta Mol. Basis Dis. 2019, 1865, 1428–1435. [Google Scholar] [CrossRef]
  174. Tapia-Maltos, M.A.; Trevino-Frenk, I.; Garcia-Gonzalez, H.B.; Rosetti, M.; Barriga-Maldonado, V.; Morales-Ramirez, F.; Lopez-Hernandez, D.C.; Rosetti, F.; Crispin, J.C. Identification of regulatory T cell molecules associated with severity of multiple sclerosis. Mult. Scler. 2021, 27, 1695–1705. [Google Scholar] [CrossRef] [PubMed]
  175. Satoh, K.; Abe-Dohmae, S.; Yokoyama, S.; St George-Hyslop, P.; Fraser, P.E. ATP-binding cassette transporter A7 (ABCA7) loss of function alters Alzheimer amyloid processing. J. Biol. Chem. 2015, 290, 24152–24165. [Google Scholar] [CrossRef] [PubMed]
  176. Frikke-Schmidt, R. Genetic variation in the ABCA1 gene, HDL cholesterol, and risk of ischemic heart disease in the general population. Atherosclerosis 2010, 208, 305–316. [Google Scholar] [CrossRef] [PubMed]
  177. Brampton, C.; Pomozi, V.; Chen, L.H.; Apana, A.; McCurdy, S.; Zoll, J.; Boisvert, W.A.; Lambert, G.; Henrion, D.; Blanchard, S.; et al. ABCC6 deficiency promotes dyslipidemia and atherosclerosis. Sci. Rep. 2021, 11, 3881. [Google Scholar] [CrossRef] [PubMed]
  178. Hardy, L.M.; Frisdal, E.; Le Goff, W. Critical Role of the Human ATP-Binding Cassette G1 Transporter in Cardiometabolic Diseases. Int. J. Mol. Sci. 2017, 18, 1892. [Google Scholar] [CrossRef] [PubMed]
  179. Pham, D.H.; Kudira, R.; Xu, L.; Valencia, C.A.; Ellis, J.L.; Shi, T.; Evason, K.J.; Osuji, I.; Matuschek, N.; Pfuhler, L.; et al. Deleterious Variants in ABCC12 are Detected in Idiopathic Chronic Cholestasis and Cause Intrahepatic Bile Duct Loss in Model Organisms. Gastroenterology 2021, 161, 287–300.e16. [Google Scholar] [CrossRef] [PubMed]
  180. Katzeff, J.S.; Lok, H.C.; Bhatia, S.; Fu, Y.; Halliday, G.M.; Kim, W.S. ATP-binding cassette transporter expression is widely dysregulated in frontotemporal dementia with TDP-43 inclusions. Front. Mol. Neurosci. 2022, 15, 1043127. [Google Scholar] [CrossRef] [PubMed]
  181. Cho, Y.Y.; Kwon, O.H.; Chung, S. Preferred Endocytosis of Amyloid Precursor Protein from Cholesterol-Enriched Lipid Raft Microdomains. Molecules 2020, 25, 5490. [Google Scholar] [CrossRef]
  182. Kojro, E.; Gimpl, G.; Lammich, S.; Marz, W.; Fahrenholz, F. Low cholesterol stimulates the nonamyloidogenic pathway by its effect on the alpha -secretase ADAM 10. Proc. Natl. Acad. Sci. USA 2001, 98, 5815–5820. [Google Scholar] [CrossRef]
  183. Wahrle, S.E.; Jiang, H.; Parsadanian, M.; Kim, J.; Li, A.; Knoten, A.; Jain, S.; Hirsch-Reinshagen, V.; Wellington, C.L.; Bales, K.R.; et al. Overexpression of ABCA1 reduces amyloid deposition in the PDAPP mouse model of Alzheimer disease. J. Clin. Investig. 2008, 118, 671–682. [Google Scholar] [CrossRef]
  184. Michaki, V.; Guix, F.X.; Vennekens, K.; Munck, S.; Dingwall, C.; Davis, J.B.; Townsend, D.M.; Tew, K.D.; Feiguin, F.; De Strooper, B.; et al. Down-regulation of the ATP-binding cassette transporter 2 (Abca2) reduces amyloid-beta production by altering Nicastrin maturation and intracellular localization. J. Biol. Chem. 2012, 287, 1100–1111. [Google Scholar] [CrossRef] [PubMed]
  185. Boehm-Cagan, A.; Bar, R.; Liraz, O.; Bielicki, J.K.; Johansson, J.O.; Michaelson, D.M. ABCA1 Agonist Reverses the ApoE4-Driven Cognitive and Brain Pathologies. J. Alzheimers Dis. 2016, 54, 1219–1233. [Google Scholar] [CrossRef]
  186. Aikawa, T.; Ren, Y.; Yamazaki, Y.; Tachibana, M.; Johnson, M.R.; Anderson, C.T.; Martens, Y.A.; Holm, M.L.; Asmann, Y.W.; Saito, T.; et al. ABCA7 haplodeficiency disturbs microglial immune responses in the mouse brain. Proc. Natl. Acad. Sci. USA 2019, 116, 23790–23796. [Google Scholar] [CrossRef] [PubMed]
  187. Aikawa, T.; Holm, M.L.; Kanekiyo, T. ABCA7 and Pathogenic Pathways of Alzheimer’s Disease. Brain Sci. 2018, 8, 27. [Google Scholar] [CrossRef] [PubMed]
  188. Khani, M.; Gibbons, E.; Bras, J.; Guerreiro, R. Challenge accepted: Uncovering the role of rare genetic variants in Alzheimer’s disease. Mol. Neurodegener. 2022, 17, 3. [Google Scholar] [CrossRef]
  189. Bossaerts, L.; Cacace, R.; Van Broeckhoven, C. The role of ATP-binding cassette subfamily A in the etiology of Alzheimer’s disease. Mol. Neurodegener. 2022, 17, 31. [Google Scholar] [CrossRef] [PubMed]
  190. Davis, W., Jr. The ATP-binding cassette transporter-2 (ABCA2) increases endogenous amyloid precursor protein expression and Abeta fragment generation. Curr. Alzheimer Res. 2010, 7, 566–577. [Google Scholar] [CrossRef] [PubMed]
  191. Fu, Y.; Hsiao, J.H.; Paxinos, G.; Halliday, G.M.; Kim, W.S. ABCA5 regulates amyloid-beta peptide production and is associated with Alzheimer’s disease neuropathology. J. Alzheimers Dis. 2015, 43, 857–869. [Google Scholar] [CrossRef] [PubMed]
  192. Bossaerts, L.; Hens, E.; Hanseeuw, B.; Vandenberghe, R.; Cras, P.; De Deyn, P.P.; Engelborghs, S.; Van Broeckhoven, C.; Consortium, B. Premature termination codon mutations in ABCA7 contribute to Alzheimer’s disease risk in Belgian patients. Neurobiol. Aging 2021, 106, 307.e1–307.e7. [Google Scholar] [CrossRef]
  193. Bossaerts, L.; Hendrickx Van de Craen, E.; Cacace, R.; Asselbergh, B.; Van Broeckhoven, C. Rare missense mutations in ABCA7 might increase Alzheimer’s disease risk by plasma membrane exclusion. Acta Neuropathol. Commun. 2022, 10, 43. [Google Scholar] [CrossRef]
  194. De Roeck, A.; Duchateau, L.; Van Dongen, J.; Cacace, R.; Bjerke, M.; Van den Bossche, T.; Cras, P.; Vandenberghe, R.; De Deyn, P.P.; Engelborghs, S.; et al. An intronic VNTR affects splicing of ABCA7 and increases risk of Alzheimer’s disease. Acta Neuropathol. 2018, 135, 827–837. [Google Scholar] [CrossRef] [PubMed]
  195. De Roeck, A.; Van Broeckhoven, C.; Sleegers, K. The role of ABCA7 in Alzheimer’s disease: Evidence from genomics, transcriptomics and methylomics. Acta Neuropathol. 2019, 138, 201–220. [Google Scholar] [CrossRef] [PubMed]
  196. Namasivayam, V.; Stefan, K.; Gorecki, L.; Korabecny, J.; Soukup, O.; Jansson, P.J.; Pahnke, J.; Stefan, S.M. Physicochemistry shapes bioactivity landscape of pan-ABC transporter modulators: Anchor point for innovative Alzheimer’s disease therapeutics. Int. J. Biol. Macromol. 2022, 217, 775–791. [Google Scholar] [CrossRef] [PubMed]
  197. Krohn, M.; Bracke, A.; Avchalumov, Y.; Schumacher, T.; Hofrichter, J.; Paarmann, K.; Frohlich, C.; Lange, C.; Bruning, T.; von Bohlen Und Halbach, O.; et al. Accumulation of murine amyloid-beta mimics early Alzheimer’s disease. Brain 2015, 138 Pt 8, 2370–2382. [Google Scholar] [CrossRef] [PubMed]
  198. Pahnke, J.; Bascunana, P.; Brackhan, M.; Stefan, K.; Namasivayam, V.; Koldamova, R.; Wu, J.; Mohle, L.; Stefan, S.M. Strategies to gain novel Alzheimer’s disease diagnostics and therapeutics using modulators of ABCA transporters. Free Neuropathol. 2021, 2, 33. [Google Scholar] [PubMed]
  199. Wolfl-Duchek, M.; Mairinger, S.; Hernandez-Lozano, I.; Filip, T.; Zoufal, V.; Lobsch, M.; Stanek, J.; Kuntner, C.; Wanek, T.; Bauer, M.; et al. Use of PET Imaging to Assess the Efficacy of Thiethylperazine to Stimulate Cerebral MRP1 Transport Activity in Wild-Type and APP/PS1-21 Mice. Int. J. Mol. Sci. 2022, 23, 6514. [Google Scholar] [CrossRef] [PubMed]
  200. Khalil, A.; Berrougui, H.; Pawelec, G.; Fulop, T. Impairment of the ABCA1 and SR-BI-mediated cholesterol efflux pathways and HDL anti-inflammatory activity in Alzheimer’s disease. Mech. Ageing Dev. 2012, 133, 20–29. [Google Scholar] [CrossRef] [PubMed]
  201. Shibata, N.; Kawarai, T.; Lee, J.H.; Lee, H.S.; Shibata, E.; Sato, C.; Liang, Y.; Duara, R.; Mayeux, R.P.; St George-Hyslop, P.H.; et al. Association studies of cholesterol metabolism genes (CH25H, ABCA1 and CH24H) in Alzheimer’s disease. Neurosci. Lett. 2006, 391, 142–146. [Google Scholar] [CrossRef] [PubMed]
  202. Koldamova, R.; Fitz, N.F.; Lefterov, I. The role of ATP-binding cassette transporter A1 in Alzheimer’s disease and neurodegeneration. Biochim. Biophys. Acta 2010, 1801, 824–830. [Google Scholar] [CrossRef]
  203. Reynolds, C.A.; Hong, M.G.; Eriksson, U.K.; Blennow, K.; Bennet, A.M.; Johansson, B.; Malmberg, B.; Berg, S.; Wiklund, F.; Gatz, M.; et al. A survey of ABCA1 sequence variation confirms association with dementia. Hum. Mutat. 2009, 30, 1348–1354. [Google Scholar] [CrossRef]
  204. Rodriguez-Rodriguez, E.; Mateo, I.; Llorca, J.; Sanchez-Quintana, C.; Infante, J.; Garcia-Gorostiaga, I.; Sanchez-Juan, P.; Berciano, J.; Combarros, O. Association of genetic variants of ABCA1 with Alzheimer’s disease risk. Am. J. Med. Genet. B Neuropsychiatr. Genet. 2007, 144B, 964–968. [Google Scholar] [CrossRef] [PubMed]
  205. Holstege, H.; Hulsman, M.; Charbonnier, C.; Grenier-Boley, B.; Quenez, O.; Grozeva, D.; van Rooij, J.G.J.; Sims, R.; Ahmad, S.; Amin, N.; et al. Exome sequencing identifies rare damaging variants in ATP8B4 and ABCA1 as risk factors for Alzheimer’s disease. Nat. Genet. 2022, 54, 1786–1794. [Google Scholar] [CrossRef]
  206. Kim, J.; Yoon, H.; Ramirez, C.M.; Lee, S.M.; Hoe, H.S.; Fernandez-Hernando, C.; Kim, J. MiR-106b impairs cholesterol efflux and increases Abeta levels by repressing ABCA1 expression. Exp. Neurol. 2012, 235, 476–483. [Google Scholar] [CrossRef] [PubMed]
  207. Ramirez, C.M.; Davalos, A.; Goedeke, L.; Salerno, A.G.; Warrier, N.; Cirera-Salinas, D.; Suarez, Y.; Fernandez-Hernando, C. MicroRNA-758 regulates cholesterol efflux through posttranscriptional repression of ATP-binding cassette transporter A1. Arterioscler. Thromb. Vasc. Biol. 2011, 31, 2707–2714. [Google Scholar] [CrossRef] [PubMed]
  208. Rayner, K.J.; Suarez, Y.; Davalos, A.; Parathath, S.; Fitzgerald, M.L.; Tamehiro, N.; Fisher, E.A.; Moore, K.J.; Fernandez-Hernando, C. MiR-33 contributes to the regulation of cholesterol homeostasis. Science 2010, 328, 1570–1573. [Google Scholar] [CrossRef] [PubMed]
  209. Horie, T.; Baba, O.; Kuwabara, Y.; Chujo, Y.; Watanabe, S.; Kinoshita, M.; Horiguchi, M.; Nakamura, T.; Chonabayashi, K.; Hishizawa, M.; et al. MicroRNA-33 deficiency reduces the progression of atherosclerotic plaque in ApoE-/- mice. J. Am. Heart Assoc. 2012, 1, e003376. [Google Scholar] [CrossRef] [PubMed]
  210. Goedeke, L.; Fernandez-Hernando, C. MicroRNAs: A connection between cholesterol metabolism and neurodegeneration. Neurobiol. Dis. 2014, 72 Pt A, 48–53. [Google Scholar] [CrossRef]
  211. Rawat, V.; Wang, S.; Sima, J.; Bar, R.; Liraz, O.; Gundimeda, U.; Parekh, T.; Chan, J.; Johansson, J.O.; Tang, C.; et al. ApoE4 Alters ABCA1 Membrane Trafficking in Astrocytes. J. Neurosci. 2019, 39, 9611–9622. [Google Scholar] [CrossRef] [PubMed]
  212. Koldamova, R.; Staufenbiel, M.; Lefterov, I. Lack of ABCA1 considerably decreases brain ApoE level and increases amyloid deposition in APP23 mice. J. Biol. Chem. 2005, 280, 43224–43235. [Google Scholar] [CrossRef]
  213. Steinberg, S.; Stefansson, H.; Jonsson, T.; Johannsdottir, H.; Ingason, A.; Helgason, H.; Sulem, P.; Magnusson, O.T.; Gudjonsson, S.A.; Unnsteinsdottir, U.; et al. Loss-of-function variants in ABCA7 confer risk of Alzheimer’s disease. Nat. Genet. 2015, 47, 445–447. [Google Scholar] [CrossRef]
  214. Hollingworth, P.; Harold, D.; Sims, R.; Gerrish, A.; Lambert, J.C.; Carrasquillo, M.M.; Abraham, R.; Hamshere, M.L.; Pahwa, J.S.; Moskvina, V.; et al. Common variants at ABCA7, MS4A6A/MS4A4E, EPHA1, CD33 and CD2AP are associated with Alzheimer’s disease. Nat. Genet. 2011, 43, 429–435. [Google Scholar] [CrossRef] [PubMed]
  215. Ghani, M.; Lang, A.E.; Zinman, L.; Nacmias, B.; Sorbi, S.; Bessi, V.; Tedde, A.; Tartaglia, M.C.; Surace, E.I.; Sato, C.; et al. Mutation analysis of patients with neurodegenerative disorders using NeuroX array. Neurobiol. Aging 2015, 36, 545.e9–e14. [Google Scholar] [CrossRef] [PubMed]
  216. Sassi, C.; Nalls, M.A.; Ridge, P.G.; Gibbs, J.R.; Ding, J.; Lupton, M.K.; Troakes, C.; Lunnon, K.; Al-Sarraj, S.; Brown, K.S.; et al. ABCA7 p.G215S as potential protective factor for Alzheimer’s disease. Neurobiol. Aging 2016, 46, 235.e1–e9. [Google Scholar] [CrossRef] [PubMed]
  217. Vasquez, J.B.; Fardo, D.W.; Estus, S. ABCA7 expression is associated with Alzheimer’s disease polymorphism and disease status. Neurosci. Lett. 2013, 556, 58–62. [Google Scholar] [CrossRef] [PubMed]
  218. Lam, F.C.; Liu, R.; Lu, P.; Shapiro, A.B.; Renoir, J.M.; Sharom, F.J.; Reiner, P.B. beta-Amyloid efflux mediated by p-glycoprotein. J. Neurochem. 2001, 76, 1121–1128. [Google Scholar] [CrossRef] [PubMed]
  219. Chai, A.B.; Leung, G.K.F.; Callaghan, R.; Gelissen, I.C. P-glycoprotein: A role in the export of amyloid-beta in Alzheimer’s disease? FEBS J. 2020, 287, 612–625. [Google Scholar] [CrossRef] [PubMed]
  220. Hartz, A.M.S.; Zhong, Y.; Shen, A.N.; Abner, E.L.; Bauer, B. Preventing P-gp Ubiquitination Lowers Abeta Brain Levels in an Alzheimer’s Disease Mouse Model. Front. Aging Neurosci. 2018, 10, 186. [Google Scholar] [CrossRef] [PubMed]
  221. Krohn, M.; Zoufal, V.; Mairinger, S.; Wanek, T.; Paarmann, K.; Bruning, T.; Eiriz, I.; Brackhan, M.; Langer, O.; Pahnke, J. Generation and Characterization of an Abcc1 Humanized Mouse Model (hABCC1(flx/flx)) with Knockout Capability. Mol. Pharmacol. 2019, 96, 138–147. [Google Scholar] [CrossRef]
  222. Zoufal, V.; Mairinger, S.; Krohn, M.; Wanek, T.; Filip, T.; Sauberer, M.; Stanek, J.; Kuntner, C.; Pahnke, J.; Langer, O. Measurement of cerebral ABCC1 transport activity in wild-type and APP/PS1-21 mice with positron emission tomography. J. Cereb. Blood Flow. Metab. 2020, 40, 954–965. [Google Scholar] [CrossRef]
  223. Xiong, H.; Callaghan, D.; Jones, A.; Bai, J.; Rasquinha, I.; Smith, C.; Pei, K.; Walker, D.; Lue, L.F.; Stanimirovic, D.; et al. ABCG2 is upregulated in Alzheimer’s brain with cerebral amyloid angiopathy and may act as a gatekeeper at the blood-brain barrier for Abeta(1–40) peptides. J. Neurosci. 2009, 29, 5463–5475. [Google Scholar] [CrossRef]
  224. Feher, A.; Juhasz, A.; Laszlo, A.; Pakaski, M.; Kalman, J.; Janka, Z. Association between the ABCG2 C421A polymorphism and Alzheimer’s disease. Neurosci. Lett. 2013, 550, 51–54. [Google Scholar] [CrossRef] [PubMed]
  225. Shen, S.; Callaghan, D.; Juzwik, C.; Xiong, H.; Huang, P.; Zhang, W. ABCG2 reduces ROS-mediated toxicity and inflammation: A potential role in Alzheimer’s disease. J. Neurochem. 2010, 114, 1590–1604. [Google Scholar] [CrossRef]
  226. Wanek, T.; Zoufal, V.; Brackhan, M.; Krohn, M.; Mairinger, S.; Filip, T.; Sauberer, M.; Stanek, J.; Pekar, T.; Pahnke, J.; et al. Brain Distribution of Dual ABCB1/ABCG2 Substrates Is Unaltered in a Beta-Amyloidosis Mouse Model. Int. J. Mol. Sci. 2020, 21, 8245. [Google Scholar] [CrossRef] [PubMed]
  227. Abuznait, A.H.; Kaddoumi, A. Role of ABC transporters in the pathogenesis of Alzheimer’s disease. ACS Chem. Neurosci. 2012, 3, 820–831. [Google Scholar] [CrossRef] [PubMed]
  228. Pereira, C.D.; Martins, F.; Wiltfang, J.; da Cruz, E.S.O.A.B.; Rebelo, S. ABC Transporters Are Key Players in Alzheimer’s Disease. J. Alzheimers Dis. 2018, 61, 463–485. [Google Scholar] [CrossRef] [PubMed]
  229. Lee, C.G.; Tang, K.; Cheung, Y.B.; Wong, L.P.; Tan, C.; Shen, H.; Zhao, Y.; Pavanni, R.; Lee, E.J.; Wong, M.C.; et al. MDR1, the blood-brain barrier transporter, is associated with Parkinson’s disease in ethnic Chinese. J. Med. Genet. 2004, 41, e60. [Google Scholar] [CrossRef] [PubMed]
  230. Tan, E.K.; Drozdzik, M.; Bialecka, M.; Honczarenko, K.; Klodowska-Duda, G.; Teo, Y.Y.; Tang, K.; Wong, L.P.; Chong, S.S.; Tan, C.; et al. Analysis of MDR1 haplotypes in Parkinson’s disease in a white population. Neurosci. Lett. 2004, 372, 240–244. [Google Scholar] [CrossRef]
  231. Furuno, T.; Landi, M.T.; Ceroni, M.; Caporaso, N.; Bernucci, I.; Nappi, G.; Martignoni, E.; Schaeffeler, E.; Eichelbaum, M.; Schwab, M.; et al. Expression polymorphism of the blood-brain barrier component P-glycoprotein (MDR1) in relation to Parkinson’s disease. Pharmacogenetics 2002, 12, 529–534. [Google Scholar] [CrossRef]
  232. Tan, E.K.; Chan, D.K.; Ng, P.W.; Woo, J.; Teo, Y.Y.; Tang, K.; Wong, L.P.; Chong, S.S.; Tan, C.; Shen, H.; et al. Effect of MDR1 haplotype on risk of Parkinson disease. Arch. Neurol. 2005, 62, 460–464. [Google Scholar] [CrossRef]
  233. Kortekaas, R.; Leenders, K.L.; van Oostrom, J.C.; Vaalburg, W.; Bart, J.; Willemsen, A.T.; Hendrikse, N.H. Blood-brain barrier dysfunction in parkinsonian midbrain in vivo. Ann. Neurol. 2005, 57, 176–179. [Google Scholar] [CrossRef]
  234. Bartels, A.L.; van Berckel, B.N.; Lubberink, M.; Luurtsema, G.; Lammertsma, A.A.; Leenders, K.L. Blood-brain barrier P-glycoprotein function is not impaired in early Parkinson’s disease. Parkinsonism Relat. Disord. 2008, 14, 505–508. [Google Scholar] [CrossRef] [PubMed]
  235. Bartels, A.L. Blood-brain barrier P-glycoprotein function in neurodegenerative disease. Curr. Pharm. Des. 2011, 17, 2771–2777. [Google Scholar] [CrossRef] [PubMed]
  236. Valenza, M.; Leoni, V.; Karasinska, J.M.; Petricca, L.; Fan, J.; Carroll, J.; Pouladi, M.A.; Fossale, E.; Nguyen, H.P.; Riess, O.; et al. Cholesterol defect is marked across multiple rodent models of Huntington’s disease and is manifest in astrocytes. J. Neurosci. 2010, 30, 10844–10850. [Google Scholar] [CrossRef] [PubMed]
  237. Schmitz, G.; Kaminski, W.E. ABC transporters and cholesterol metabolism. Front. Biosci. 2001, 6, D505–D514. [Google Scholar] [CrossRef] [PubMed]
  238. Gonzalez-Guevara, E.; Cardenas, G.; Perez-Severiano, F.; Martinez-Lazcano, J.C. Dysregulated Brain Cholesterol Metabolism Is Linked to Neuroinflammation in Huntington’s Disease. Mov. Disord. 2020, 35, 1113–1127. [Google Scholar] [CrossRef] [PubMed]
  239. Wu, J.; Mohle, L.; Bruning, T.; Eiriz, I.; Rafehi, M.; Stefan, K.; Stefan, S.M.; Pahnke, J. A Novel Huntington’s Disease Assessment Platform to Support Future Drug Discovery and Development. Int. J. Mol. Sci. 2022, 23, 14763. [Google Scholar] [CrossRef] [PubMed]
  240. Galea, E.; Launay, N.; Portero-Otin, M.; Ruiz, M.; Pamplona, R.; Aubourg, P.; Ferrer, I.; Pujol, A. Oxidative stress underlying axonal degeneration in adrenoleukodystrophy: A paradigm for multifactorial neurodegenerative diseases? Biochim. Biophys. Acta 2012, 1822, 1475–1488. [Google Scholar] [CrossRef] [PubMed]
  241. Wright, G.E.B.; Caron, N.S.; Ng, B.; Casal, L.; Casazza, W.; Xu, X.; Ooi, J.; Pouladi, M.A.; Mostafavi, S.; Ross, C.J.D.; et al. Gene expression profiles complement the analysis of genomic modifiers of the clinical onset of Huntington disease. Hum. Mol. Genet. 2020, 29, 2788–2802. [Google Scholar] [CrossRef]
  242. Block, R.C.; Dorsey, E.R.; Beck, C.A.; Brenna, J.T.; Shoulson, I. Altered cholesterol and fatty acid metabolism in Huntington disease. J. Clin. Lipidol. 2010, 4, 17–23. [Google Scholar] [CrossRef]
  243. Ma, C.Y.; Li, C.; Zhou, X.; Zhang, Z.; Jiang, H.; Liu, H.; Chen, H.J.; Tse, H.F.; Liao, C.; Lian, Q. Management of adrenoleukodystrophy: From pre-clinical studies to the development of new therapies. Biomed. Pharmacother. 2021, 143, 112214. [Google Scholar] [CrossRef]
  244. Andreoletti, P.; Raas, Q.; Gondcaille, C.; Cherkaoui-Malki, M.; Trompier, D.; Savary, S. Predictive Structure and Topology of Peroxisomal ATP-Binding Cassette (ABC) Transporters. Int. J. Mol. Sci. 2017, 18, 1593. [Google Scholar] [CrossRef]
  245. Zhu, J.; Eichler, F.; Biffi, A.; Duncan, C.N.; Williams, D.A.; Majzoub, J.A. The Changing Face of Adrenoleukodystrophy. Endocr. Rev. 2020, 41, 577–593. [Google Scholar] [CrossRef] [PubMed]
  246. Lazarowski, A.; Czornyj, L. Role of ABC-Transporters in Epileptogenesis and Pharmacoresistant Epilepsy. In Epileptology—The Modern State of Science; InTechOpen: London, UK, 2016. [Google Scholar]
  247. Deng, X.; Xie, Y.; Chen, Y. Effect of Neuroinflamm. on ABC Transporters: Possible Contribution to Refractory Epilepsy. CNS Neurol. Disord. Drug Targets 2018, 17, 728–735. [Google Scholar] [CrossRef] [PubMed]
  248. Pan, W.; Yu, C.; Hsuchou, H.; Kastin, A.J. The role of cerebral vascular NFkappaB in LPS-induced inflammation: Differential regulation of efflux transporter and transporting cytokine receptors. Cell Physiol. Biochem. 2010, 25, 623–630. [Google Scholar] [CrossRef]
  249. Leandro, K.; Bicker, J.; Alves, G.; Falcao, A.; Fortuna, A. ABC transporters in drug-resistant epilepsy: Mechanisms of upregulation and therapeutic approaches. Pharmacol. Res. 2019, 144, 357–376. [Google Scholar] [CrossRef]
  250. Sisodiya, S.M.; Lint, W.R.; Harding, B.N.; Squier, M.V.; Thom, M. Drug resistance in epilepsy: Human epilepsy. Novartis Found. Symp. 2002, 243, 167–174, Discussion 174–179, 180–185. [Google Scholar]
  251. Aronica, E.; Gorter, J.A.; Redeker, S.; van Vliet, E.A.; Ramkema, M.; Scheffer, G.L.; Scheper, R.J.; van der Valk, P.; Leenstra, S.; Baayen, J.C.; et al. Localization of breast cancer resistance protein (BCRP) in microvessel endothelium of human control and epileptic brain. Epilepsia 2005, 46, 849–857. [Google Scholar] [CrossRef] [PubMed]
  252. Seegers, U.; Potschka, H.; Loscher, W. Transient increase of P-glycoprotein expression in endothelium and parenchyma of limbic brain regions in the kainate model of temporal lobe epilepsy. Epilepsy Res. 2002, 51, 257–268. [Google Scholar] [CrossRef]
  253. van Vliet, E.A.; Redeker, S.; Aronica, E.; Edelbroek, P.M.; Gorter, J.A. Expression of multidrug transporters MRP1, MRP2, and BCRP shortly after status epilepticus, during the latent period, and in chronic epileptic rats. Epilepsia 2005, 46, 1569–1580. [Google Scholar] [CrossRef]
  254. Volk, H.A.; Burkhardt, K.; Potschka, H.; Chen, J.; Becker, A.; Loscher, W. Neuronal expression of the drug efflux transporter P-glycoprotein in the rat hippocampus after limbic seizures. Neuroscience 2004, 123, 751–759. [Google Scholar] [CrossRef]
  255. Mohi-Ud-Din, R.; Mir, R.H.; Mir, P.A.; Banday, N.; Shah, A.J.; Sawhney, G.; Bhat, M.M.; Batiha, G.E.; Pottoo, F.H. Dysfunction of ABC Transporters at the Surface of BBB: Potential Implications in Intractable Epilepsy and Applications of Nanotechnology Enabled Drug Delivery. Curr. Drug Metab. 2022, 23, 735–756. [Google Scholar] [CrossRef] [PubMed]
  256. Banerjee Dixit, A.; Sharma, D.; Srivastava, A.; Banerjee, J.; Tripathi, M.; Prakash, D.; Sarat Chandra, P. Upregulation of breast cancer resistance protein and major vault protein in drug resistant epilepsy. Seizure 2017, 47, 9–12. [Google Scholar] [CrossRef] [PubMed]
  257. Iannetti, P.; Spalice, A.; Parisi, P. Calcium-channel blocker verapamil administration in prolonged and refractory status epilepticus. Epilepsia 2005, 46, 967–969. [Google Scholar] [CrossRef] [PubMed]
  258. Summers, M.A.; Moore, J.L.; McAuley, J.W. Use of verapamil as a potential P-glycoprotein inhibitor in a patient with refractory epilepsy. Ann. Pharmacother. 2004, 38, 1631–1634. [Google Scholar] [CrossRef] [PubMed]
  259. Brandt, C.; Bethmann, K.; Gastens, A.M.; Loscher, W. The multidrug transporter hypothesis of drug resistance in epilepsy: Proof-of-principle in a rat model of temporal lobe epilepsy. Neurobiol. Dis. 2006, 24, 202–211. [Google Scholar] [CrossRef] [PubMed]
  260. Bauer, M.; Karch, R.; Zeitlinger, M.; Liu, J.; Koepp, M.J.; Asselin, M.C.; Sisodiya, S.M.; Hainfellner, J.A.; Wadsak, W.; Mitterhauser, M.; et al. In vivo P-glycoprotein function before and after epilepsy surgery. Neurology 2014, 83, 1326–1331. [Google Scholar] [CrossRef] [PubMed]
  261. Clinckers, R.; Smolders, I.; Meurs, A.; Ebinger, G.; Michotte, Y. Quantitative in vivo microdialysis study on the influence of multidrug transporters on the blood-brain barrier passage of oxcarbazepine: Concomitant use of hippocampal monoamines as pharmacodynamic markers for the anticonvulsant activity. J. Pharmacol. Exp. Ther. 2005, 314, 725–731. [Google Scholar] [CrossRef] [PubMed]
  262. Aronica, E.; Gorter, J.A.; Jansen, G.H.; van Veelen, C.W.; van Rijen, P.C.; Leenstra, S.; Ramkema, M.; Scheffer, G.L.; Scheper, R.J.; Troost, D. Expression and cellular distribution of multidrug transporter proteins in two major causes of medically intractable epilepsy: Focal cortical dysplasia and glioneuronal tumors. Neuroscience 2003, 118, 417–429. [Google Scholar] [CrossRef] [PubMed]
  263. Thom, M. Review: Hippocampal sclerosis in epilepsy: A neuropathology review. Neuropathol. Appl. Neurobiol. 2014, 40, 520–543. [Google Scholar] [CrossRef]
  264. Sisodiya, S.M.; Lin, W.R.; Harding, B.N.; Squier, M.V.; Thom, M. Drug resistance in epilepsy: Expression of drug resistance proteins in common causes of refractory epilepsy. Brain 2002, 125 Pt 1, 22–31. [Google Scholar] [CrossRef]
  265. Sisodiya, S.M.; Martinian, L.; Scheffer, G.L.; van der Valk, P.; Scheper, R.J.; Harding, B.N.; Thom, M. Vascular colocalization of P-glycoprotein, multidrug-resistance associated protein 1, breast cancer resistance protein and major vault protein in human epileptogenic pathologies. Neuropathol. Appl. Neurobiol. 2006, 32, 51–63. [Google Scholar] [CrossRef] [PubMed]
  266. Lachos, J.; Zattoni, M.; Wieser, H.G.; Fritschy, J.M.; Langmann, T.; Schmitz, G.; Errede, M.; Virgintino, D.; Yonekawa, Y.; Frei, K. Characterization of the gene expression profile of human hippocampus in mesial temporal lobe epilepsy with hippocampal sclerosis. Epilepsy Res. Treat. 2011, 2011, 758407. [Google Scholar] [CrossRef] [PubMed]
  267. Nelson, P.T.; Jicha, G.A.; Wang, W.X.; Ighodaro, E.; Artiushin, S.; Nichols, C.G.; Fardo, D.W. ABCC9/SUR2 in the brain: Implications for hippocampal sclerosis of aging and a potential therapeutic target. Ageing Res. Rev. 2015, 24 Pt B, 111–125. [Google Scholar] [CrossRef]
  268. Nelson, P.T.; Wang, W.X.; Partch, A.B.; Monsell, S.E.; Valladares, O.; Ellingson, S.R.; Wilfred, B.R.; Naj, A.C.; Wang, L.S.; Kukull, W.A.; et al. Reassessment of risk genotypes (GRN, TMEM106B, and ABCC9 variants) associated with hippocampal sclerosis of aging pathology. J. Neuropathol. Exp. Neurol. 2015, 74, 75–84. [Google Scholar] [CrossRef] [PubMed]
  269. Guerrini, R.; Barba, C. Focal cortical dysplasia: An update on diagnosis and treatment. Expert Rev. Neurother. 2021, 21, 1213–1224. [Google Scholar] [CrossRef] [PubMed]
  270. Schumacher, T.; Krohn, M.; Hofrichter, J.; Lange, C.; Stenzel, J.; Steffen, J.; Dunkelmann, T.; Paarmann, K.; Frohlich, C.; Uecker, A.; et al. ABC transporters B1, C1 and G2 differentially regulate neuroregeneration in mice. PLoS ONE 2012, 7, e35613. [Google Scholar] [CrossRef] [PubMed]
  271. Yamamoto, A.; Shofuda, T.; Islam, M.O.; Nakamura, Y.; Yamasaki, M.; Okano, H.; Kanemura, Y. ABCB1 is predominantly expressed in human fetal neural stem/progenitor cells at an early development stage. J. Neurosci. Res. 2009, 87, 2615–2623. [Google Scholar] [CrossRef] [PubMed]
  272. Czornyj, L.; Lazarowski, A. ABC-transporters as stem-cell markers in brain dysplasia/tumor epilepsies. Front. Biosci. 2014, 19, 1425–1435. [Google Scholar] [CrossRef] [PubMed]
  273. Sisodiya, S.M.; Lin, W.R.; Squier, M.V.; Thom, M. Multidrug-resistance protein 1 in focal cortical dysplasia. Lancet 2001, 357, 42–43. [Google Scholar] [CrossRef]
  274. Marchi, N.; Guiso, G.; Rizzi, M.; Pirker, S.; Novak, K.; Czech, T.; Baumgartner, C.; Janigro, D.; Caccia, S.; Vezzani, A. A pilot study on brain-to-plasma partition of 10,11-dyhydro-10-hydroxy-5H-dibenzo(b,f)azepine-5-carboxamide and MDR1 brain expression in epilepsy patients not responding to oxcarbazepine. Epilepsia 2005, 46, 1613–1619. [Google Scholar] [CrossRef]
  275. Potschka, H.; Loscher, W. In vivo evidence for P-glycoprotein-mediated transport of phenytoin at the blood-brain barrier of rats. Epilepsia 2001, 42, 1231–1240. [Google Scholar] [CrossRef] [PubMed]
  276. Potschka, H.; Fedrowitz, M.; Loscher, W. P-glycoprotein and multidrug resistance-associated protein are involved in the regulation of extracellular levels of the major antiepileptic drug carbamazepine in the brain. Neuroreport 2001, 12, 3557–3560. [Google Scholar] [CrossRef] [PubMed]
  277. Sills, G.J.; Kwan, P.; Butler, E.; de Lange, E.C.; van den Berg, D.J.; Brodie, M.J. P-glycoprotein-mediated efflux of antiepileptic drugs: Preliminary studies in mdr1a knockout mice. Epilepsy Behav. 2002, 3, 427–432. [Google Scholar] [CrossRef]
  278. Potschka, H.; Fedrowitz, M.; Loscher, W. P-Glycoprotein-mediated efflux of phenobarbital, lamotrigine, and felbamate at the blood-brain barrier: Evidence from microdialysis experiments in rats. Neurosci. Lett. 2002, 327, 173–176. [Google Scholar] [CrossRef] [PubMed]
  279. Rizzi, M.; Caccia, S.; Guiso, G.; Richichi, C.; Gorter, J.A.; Aronica, E.; Aliprandi, M.; Bagnati, R.; Fanelli, R.; D’Incalci, M.; et al. Limbic seizures induce P-glycoprotein in rodent brain: Functional implications for pharmacoresistance. J. Neurosci. 2002, 22, 5833–5839. [Google Scholar] [CrossRef] [PubMed]
  280. Tishler, D.M.; Weinberg, K.I.; Hinton, D.R.; Barbaro, N.; Annett, G.M.; Raffel, C. MDR1 gene expression in brain of patients with medically intractable epilepsy. Epilepsia 1995, 36, 1–6. [Google Scholar] [CrossRef]
  281. Potschka, H.; Loscher, W. Multidrug resistance-associated protein is involved in the regulation of extracellular levels of phenytoin in the brain. Neuroreport 2001, 12, 2387–2389. [Google Scholar] [CrossRef] [PubMed]
  282. Potschka, H.; Fedrowitz, M.; Loscher, W. Multidrug resistance protein MRP2 contributes to blood-brain barrier function and restricts antiepileptic drug activity. J. Pharmacol. Exp. Ther. 2003, 306, 124–131. [Google Scholar] [CrossRef] [PubMed]
  283. Wen, T.; Liu, Y.C.; Yang, H.W.; Liu, H.Y.; Liu, X.D.; Wang, G.J.; Xie, L. Effect of 21-day exposure of phenobarbital, carbamazepine and phenytoin on P-glycoprotein expression and activity in the rat brain. J. Neurol. Sci. 2008, 270, 99–106. [Google Scholar] [CrossRef]
  284. Wang-Tilz, Y.; Tilz, C.; Wang, B.; Tilz, G.P.; Stefan, H. Influence of lamotrigine and topiramate on MDR1 expression in difficult-to-treat temporal lobe epilepsy. Epilepsia 2006, 47, 233–239. [Google Scholar] [CrossRef]
  285. Yang, H.W.; Liu, H.Y.; Liu, X.; Zhang, D.M.; Liu, Y.C.; Liu, X.D.; Wang, G.J.; Xie, L. Increased P-glycoprotein function and level after long-term exposure of four antiepileptic drugs to rat brain microvascular endothelial cells in vitro. Neurosci. Lett. 2008, 434, 299–303. [Google Scholar] [CrossRef] [PubMed]
  286. Ludwin, S.K. The pathogenesis of multiple sclerosis: Relating human pathology to experimental studies. J. Neuropathol. Exp. Neurol. 2006, 65, 305–318. [Google Scholar] [CrossRef] [PubMed]
  287. Giannopapas, V.; Kitsos, D.; Tsogka, A.; Tzartos, J.S.; Paraskevas, G.; Tsivgoulis, G.; Voumvourakis, K.; Giannopoulos, S.; Bakalidou, D. Sexual dysfunction therapeutic approaches in patients with multiple sclerosis: A systematic review. Neurol. Sci. 2023, 44, 873–880. [Google Scholar] [CrossRef] [PubMed]
  288. Filippi, M.; Bar-Or, A.; Piehl, F.; Preziosa, P.; Solari, A.; Vukusic, S.; Rocca, M.A. Multiple sclerosis. Nat. Rev. Dis. Primers 2018, 4, 43. [Google Scholar] [CrossRef] [PubMed]
  289. Karussis, D. The diagnosis of multiple sclerosis and the various related demyelinating syndromes: A critical review. J. Autoimmun. 2014, 48–49, 134–142. [Google Scholar] [CrossRef] [PubMed]
  290. Olek, M.J. Multiple Sclerosis. Ann. Intern. Med. 2021, 174, ITC81–ITC96. [Google Scholar] [CrossRef]
  291. Oh, J.; Vidal-Jordana, A.; Montalban, X. Multiple sclerosis: Clinical aspects. Curr. Opin. Neurol. 2018, 31, 752–759. [Google Scholar] [CrossRef] [PubMed]
  292. Compston, A.; Coles, A. Multiple sclerosis. Lancet 2008, 372, 1502–1517. [Google Scholar] [CrossRef] [PubMed]
  293. Thompson, A.J.; Baranzini, S.E.; Geurts, J.; Hemmer, B.; Ciccarelli, O. Multiple sclerosis. Lancet 2018, 391, 1622–1636. [Google Scholar] [CrossRef]
  294. Constantinescu, C.S.; Farooqi, N.; O’Brien, K.; Gran, B. Experimental autoimmune encephalomyelitis (EAE) as a model for multiple sclerosis (MS). Br. J. Pharmacol. 2011, 164, 1079–1106. [Google Scholar] [CrossRef]
  295. Mahad, D.H.; Trapp, B.D.; Lassmann, H. Pathological mechanisms in progressive multiple sclerosis. Lancet Neurol. 2015, 14, 183–193. [Google Scholar] [CrossRef]
  296. Kooij, G.; van Horssen, J.; de Lange, E.C.; Reijerkerk, A.; van der Pol, S.M.; van Het Hof, B.; Drexhage, J.; Vennegoor, A.; Killestein, J.; Scheffer, G.; et al. T lymphocytes impair P-glycoprotein function during neuroinflammation. J. Autoimmun. 2010, 34, 416–425. [Google Scholar] [CrossRef]
  297. Gray, E.; Rice, C.; Hares, K.; Redondo, J.; Kemp, K.; Williams, M.; Brown, A.; Scolding, N.; Wilkins, A. Reductions in neuronal peroxisomes in multiple sclerosis grey matter. Mult. Scler. 2014, 20, 651–659. [Google Scholar] [CrossRef] [PubMed]
  298. Zhang, W.; Liu, M.; Yang, L.; Huang, F.; Lan, Y.; Li, H.; Wu, H.; Zhang, B.; Shi, H.; Wu, X. P-glycoprotein Inhibitor Tariquidar Potentiates Efficacy of Astragaloside IV in Experimental Autoimmune Encephalomyelitis Mice. Molecules 2019, 24, 561. [Google Scholar] [CrossRef] [PubMed]
  299. Bogie, J.F.J.; Grajchen, E.; Wouters, E.; Corrales, A.G.; Dierckx, T.; Vanherle, S.; Mailleux, J.; Gervois, P.; Wolfs, E.; Dehairs, J.; et al. Stearoyl-CoA desaturase-1 impairs the reparative properties of macrophages and microglia in the brain. J. Exp. Med. 2020, 217, e20191660. [Google Scholar] [CrossRef] [PubMed]
  300. Al-Izki, S.; Pryce, G.; Hankey, D.J.; Lidster, K.; von Kutzleben, S.M.; Browne, L.; Clutterbuck, L.; Posada, C.; Edith Chan, A.W.; Amor, S.; et al. Lesional-targeting of neuroprotection to the inflammatory penumbra in experimental multiple sclerosis. Brain 2014, 137 Pt 1, 92–108. [Google Scholar] [CrossRef] [PubMed]
  301. Gerzanich, V.; Makar, T.K.; Guda, P.R.; Kwon, M.S.; Stokum, J.A.; Woo, S.K.; Ivanova, S.; Ivanov, A.; Mehta, R.I.; Morris, A.B.; et al. Salutary effects of glibenclamide during the chronic phase of murine experimental autoimmune encephalomyelitis. J. Neuroinflamm. 2017, 14, 177. [Google Scholar] [CrossRef] [PubMed]
  302. Jorissen, W.; Wouters, E.; Bogie, J.F.; Vanmierlo, T.; Noben, J.P.; Sviridov, D.; Hellings, N.; Somers, V.; Valcke, R.; Vanwijmeersch, B.; et al. Relapsing-remitting multiple sclerosis patients display an altered lipoprotein profile with dysfunctional HDL. Sci. Rep. 2017, 7, 43410. [Google Scholar] [CrossRef] [PubMed]
  303. Thiele Nee Schrewe, L.; Guse, K.; Tietz, S.; Remlinger, J.; Demir, S.; Pedreiturria, X.; Hoepner, R.; Salmen, A.; Pistor, M.; Turner, T.; et al. Functional relevance of the multi-drug transporter abcg2 on teriflunomide therapy in an animal model of multiple sclerosis. J. Neuroinflamm. 2020, 17, 9. [Google Scholar] [CrossRef]
  304. Cotte, S.; von Ahsen, N.; Kruse, N.; Huber, B.; Winkelmann, A.; Zettl, U.K.; Starck, M.; Konig, N.; Tellez, N.; Dorr, J.; et al. ABC-transporter gene-polymorphisms are potential pharmacogenetic markers for mitoxantrone response in multiple sclerosis. Brain 2009, 132 Pt 9, 2517–2530. [Google Scholar] [CrossRef]
  305. Gorska, A.; Santos-Garcia, I.; Villa-Cruz, M.; Wu, J.; van Genderen, S.; Pahnke, J. Recent Developments to Tackle Dementia and Movement Disorders; Soria-Moria Patologi Årsmøte, Soria-Moria Conference Center: Oslo, Norway, 2023. [Google Scholar]
  306. Mahringer, A.; Fricker, G. ABC transporters at the blood-brain barrier. Expert Opin. Drug Metab. Toxicol. 2016, 12, 499–508. [Google Scholar] [CrossRef] [PubMed]
  307. Osgood, D.; Miller, M.C.; Messier, A.A.; Gonzalez, L.; Silverberg, G.D. Aging alters mRNA expression of amyloid transporter genes at the blood-brain barrier. Neurobiol. Aging 2017, 57, 178–185. [Google Scholar] [CrossRef] [PubMed]
  308. Kawatani, K.; Holm, M.L.; Starling, S.C.; Martens, Y.A.; Zhao, J.; Lu, W.; Ren, Y.; Li, Z.; Jiang, P.; Jiang, Y.; et al. ABCA7 deficiency causes neuronal dysregulation by altering mitochondrial lipid metabolism. Mol. Psychiatry, 2023; ahead of print. [Google Scholar] [CrossRef]
  309. Vogelgesang, S.; Glatzel, M.; Walker, L.C.; Kroemer, H.K.; Aguzzi, A.; Warzok, R.W. Cerebrovascular P-glycoprotein expression is decreased in Creutzfeldt-Jakob disease. Acta Neuropathol. 2006, 111, 436–443. [Google Scholar] [CrossRef] [PubMed]
  310. Kaas, B.; Hillis, A.E.; Pantelyat, A. Progressive supranuclear palsy and pawpaw. Neurol. Clin. Pract. 2020, 10, e17–e18. [Google Scholar] [CrossRef] [PubMed]
  311. Manoharan, J.P.; Palanisamy, H.; Vidyalakshmi, S. Overcoming multi drug resistance mediated by ABC transporters by a novel acetogenin- annonacin from Annona muricata L. J. Ethnopharmacol. 2024, 322, 117598. [Google Scholar] [CrossRef] [PubMed]
  312. Mosser, J.; Douar, A.M.; Sarde, C.O.; Kioschis, P.; Feil, R.; Moser, H.; Poustka, A.M.; Mandel, J.L.; Aubourg, P. Putative X-linked adrenoleukodystrophy gene shares unexpected homology with ABC transporters. Nature 1993, 361, 726–730. [Google Scholar] [CrossRef]
  313. Coelho, D.; Kim, J.C.; Miousse, I.R.; Fung, S.; du Moulin, M.; Buers, I.; Suormala, T.; Burda, P.; Frapolli, M.; Stucki, M.; et al. Mutations in ABCD4 cause a new inborn error of vitamin B12 metabolism. Nat. Genet. 2012, 44, 1152–1155. [Google Scholar] [CrossRef]
  314. Zollmann, T.; Moiset, G.; Tumulka, F.; Tampe, R.; Poolman, B.; Abele, R. Single liposome analysis of peptide translocation by the ABC transporter TAPL. Proc. Natl. Acad. Sci. USA 2015, 112, 2046–2051. [Google Scholar] [CrossRef]
  315. Krishnamurthy, P.C.; Du, G.; Fukuda, Y.; Sun, D.; Sampath, J.; Mercer, K.E.; Wang, J.; Sosa-Pineda, B.; Murti, K.G.; Schuetz, J.D. Identification of a mammalian mitochondrial porphyrin transporter. Nature 2006, 443, 586–589. [Google Scholar] [CrossRef]
  316. Kim, S.; Lee, S.S.; Park, J.G.; Kim, J.W.; Ju, S.; Choi, S.H.; Kim, S.; Kim, N.J.; Hong, S.; Kang, J.Y.; et al. Structural Insights into Porphyrin Recognition by the Human ATP-Binding Cassette Transporter ABCB6. Mol. Cells 2022, 45, 575–587. [Google Scholar] [CrossRef]
  317. Ulrich, D.L.; Lynch, J.; Wang, Y.; Fukuda, Y.; Nachagari, D.; Du, G.; Sun, D.; Fan, Y.; Tsurkan, L.; Potter, P.M.; et al. ATP-dependent mitochondrial porphyrin importer ABCB6 protects against phenylhydrazine toxicity. J. Biol. Chem. 2012, 287, 12679–12690. [Google Scholar] [CrossRef] [PubMed]
  318. Allikmets, R.; Raskind, W.H.; Hutchinson, A.; Schueck, N.D.; Dean, M.; Koeller, D.M. Mutation of a putative mitochondrial iron transporter gene (ABC7) in X-linked sideroblastic anemia and ataxia (XLSA/A). Hum. Mol. Genet. 1999, 8, 743–749. [Google Scholar] [CrossRef] [PubMed]
  319. Bekri, S.; Kispal, G.; Lange, H.; Fitzsimons, E.; Tolmie, J.; Lill, R.; Bishop, D.F. Human ABC7 transporter: Gene structure and mutation causing X-linked sideroblastic anemia with ataxia with disruption of cytosolic iron-sulfur protein maturation. Blood 2000, 96, 3256–3264. [Google Scholar] [CrossRef] [PubMed]
  320. Pondarre, C.; Antiochos, B.B.; Campagna, D.R.; Clarke, S.L.; Greer, E.L.; Deck, K.M.; McDonald, A.; Han, A.P.; Medlock, A.; Kutok, J.L.; et al. The mitochondrial ATP-binding cassette transporter Abcb7 is essential in mice and participates in cytosolic iron-sulfur cluster biogenesis. Hum. Mol. Genet. 2006, 15, 953–964. [Google Scholar] [CrossRef] [PubMed]
  321. Pondarre, C.; Campagna, D.R.; Antiochos, B.; Sikorski, L.; Mulhern, H.; Fleming, M.D. Abcb7, the gene responsible for X-linked sideroblastic anemia with ataxia, is essential for hematopoiesis. Blood 2007, 109, 3567–3569. [Google Scholar] [CrossRef] [PubMed]
  322. Ichikawa, Y.; Bayeva, M.; Ghanefar, M.; Potini, V.; Sun, L.; Mutharasan, R.K.; Wu, R.; Khechaduri, A.; Jairaj Naik, T.; Ardehali, H. Disruption of ATP-binding cassette B8 in mice leads to cardiomyopathy through a decrease in mitochondrial iron export. Proc. Natl. Acad. Sci. USA 2012, 109, 4152–4157. [Google Scholar] [CrossRef] [PubMed]
  323. Liesa, M.; Qiu, W.; Shirihai, O.S. Mitochondrial ABC transporters function: The role of ABCB10 (ABC-me) as a novel player in cellular handling of reactive oxygen species. Biochim. Biophys. Acta 2012, 1823, 1945–1957. [Google Scholar] [CrossRef] [PubMed]
  324. Bear, C.E.; Li, C.H.; Kartner, N.; Bridges, R.J.; Jensen, T.J.; Ramjeesingh, M.; Riordan, J.R. Purification and functional reconstitution of the cystic fibrosis transmembrane conductance regulator (CFTR). Cell 1992, 68, 809–818. [Google Scholar] [CrossRef] [PubMed]
  325. Inagaki, N.; Gonoi, T.; Clement, J.P.; Wang, C.Z.; Aguilar-Bryan, L.; Bryan, J.; Seino, S. A family of sulfonylurea receptors determines the pharmacological properties of ATP-sensitive K+ channels. Neuron 1996, 16, 1011–1017. [Google Scholar] [CrossRef]
  326. Inagaki, N.; Gonoi, T.; Clement, J.P.T.; Namba, N.; Inazawa, J.; Gonzalez, G.; Aguilar-Bryan, L.; Seino, S.; Bryan, J. Reconstitution of IKATP: An inward rectifier subunit plus the sulfonylurea receptor. Science 1995, 270, 1166–1170. [Google Scholar] [CrossRef]
  327. Yamada, M.; Isomoto, S.; Matsumoto, S.; Kondo, C.; Shindo, T.; Horio, Y.; Kurachi, Y. Sulphonylurea receptor 2B and Kir6.1 form a sulphonylurea-sensitive but ATP-insensitive K+ channel. J. Physiol. 1997, 499 Pt 3, 715–720. [Google Scholar] [CrossRef] [PubMed]
  328. Neumann, J.; Rose-Sperling, D.; Hellmich, U.A. Diverse relations between ABC transporters and lipids: An overview. Biochim. Biophys. Acta Biomembr. 2017, 1859, 605–618. [Google Scholar] [CrossRef] [PubMed]
  329. Plummer, A.M.; Culbertson, A.T.; Liao, M. The ABCs of Sterol Transport. Annu. Rev. Physiol. 2021, 83, 153–181. [Google Scholar] [CrossRef] [PubMed]
  330. Fitzgerald, M.L.; Morris, A.L.; Chroni, A.; Mendez, A.J.; Zannis, V.I.; Freeman, M.W. ABCA1 and amphipathic apolipoproteins form high-affinity molecular complexes required for cholesterol efflux. J. Lipid Res. 2004, 45, 287–294. [Google Scholar] [CrossRef] [PubMed]
  331. Jacobo-Albavera, L.; Dominguez-Perez, M.; Medina-Leyte, D.J.; Gonzalez-Garrido, A.; Villarreal-Molina, T. The Role of the ATP-Binding Cassette A1 (ABCA1) in Human Disease. Int. J. Mol. Sci. 2021, 22, 1593. [Google Scholar] [CrossRef] [PubMed]
  332. Kaminski, W.E.; Piehler, A.; Pullmann, K.; Porsch-Ozcurumez, M.; Duong, C.; Bared, G.M.; Buchler, C.; Schmitz, G. Complete coding sequence, promoter region, and genomic structure of the human ABCA2 gene and evidence for sterol-dependent regulation in macrophages. Biochem. Biophys. Res. Commun. 2001, 281, 249–258. [Google Scholar] [CrossRef] [PubMed]
  333. Garmany, T.H.; Moxley, M.A.; White, F.V.; Dean, M.; Hull, W.M.; Whitsett, J.A.; Nogee, L.M.; Hamvas, A. Surfactant composition and function in patients with ABCA3 mutations. Pediatr. Res. 2006, 59, 801–805. [Google Scholar] [CrossRef] [PubMed]
  334. Mulugeta, S.; Gray, J.M.; Notarfrancesco, K.L.; Gonzales, L.W.; Koval, M.; Feinstein, S.I.; Ballard, P.L.; Fisher, A.B.; Shuman, H. Identification of LBM180, a lamellar body limiting membrane protein of alveolar type II cells, as the ABC transporter protein ABCA3. J. Biol. Chem. 2002, 277, 22147–22155. [Google Scholar] [CrossRef]
  335. Cheong, N.; Madesh, M.; Gonzales, L.W.; Zhao, M.; Yu, K.; Ballard, P.L.; Shuman, H. Functional and trafficking defects in ATP binding cassette A3 mutants associated with respiratory distress syndrome. J. Biol. Chem. 2006, 281, 9791–9800. [Google Scholar] [CrossRef]
  336. Vegeto, E.; Villa, A.; Della Torre, S.; Crippa, V.; Rusmini, P.; Cristofani, R.; Galbiati, M.; Maggi, A.; Poletti, A. The Role of Sex and Sex Hormones in Neurodegenerative Diseases. Endocr. Rev. 2020, 41, 273–319. [Google Scholar]
  337. Vaidya, B.; Dhamija, K.; Guru, P.; Sharma, S.S. Parkinson’s disease in women: Mechanisms underlying sex differences. Eur. J. Pharmacol. 2021, 895, 173862. [Google Scholar] [CrossRef] [PubMed]
  338. Atwood, C.S.; Bowen, R.L. The endocrine dyscrasia that accompanies menopause and andropause induces aberrant cell cycle signaling that triggers re-entry of post-mitotic neurons into the cell cycle, neurodysfunction, neurodegeneration and cognitive disease. Horm. Behav. 2015, 76, 63–80. [Google Scholar] [CrossRef] [PubMed]
  339. Cheng, Y.J.; Lin, C.H.; Lane, H.Y. From Menopause to Neurodegeneration-Molecular Basis and Potential Therapy. Int. J. Mol. Sci. 2021, 22, 8654. [Google Scholar] [CrossRef]
  340. Sumien, N.; Cunningham, J.T.; Davis, D.L.; Engelland, R.; Fadeyibi, O.; Farmer, G.E.; Mabry, S.; Mensah-Kane, P.; Trinh, O.T.P.; Vann, P.H.; et al. Neurodegenerative Disease: Roles for Sex, Hormones, and Oxidative Stress. Endocrinology 2021, 162, bqab185. [Google Scholar] [CrossRef] [PubMed]
  341. Barth, C.; Crestol, A.; de Lange, A.G.; Galea, L.A.M. Sex steroids and the female brain across the lifespan: Insights into risk of depression and Alzheimer’s disease. Lancet Diabetes Endocrinol. 2023, 11, 926–941. [Google Scholar] [CrossRef] [PubMed]
  342. Cho, E.; Montgomery, R.B.; Mostaghel, E.A. Minireview: SLCO and ABC transporters: A role for steroid transport in prostate cancer progression. Endocrinology 2014, 155, 4124–4132. [Google Scholar] [CrossRef] [PubMed]
  343. Samant, M.D.; Jackson, C.M.; Felix, C.L.; Jones, A.J.; Goodrich, D.W.; Foster, B.A.; Huss, W.J. Multi-Drug Resistance ABC Transporter Inhibition Enhances Murine Ventral Prostate Stem/Progenitor Cell Differentiation. Stem Cells Dev. 2015, 24, 1236–1251. [Google Scholar] [CrossRef] [PubMed]
  344. Ueda, K.; Okamura, N.; Hirai, M.; Tanigawara, Y.; Saeki, T.; Kioka, N.; Komano, T.; Hori, R. Human P-glycoprotein transports cortisol, aldosterone, and dexamethasone, but not progesterone. J. Biol. Chem. 1992, 267, 24248–24252. [Google Scholar] [CrossRef]
  345. Sivils, J.C.; Gonzalez, I.; Bain, L.J. Mice lacking Mrp1 have reduced testicular steroid hormone levels and alterations in steroid biosynthetic enzymes. Gen. Comp. Endocrinol. 2010, 167, 51–59. [Google Scholar] [CrossRef]
  346. Qian, Y.M.; Song, W.C.; Cui, H.; Cole, S.P.; Deeley, R.G. Glutathione stimulates sulfated estrogen transport by multidrug resistance protein 1. J. Biol. Chem. 2001, 276, 6404–6411. [Google Scholar] [CrossRef]
  347. Zelcer, N.; Reid, G.; Wielinga, P.; Kuil, A.; van der Heijden, I.; Schuetz, J.D.; Borst, P. Steroid and bile acid conjugates are substrates of human multidrug-resistance protein (MRP) 4 (ATP-binding cassette C4). Biochem. J. 2003, 371 Pt 2, 361–367. [Google Scholar] [CrossRef] [PubMed]
  348. Li, C.Y.; Basit, A.; Gupta, A.; Gaborik, Z.; Kis, E.; Prasad, B. Major glucuronide metabolites of testosterone are primarily transported by MRP2 and MRP3 in human liver, intestine and kidney. J. Steroid Biochem. Mol. Biol. 2019, 191, 105350. [Google Scholar] [CrossRef] [PubMed]
  349. Jarvinen, E.; Kidron, H.; Finel, M. Human efflux transport of testosterone, epitestosterone and other androgen glucuronides. J. Steroid Biochem. Mol. Biol. 2020, 197, 105518. [Google Scholar] [CrossRef] [PubMed]
  350. Lee, Y.M.; Cui, Y.; Konig, J.; Risch, A.; Jager, B.; Drings, P.; Bartsch, H.; Keppler, D.; Nies, A.T. Identification and functional characterization of the natural variant MRP3-Arg1297His of human multidrug resistance protein 3 (MRP3/ABCC3). Pharmacogenetics 2004, 14, 213–223. [Google Scholar] [CrossRef] [PubMed]
  351. Wang, J.; Li, X.; Wang, F.F.; Cheng, M.T.; Mao, Y.X.; Fang, S.C.; Wang, L.; Zhou, C.Z.; Hou, W.T.; Chen, Y. Placing steroid hormones within the human ABCC3 transporter reveals a compatible amphiphilic substrate-binding pocket. EMBO J. 2023, 42, e113415. [Google Scholar] [CrossRef] [PubMed]
  352. Chen, Z.S.; Guo, Y.; Belinsky, M.G.; Kotova, E.; Kruh, G.D. Transport of bile acids, sulfated steroids, estradiol 17-beta-D-glucuronide, and leukotriene C4 by human multidrug resistance protein 8 (ABCC11). Mol. Pharmacol. 2005, 67, 545–557. [Google Scholar] [CrossRef]
  353. Imai, Y.; Asada, S.; Tsukahara, S.; Ishikawa, E.; Tsuruo, T.; Sugimoto, Y. Breast cancer resistance protein exports sulfated estrogens but not free estrogens. Mol. Pharmacol. 2003, 64, 610–618. [Google Scholar] [CrossRef] [PubMed]
  354. Goicoechea, L.; Conde de la Rosa, L.; Torres, S.; Garcia-Ruiz, C.; Fernandez-Checa, J.C. Mitochondrial cholesterol: Metabolism and impact on redox biology and disease. Redox Biol. 2023, 61, 102643. [Google Scholar] [CrossRef]
Figure 4. Clinical course of EAE in ABCB1a/b-ko mice (purple) compared to C57BL/6J control mice (grey). Data presented in the figure (mean +/− SEM) were statistically analyzed using a Mann–Whitney U test and Wilcoxon rank-sum test. * p < 0.05; ** p < 0.01. ABCB1-ko n = 15; C57BL/6J n = 13.
Figure 4. Clinical course of EAE in ABCB1a/b-ko mice (purple) compared to C57BL/6J control mice (grey). Data presented in the figure (mean +/− SEM) were statistically analyzed using a Mann–Whitney U test and Wilcoxon rank-sum test. * p < 0.05; ** p < 0.01. ABCB1-ko n = 15; C57BL/6J n = 13.
Cells 13 00740 g004
Figure 5. Clinical course of EAE in ABCC1-ko mice (blue) compared to C57BL/6J control mice (grey). Data presented in the figure (mean +/− SEM) were statistically analyzed using a Mann–Whitney U test and Wilcoxon rank-sum test. * p < 0.05; ** p < 0.01. ABCC1-ko n = 22; C57BL/6J n = 25.
Figure 5. Clinical course of EAE in ABCC1-ko mice (blue) compared to C57BL/6J control mice (grey). Data presented in the figure (mean +/− SEM) were statistically analyzed using a Mann–Whitney U test and Wilcoxon rank-sum test. * p < 0.05; ** p < 0.01. ABCC1-ko n = 22; C57BL/6J n = 25.
Cells 13 00740 g005
Figure 6. Clinical course of EAE in ABCA7-ko mice (red) compared to C57BL/6J control mice (grey). Data presented in the figure (mean +/− SEM) were statistically analyzed using a Mann–Whitney U test and Wilcoxon rank-sum test. * p < 0.05; ** p < 0.01; *** p < 0.001. ABCA7-ko n = 31 until day 27 and n = 22 from day 28; C57BL/6J n = 34 until day 27 and n = 28 from day 28.
Figure 6. Clinical course of EAE in ABCA7-ko mice (red) compared to C57BL/6J control mice (grey). Data presented in the figure (mean +/− SEM) were statistically analyzed using a Mann–Whitney U test and Wilcoxon rank-sum test. * p < 0.05; ** p < 0.01; *** p < 0.001. ABCA7-ko n = 31 until day 27 and n = 22 from day 28; C57BL/6J n = 34 until day 27 and n = 28 from day 28.
Cells 13 00740 g006
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

Villa, M.; Wu, J.; Hansen, S.; Pahnke, J. Emerging Role of ABC Transporters in Glia Cells in Health and Diseases of the Central Nervous System. Cells 2024, 13, 740. https://doi.org/10.3390/cells13090740

AMA Style

Villa M, Wu J, Hansen S, Pahnke J. Emerging Role of ABC Transporters in Glia Cells in Health and Diseases of the Central Nervous System. Cells. 2024; 13(9):740. https://doi.org/10.3390/cells13090740

Chicago/Turabian Style

Villa, Maria, Jingyun Wu, Stefanie Hansen, and Jens Pahnke. 2024. "Emerging Role of ABC Transporters in Glia Cells in Health and Diseases of the Central Nervous System" Cells 13, no. 9: 740. https://doi.org/10.3390/cells13090740

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