Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 6 de 6
1.
Int J Mol Sci ; 23(23)2022 Nov 24.
Article En | MEDLINE | ID: mdl-36498995

Maintenance of the tightly regulated homeostatic environment of the brain is facilitated by the blood-brain barrier (BBB). P-glycoprotein (P-gp), an ATP-binding cassette transporter, is expressed on the luminal surface of the endothelial cells in the BBB, and actively exports a wide variety of substrates to limit exposure of the vulnerable brain environment to waste buildup and neurotoxic compounds. Downregulation of P-gp expression and activity at the BBB have been reported with ageing and in neurodegenerative diseases. Upregulation of P-gp at the BBB contributes to poor therapeutic outcomes due to altered pharmacokinetics of CNS-acting drugs. The regulation of P-gp is highly complex, but unravelling the mechanisms involved may help the development of novel and nuanced strategies to modulate P-gp expression for therapeutic benefit. This review summarises the current understanding of P-gp regulation in the brain, encompassing the transcriptional, post-transcriptional and post-translational mechanisms that have been identified to affect P-gp expression and transport activity.


ATP Binding Cassette Transporter, Subfamily B, Member 1 , Endothelial Cells , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Endothelial Cells/metabolism , ATP Binding Cassette Transporter, Subfamily B/metabolism , Brain/metabolism , Blood-Brain Barrier/metabolism , Central Nervous System Agents
2.
Int J Mol Sci ; 23(3)2022 Jan 18.
Article En | MEDLINE | ID: mdl-35162941

The ATP-binding cassette transporter, P-glycoprotein (P-gp), has been demonstrated to facilitate the clearance of amyloid-beta (Aß) peptides, exporting the neurotoxic entity out of neurons and out of the brain via the blood-brain barrier. However, its expression and function diminish with age and in Alzheimer's disease. P-gp is known to undergo ubiquitination, a post-translational modification that results in internalisation and/or degradation of the protein. NEDD4-1 is a ubiquitin E3 ligase that has previously been shown to ubiquitinate P-gp and reduce its cell surface expression. However, whether this effect translates into altered P-gp activity remains to be determined. siRNA was used to knockdown the expression of Nedd4 in CHO-APP cells. Western blot analysis confirmed that absence of Nedd4 was associated with increased P-gp protein expression. This was accompanied by increased transport activity, as shown by export of the P-gp substrate calcein-AM, as well as enhanced secretion of Aß peptides, as shown by ELISA. These results implicate Nedd4 in the regulation of P-gp, and highlight a potential approach for restoring or augmenting P-gp expression and function to facilitate Aß clearance from the brain.


Amyloid beta-Protein Precursor/metabolism , Cloning, Molecular/methods , Nedd4 Ubiquitin Protein Ligases/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Biological Transport , CHO Cells , Cricetulus , Fluoresceins/metabolism , Gene Expression Regulation , Gene Knockdown Techniques , Humans , Nedd4 Ubiquitin Protein Ligases/metabolism , Ubiquitination
3.
Article En | MEDLINE | ID: mdl-34044125

Since the identification of the apolipoprotein E (apoE) *ε4 allele as a major genetic risk factor for late-onset Alzheimer's disease, significant efforts have been aimed at elucidating how apoE4 expression confers greater brain amyloid-ß (Aß) burden, earlier disease onset and worse clinical outcomes compared to apoE2 and apoE3. ApoE primarily functions as a lipid carrier to regulate cholesterol metabolism in circulation as well as in the brain. However, it has also been suggested to interact with hydrophobic Aß peptides to influence their processing in an isoform-dependent manner. Here, we review evidence from in vitro and in vivo studies extricating the effects of the three apoE isoforms, on different stages of the Aß processing pathway including synthesis, aggregation, deposition, clearance and degradation. ApoE4 consistently correlates with impaired Aß clearance, however data regarding Aß synthesis and aggregation are conflicting and likely reflect inconsistencies in experimental approaches across studies. We further discuss the physical and chemical properties of apoE that may explain the inherent differences in activity between the isoforms. The lipidation status and lipid transport function of apoE are intrinsically linked with its ability to interact with Aß. Traditionally, apoE-oriented therapeutic strategies for Alzheimer's disease have been proposed to non-specifically enhance or inhibit apoE activity. However, given the wide-ranging physiological functions of apoE in the brain and periphery, a more viable approach may be to specifically target and neutralise the pathological apoE4 isoform.


Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Apolipoproteins E/metabolism , Alleles , Alzheimer Disease/genetics , Brain/metabolism , Humans , Protein Isoforms/metabolism
4.
Int J Mol Sci ; 22(1)2020 Dec 29.
Article En | MEDLINE | ID: mdl-33383667

Defective clearance mechanisms lead to the accumulation of amyloid-beta (Aß) peptides in the Alzheimer's brain. Though predominantly generated in neurons, little is known about how these hydrophobic, aggregation-prone, and tightly membrane-associated peptides exit into the extracellular space where they deposit and propagate neurotoxicity. The ability for P-glycoprotein (P-gp), an ATP-binding cassette (ABC) transporter, to export Aß across the blood-brain barrier (BBB) has previously been reported. However, controversies surrounding the P-gp-Aß interaction persist. Here, molecular data affirm that both Aß40 and Aß42 peptide isoforms directly interact with and are substrates of P-gp. This was reinforced ex vivo by the inhibition of Aß42 transport in brain capillaries from P-gp-knockout mice. Moreover, we explored whether P-gp could exert the same role in neurons. Comparison between non-neuronal CHO-APP and human neuroblastoma SK-N-SH cells revealed that P-gp is expressed and active in both cell types. Inhibiting P-gp activity using verapamil and nicardipine impaired Aß40 and Aß42 secretion from both cell types, as determined by ELISA. Collectively, these findings implicate P-gp in Aß export from neurons, as well as across the BBB endothelium, and suggest that restoring or enhancing P-gp function could be a viable therapeutic approach for removing excess Aß out of the brain in Alzheimer's disease.


ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Blood-Brain Barrier/metabolism , Neurons/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Brain/metabolism , CHO Cells , Capillaries/metabolism , Cell Line, Tumor , Cell Survival , Cricetulus , Gene Expression , Humans , Peptide Fragments/metabolism , Protein Binding , Protein Transport
5.
FEBS J ; 287(4): 612-625, 2020 02.
Article En | MEDLINE | ID: mdl-31750987

The accumulation of amyloid-ß (Aß) peptides is a key histopathological feature of the Alzheimer's brain. Defective clearance mechanisms result in toxic levels of soluble Aß40 and Aß42 oligomers, leading to impaired synaptic function, neurodegeneration and cognitive decline. Growing evidence points to the involvement of P-glycoprotein (P-gp or ABCB1), an ATP-binding cassette transporter highly expressed on the luminal side of the blood-brain barrier, in facilitating the clearance of Aß from the brain. In this review, we summarise evidence from human, animal and in vitro studies examining the contribution of P-gp to Aß clearance, and discuss the potential for P-gp as a novel pharmacological target in Alzheimer's disease (AD). P-gp expression and activity in the brain are inversely correlated with ageing, Aß deposition and AD. Moreover, Aß itself has been found to compromise the expression of P-gp, thereby exacerbating Aß deposition and disease. Despite decades of research, the pathophysiology of AD remains elusive. Understanding the normal versus impaired processing and clearance mechanisms affecting Aß peptides will assist the development of more effective therapeutic agents to combat this progressive neurodegenerative condition that continues to devastate millions of patients globally.


Alzheimer Disease/genetics , Amyloid beta-Peptides/genetics , Blood-Brain Barrier/metabolism , Brain/metabolism , Peptide Fragments/genetics , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Animals , Blood-Brain Barrier/pathology , Brain/pathology , Disease Models, Animal , Gene Expression Regulation , Humans , Neurons/metabolism , Neurons/pathology , Peptide Fragments/metabolism , Presenilin-1/genetics , Presenilin-1/metabolism , Presenilin-2/genetics , Presenilin-2/metabolism , Protein Aggregates , Protein Transport , Proteolysis , Signal Transduction
6.
Respir Res ; 18(1): 41, 2017 02 28.
Article En | MEDLINE | ID: mdl-28241820

Respiratory diseases including asthma and chronic obstructive pulmonary disease (COPD) are characterised by excessive and persistent inflammation. Current treatments are often inadequate for symptom and disease control, and hence new therapies are warranted. Recent emerging research has implicated dyslipidaemia in pulmonary inflammation. Three ATP-binding cassette (ABC) transporters are found in the mammalian lung - ABCA1, ABCG1 and ABCA3 - that are involved in movement of cholesterol and phospholipids from lung cells. The aim of this review is to corroborate the current evidence for the role of ABC lipid transporters in pulmonary lipid homeostasis and inflammation. Here, we summarise results from murine knockout studies, human diseases associated with ABC transporter mutations, and in vitro studies. Disruption to ABC transporter activity results in lipid accumulation and elevated levels of inflammatory cytokines in lung tissue. Furthermore, these ABC-knockout mice exhibit signs of respiratory distress. ABC lipid transporters appear to have a crucial and protective role in the lung. However, our knowledge of the underlying molecular mechanisms for these benefits requires further attention. Understanding the relationship between cholesterol and inflammation in the lung, and the role that ABC transporters play in this may illuminate new pathways to target for the treatment of inflammatory lung diseases.


ATP Binding Cassette Transporter, Subfamily A/immunology , Lipid Metabolism/immunology , Lipids/immunology , Lung/immunology , Models, Immunological , Pneumonia/immunology , Animals , Cytokines/immunology , Homeostasis/immunology , Humans , Mice
...