51
|
Gil-Martins E, Barbosa DJ, Silva V, Remião F, Silva R. Dysfunction of ABC transporters at the blood-brain barrier: Role in neurological disorders. Pharmacol Ther 2020; 213:107554. [PMID: 32320731 DOI: 10.1016/j.pharmthera.2020.107554] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 04/07/2020] [Indexed: 12/14/2022]
Abstract
ABC (ATP-binding cassette) transporters represent one of the largest and most diverse superfamily of proteins in living species, playing an important role in many biological processes such as cell homeostasis, cell signaling, drug metabolism and nutrient uptake. Moreover, using the energy generated from ATP hydrolysis, they mediate the efflux of endogenous and exogenous substrates from inside the cells, thereby reducing their intracellular accumulation. At present, 48 ABC transporters have been identified in humans, which were classified into 7 different subfamilies (A to G) according to their phylogenetic analysis. Nevertheless, the most studied members with importance in drug therapeutic efficacy and toxicity include P-glycoprotein (P-gp), a member of the ABCB subfamily, the multidrug-associated proteins (MPRs), members of the ABCC subfamily, and breast cancer resistance protein (BCRP), a member of the ABCG subfamily. They exhibit ubiquitous expression throughout the human body, with a special relevance in barrier tissues like the blood-brain barrier (BBB). At this level, they play a physiological function in tissue protection by reducing or limiting the brain accumulation of neurotoxins. Furthermore, dysfunction of ABC transporters, at expression and/or activity level, has been associated with many neurological diseases, including epilepsy, multiple sclerosis, Alzheimer's disease, and amyotrophic lateral sclerosis. Additionally, these transporters are strikingly associated with the pharmacoresistance to central nervous system (CNS) acting drugs, because they contribute to the decrease in drug bioavailability. This article reviews the signaling pathways that regulate the expression and activity of P-gp, BCRP and MRPs subfamilies of transporters, with particular attention at the BBB level, and their mis-regulation in neurological disorders.
Collapse
Affiliation(s)
- Eva Gil-Martins
- UCIBIO-REQUIMTE, Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Daniel José Barbosa
- Instituto de Biologia Molecular e Celular (IBMC), Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal.
| | - Vera Silva
- UCIBIO-REQUIMTE, Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Fernando Remião
- UCIBIO-REQUIMTE, Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| | - Renata Silva
- UCIBIO-REQUIMTE, Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| |
Collapse
|
52
|
Oikari LE, Pandit R, Stewart R, Cuní-López C, Quek H, Sutharsan R, Rantanen LM, Oksanen M, Lehtonen S, de Boer CM, Polo JM, Götz J, Koistinaho J, White AR. Altered Brain Endothelial Cell Phenotype from a Familial Alzheimer Mutation and Its Potential Implications for Amyloid Clearance and Drug Delivery. Stem Cell Reports 2020; 14:924-939. [PMID: 32275861 PMCID: PMC7220857 DOI: 10.1016/j.stemcr.2020.03.011] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 03/11/2020] [Accepted: 03/12/2020] [Indexed: 12/30/2022] Open
Abstract
The blood-brain barrier (BBB) presents a barrier for circulating factors, but simultaneously challenges drug delivery. How the BBB is altered in Alzheimer disease (AD) is not fully understood. To facilitate this analysis, we derived brain endothelial cells (iBECs) from human induced pluripotent stem cells (hiPSCs) of several patients carrying the familial AD PSEN1 mutation. We demonstrate that, compared with isogenic PSEN1 corrected and control iBECs, AD-iBECs exhibit altered tight and adherens junction protein expression as well as efflux properties. Furthermore, by applying focused ultrasound (FUS) that transiently opens the BBB and achieves multiple therapeutic effects in AD mouse models, we found an altered permeability to 3–5 kDa dextran as a model cargo and the amyloid-β (Aβ) peptide in AD-iBECs compared with control iBECs. This presents human-derived in vitro models of the BBB as a valuable tool to understand its role and properties in a disease context, with possible implications for drug delivery. iBECs with familial AD mutation express altered levels of tight junction proteins AD-iBECs exhibit altered efflux transporter expression and function to control iBECs Focused ultrasound disrupts iBEC monolayer indicating effects of BBB opening AD-iBECs respond differently to control iBECs to effects of focused ultrasound
Collapse
Affiliation(s)
- Lotta E Oikari
- Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Rucha Pandit
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Romal Stewart
- Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Carla Cuní-López
- Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Hazel Quek
- Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Ratneswary Sutharsan
- Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia; Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Laura M Rantanen
- Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Minna Oksanen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Sarka Lehtonen
- Neuroscience Center, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland; A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Carmela Maria de Boer
- Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia
| | - Jose M Polo
- Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia; Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
| | - Jürgen Götz
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Jari Koistinaho
- Neuroscience Center, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland; A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Anthony R White
- Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.
| |
Collapse
|
53
|
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. [PMID: 32257549 PMCID: PMC7069460 DOI: 10.14336/ad.2019.0519] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 05/19/2019] [Indexed: 12/17/2022] Open
Abstract
The neurovascular unit (NVU) plays an important role in maintaining the function of the central nervous system (CNS). Emerging evidence has indicated that the NVU changes function and molecules at the early stage of Alzheimer’s disease (AD), which initiates multiple pathways of neurodegeneration. Cell types in the NVU have become attractive targets in the interventional treatment of AD. The NVU transportation system contains a variety of proteins involved in compound transport and neurotransmission. Brain transporters can be classified as members of the solute carrier (SLC) and ATP-binding cassette (ABC) families in the NVU. Moreover, the transporters can regulate both endogenous toxins, including amyloid-beta (Aβ) and xenobiotic homeostasis, in the brains of AD patients. Genome-wide association studies (GWAS) have identified some transporter gene variants as susceptibility loci for late-onset AD. Therefore, the present study summarizes changes in blood-brain barrier (BBB) permeability in AD, identifies the location of SLC and ABC transporters in the brain and focuses on major SLC and ABC transporters that contribute to AD pathology.
Collapse
Affiliation(s)
- Yongming Jia
- 1Department of Neuropharmacology, College of Pharmacy, Qiqihar Medical University, Qiqihar, China
| | - Na Wang
- 2Department of Pathophysiology, Basic Medical Science College, Qiqihar Medical University, Qiqihar, China
| | - Yingbo Zhang
- 3College of Pathology, Qiqihar Medical University, Qiqihar, China
| | - Di Xue
- 1Department of Neuropharmacology, College of Pharmacy, Qiqihar Medical University, Qiqihar, China
| | - Haoming Lou
- 4Department of Medicinal Chemistry and Chemistry of Chinese Materia Medica, School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Xuewei Liu
- 1Department of Neuropharmacology, College of Pharmacy, Qiqihar Medical University, Qiqihar, China
| |
Collapse
|
54
|
Shubbar MH, Penny JI. Therapeutic drugs modulate ATP-Binding cassette transporter-mediated transport of amyloid beta (1-42) in brain microvascular endothelial cells. Eur J Pharmacol 2020; 874:173009. [PMID: 32061744 DOI: 10.1016/j.ejphar.2020.173009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 01/28/2020] [Accepted: 02/10/2020] [Indexed: 01/07/2023]
Abstract
Deposition of amyloid-β peptide (Aβ(1-42)) is a hallmark of Alzheimer's disease. Clearance of Aβ(1-42), across the blood-brain barrier (BBB), is mediated by ATP-binding Cassette (ABC) efflux transporters. Many therapeutic drugs inhibit ABC transporters, but little is known of the effect of therapeutic drugs on Aβ(1-42) transport across BBB endothelial cells. The effects of selected, widely prescribed, therapeutic drugs on ABCB1, ABCC5 and ABCG2 activities were determined by measuring intracellular levels of calcein, GS-MF, and Hoechst 33342 respectively in primary porcine brain endothelial cells (PBECs). The ability of ABCB1, ABCC5 and ABCG2 to transport Aβ(1-42) was determined using fluorescent Aβ(1-42). The ability of the ABCB1, ABCC5 and ABCG2 inhibitor telmisartan to modify transcellular Aβ(1-42) transport was investigated using PBEC monolayers housed in Transwell® inserts. Treatment of PBECs with ABC transporter inhibitory drugs (indomethacin, olanzapine, chlorpromazine, telmisartan, pantoprazole, quinidine, sulfasalazine and nefazodone) increased Aβ(1-42) intracellular accumulation. Inhibition of ABCB1, ABCC5 and ABCG2 by telmisartan increased Aβ(1-42) transport in the apical to basal direction and reduced its transport in basal to apical direction in PBEC monolayers. ABCB1, ABCC5 and ABCG2 mediate the efflux transport of Aβ(1-42) in BBB endothelial cells. Inhibition of ABC transporters by therapeutic drugs, at plasma concentrations, could decrease Aβ(1-42) clearance from brain, across BBB endothelial cells into blood, and potentially influence levels of the Aβ(1-42) peptide within the brain.
Collapse
Affiliation(s)
- Maryam H Shubbar
- Division of Pharmacy & Optometry, University of Manchester, Manchester, M13 9PT, UK.
| | - Jeffrey I Penny
- Division of Pharmacy & Optometry, University of Manchester, Manchester, M13 9PT, UK
| |
Collapse
|
55
|
Zoufal V, Wanek T, Krohn M, Mairinger S, Filip T, Sauberer M, Stanek J, Pekar T, Bauer M, Pahnke J, Langer O. Age dependency of cerebral P-glycoprotein function in wild-type and APPPS1 mice measured with PET. J Cereb Blood Flow Metab 2020; 40:150-162. [PMID: 30354871 PMCID: PMC6928546 DOI: 10.1177/0271678x18806640] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
P-glycoprotein (P-gp, ABCB1) is an efflux transporter at the blood-brain barrier (BBB), which mediates clearance of beta-amyloid (Aβ) from brain into blood. We used (R)-[11C]verapamil PET in combination with partial P-gp inhibition with tariquidar to measure cerebral P-gp function in a beta-amyloidosis mouse model (APPtg) and in control mice at three different ages (50, 200 and 380 days). Following tariquidar pre-treatment (4 mg/kg), whole brain-to-plasma radioactivity concentration ratios (Kp,brain) were significantly higher in APPtg than in wild-type mice aged 50 days, pointing to decreased cerebral P-gp function. Moreover, we found an age-dependent decrease in cerebral P-gp function in both wild-type and APPtg mice of up to -50%. Alterations in P-gp function were more pronounced in Aβ-rich brain regions (hippocampus, cortex) than in a control region with negligible Aβ load (cerebellum). PET results were confirmed by immunohistochemical staining of P-gp in brain microvessels. Our results confirm previous findings of reduced P-gp function in Alzheimer's disease mouse models and show that our PET protocol possesses adequate sensitivity to measure these functional changes in vivo. Our PET protocol may find use in clinical studies to test the efficacy of drugs to induce P-gp function at the human BBB to enhance Aβ clearance.
Collapse
Affiliation(s)
- Viktoria Zoufal
- Center for Health & Bioresources, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Thomas Wanek
- Center for Health & Bioresources, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Markus Krohn
- Department of Neuro/Pathology, University of Oslo (UiO) and Oslo University Hospital (OUS), Oslo, Norway
| | - Severin Mairinger
- Center for Health & Bioresources, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Thomas Filip
- Center for Health & Bioresources, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Michael Sauberer
- Center for Health & Bioresources, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Johann Stanek
- Center for Health & Bioresources, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Thomas Pekar
- University of Applied Sciences, Wiener Neustadt, Austria
| | - Martin Bauer
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Jens Pahnke
- Department of Neuro/Pathology, University of Oslo (UiO) and Oslo University Hospital (OUS), Oslo, Norway.,LIED, University of Lübeck, Lübeck, Germany.,Leibniz-Institute of Plant Biochemistry, Halle, Germany.,Department of Pharmacology, University of Latvia, Rīga, Latvia
| | - Oliver Langer
- Center for Health & Bioresources, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria.,Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria.,Department of Biomedical Imaging und Image-Guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
56
|
Sekhar GN, Fleckney AL, Boyanova ST, Rupawala H, Lo R, Wang H, Farag DB, Rahman KM, Broadstock M, Reeves S, Thomas SA. Region-specific blood-brain barrier transporter changes leads to increased sensitivity to amisulpride in Alzheimer's disease. Fluids Barriers CNS 2019; 16:38. [PMID: 31842924 PMCID: PMC6915870 DOI: 10.1186/s12987-019-0158-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 12/02/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Research into amisulpride use in Alzheimer's disease (AD) implicates blood-brain barrier (BBB) dysfunction in antipsychotic sensitivity. Research into BBB transporters has been mainly directed towards the ABC superfamily, however, solute carrier (SLC) function in AD has not been widely studied. This study tests the hypothesis that transporters for organic cations contribute to the BBB delivery of the antipsychotics (amisulpride and haloperidol) and is disrupted in AD. METHODS The accumulation of [3H]amisulpride (3.7-7.7 nM) and [3H]haloperidol (10 nM) in human (hCMEC/D3) and mouse (bEnd.3) brain endothelial cell lines was explored. Computational approaches examined molecular level interactions of both drugs with the SLC transporters [organic cation transporter 1 (OCT1), plasma membrane monoamine transporter (PMAT) and multi-drug and toxic compound extrusion proteins (MATE1)] and amisulpride with the ABC transporter (P-glycoprotein). The distribution of [3H]amisulpride in wildtype and 3×transgenic AD mice was examined using in situ brain perfusion experiments. Western blots determined transporter expression in mouse and human brain capillaries . RESULTS In vitro BBB and in silico transporter studies indicated that [3H]amisulpride and [3H]haloperidol were transported by the influx transporter, OCT1, and efflux transporters MATE1 and PMAT. Amisulpride did not have a strong interaction with OCTN1, OCTN2, P-gp, BCRP or MRP and could not be described as a substrate for these transporters. Amisulpride brain uptake was increased in AD mice compared to wildtype mice, but vascular space was unaffected. There were no measurable changes in the expression of MATE1, MATE2, PMAT OCT1, OCT2, OCT3, OCTN1, OCTN2 and P-gp in capillaries isolated from whole brain homogenates from the AD mice compared to wildtype mice. Although, PMAT and MATE1 expression was reduced in capillaries obtained from specific human brain regions (i.e. putamen and caudate) from AD cases (Braak stage V-VI) compared to age matched controls (Braak stage 0-II). CONCLUSIONS Together our research indicates that the increased sensitivity of individuals with Alzheimer's to amisulpride is related to previously unreported changes in function and expression of SLC transporters at the BBB (in particular PMAT and MATE1). Dose adjustments may be required for drugs that are substrates of these transporters when prescribing for individuals with AD.
Collapse
Affiliation(s)
- Gayathri Nair Sekhar
- Faculty of Life Sciences and Medicine, School of Cancer and Pharmaceutical Sciences, King's College London, Franklin-Wilkins Building, 150 Stamford Street, Waterloo, London, SE1 9NH, UK
| | - Alice L Fleckney
- Faculty of Life Sciences and Medicine, School of Cancer and Pharmaceutical Sciences, King's College London, Franklin-Wilkins Building, 150 Stamford Street, Waterloo, London, SE1 9NH, UK
| | - Sevda Tomova Boyanova
- Faculty of Life Sciences and Medicine, School of Cancer and Pharmaceutical Sciences, King's College London, Franklin-Wilkins Building, 150 Stamford Street, Waterloo, London, SE1 9NH, UK
| | - Huzefa Rupawala
- Faculty of Life Sciences and Medicine, School of Cancer and Pharmaceutical Sciences, King's College London, Franklin-Wilkins Building, 150 Stamford Street, Waterloo, London, SE1 9NH, UK
| | - Rachel Lo
- Faculty of Life Sciences and Medicine, School of Cancer and Pharmaceutical Sciences, King's College London, Franklin-Wilkins Building, 150 Stamford Street, Waterloo, London, SE1 9NH, UK
| | - Hao Wang
- Faculty of Life Sciences and Medicine, School of Cancer and Pharmaceutical Sciences, King's College London, Franklin-Wilkins Building, 150 Stamford Street, Waterloo, London, SE1 9NH, UK
| | - Doaa B Farag
- Faculty of Life Sciences and Medicine, School of Cancer and Pharmaceutical Sciences, King's College London, Franklin-Wilkins Building, 150 Stamford Street, Waterloo, London, SE1 9NH, UK
- Faculty of Pharmacy, Misr International University, Cairo, 11431, Egypt
| | - Khondaker Miraz Rahman
- Faculty of Life Sciences and Medicine, School of Cancer and Pharmaceutical Sciences, King's College London, Franklin-Wilkins Building, 150 Stamford Street, Waterloo, London, SE1 9NH, UK
| | - Martin Broadstock
- Wolfson Centre for Age-Related Diseases, King's College London, Guy's Campus, London, SE1 1UL, UK
- Maurice Wohl Clinical Neuroscience Institute, King's College London, 125 Coldharbour Lane, Camberwell, London, SE5 9N, UK
| | - Suzanne Reeves
- Division of Psychiatry, Faculty of Brain Sciences, University College London, 149 Tottenham Court Road, London, W1T 7NF, UK
| | - Sarah Ann Thomas
- Faculty of Life Sciences and Medicine, School of Cancer and Pharmaceutical Sciences, King's College London, Franklin-Wilkins Building, 150 Stamford Street, Waterloo, London, SE1 9NH, UK.
| |
Collapse
|
57
|
Chai AB, Leung GKF, Callaghan R, Gelissen IC. P‐glycoprotein: a role in the export of amyloid‐β in Alzheimer's disease? FEBS J 2019; 287:612-625. [DOI: 10.1111/febs.15148] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 10/09/2019] [Accepted: 11/19/2019] [Indexed: 12/25/2022]
Affiliation(s)
- Amanda B. Chai
- School of Pharmacy Faculty of Medicine and Health University of Sydney Australia
| | - Gavin K. F. Leung
- School of Pharmacy Faculty of Medicine and Health University of Sydney Australia
| | - Richard Callaghan
- Research School of Biology and Medical School Australian National University Canberra Australia
| | - Ingrid C. Gelissen
- School of Pharmacy Faculty of Medicine and Health University of Sydney Australia
| |
Collapse
|
58
|
Jha NK, Kar R, Niranjan R. ABC Transporters in Neurological Disorders: An Important Gateway for Botanical Compounds Mediated Neuro-Therapeutics. Curr Top Med Chem 2019; 19:795-811. [PMID: 30977450 DOI: 10.2174/1568026619666190412121811] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 02/27/2019] [Accepted: 03/07/2019] [Indexed: 12/13/2022]
Abstract
Neurodegeneration is a distinguishing feature of many age related disorders and other vector borne neuroinflammatory diseases. There are a number of factors that can modulate the pathology of these disorders. ATP-binding cassette (ABC) transporters are primarily involved in the maintenance of normal brain homeostasis by eliminating toxic peptides and compounds from the brain. Also, ABC transporters protect the brain from the unwanted effects of endogenous and exogenous toxins that can enter the brain parenchyma. Therefore, these transporters have the ability to determine the pathological outcomes of several neurological disorders. For instance, ABC transporters like P-glycoprotein (ABCB1), and BCRP (ABCG2) have been reported to facilitate the clearance of peptides such as amyloid-β (Aβ) that accumulate in the brain during Alzheimer's disease (AD) progression. Other members such as ABCA1, ABCA2, ABCC8, ABCC9, ABCG1 and ABCG4 also have been reported to be involved in the progression of various brain disorders such as HIV-associated dementia, Multiple sclerosis (MS), Ischemic stroke, Japanese encephalitis (JE) and Epilepsy. However, these defective transporters can be targeted by numerous botanical compounds such as Verapamil, Berberine and Fascalpsyn as a therapeutic target to treat these neurological outcomes. These compounds are already reported to modulate ABC transporter activity in the CNS. Nonetheless, the exact mechanisms involving the ABC transporters role in normal brain functioning, their role in neuronal dysfunction and how these botanical compounds ensure and facilitate their therapeutic action in association with defective transporters still remain elusive. This review therefore, summarizes the role of ABC transporters in neurological disorders, with a special emphasis on its role in AD brains. The prospect of using botanical/natural compounds as modulators of ABC transporters in neurological disorders is discussed in the latter half of the article.
Collapse
Affiliation(s)
- Niraj Kumar Jha
- Department of Biotechnology, Noida Institute of Engineering & Technology (NIET), Greater Noida, India
| | - Rohan Kar
- Department of Biotechnology, Noida Institute of Engineering & Technology (NIET), Greater Noida, India
| | - Rituraj Niranjan
- Unit of Microbiology and Molecular Biology, ICMR-Vector Control Research Center, Puducherry-605006, India
| |
Collapse
|
59
|
Katt ME, Mayo LN, Ellis SE, Mahairaki V, Rothstein JD, Cheng L, Searson PC. The role of mutations associated with familial neurodegenerative disorders on blood-brain barrier function in an iPSC model. Fluids Barriers CNS 2019; 16:20. [PMID: 31303172 PMCID: PMC6628493 DOI: 10.1186/s12987-019-0139-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 05/27/2019] [Indexed: 01/23/2023] Open
Abstract
Background Blood–brain barrier dysfunction is associated with many late-stage neurodegenerative diseases. An emerging question is whether the mutations associated with neurodegenerative diseases can independently lead to blood–brain barrier (BBB) dysfunction. Studies from patient-derived induced pluripotent stem cells suggest that mutations associated with neurodegenerative disease are non-cell autonomous, resulting in gain of toxic function in derived neurons and astrocytes. Here we assess whether selected mutations associated with neurodegenerative diseases can contribute to impairment of the blood–brain barrier. Methods We assessed barrier function of confluent monolayers of human brain microvascular endothelial cells (hBMECs) derived from induced pluripotent stem cells (iPSC) from three healthy individuals and eight individuals with neurodegenerative disease. We systematically assessed protein and gene expression of BBB biomarkers, transendothelial resistance (TEER), permeability of Lucifer yellow, permeability of d-glucose, permeability of rhodamine 123, the efflux ratio of rhodamine 123, and P-gp inhibition using Tariquidar for confluent monolayers of human brain microvascular endothelial cell (hBMECs). Results We provide evidence supporting the hypothesis that mutations associated with neurodegenerative disease can independently cause BBB dysfunction. These functional changes are not catastrophic since barrier breakdown would result in BBB impairment during development. Synergistic interactions between non-cell autonomous cerebrovascular dysfunction and the effects of gain-of-toxic function in neurons (e.g. toxic oligomers) are likely to increase disease burden through a positive feedback mechanism. Conclusions These results suggest that the accumulation of defects in brain microvascular endothelial cells may ultimately lead to impairment of the BBB. Small changes in barrier function over time could lead to accumulated defects that result in positive feedback to unrelated central nervous system diseases. Electronic supplementary material The online version of this article (10.1186/s12987-019-0139-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Moriah E Katt
- Institute for Nanobiotechnology, Johns Hopkins University, 100 Croft Hall, 3400 North Charles Street, Baltimore, MD, 21218, USA.,Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Lakyn N Mayo
- Institute for Nanobiotechnology, Johns Hopkins University, 100 Croft Hall, 3400 North Charles Street, Baltimore, MD, 21218, USA.,Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Shannon E Ellis
- Department of Biostatistics, Johns Hopkins University School of Public Health, Baltimore, MD, USA
| | - Vasiliki Mahairaki
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jeffrey D Rothstein
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Linzhao Cheng
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Peter C Searson
- Institute for Nanobiotechnology, Johns Hopkins University, 100 Croft Hall, 3400 North Charles Street, Baltimore, MD, 21218, USA. .,Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA. .,Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
60
|
Vilar S, Sobarzo-Sánchez E, Uriarte E. In Silico Prediction of P-glycoprotein Binding: Insights from Molecular Docking Studies. Curr Med Chem 2019; 26:1746-1760. [DOI: 10.2174/0929867325666171129121924] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 11/10/2017] [Accepted: 11/21/2017] [Indexed: 12/21/2022]
Abstract
The P-glycoprotein is an efflux transporter that expels substances out of the
cells and has an important impact on the pharmacokinetic and pharmacodynamic properties
of drugs. The study of the interactions between ligands and the P-glycoprotein has
implications in the design of Central Nervous System drugs and their transport across the
blood-brain barrier. Moreover, since the P-glycoprotein is overexpressed in some types of
cancers, the protein is responsible for expelling the drug therapies from the cells, and
hence, for drug resistance. In this review, we describe different P-glycoprotein binding
sites reported for substrates, inhibitors and modulators, and focus on molecular docking
studies that provide useful information about drugs and P-glycoprotein interactions.
Docking in crystallized structures and homology models showed potential in the detection
of the binding site and key residues responsible for ligand recognition. Moreover, virtual
screening through molecular docking discriminates P-glycoprotein ligands from decoys.
We also discuss challenges and limitations of molecular docking simulations applied to
this particular protein. Computational structure-based approaches are very helpful in the
study of novel ligands that interact with the P-glycoprotein and provide insights to understand
the P-glycoprotein molecular mechanism of action.
Collapse
Affiliation(s)
- Santiago Vilar
- Departamento de Quimica Organica, Facultad de Farmacia, Universidad de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Eduardo Sobarzo-Sánchez
- Departamento de Quimica Organica, Facultad de Farmacia, Universidad de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Eugenio Uriarte
- Departamento de Quimica Organica, Facultad de Farmacia, Universidad de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| |
Collapse
|
61
|
Bourassa P, Tremblay C, Schneider JA, Bennett DA, Calon F. Beta-amyloid pathology in human brain microvessel extracts from the parietal cortex: relation with cerebral amyloid angiopathy and Alzheimer's disease. Acta Neuropathol 2019; 137:801-823. [PMID: 30729296 DOI: 10.1007/s00401-019-01967-4] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 01/24/2019] [Accepted: 01/24/2019] [Indexed: 01/13/2023]
Abstract
Several pieces of evidence suggest that blood-brain barrier (BBB) dysfunction is implicated in the pathophysiology of Alzheimer's disease (AD), exemplified by the frequent occurrence of cerebral amyloid angiopathy (CAA) and the defective clearance of Aβ peptides. However, the specific role of brain microvascular cells in these anomalies remains elusive. In this study, we validated by Western, ELISA and immunofluorescence analyses a procedure to generate microvasculature-enriched fractions from frozen samples of human cerebral cortex. We then investigated Aβ and proteins involved in its clearance or production in microvessel extracts generated from the parietal cortex of 60 volunteers in the Religious Orders Study. Volunteers were categorized as AD (n = 38) or controls (n = 22) based on the ABC scoring method presented in the revised guidelines for the neuropathological diagnosis of AD. Higher ELISA-determined concentrations of vascular Aβ40 and Aβ42 were found in persons with a neuropathological diagnosis of AD, in apoE4 carriers and in participants with advanced parenchymal CAA, compared to respective age-matched controls. Vascular levels of two proteins involved in Aβ clearance, ABCB1 and neprilysin, were lower in persons with AD and positively correlated with cognitive function, while being inversely correlated to vascular Aβ40. In contrast, BACE1, a protein necessary for Aβ production, was increased in individuals with AD and in apoE4 carriers, negatively correlated to cognitive function and positively correlated to Aβ40 in microvessel extracts. The present report indicates that concentrating microvessels from frozen human brain samples facilitates the quantitative biochemical analysis of cerebrovascular dysfunction in CNS disorders. Data generated overall show that microvessels extracted from individuals with parenchymal CAA-AD contained more Aβ and BACE1 and less ABCB1 and neprilysin, evidencing a pattern of dysfunction in brain microvascular cells contributing to CAA and AD pathology and symptoms.
Collapse
Affiliation(s)
- Philippe Bourassa
- Faculté de pharmacie, Université Laval, Quebec, QC, Canada
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, 2705, Boulevard Laurier, Room T2-67, Quebec, QC, G1V 4G2, Canada
| | - Cyntia Tremblay
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, 2705, Boulevard Laurier, Room T2-67, Quebec, QC, G1V 4G2, Canada
| | - Julie A Schneider
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Frédéric Calon
- Faculté de pharmacie, Université Laval, Quebec, QC, Canada.
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, 2705, Boulevard Laurier, Room T2-67, Quebec, QC, G1V 4G2, Canada.
| |
Collapse
|
62
|
Wiese M, Stefan SM. The A‐B‐C of small‐molecule ABC transport protein modulators: From inhibition to activation—a case study of multidrug resistance‐associated protein 1 (ABCC1). Med Res Rev 2019; 39:2031-2081. [DOI: 10.1002/med.21573] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 02/26/2019] [Accepted: 03/05/2019] [Indexed: 12/17/2022]
Affiliation(s)
- Michael Wiese
- Pharmaceutical Institute, Rheinische Friedrich‐Wilhelms‐University of Bonn Bonn Germany
| | - Sven Marcel Stefan
- Pharmaceutical Institute, Rheinische Friedrich‐Wilhelms‐University of Bonn Bonn Germany
| |
Collapse
|
63
|
Moura RP, Martins C, Pinto S, Sousa F, Sarmento B. Blood-brain barrier receptors and transporters: an insight on their function and how to exploit them through nanotechnology. Expert Opin Drug Deliv 2019; 16:271-285. [PMID: 30767695 DOI: 10.1080/17425247.2019.1583205] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION The blood-brain barrier (BBB) is a highly limiting barrier that prevents the brain from contacting with several circulating molecules, including harmful agents. However, certain systemic nutrients and macromolecules are able to cross the BBB and reach the brain parenchyma, involving the interaction with multiple receptors and/or transporters at the BBB surface. Nanotechnology allows the creation of drug vehicles, functionalized with targeting ligands for binding specific BBB receptors and/or transporters, hence triggering the transport through this biobarrier. AREAS COVERED This review focuses the BBB receptors/transporters to be exploited in regard to their overall structure and biologic function, as well as their role in the development of strategies envisaging drug delivery to the brain. Then, the interplay between the targeting of these BBB receptors/transporters and nanotechnology is explored, as they can increase by several-fold the effectiveness of brain-targeted therapies. EXPERT OPINION Nanomedicine may be particularly useful in brain drug delivery, mainly due to the possibility of functionalizing nanoparticles to target specific receptors/transporters. Since the BBB is endowed with numerous receptors and transporters responsible for regulating the proper metabolic activity of the brain, their targeting can be a promising bypass strategy to circumvent the hurdle that the BBB represents for brain drug delivery.
Collapse
Affiliation(s)
- Rui Pedro Moura
- a CESPU - Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde , Gandra , Portugal
| | - Cláudia Martins
- b I3S - Instituto de Investigação e Inovação em Saúde , Universidade do Porto , Porto , Portugal.,c INEB - Instituto de Engenharia Biomédica , Universidade do Porto , Porto , Portugal.,d ICBAS - Instituto de Ciências Biomédicas Abel Salazar , Universidade do Porto , Porto , Portugal
| | - Soraia Pinto
- b I3S - Instituto de Investigação e Inovação em Saúde , Universidade do Porto , Porto , Portugal.,c INEB - Instituto de Engenharia Biomédica , Universidade do Porto , Porto , Portugal
| | - Flávia Sousa
- a CESPU - Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde , Gandra , Portugal.,b I3S - Instituto de Investigação e Inovação em Saúde , Universidade do Porto , Porto , Portugal.,c INEB - Instituto de Engenharia Biomédica , Universidade do Porto , Porto , Portugal.,d ICBAS - Instituto de Ciências Biomédicas Abel Salazar , Universidade do Porto , Porto , Portugal
| | - Bruno Sarmento
- a CESPU - Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde , Gandra , Portugal.,b I3S - Instituto de Investigação e Inovação em Saúde , Universidade do Porto , Porto , Portugal.,c INEB - Instituto de Engenharia Biomédica , Universidade do Porto , Porto , Portugal
| |
Collapse
|
64
|
Liu L, Liu X. Contributions of Drug Transporters to Blood-Brain Barriers. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1141:407-466. [PMID: 31571171 DOI: 10.1007/978-981-13-7647-4_9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Blood-brain interfaces comprise the cerebral microvessel endothelium forming the blood-brain barrier (BBB) and the epithelium of the choroid plexuses forming the blood-cerebrospinal fluid barrier (BCSFB). Their main functions are to impede free diffusion between brain fluids and blood; to provide transport processes for essential nutrients, ions, and metabolic waste products; and to regulate the homeostasis of central nervous system (CNS), all of which are attributed to absent fenestrations, high expression of tight junction proteins at cell-cell contacts, and expression of multiple transporters, receptors, and enzymes. Existence of BBB is an important reason that systemic drug administration is not suitable for the treatment of CNS diseases. Some diseases, such epilepsy, Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and diabetes, alter BBB function via affecting tight junction proteins or altering expression and function of these transporters. This chapter will illustrate function of BBB, expression of transporters, as well as their alterations under disease status.
Collapse
Affiliation(s)
- Li Liu
- China Pharmaceutical University, Nanjing, China
| | - Xiaodong Liu
- China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
65
|
Storck SE, Hartz AM, Bernard J, Wolf A, Kachlmeier A, Mahringer A, Weggen S, Pahnke J, Pietrzik CU. The concerted amyloid-beta clearance of LRP1 and ABCB1/P-gp across the blood-brain barrier is linked by PICALM. Brain Behav Immun 2018; 73:21-33. [PMID: 30041013 PMCID: PMC7748946 DOI: 10.1016/j.bbi.2018.07.017] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 07/10/2018] [Accepted: 07/20/2018] [Indexed: 01/18/2023] Open
Abstract
The accumulation of neurotoxic amyloid-beta (Aβ) in the brain is a characteristic hallmark of Alzheimer's disease (AD). The blood-brain barrier (BBB) provides a large surface area and has been shown to be an important mediator for removal of brain Aβ. Both, the ABC transporter P-glycoprotein (ABCB1/P-gp) and the receptor low-density lipoprotein receptor-related protein 1 (LRP1) have been implicated to play crucial roles in Aβ efflux from brain. Here, with immunoprecipitation experiments, co-immunostainings and dual inhibition of ABCB1/P-gp and LRP1, we show that both proteins are functionally linked, mediating a concerted transcytosis of Aβ through endothelial cells. Late-onset AD risk factor Phosphatidylinositol binding clathrin assembly protein (PICALM) is associated with both ABCB1/P-gp and LRP1 representing a functional link and guiding both proteins through the brain endothelium. Together, our results give more mechanistic insight on Aβ transport across the BBB and show that the functional interplay of different clearance proteins is needed for the rapid removal of Aβ from the brain.
Collapse
Affiliation(s)
- Steffen E. Storck
- Institute for Pathobiochemistry, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Anika M.S. Hartz
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States,Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Jessica Bernard
- Institute for Pathobiochemistry, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Andrea Wolf
- Department of Pharmacy Practice and Pharmaceutical Sciences, College of Pharmacy, University of Minnesota, Duluth, MN, United States
| | - André Kachlmeier
- Institute for Pathobiochemistry, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Anne Mahringer
- Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, Germany
| | - Sascha Weggen
- Department of Neuropathology, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| | - Jens Pahnke
- University of Oslo (UiO) & Oslo University Hospital (OUS), Department of Neuro-/Pathology, Oslo, Norway,University of Lübeck (UzL), LIED, Lübeck, Germany,Leibniz-Institute of Plant Biochemistry (IPB), Department for Bioorganic Chemistry, Halle, Germany,University of Latvia (UL), Department of Pharmacology, Riga, Latvia
| | - Claus U. Pietrzik
- Institute for Pathobiochemistry, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany,Corresponding author at: University Medical Center of the Johannes Gutenberg-University of Mainz, Institute for Pathobiochemistry, Molecular Neurodegeneration, Duesbergweg 6, 55099 Mainz, Germany. (C.U. Pietrzik)
| |
Collapse
|
66
|
Pan Y, Nicolazzo JA. Impact of aging, Alzheimer's disease and Parkinson's disease on the blood-brain barrier transport of therapeutics. Adv Drug Deliv Rev 2018; 135:62-74. [PMID: 29665383 DOI: 10.1016/j.addr.2018.04.009] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 01/17/2018] [Accepted: 04/07/2018] [Indexed: 01/01/2023]
Abstract
Older people are at a greater risk of medicine-induced toxicity resulting from either increased drug sensitivity or age-related pharmacokinetic changes. The scenario is further complicated with the two most prevalent age-related neurodegenerative diseases, Alzheimer's disease (AD) and Parkinson's disease (PD). With aging, AD and PD, there is growing evidence of altered structure and function of the blood-brain barrier (BBB), including modifications to tight junctions and efflux transporters, such as P-glycoprotein. The subsequent impact on CNS drug exposure and risk of neurotoxicity from systemically-acting medicines is less well characterized. The purpose of this review, therefore, is to provide an overview of the multiple changes that occur to the BBB as a result of aging, AD and PD, and the impact that such changes have on CNS exposure of drugs, based on studies conducted in aged rodents or rodent models of disease, and in elderly people with and without AD or PD.
Collapse
Affiliation(s)
- Yijun Pan
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, Victoria 3052, Australia
| | - Joseph A Nicolazzo
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, Victoria 3052, Australia.
| |
Collapse
|
67
|
Bharate S, Kumar V, Singh G, Singh A, Gupta M, Singh D, Kumar A, Vishwakarma RA, Bharate SB. Preclinical Development of Crocus sativus-Based Botanical Lead IIIM-141 for Alzheimer's Disease: Chemical Standardization, Efficacy, Formulation Development, Pharmacokinetics, and Safety Pharmacology. ACS OMEGA 2018; 3:9572-9585. [PMID: 31459089 PMCID: PMC6644748 DOI: 10.1021/acsomega.8b00841] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 08/03/2018] [Indexed: 05/03/2023]
Abstract
Crocus sativus L. (family: Iridaceae) has been documented in traditional medicine with numerous medicinal properties. Recently, we have shown that C. sativus extract (IIIM-141) displays promising efficacy in a genetic mice (5XFAD) model of Alzheimer's disease (AD) (ACS Chem. Neurosci. 2017, 16, 1756). To translate the available traditional knowledge and the scientifically validated results into modern medicine, herein we aimed to carry out its preclinical development. IIIM-141 is primarily a mixture of crocins containing trans-4-GG-crocin (36 % w/w) as the principal component. The in vitro studies show that IIIM-141 has protective as well as therapeutic properties in assays related to AD. It induces the expression of P-gp, thereby enhancing the amyloid-β clearance from an AD brain. It also inhibits NLRP3 inflammasome and protects SH-SY5Y cells against amyloid-β- and glutamate-induced neurotoxicities. In behavioral models, it decreased the streptozotocin-induced memory impairment in rats and recovered the scopolamine-induced memory deficit in Swiss albino mice at 100 mg/kg dose. The acute oral toxicity study shows that IIIM-141 is safe up to the dose of 2000 mg/kg, with no effect on the body weight and on the biochemical/hematological parameters of the rats. The repeated oral administration of IIIM-141 for 28 days at 100 mg/kg dose did not cause any preterminal deaths and abnormalities in Wistar rats. The pharmacokinetic analysis indicated that after oral administration of IIIM-141, the majority of crocin gets hydrolyzed to its aglycone crocetin. The sustained release (SR) capsule formulation was developed, which showed an improved in vitro dissolution profile and a significantly enhanced plasma exposure in the pharmacokinetic study. The SR formulation resulted in 3.3-fold enhancement in the area under the curve of crocetin and doubling of the crocetin/crocin ratio in plasma compared with the extract. The data presented herein will serve as the benchmark for further research on this botanical candidate.
Collapse
Affiliation(s)
- Sonali
S. Bharate
- Preformulaion
Laboratory, PK-PD Toxicology and Formulation Division, Academy of Scientific
& Innovative Research, PK-PD Toxicology and Formulation Division, QC-QA Division, and Medicinal Chemistry
Division, CSIR-Indian Institute of Integrative
Medicine, Canal Road, Jammu 180001, India
| | - Vikas Kumar
- Preformulaion
Laboratory, PK-PD Toxicology and Formulation Division, Academy of Scientific
& Innovative Research, PK-PD Toxicology and Formulation Division, QC-QA Division, and Medicinal Chemistry
Division, CSIR-Indian Institute of Integrative
Medicine, Canal Road, Jammu 180001, India
| | - Gurdarshan Singh
- Preformulaion
Laboratory, PK-PD Toxicology and Formulation Division, Academy of Scientific
& Innovative Research, PK-PD Toxicology and Formulation Division, QC-QA Division, and Medicinal Chemistry
Division, CSIR-Indian Institute of Integrative
Medicine, Canal Road, Jammu 180001, India
| | - Amarinder Singh
- Preformulaion
Laboratory, PK-PD Toxicology and Formulation Division, Academy of Scientific
& Innovative Research, PK-PD Toxicology and Formulation Division, QC-QA Division, and Medicinal Chemistry
Division, CSIR-Indian Institute of Integrative
Medicine, Canal Road, Jammu 180001, India
| | - Mehak Gupta
- Preformulaion
Laboratory, PK-PD Toxicology and Formulation Division, Academy of Scientific
& Innovative Research, PK-PD Toxicology and Formulation Division, QC-QA Division, and Medicinal Chemistry
Division, CSIR-Indian Institute of Integrative
Medicine, Canal Road, Jammu 180001, India
| | - Deepika Singh
- Preformulaion
Laboratory, PK-PD Toxicology and Formulation Division, Academy of Scientific
& Innovative Research, PK-PD Toxicology and Formulation Division, QC-QA Division, and Medicinal Chemistry
Division, CSIR-Indian Institute of Integrative
Medicine, Canal Road, Jammu 180001, India
| | - Ajay Kumar
- Preformulaion
Laboratory, PK-PD Toxicology and Formulation Division, Academy of Scientific
& Innovative Research, PK-PD Toxicology and Formulation Division, QC-QA Division, and Medicinal Chemistry
Division, CSIR-Indian Institute of Integrative
Medicine, Canal Road, Jammu 180001, India
| | - Ram A. Vishwakarma
- Preformulaion
Laboratory, PK-PD Toxicology and Formulation Division, Academy of Scientific
& Innovative Research, PK-PD Toxicology and Formulation Division, QC-QA Division, and Medicinal Chemistry
Division, CSIR-Indian Institute of Integrative
Medicine, Canal Road, Jammu 180001, India
- E-mail: . Phone: +91 191 2569111. Fax: +91 191 2569333 (R.A.V.)
| | - Sandip B. Bharate
- Preformulaion
Laboratory, PK-PD Toxicology and Formulation Division, Academy of Scientific
& Innovative Research, PK-PD Toxicology and Formulation Division, QC-QA Division, and Medicinal Chemistry
Division, CSIR-Indian Institute of Integrative
Medicine, Canal Road, Jammu 180001, India
- E-mail: . Phone: +91 191 2569006. Fax: +91 191 2569333 (S.B.B.)
| |
Collapse
|
68
|
Wei Y, Shin MR, Sesti F. Oxidation of KCNB1 channels in the human brain and in mouse model of Alzheimer's disease. Cell Death Dis 2018; 9:820. [PMID: 30050035 PMCID: PMC6062629 DOI: 10.1038/s41419-018-0886-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 06/23/2018] [Accepted: 07/16/2018] [Indexed: 01/02/2023]
Abstract
Oxidative modification of the voltage-gated K+ channel subfamily B member 1 (KCNB1, Kv2.1) is emerging as a mechanism of neuronal vulnerability potentially capable of affecting multiple conditions associated with oxidative stress, from normal aging to neurodegenerative disease. In this study we report that oxidation of KCNB1 channels is exacerbated in the post mortem brains of Alzheimer’s disease (AD) donors compared to age-matched controls. In addition, phosphorylation of Focal Adhesion kinases (FAK) and Src tyrosine kinases, two key signaling steps that follow KCNB1 oxidation, is also strengthened in AD vs. control brains. Quadruple transgenic mice expressing a non-oxidizable form of KCNB1 in the 3xTg-AD background (APPSWE, PS1M146V, and tauP301L), exhibit improved working memory along with reduced brain inflammation, protein carbonylation and intraneuronal β-amyloid (Aβ) compared to 3xTg-AD mice or mice expressing the wild type (WT) KCNB1 channel. We conclude that oxidation of KCNB1 channels is a mechanism of neuronal vulnerability that is pervasive in the vertebrate brain.
Collapse
Affiliation(s)
- Yu Wei
- Department of Neuroscience and Cell Biology, Rutgers University Robert Wood Johnson Medical School, 683 Hoes Lane West, Piscataway, NJ, 08854, USA
| | - Mi Ryung Shin
- Department of Neuroscience and Cell Biology, Rutgers University Robert Wood Johnson Medical School, 683 Hoes Lane West, Piscataway, NJ, 08854, USA
| | - Federico Sesti
- Department of Neuroscience and Cell Biology, Rutgers University Robert Wood Johnson Medical School, 683 Hoes Lane West, Piscataway, NJ, 08854, USA.
| |
Collapse
|
69
|
Exposure of human neurons to silver nanoparticles induces similar pattern of ABC transporters gene expression as differentiation: Study on proliferating and post-mitotic LUHMES cells. Mech Ageing Dev 2018; 171:7-14. [DOI: 10.1016/j.mad.2018.02.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 01/26/2018] [Accepted: 02/22/2018] [Indexed: 11/17/2022]
|
70
|
McInerney MP, Volitakis I, Bush AI, Banks WA, Short JL, Nicolazzo JA. Ionophore and Biometal Modulation of P-glycoprotein Expression and Function in Human Brain Microvascular Endothelial Cells. Pharm Res 2018; 35:83. [DOI: 10.1007/s11095-018-2377-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 02/26/2018] [Indexed: 11/30/2022]
|
71
|
Yulug B, Hanoglu L, Ozansoy M, Isık D, Kilic U, Kilic E, Schabitz WR. Therapeutic role of rifampicin in Alzheimer's disease. Psychiatry Clin Neurosci 2018; 72:152-159. [PMID: 29315976 DOI: 10.1111/pcn.12637] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 12/06/2017] [Accepted: 12/17/2017] [Indexed: 12/11/2022]
Abstract
Rifampicin exerts significant brain protective functions in multiple experimental models. Here we summarize the underlying mechanisms of the neuroprotective and pro-cognitive effects of rifampicin that are mediated by its anti-inflammatory, anti-tau, anti-amyloid, and cholinergic effects. Beyond suggesting that rifampicin shows strong brain protective effects in preclinical models of Alzheimer's disease, we also provide substantial clinical evidence for the neuroprotective and pro-cognitive effects of rifampicin. Future neuroimaging studies combined with clinical assessment scores are the following steps to be taken in this field of research.
Collapse
Affiliation(s)
- Burak Yulug
- Department of Neurology, Istanbul Medipol University, Istanbul, Turkey.,Department of Restorative and Regenerative Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Lütfü Hanoglu
- Department of Neurology, Istanbul Medipol University, Istanbul, Turkey.,Department of Restorative and Regenerative Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Mehmet Ozansoy
- Department of Restorative and Regenerative Medicine, Istanbul Medipol University, Istanbul, Turkey.,Department of Physiology, International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Dogan Isık
- Department of Psychiatry, Istanbul Medipol University, Istanbul, Turkey
| | - Ulkan Kilic
- Department of Medical Biology, Faculty of Medicine, University of Health Sciences, Istanbul, Turkey
| | - Ertugrul Kilic
- Department of Restorative and Regenerative Medicine, Istanbul Medipol University, Istanbul, Turkey.,Department of Physiology, International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Wolf Rüdiger Schabitz
- Department of Neurology, Bethel, EVKB, Bielefeld, University of Münster, Münster, Germany
| |
Collapse
|
72
|
Banks WA, Kovac A, Majerova P, Bullock KM, Shi M, Zhang J. Tau Proteins Cross the Blood-Brain Barrier. J Alzheimers Dis 2018; 55:411-419. [PMID: 27662303 DOI: 10.3233/jad-160542] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Tauopathies are a hallmark of many neurodegenerative diseases, including Alzheimer's disease and traumatic brain injuries. It has been demonstrated that amyloid-beta peptides, alpha-synuclein, and prion proteins cross the blood-brain barrier (BBB), contributing to their abilities to induce disease. Very little is known about whether tau proteins can cross the BBB. Here we systematically characterized several key forms of tau proteins to cross the BBB, including Tau-441 (2N4R), Tau-410 (2N3R), truncated tau 151-391 (0N4R), and truncated tau 121-227. All of these tau proteins crossed the BBB readily and bidirectonally; however, only Tau-410 had a saturable component to its influx. The tau proteins also entered the blood after their injection into the brain, with Tau 121-227 having the slowest exit from brain. The tau proteins varied in regards to their enzymatic stability in brain and blood and in their peripheral pharmacokinetics. These results show that blood-borne tau proteins could contribute to brain tauopathies. The result also suggest that the CNS can contribute to blood levels of tau, raising the possibility that, as suggested for other misfolded proteins, blood levels of tau proteins could be used as a biomarker of CNS disease.
Collapse
Affiliation(s)
- William A Banks
- Research and Development, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA.,Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Andrej Kovac
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovak Republic.,Department of Pharmacology and Toxicology, The University of Veterinary Medicine and Pharmacy, Kosice, Slovak Republic
| | - Petra Majerova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovak Republic.,AXON Neuroscience SE, Bratislava, Slovak Republic
| | - Kristin M Bullock
- Research and Development, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
| | - Min Shi
- Department of Pathology, University of Washington School of Medicine, Seattle, WA, USA
| | - Jing Zhang
- Department of Pathology, University of Washington School of Medicine, Seattle, WA, USA.,Department of Pathology, Peking University Health Science Center and Peking University Third Hospital, Beijing, China
| |
Collapse
|
73
|
Gustafsson S, Lindström V, Ingelsson M, Hammarlund-Udenaes M, Syvänen S. Intact blood-brain barrier transport of small molecular drugs in animal models of amyloid beta and alpha-synuclein pathology. Neuropharmacology 2018; 128:482-491. [DOI: 10.1016/j.neuropharm.2017.08.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 07/04/2017] [Accepted: 08/03/2017] [Indexed: 01/19/2023]
|
74
|
Pereira CD, Martins F, Wiltfang J, da Cruz e Silva OA, Rebelo S. ABC Transporters Are Key Players in Alzheimer’s Disease. J Alzheimers Dis 2017; 61:463-485. [DOI: 10.3233/jad-170639] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Cátia D. Pereira
- Department of Medical Sciences, Neuroscience and Signalling Laboratory, Institute for Biomedicine – iBiMED, University of Aveiro, Aveiro, Portugal
| | - Filipa Martins
- Department of Medical Sciences, Neuroscience and Signalling Laboratory, Institute for Biomedicine – iBiMED, University of Aveiro, Aveiro, Portugal
| | - Jens Wiltfang
- Department of Medical Sciences, Neuroscience and Signalling Laboratory, Institute for Biomedicine – iBiMED, University of Aveiro, Aveiro, Portugal
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen (UMG), Göttingen, Germany
| | - Odete A.B. da Cruz e Silva
- Department of Medical Sciences, Neuroscience and Signalling Laboratory, Institute for Biomedicine – iBiMED, University of Aveiro, Aveiro, Portugal
| | - Sandra Rebelo
- Department of Medical Sciences, Neuroscience and Signalling Laboratory, Institute for Biomedicine – iBiMED, University of Aveiro, Aveiro, Portugal
| |
Collapse
|
75
|
P-glycoprotein (ABCB1) and Oxidative Stress: Focus on Alzheimer's Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:7905486. [PMID: 29317984 PMCID: PMC5727796 DOI: 10.1155/2017/7905486] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 10/30/2017] [Indexed: 11/23/2022]
Abstract
ATP-binding cassette (ABC) transporters, in particular P-glycoprotein (encoded by ABCB1), are important and selective elements of the blood-brain barrier (BBB), and they actively contribute to brain homeostasis. Changes in ABCB1 expression and/or function at the BBB may not only alter the expression and function of other molecules at the BBB but also affect brain environment. Over the last decade, a number of reports have shown that ABCB1 actively mediates the transport of beta amyloid (Aβ) peptide. This finding has opened up an entirely new line of research in the field of Alzheimer's disease (AD). Indeed, despite intense research efforts, AD remains an unsolved pathology and effective therapies are still unavailable. Here, we review the crucial role of ABCB1 in the Aβ transport and how oxidative stress may interfere with this process. A detailed understanding of ABCB1 regulation can provide the basis for improved neuroprotection in AD and also enhanced therapeutic drug delivery to the brain.
Collapse
|
76
|
de Wit NM, Vanmol J, Kamermans A, Hendriks JJA, de Vries HE. Inflammation at the blood-brain barrier: The role of liver X receptors. Neurobiol Dis 2017; 107:57-65. [DOI: 10.1016/j.nbd.2016.09.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 09/11/2016] [Accepted: 09/17/2016] [Indexed: 02/05/2023] Open
|
77
|
Hindle SJ, Munji RN, Dolghih E, Gaskins G, Orng S, Ishimoto H, Soung A, DeSalvo M, Kitamoto T, Keiser MJ, Jacobson MP, Daneman R, Bainton RJ. Evolutionarily Conserved Roles for Blood-Brain Barrier Xenobiotic Transporters in Endogenous Steroid Partitioning and Behavior. Cell Rep 2017; 21:1304-1316. [PMID: 29091768 PMCID: PMC5774027 DOI: 10.1016/j.celrep.2017.10.026] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 08/25/2017] [Accepted: 10/05/2017] [Indexed: 12/30/2022] Open
Abstract
Central nervous system (CNS) chemical protection depends upon discrete control of small-molecule access by the blood-brain barrier (BBB). Curiously, some drugs cause CNS side-effects despite negligible transit past the BBB. To investigate this phenomenon, we asked whether the highly BBB-enriched drug efflux transporter MDR1 has dual functions in controlling drug and endogenous molecule CNS homeostasis. If this is true, then brain-impermeable drugs could induce behavioral changes by affecting brain levels of endogenous molecules. Using computational, genetic, and pharmacologic approaches across diverse organisms, we demonstrate that BBB-localized efflux transporters are critical for regulating brain levels of endogenous steroids and steroid-regulated behaviors (sleep in Drosophila and anxiety in mice). Furthermore, we show that MDR1-interacting drugs are associated with anxiety-related behaviors in humans. We propose a general mechanism for common behavioral side effects of prescription drugs: pharmacologically challenging BBB efflux transporters disrupts brain levels of endogenous substrates and implicates the BBB in behavioral regulation.
Collapse
Affiliation(s)
- Samantha J Hindle
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, USA
| | - Roeben N Munji
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, USA; Division of Clinical Pharmacology and Experimental Therapeutics, University of California San Francisco, San Francisco, CA, USA; Department of Anatomy, University of California San Francisco, San Francisco, CA, USA; Department of Pharmacology, University of California San Diego, La Jolla, CA, USA
| | - Elena Dolghih
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
| | - Garrett Gaskins
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA; Institute for Neurodegenerative Disease, University of California San Francisco, San Francisco, CA, USA; Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Souvinh Orng
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, USA
| | - Hiroshi Ishimoto
- Division of Biological Science, Graduate School of Science, Nagoya University, Japan
| | - Allison Soung
- Department of Anatomy, University of California San Francisco, San Francisco, CA, USA; Department of Pharmacology, University of California San Diego, La Jolla, CA, USA
| | - Michael DeSalvo
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, USA
| | | | - Michael J Keiser
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA; Institute for Neurodegenerative Disease, University of California San Francisco, San Francisco, CA, USA; Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Matthew P Jacobson
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
| | - Richard Daneman
- Department of Anatomy, University of California San Francisco, San Francisco, CA, USA; Department of Pharmacology, University of California San Diego, La Jolla, CA, USA.
| | - Roland J Bainton
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
78
|
Disease-Induced Alterations in Brain Drug Transporters in Animal Models of Alzheimer’s Disease. Pharm Res 2017; 34:2652-2662. [DOI: 10.1007/s11095-017-2263-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 09/12/2017] [Indexed: 12/12/2022]
|
79
|
Yamazaki Y, Kanekiyo T. Blood-Brain Barrier Dysfunction and the Pathogenesis of Alzheimer's Disease. Int J Mol Sci 2017; 18:ijms18091965. [PMID: 28902142 PMCID: PMC5618614 DOI: 10.3390/ijms18091965] [Citation(s) in RCA: 258] [Impact Index Per Article: 36.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 09/06/2017] [Accepted: 09/07/2017] [Indexed: 01/22/2023] Open
Abstract
Brain capillary endothelial cells form the blood-brain barrier (BBB), which is covered with basement membranes and is also surrounded by pericytes and astrocyte end-feet in the neurovascular unit. The BBB tightly regulates the molecular exchange between the blood flow and brain parenchyma, thereby regulating the homeostasis of the central nervous system (CNS). Thus, dysfunction of the BBB is likely involved in the pathogenesis of several neurological diseases, including Alzheimer’s disease (AD). While amyloid-β (Aβ) deposition and neurofibrillary tangle formation in the brain are central pathological hallmarks in AD, cerebrovascular lesions and BBB alteration have also been shown to frequently coexist. Although further clinical studies should clarify whether BBB disruption is a specific feature of AD pathogenesis, increasing evidence indicates that each component of the neurovascular unit is significantly affected in the presence of AD-related pathologies in animal models and human patients. Conversely, since some portions of Aβ are eliminated along the neurovascular unit and across the BBB, disturbing the pathways may result in exacerbated Aβ accumulation in the brain. Thus, current evidence suggests that BBB dysfunction may causatively and consequently contribute to AD pathogenesis, forming a vicious cycle between brain Aβ accumulation and neurovascular unit impairments during disease progression.
Collapse
Affiliation(s)
- Yu Yamazaki
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA.
| | - Takahisa Kanekiyo
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA.
| |
Collapse
|
80
|
Erbayraktar Z, Evlice A, Yener G, Ulusu NN. Effects of donepezil on liver and kidney functions for the treatment of Alzheimer's disease. J Integr Neurosci 2017; 16:335-346. [PMID: 28891516 DOI: 10.3233/jin-170020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Aim of the present study is to investigate the effects of medication with donepezil (acetylcholinesterase inhibitor) on the liver and kidney function in Alzheimer's disease (AD) and to compare the effects of donepezil medication during short (one month) and long term (six years) follow-ups. We evaluated female and male patients from Cukurova [42 AD patients; short term (5 mg/day)] and Dokuz Eylul [68 AD patients; long term (10 mg/day)] University Hospital. The results compared with the geriatric population without dementia in other words who are not in medication with donepezil. For short term evaluation all subjects underwent periodic examination with tests regarding hepatic and renal functions; firstly, before starting treatment and then repeated one month later. For long term evaluation all subjects underwent periodic examination with tests regarding hepatic and renal functions; three times at the end of each two consecutive years of treatment with donepezil. AD patients' results were also compared with 79 neurologically healthy geriatric patients without dementia who were over 65 years of age and were not receiving medication with donepezil. For this task, serum aspartate aminotransferase (AST) and alanine aminotransferase (ALT) levels are used to predict possible liver damage, while the blood urea nitrogen (BUN) and creatinine (CRE) levels for kidney damage. No significant difference between the groups regarding the routine control of biochemical parameters was observed in short term drug medication. In long term patients' group; the effects of two years use of donepezil on renal and hepatic function were also evaluated and levels of AST, ALT, BUN and CRE were found to be increased significantly compared to pretreatment levels. But, they remained in the reference intervals. However, levels of AST and ALT at the end of the fourth year of therapy were similar to those measured at the end of the second year, levels of BUN and CRE continuing to increase with staying below the reference limits. Functional markers obtained at the end of the sixth year of therapy were not differing from those of the fourth year. No significant difference was found during comparisons within the results of the neurologically healthy geriatric patient group. During comparisons between the two groups, measurements obtained at all-time points were significantly high in donepezil treated AD patients. We concluded that customized dosage according to hepatic and renal functions is necessary for using acetylcholinesterase inhibitor in AD patients.
Collapse
Affiliation(s)
- Zübeyde Erbayraktar
- Department of Biochemistry, Faculty of Medicine, Dokuz Eylul University, İzmir, Turkey
| | - Ahmet Evlice
- Department of Neurology, Faculty of Medicine, Çukurova University, Adana, Turkey
| | - Görsev Yener
- Department of Neurology, Faculty of Medicine, Dokuz Eylul University, İzmir, Turkey
| | - N Nuray Ulusu
- School of Medicine, Koc University, Rumelifeneri Yolu, Sariyer, Istanbul, Turkey
| |
Collapse
|
81
|
McInerney MP, Pan Y, Short JL, Nicolazzo JA. Development and Validation of an In-Cell Western for Quantifying P-Glycoprotein Expression in Human Brain Microvascular Endothelial (hCMEC/D3) Cells. J Pharm Sci 2017; 106:2614-2624. [DOI: 10.1016/j.xphs.2016.12.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 12/06/2016] [Accepted: 12/07/2016] [Indexed: 10/20/2022]
|
82
|
Osgood D, Miller MC, Messier AA, Gonzalez L, Silverberg GD. Aging alters mRNA expression of amyloid transporter genes at the blood-brain barrier. Neurobiol Aging 2017; 57:178-185. [PMID: 28654861 PMCID: PMC5728118 DOI: 10.1016/j.neurobiolaging.2017.05.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 05/11/2017] [Accepted: 05/12/2017] [Indexed: 12/22/2022]
Abstract
Decreased clearance of potentially toxic metabolites, due to aging changes, likely plays a significant role in the accumulation of amyloid-beta (Aβ) peptides and other macromolecules in the brain of the elderly and in the patients with Alzheimer's disease (AD). Aging is the single most important risk factor for AD development. Aβ transport receptor proteins expressed at the blood-brain barrier are significantly altered with age: the efflux transporters lipoprotein receptor-related protein 1 and P-glycoprotein are reduced, whereas the influx transporter receptor for advanced glycation end products is increased. These receptors play an important role in maintaining brain biochemical homeostasis. We now report that, in a rat model of aging, gene transcription is altered in aging, as measured by Aβ receptor gene messenger RNA (mRNA) at 3, 6, 9, 12, 15, 20, 30, and 36 months. Gene mRNA expression from isolated cerebral microvessels was measured by quantitative polymerase chain reaction. Lipoprotein receptor-related protein 1 and P-glycoprotein mRNA were significantly reduced in aging, and receptor for advanced glycation end products was increased, in parallel with the changes seen in receptor protein expression. Transcriptional changes appear to play a role in aging alterations in blood-brain barrier receptor expression and Aβ accumulation.
Collapse
Affiliation(s)
- Doreen Osgood
- Department of Neurosurgery, Warren Alpert Medical School, Brown University, Providence, RI, USA; The Aldrich Laboratories, Rhode Island Hospital, Providence, RI, USA
| | - Miles C Miller
- Department of Neurosurgery, Warren Alpert Medical School, Brown University, Providence, RI, USA; The Aldrich Laboratories, Rhode Island Hospital, Providence, RI, USA
| | - Arthur A Messier
- Department of Neurosurgery, Warren Alpert Medical School, Brown University, Providence, RI, USA; The Aldrich Laboratories, Rhode Island Hospital, Providence, RI, USA
| | - Liliana Gonzalez
- Department of Computer Science and Statistics, University of Rhode Island, Kingston, RI, USA
| | - Gerald D Silverberg
- Department of Neurosurgery, Warren Alpert Medical School, Brown University, Providence, RI, USA; The Aldrich Laboratories, Rhode Island Hospital, Providence, RI, USA.
| |
Collapse
|
83
|
Morris ME, Rodriguez-Cruz V, Felmlee MA. SLC and ABC Transporters: Expression, Localization, and Species Differences at the Blood-Brain and the Blood-Cerebrospinal Fluid Barriers. AAPS JOURNAL 2017; 19:1317-1331. [PMID: 28664465 DOI: 10.1208/s12248-017-0110-8] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 06/05/2017] [Indexed: 12/11/2022]
Abstract
The blood-brain barrier (BBB) and the blood-cerebrospinal fluid barrier (BCSFB) separate the brain and cerebrospinal fluid (CSF) from the systemic circulation and represent a barrier to the uptake of both endogenous compounds and xenobiotics into the brain. For compounds whose passive diffusion is limited due to their ionization or hydrophilicity, membrane transporters can facilitate their uptake across the BBB or BCSFB. Members of the solute carrier (SLC) and ATP-binding case (ABC) families are present on these barriers. Differences exist in the localization and expression of transport proteins between the BBB and BCSFB, resulting in functional differences in transport properties. This review focuses on the expression, membrane localization, and different isoforms present at each barrier. Diseases that affect the central nervous system including brain tumors, HIV, Alzheimer's disease, Parkinson's disease, and stroke affect the integrity and expression of transporters at the BBB and BCSFB and will be briefly reviewed.
Collapse
Affiliation(s)
- Marilyn E Morris
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York at Buffalo, Buffalo, New York, 14214-8033, USA.
| | - Vivian Rodriguez-Cruz
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York at Buffalo, Buffalo, New York, 14214-8033, USA
| | - Melanie A Felmlee
- Department of Pharmaceutics and Medicinal Chemistry, Thomas J Long School of Pharmacy and Health Sciences, University of the Pacific, 3601 Pacific Ave, Stockton, California, 95211, USA
| |
Collapse
|
84
|
Desai BS, Monahan AJ, Carvey PM, Hendey B. Blood–Brain Barrier Pathology in Alzheimer's and Parkinson's Disease: Implications for Drug Therapy. Cell Transplant 2017; 16:285-99. [PMID: 17503739 DOI: 10.3727/000000007783464731] [Citation(s) in RCA: 229] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The blood–brain barrier (BBB) is a tightly regulated barrier in the central nervous system. Though the BBB is thought to be intact during neurodegenerative diseases such as Alzheimer's (AD) and Parkinson's disease (PD), recent evidence argues otherwise. Dysfunction of the BBB may be involved in disease progression, eliciting of peripheral immune response, and, most importantly, altered drug efficacy. In this review, we will give a brief overview of the BBB, its components, and their functions. We will critically evaluate the current literature in AD and PD BBB pathology resulting from insult, neuroinflammation, and neurodegeneration. Specifically, we will discuss alterations in tight junction, transport and endothelial cell surface proteins, and vascular density changes, all of which result in altered permeability. Finally, we will discuss the implications of BBB dysfunction in current and future therapeutics. Developing a better appreciation of BBB dysfunction in AD and PD may not only provide novel strategies in treatment, but will prove an interesting milestone in understanding neurodegenerative disease etiology and progression.
Collapse
Affiliation(s)
- Brinda S Desai
- Department of Pharmacology, Rush University Medical Center, Chicago, IL 60612, USA.
| | | | | | | |
Collapse
|
85
|
Kannan P, Schain M, Kretzschmar WW, Weidner L, Mitsios N, Gulyás B, Blom H, Gottesman MM, Innis RB, Hall MD, Mulder J. An automated method measures variability in P-glycoprotein and ABCG2 densities across brain regions and brain matter. J Cereb Blood Flow Metab 2017; 37:2062-2075. [PMID: 27488911 PMCID: PMC5464701 DOI: 10.1177/0271678x16660984] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 06/01/2016] [Accepted: 06/22/2016] [Indexed: 12/13/2022]
Abstract
Changes in P-glycoprotein and ABCG2 densities may play a role in amyloid-beta accumulation in Alzheimer's disease. However, previous studies report conflicting results from different brain regions, without correcting for changes in vessel density. We developed an automated method to measure transporter density exclusively within the vascular space, thereby correcting for vessel density. We then examined variability in transporter density across brain regions, matter, and disease using two cohorts of post-mortem brains from Alzheimer's disease patients and age-matched controls. Changes in transporter density were also investigated in capillaries near plaques and on the mRNA level. P-glycoprotein density varied with brain region and matter, whereas ABCG2 density varied with brain matter. In temporal cortex, P-glycoprotein density was 53% lower in Alzheimer's disease samples than in controls, and was reduced by 35% in capillaries near plaque deposits within Alzheimer's disease samples. ABCG2 density was unaffected in Alzheimer's disease. No differences were detected at the transcript level. Our study indicates that region-specific changes in transporter densities can occur globally and locally near amyloid-beta deposits in Alzheimer's disease, providing an explanation for conflicting results in the literature. When differences in region and matter are accounted for, changes in density can be reproducibly measured using our automated method.
Collapse
Affiliation(s)
- Pavitra Kannan
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, USA
- Karolinska Institutet, Department of Clinical Neuroscience, Stockholm, Sweden
- CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, UK
| | - Martin Schain
- Karolinska Institutet, Department of Clinical Neuroscience, Stockholm, Sweden
| | | | - Lora Weidner
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, USA
- Karolinska Institutet, Department of Neuroscience, Science for Life Laboratory, Stockholm, Sweden
| | - Nicholas Mitsios
- Karolinska Institutet, Department of Neuroscience, Science for Life Laboratory, Stockholm, Sweden
| | - Balázs Gulyás
- Karolinska Institutet, Department of Clinical Neuroscience, Stockholm, Sweden
| | - Hans Blom
- Karolinska Institutet, Department of Neuroscience, Science for Life Laboratory, Stockholm, Sweden
| | | | - Robert B Innis
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, USA
| | - Matthew D Hall
- Laboratory of Cell Biology, National Cancer Institute, Bethesda, MD, USA
| | - Jan Mulder
- Karolinska Institutet, Department of Neuroscience, Science for Life Laboratory, Stockholm, Sweden
| |
Collapse
|
86
|
Ban JJ, Chung JY, Lee M, Im W, Kim M. MicroRNA-27a reduces mutant hutingtin aggregation in an in vitro model of Huntington's disease. Biochem Biophys Res Commun 2017; 488:316-321. [PMID: 28495533 DOI: 10.1016/j.bbrc.2017.05.040] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 05/07/2017] [Indexed: 01/28/2023]
Abstract
Huntington's disease (HD) is a fatal genetic disease caused by abnormal aggregation of mutant huntingtin protein (mHtt). Reduction of mHtt aggregation decreases cell death of the brain and is a promising therapeutic strategy of HD. MicroRNAs are short non-coding nucleotides which modulate various genes and dysregulated in many diseases including HD. MicroRNA miR-27a was reported to be reduced in the brain of R6/2 HD mouse model and modulate multidrug resistance protein-1 (MDR-1). Using subventricular zone-derived neuronal stem cells (NSCs), we used in vitro HD model to test the effect of miR-27a on MDR-1 and mHtt aggregation. R6/2-derived NSCs can be differentiated under condition of growth factor deprivation, and the progression of differentiation leads to a decrease of MDR-1 level and efflux function of cells. Immunocytochemistry result also confirmed that mHtt aggregation was increased with differentiation. We transfected miR-27a in the R6/2-derived differentiated NSCs, and examined phenotype of HD, mHtt aggregation. As a result, miR-27a transfection resulted in reduction of mHtt aggregation in HD cells. In addition, MDR-1, which can transport mHtt, protein level was increased by miR-27a transfection. Conversely, knock-down of MDR-1 through MDR-1 siRNA increased mHtt aggregation in vitro. Our results indicate that miR-27a could reduce mHtt level of the HD cell by augmenting MDR-1 function.
Collapse
Affiliation(s)
- Jae-Jun Ban
- Department of Neurology, Seoul National University Hospital, Seoul, South Korea
| | - Jin-Young Chung
- Department of Veterinary Internal Medicine and Geriatrics, College of Veterinary Medicine, Kangwon National University, Gangwon, South Korea
| | - Mijung Lee
- Department of Neurology, Seoul National University Hospital, Seoul, South Korea
| | - Wooseok Im
- Department of Neurology, Seoul National University Hospital, Seoul, South Korea; Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, South Korea.
| | - Manho Kim
- Department of Neurology, Seoul National University Hospital, Seoul, South Korea; Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, South Korea; Protein Metabolism Medical Research Center, College of Medicine, Seoul National University, Seoul, South Korea.
| |
Collapse
|
87
|
McInerney MP, Short JL, Nicolazzo JA. Neurovascular Alterations in Alzheimer's Disease: Transporter Expression Profiles and CNS Drug Access. AAPS JOURNAL 2017; 19:940-956. [PMID: 28462473 DOI: 10.1208/s12248-017-0077-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 03/15/2017] [Indexed: 01/05/2023]
Abstract
Despite a century of steady and incremental progress toward understanding the underlying biochemical mechanisms, Alzheimer's disease (AD) remains a complicated and enigmatic disease, and greater insight will be necessary before substantive clinical success is realised. Over the last decade in particular, a large body of work has highlighted the cerebral microvasculature as an anatomical region with an increasingly apparent role in the pathogenesis of AD. The causative interplay and temporal cascade that manifest between the brain vasculature and the wider disease progression of AD are not yet fully understood, and further inquiry is required to properly characterise these relationships. The purpose of this review is to highlight the recent advancements in research implicating neurovascular factors in AD, at both the molecular and anatomical levels. We begin with a brief introduction of the biochemical and genetic aspects of AD, before reviewing the essential concepts of the blood-brain barrier (BBB) and the neurovascular unit (NVU). In detail, we then examine the evidence demonstrating involvement of BBB dysfunction in AD pathogenesis, highlighting the importance of neurovascular components in AD. Lastly, we include within this review research that focuses on how altered properties of the BBB in AD impact upon CNS exposure of therapeutic agents and the potential clinical impact that this may have on people with this disease.
Collapse
Affiliation(s)
- Mitchell P McInerney
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
| | - Jennifer L Short
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, 3052, VIC, Australia
| | - Joseph A Nicolazzo
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia.
| |
Collapse
|
88
|
Cellular Models and In Vitro Assays for the Screening of modulators of P-gp, MRP1 and BCRP. Molecules 2017; 22:molecules22040600. [PMID: 28397762 PMCID: PMC6153761 DOI: 10.3390/molecules22040600] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 03/24/2017] [Accepted: 03/28/2017] [Indexed: 12/12/2022] Open
Abstract
Adenosine triphosphate (ATP)-binding cassette (ABC) transporters are highly expressed in tumor cells, as well as in organs involved in absorption and secretion processes, mediating the ATP-dependent efflux of compounds, both endogenous substances and xenobiotics, including drugs. Their expression and activity levels are modulated by the presence of inhibitors, inducers and/or activators. In vitro, ex vivo and in vivo studies with both known and newly synthesized P-glycoprotein (P-gp) inducers and/or activators have shown the usefulness of these transport mechanisms in reducing the systemic exposure and specific tissue access of potentially harmful compounds. This article focuses on the main ABC transporters involved in multidrug resistance [P-gp, multidrug resistance-associated protein 1 (MRP1) and breast cancer resistance protein (BCRP)] expressed in tissues of toxicological relevance, such as the blood-brain barrier, cardiovascular system, liver, kidney and intestine. Moreover, it provides a review of the available cellular models, in vitro and ex vivo assays for the screening and selection of safe and specific inducers and activators of these membrane transporters. The available cellular models and in vitro assays have been proposed as high throughput and low-cost alternatives to excessive animal testing, allowing the evaluation of a large number of compounds.
Collapse
|
89
|
Ueno M. Elucidation of mechanism of blood-brain barrier damage for prevention and treatment of vascular dementia. Rinsho Shinkeigaku 2017; 57:95-109. [PMID: 28228623 DOI: 10.5692/clinicalneurol.cn-001004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
It is well-known that the blood-brain barrier (BBB) plays significant roles in transporting intravascular substances into the brain. The BBB in cerebral capillaries essentially impedes the influx of intravascular compounds from the blood to the brain, while nutritive substances, such as glucose, can be selectively transported through several types of influx transporters in endothelial cells. In the choroid plexus, intravascular substances can invade the parenchyma as fenestrations exist in endothelial cells of capillaries. However, the substances cannot invade the ventricles easily as there are tight junctions between epithelial cells in the choroid plexus. This restricted movement of the substances across the cytoplasm of the epithelial cells constitutes a blood-cerebrospinal fluid barrier (BCSFB). In the brain, there are circumventricular organs, in which the barrier function is imperfect in capillaries. Accordingly, it is reasonable to consider that intravascular substances can move in and around the parenchyma of the organs. Actually, it was reported in mice that intravascular substances moved in the corpus callosum, medial portions of the hippocampus, and periventricular areas via the subfornical organs or the choroid plexus. Regarding pathways of intracerebral interstitial and cerebrospinal fluids to the outside of the brain, two representative drainage pathways, or perivascular drainage and glymphatic pathways, are being established. The first is the pathway in a retrograde direction to the blood flow through the basement membrane in walls of cerebral capillaries, the tunica media of arteries, and the vessels walls of the internal carotid artery. The second is in an anterograde direction to blood flow through the para-arterial routes, aquaporin 4-dependent transport through the astroglial cytoplasm, and para-venous routes, and then the fluids drain into the subarachnoid CSF. These fluids are finally considered to drain into the cervical lymph nodes or veins. These clearance pathways may play a role in maintenance of the barrier in the entire brain. Obstruction of the passage of fluids through the perivascular drainage and glymphatic pathways as well as damage of the BBB and BCSFB may induce several kinds of brain disorders, such as vascular dementia. In this review, we focus on the relationship between damage of the barriers and the pathogenesis of vascular dementia and introduce recent findings including our experimental data using animal models.
Collapse
Affiliation(s)
- Masaki Ueno
- Inflammation Pathology, Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University
| |
Collapse
|
90
|
Wang W, Bodles-Brakhop AM, Barger SW. A Role for P-Glycoprotein in Clearance of Alzheimer Amyloid β -Peptide from the Brain. Curr Alzheimer Res 2017; 13:615-20. [PMID: 26971931 DOI: 10.2174/1567205013666160314151012] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 03/07/2016] [Indexed: 12/20/2022]
Abstract
Most data indicates that Alzheimer's disease involves an accumulation of amyloid β - peptide (Aβ) in the CNS and that sporadic cases arise from a deficiency in Aβ clearance. Considerable attention has been given to mechanisms by which Aβ might be transported between the brain and blood, and evidence suggests that p-glycoprotein, also known as the multi-drug resistance (MDR) protein (product of the ABCB1 gene), plays a role in Aβ transport across the blood-brain barrier (BBB). We tested this possibility through two approaches: First, wild-type and MDR1A-knockout mice were compared after intravenous injection of [(125)I]-labeled Aβ; after 60 min, homogenates of brain parenchyma were subjected to γ-counting of TCA-precipitable material, and histological sections of brain were subjected to autoradiography. Second, MDR1Aknockout mice were crossed with Tg2576 APP transgenic mice, a line that routinely accumulates Aβ in the brain; SDS and formic acid extracts of brain homogenates were assessed for Aβ levels by ELISA. Each of these approaches yielded data indicating that Aβ accumulates to a greater degree in mice lacking MDR1A. These findings confirm other reports linking p-glycoprotein to Aβ clearance across the BBB and have important implications for Alzheimer's disease genetics, pharmacology, and epidemiology.
Collapse
Affiliation(s)
| | | | - Steven W Barger
- Reynolds Institute on Aging, #807, 629 Jack Stephens Drive, Little Rock AR 72205, USA.
| |
Collapse
|
91
|
Synthesis and In Vivo Imaging of N-(3-[11C]Methoxybenzyl)-2-(3-Methoxyphenyl)ethylaniline as a Potential Targeting Agent for P-glycoprotein. Mol Imaging Biol 2016; 18:916-923. [DOI: 10.1007/s11307-016-0965-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
92
|
Duarte AC, Hrynchak MV, Gonçalves I, Quintela T, Santos CRA. Sex Hormone Decline and Amyloid β Synthesis, Transport and Clearance in the Brain. J Neuroendocrinol 2016; 28. [PMID: 27632792 DOI: 10.1111/jne.12432] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 09/12/2016] [Accepted: 09/12/2016] [Indexed: 12/15/2022]
Abstract
Sex hormones (SH) are essential regulators of the central nervous system. The decline in SH levels along with ageing may contribute to compromised neuroprotection and set the grounds for neurodegeneration and cognitive impairments. In Alzheimer's disease, besides other pathological features, there is an imbalance between amyloid β (Aβ) production and clearance, leading to its accumulation in the brain of older subjects. Aβ accumulation is a primary cause for brain inflammation and degeneration, as well as concomitant cognitive decline. There is mounting evidence that SH modulate Aβ production, transport and clearance. Importantly, SH regulate most of the molecules involved in the amyloidogenic pathway, their transport across brain barriers for elimination, and their degradation in the brain interstitial fluid. This review brings together data on the regulation of Aβ production, metabolism, degradation and clearance by SH.
Collapse
Affiliation(s)
- A C Duarte
- Health Sciences Research Centre - CICS-UBI, University of Beira Interior, Covilhã, Portugal
| | - M V Hrynchak
- Health Sciences Research Centre - CICS-UBI, University of Beira Interior, Covilhã, Portugal
| | - I Gonçalves
- Health Sciences Research Centre - CICS-UBI, University of Beira Interior, Covilhã, Portugal
| | - T Quintela
- Health Sciences Research Centre - CICS-UBI, University of Beira Interior, Covilhã, Portugal
| | - C R A Santos
- Health Sciences Research Centre - CICS-UBI, University of Beira Interior, Covilhã, Portugal
| |
Collapse
|
93
|
Do TM, Dodacki A, Alata W, Calon F, Nicolic S, Scherrmann JM, Farinotti R, Bourasset F. Age-Dependent Regulation of the Blood-Brain Barrier Influx/Efflux Equilibrium of Amyloid-β Peptide in a Mouse Model of Alzheimer's Disease (3xTg-AD). J Alzheimers Dis 2016; 49:287-300. [PMID: 26484906 DOI: 10.3233/jad-150350] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The involvement of transporters located at the blood-brain barrier (BBB) has been suggested in the control of cerebral Aβ levels, and thereby in Alzheimer's disease (AD). However, little is known about the regulation of these transporters at the BBB in animal models of AD. In this study, we investigated the BBB expression of Aβ influx (Rage) and efflux (Abcb1-Abcg2-Abcg4-Lrp-1) transporters and cholesterol transporter (Abca1) in 3-18-month-old 3xTg-AD and control mice. The age-dependent effect of BBB transporters regulation on the brain uptake clearance (Clup) of [3H]cholesterol and [3H]Aβ1 - 40 was then evaluated in these mice, using the in situ brain perfusion technique. Our data suggest that transgenes expression led to the BBB increase in Aβ influx receptor (Rage) and decrease in efflux receptor (Lrp-1). Our data also indicate that mice have mechanisms counteracting this increased net influx. Indeed, Abcg4 and Abca1 are up regulated in 3- and 3/6-month-old 3xTg-AD mice, respectively. Our data show that the balance between the BBB influx and efflux of Aβ is maintained in 3 and 6-month-old 3xTg-AD mice, suggesting that Abcg4 and Abca1 control the efflux of Aβ through the BBB by a direct (Abcg4) or indirect (Abca1) mechanism. At 18 months, the BBB Aβ efflux is significantly increased in 3xTg-AD mice compared to controls. This could result from the significant up-regulation of both Abcg2 and Abcb1 in 3xTg-AD mice compared to control mice. Thus, age-dependent regulation of several Aβ and cholesterol transporters at the BBB could ultimately limit the brain accumulation of Aβ.
Collapse
Affiliation(s)
- Tuan Minh Do
- Laboratoire de Pharmacie Clinique et pharmacocinétique, EA 4123, Université Paris-Sud 11, Faculté de Pharmacie, Châtenay-Malabry, France
| | - Agnès Dodacki
- Inserm UMR-S1144, Paris, F 75006, France.,Université Paris Descartes, Paris, France.,Université Paris Diderot, Paris, France
| | - Wael Alata
- Faculty of Pharmacy, Laval University, Quebec (QC), Canada
| | - Frederic Calon
- Faculty of Pharmacy, Laval University, Quebec (QC), Canada
| | - Sophie Nicolic
- Inserm UMR-S1144, Paris, F 75006, France.,Université Paris Descartes, Paris, France.,Université Paris Diderot, Paris, France
| | - Jean-Michel Scherrmann
- Inserm UMR-S1144, Paris, F 75006, France.,Université Paris Descartes, Paris, France.,Université Paris Diderot, Paris, France
| | - Robert Farinotti
- Laboratoire de Pharmacie Clinique et pharmacocinétique, EA 4123, Université Paris-Sud 11, Faculté de Pharmacie, Châtenay-Malabry, France
| | - Fanchon Bourasset
- Inserm UMR-S1144, Paris, F 75006, France.,Université Paris Descartes, Paris, France.,Université Paris Diderot, Paris, France
| |
Collapse
|
94
|
Cucullo L, Liles T. Membrane Transporters and Pharmacological Implications. JOURNAL OF PHARMACOVIGILANCE 2016; 4. [PMID: 27610401 PMCID: PMC5012531 DOI: 10.4172/2329-6887.1000e155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Affiliation(s)
- Luca Cucullo
- Department of Pharmaceutical Sciences, 1300 S Coulter St, Amarillo TX 79106-1712, USA; Center for Blood-Brain Barrier Research, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Taylor Liles
- Department of Pharmaceutical Sciences, 1300 S Coulter St, Amarillo TX 79106-1712, USA
| |
Collapse
|
95
|
The choroid plexus in health and in disease: dialogues into and out of the brain. Neurobiol Dis 2016; 107:32-40. [PMID: 27546055 DOI: 10.1016/j.nbd.2016.08.011] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 07/20/2016] [Accepted: 08/17/2016] [Indexed: 12/31/2022] Open
Abstract
This article brings the choroid plexus into the context of health and disease. It is remarkable that the choroid plexus, composed by a monolayer of epithelial cells that lie in a highly vascularized stroma, floating within the brain ventricles, gets so little attention in major physiology and medicine text books and in the scientific literature in general. Consider that it is responsible for producing most of the about 150mL of cerebrospinal fluid that fills the brain ventricles and the subarachnoid space and surrounds the spinal cord in the adult human central nervous system, which is renewed approximately 2-3 times daily. As such, its activity influences brain metabolism and function, which will be addressed. Reflect that it contains an impressive number of receptors and transporters, both in the apical and basolateral sides of the epithelial cells, and as such is a key structure for the communication between the brain and the periphery. This will be highlighted in the context of neonatal jaundice, multiple sclerosis and Alzheimer's disease. Realize that the capillaries that irrigate the choroid plexus stroma do not possess tight junctions and that the blood flow to the choroid plexus is five times higher than that in the brain parenchyma, allowing for a rapid sensing system and delivery of molecules such as nutrients and metals as will be revised. Recognize that certain drugs reach the brain parenchyma solely through the choroid plexus epithelia, which has potential to be manipulated in diseases such as neonatal jaundice and Alzheimer's disease as will be discussed. Without further notice, it must be now clear that understanding the choroid plexus is necessary for comprehending the brain and how the brain is modulated and modulates all other systems, in health and in disease. This review article intends to address current knowledge on the choroid plexus, and to motivate the scientific community to consider it when studying normal brain physiology and diseases of the central nervous system. It will guide the reader through several aspects of the choroid plexus in normal physiology, in diseases characteristic of various periods of life (newborns-kernicterus, young adults-multiple sclerosis and the elder-Alzheimer's disease), and how sex-differences may relate to disease susceptibility.
Collapse
|
96
|
Aβ40 Reduces P-Glycoprotein at the Blood-Brain Barrier through the Ubiquitin-Proteasome Pathway. J Neurosci 2016; 36:1930-41. [PMID: 26865616 DOI: 10.1523/jneurosci.0350-15.2016] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED Failure to clear amyloid-β (Aβ) from the brain is in part responsible for Aβ brain accumulation in Alzheimer's disease (AD). A critical protein for clearing Aβ across the blood-brain barrier is the efflux transporter P-glycoprotein (P-gp) in the luminal plasma membrane of the brain capillary endothelium. P-gp is reduced at the blood-brain barrier in AD, which has been shown to be associated with Aβ brain accumulation. However, the mechanism responsible for P-gp reduction in AD is not well understood. Here we focused on identifying critical mechanistic steps involved in reducing P-gp in AD. We exposed isolated rat brain capillaries to 100 nm Aβ40, Aβ40, aggregated Aβ40, and Aβ42. We observed that only Aβ40 triggered reduction of P-gp protein expression and transport activity levels; this occurred in a dose- and time-dependent manner. To identify the steps involved in Aβ-mediated P-gp reduction, we inhibited protein ubiquitination, protein trafficking, and the ubiquitin-proteasome system, and monitored P-gp protein expression, transport activity, and P-gp-ubiquitin levels. Thus, exposing brain capillaries to Aβ40 triggers ubiquitination, internalization, and proteasomal degradation of P-gp. These findings may provide potential therapeutic targets within the blood-brain barrier to limit P-gp degradation in AD and improve Aβ brain clearance. SIGNIFICANCE STATEMENT The mechanism reducing blood-brain barrier P-glycoprotein (P-gp) in Alzheimer's disease is poorly understood. In the present study, we focused on defining this mechanism. We demonstrate that Aβ40 drives P-gp ubiquitination, internalization, and proteasome-dependent degradation, reducing P-gp protein expression and transport activity in isolated brain capillaries. These findings may provide potential therapeutic avenues within the blood-brain barrier to limit P-gp degradation in Alzheimer's disease and improve Aβ brain clearance.
Collapse
|
97
|
Yuede CM, Lee H, Restivo JL, Davis TA, Hettinger JC, Wallace CE, Young KL, Hayne MR, Bu G, Li CZ, Cirrito JR. Rapid in vivo measurement of β-amyloid reveals biphasic clearance kinetics in an Alzheimer's mouse model. J Exp Med 2016; 213:677-85. [PMID: 27069115 PMCID: PMC4854730 DOI: 10.1084/jem.20151428] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 03/04/2016] [Indexed: 01/03/2023] Open
Abstract
Accumulation of β-amyloid peptide is a key step in Alzheimer’s disease pathogenesis. Yuede et al. propose a novel method to track β-amyloid levels in vivo. Findings from genetic, animal model, and human studies support the observation that accumulation of the β-amyloid (Aβ) peptide in the brain plays a central role in the pathogenic cascade of Alzheimer’s disease (AD). Human studies suggest that one key factor leading to accumulation is a defect in brain Aβ clearance. We have developed a novel microimmunoelectrode (MIE) to study the kinetics of Aβ clearance using an electrochemical approach. This is the first study using MIEs in vivo to measure rapid changes in Aβ levels in the brains of living mice. Extracellular, interstitial fluid (ISF) Aβ levels were measured in the hippocampus of APP/PS1 mice. Baseline levels of Aβ40 in the ISF are relatively stable and begin to decline within minutes of blocking Aβ production with a γ-secretase inhibitor. Pretreatment with a P-glycoprotein inhibitor, which blocks blood–brain barrier transport of Aβ, resulted in significant prolongation of Aβ40 half-life, but only in the latter phase of Aβ clearance from the ISF.
Collapse
Affiliation(s)
- Carla M Yuede
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110
| | - Hyo Lee
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110
| | - Jessica L Restivo
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110
| | - Todd A Davis
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110
| | - Jane C Hettinger
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110
| | - Clare E Wallace
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110
| | - Katherine L Young
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110
| | - Margaret R Hayne
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224
| | - Chen-Zhong Li
- Nanobioengineering/Bioelectronics Laboratory, Department of Biomedical Engineering, Florida International University, Miami, FL 33199
| | - John R Cirrito
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
98
|
Abstract
INTRODUCTION The blood-brain barrier (BBB) possesses an outstanding ability to protect the brain against xenobiotics and potentially poisonous metabolites. Owing to this, ATP binding cassette (ABC) export proteins have garnered significant interest in the research community. These transport proteins are predominantly localized to the luminal membrane of brain microvessels, where they recognize a wide range of different substrates and transport them back into the blood circulation. AREAS COVERED This review summarizes recent findings on these transport proteins, including their expression in the endothelial cell membrane and their substrate recognition. Signaling cascades underlying the expression and function of these proteins will be discussed as well as their role in diseases such as Alzheimer's disease, epilepsy, amyotrophic lateral sclerosis and brain tumors. EXPERT OPINION ABC transporters represent an integral part of the human transportome and are of particular interest at the blood-brain barrier they as they significantly contribute to brain homeostasis. In addition, they appear to be involved in myriad CNS diseases. Therefore studying their mechanisms of action as well as their signaling cascades and responses to internal and external stimuli will help us understand the pathogenesis of these diseases.
Collapse
Affiliation(s)
- Anne Mahringer
- a Institute of Pharmacy and Molecular Biotechnology , Ruprecht-Karls University , Heidelberg , Germany
| | - Gert Fricker
- a Institute of Pharmacy and Molecular Biotechnology , Ruprecht-Karls University , Heidelberg , Germany
| |
Collapse
|
99
|
Mohamed LA, Keller JN, Kaddoumi A. Role of P-glycoprotein in mediating rivastigmine effect on amyloid-β brain load and related pathology in Alzheimer's disease mouse model. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1862:778-787. [PMID: 26780497 PMCID: PMC4788561 DOI: 10.1016/j.bbadis.2016.01.013] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2015] [Revised: 01/13/2016] [Accepted: 01/14/2016] [Indexed: 01/01/2023]
Abstract
Recently, we showed that rivastigmine decreased amyloid-β (Aβ) brain load in aged rats by enhancing its clearance across the blood-brain barrier (BBB) via upregulation of P-glycoprotein (P-gp) and low-density lipoprotein receptor-related protein 1 (LRP1). Here, we extend our previous work to clarify P-gp role in mediating rivastigmine effect on Aβ brain levels and neuroprotection in a mouse model of Alzheimer's disease (AD) that expresses different levels of P-gp. APPSWE mice were bred with mdr1a/b knockout mice to produce littermates that were divided into three groups; APP(+)/mdr1(+/+), APP(+)/mdr1(+/-) and APP(+)/mdr1(-/-). Animals received rivastigmine treatment (0.3mg/kg/day) or vehicle for 8weeks using Alzet osmotic mini-pumps. ELISA analysis of brain homogenates for Aβ showed rivastigmine treatment to significantly decrease Aβ brain load in APP(+)/mdr1(+/+) by 25% and in APP(+)/mdr1(+/-) mice by 21% compared to their vehicle treated littermates, but not in APP(+)/mdr1(-/-) mice. In addition, rivastigmine reduced GFAP immunostaining of astrocytes by 50% and IL-1β brain level by 43% in APP(+)/mdr1(+/+) mice, however its effect was less pronounced in P-gp knockout mice. Moreover, rivastigmine demonstrated a P-gp expression dependent neuroprotective effect that was highest in APP(+)/mdr1(+/+)>APP(+)/mdr1(+/-)>APP(+)/mdr1(-/-) as determined by expression of synaptic markers PSD-95 and SNAP-25 using Western blot analysis. Collectively, our results suggest that P-gp plays important role in mediating rivastigmine non-cholinergic beneficial effects, including Aβ brain load reduction, neuroprotective and anti-inflammatory effects in the AD mouse models.
Collapse
Affiliation(s)
- Loqman A Mohamed
- Department of Basic Pharmaceutical Science, School of Pharmacy, University of Louisiana at Monroe, 1800 Bienville Dr., Monroe, LA 71201, United States
| | - Jeffrey N Keller
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA 70808, United States
| | - Amal Kaddoumi
- Department of Basic Pharmaceutical Science, School of Pharmacy, University of Louisiana at Monroe, 1800 Bienville Dr., Monroe, LA 71201, United States.
| |
Collapse
|
100
|
Prachayasittikul V, Prachayasittikul V. P-glycoprotein transporter in drug development. EXCLI JOURNAL 2016; 15:113-8. [PMID: 27047321 PMCID: PMC4817426 DOI: 10.17179/excli2015-768] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 02/02/2016] [Indexed: 12/13/2022]
Affiliation(s)
- Veda Prachayasittikul
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok 10700, Thailand; Dental Hospital Mahidol University Faculty of Dentistry, Faculty of Dentistry, Mahidol University, Bangkok 10400, Thailand
| | - Virapong Prachayasittikul
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok 10700, Thailand
| |
Collapse
|