1
|
Benarroch E. What Are the Functions of Caveolins and Their Role in Neurologic Disorders? Neurology 2025; 104:e213341. [PMID: 39805058 DOI: 10.1212/wnl.0000000000213341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 12/02/2024] [Indexed: 01/16/2025] Open
|
2
|
Badaut J, Blochet C, Obenaus A, Hirt L. Physiological and pathological roles of caveolins in the central nervous system. Trends Neurosci 2024; 47:651-664. [PMID: 38972795 PMCID: PMC11324375 DOI: 10.1016/j.tins.2024.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 05/14/2024] [Accepted: 06/12/2024] [Indexed: 07/09/2024]
Abstract
Caveolins are a family of transmembrane proteins located in caveolae, small lipid raft invaginations of the plasma membrane. The roles of caveolin-enriched lipid rafts are diverse, and include mechano-protection, lipid homeostasis, metabolism, transport, and cell signaling. Caveolin-1 (Cav-1) and other caveolins were described in endothelial cells and later in other cell types of the central nervous system (CNS), including neurons, astrocytes, oligodendrocytes, microglia, and pericytes. This pancellular presence of caveolins demands a better understanding of their functional roles in each cell type. In this review we describe the various functions of Cav-1 in the cells of normal and pathological brains. Several emerging preclinical findings suggest that Cav-1 could represent a potential therapeutic target in brain disorders.
Collapse
Affiliation(s)
- Jérôme Badaut
- CNRS UMR 5536 RMSB-University of Bordeaux, Bordeaux, France; Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA.
| | - Camille Blochet
- Department of Clinical Neurosciences, CHUV, Lausanne, Switzerland; Department of Fundamental Neuroscience, University of Lausanne, Lausanne, Switzerland
| | - André Obenaus
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA; Division of Biomedical Sciences, University of California Riverside, Riverside, CA, USA
| | - Lorenz Hirt
- Department of Clinical Neurosciences, CHUV, Lausanne, Switzerland; Department of Fundamental Neuroscience, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
3
|
Lau K, Kotzur R, Richter F. Blood-brain barrier alterations and their impact on Parkinson's disease pathogenesis and therapy. Transl Neurodegener 2024; 13:37. [PMID: 39075566 PMCID: PMC11285262 DOI: 10.1186/s40035-024-00430-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 07/11/2024] [Indexed: 07/31/2024] Open
Abstract
There is increasing evidence for blood-brain barrier (BBB) alterations in Parkinson's disease (PD), the second most common neurodegenerative disorder with rapidly rising prevalence. Altered tight junction and transporter protein levels, accumulation of α-synuclein and increase in inflammatory processes lead to extravasation of blood molecules and vessel degeneration. This could result in a self-perpetuating pathophysiology of inflammation and BBB alteration, which contribute to neurodegeneration. Toxin exposure or α-synuclein over-expression in animal models has been shown to initiate similar pathologies, providing a platform to study underlying mechanisms and therapeutic interventions. Here we provide a comprehensive review of the current knowledge on BBB alterations in PD patients and how rodent models that replicate some of these changes can be used to study disease mechanisms. Specific challenges in assessing the BBB in patients and in healthy controls are discussed. Finally, a potential role of BBB alterations in disease pathogenesis and possible implications for therapy are explored. The interference of BBB alterations with current and novel therapeutic strategies requires more attention. Brain region-specific BBB alterations could also open up novel opportunities to target specifically vulnerable neuronal subpopulations.
Collapse
Affiliation(s)
- Kristina Lau
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Foundation, Bünteweg 17, 30559, Hannover, Germany
- Center for Systems Neuroscience, Hannover, Germany
| | - Rebecca Kotzur
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Foundation, Bünteweg 17, 30559, Hannover, Germany
| | - Franziska Richter
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Foundation, Bünteweg 17, 30559, Hannover, Germany.
- Center for Systems Neuroscience, Hannover, Germany.
| |
Collapse
|
4
|
Ni C, Hong M. Oligomerization of drug transporters: Forms, functions, and mechanisms. Acta Pharm Sin B 2024; 14:1924-1938. [PMID: 38799641 PMCID: PMC11119549 DOI: 10.1016/j.apsb.2024.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 12/07/2023] [Accepted: 01/05/2024] [Indexed: 05/29/2024] Open
Abstract
Drug transporters are essential players in the transmembrane transport of a wide variety of clinical drugs. The broad substrate spectra and versatile distribution pattern of these membrane proteins infer their pharmacological and clinical significance. With our accumulating knowledge on the three-dimensional structure of drug transporters, their oligomerization status has become a topic of intense study due to the possible functional roles carried out by such kind of post-translational modification (PTM). In-depth studies of oligomeric complexes formed among drug transporters as well as their interactions with other regulatory proteins can help us better understand the regulatory mechanisms of these membrane proteins, provide clues for the development of novel drugs, and improve the therapeutic efficacy. In this review, we describe different oligomerization forms as well as their structural basis of major drug transporters in the ATP-binding cassette and solute carrier superfamilies, summarize our current knowledge on the influence of oligomerization for protein expression level and transport function of these membrane proteins, and discuss the regulatory mechanisms of oligomerization. Finally, we highlight the challenges associated with the current oligomerization studies and propose some thoughts on the pharmaceutical application of this important drug transporter PTM.
Collapse
Affiliation(s)
- Chunxu Ni
- College of Life Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Mei Hong
- College of Life Sciences, South China Agricultural University, Guangzhou 510642, China
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
5
|
Ávila-Gálvez MÁ, Marques D, Figueira I, Cankar K, Bosch D, Brito MA, Dos Santos CN. Costunolide and parthenolide: Novel blood-brain barrier permeable sesquiterpene lactones to improve barrier tightness. Biomed Pharmacother 2023; 167:115413. [PMID: 37683593 DOI: 10.1016/j.biopha.2023.115413] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 08/21/2023] [Accepted: 08/29/2023] [Indexed: 09/10/2023] Open
Abstract
Sesquiterpene lactones - such as those found in chicory - are considered promising bioactive compounds. These small molecules have shown several health benefits for various diseases, including brain disorders. However, it is unknown whether these compounds can cross the blood-brain barrier (BBB), and which could be the effects on brain microvascular endothelial cells. We show that six sesquiterpene lactones evaluated in an in vitro model of the BBB have different capacities to be transported through the barrier. Costunolide presented more than 20 % of transport while lactucin, 11β-13-dihydrolactucin, 11β-13-dihydrolactucopicrin, and parthenolide presented between 10 % and 20 %, whilst almost no transport was detected for lactucopicrin. Furthermore, costunolide and parthenolide reduced P-gp ABC transporter expression alongside an increase in caveolin-1, the main protein of caveolae. Remarkably, these two compounds improved barrier tightness by increasing the expression of both tight and adherens junctions. These findings open a new avenue to explore costunolide and parthenolide as promising compounds for brain therapies.
Collapse
Affiliation(s)
- María Ángeles Ávila-Gálvez
- Instituto de Biologia Experimental e Tecnológica (iBET), Av. República, Qta. Marquês, 2780-157 Oeiras, Portugal; iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Lisboa, Portugal
| | - Daniela Marques
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Lisboa, Portugal
| | - Inês Figueira
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Lisboa, Portugal
| | - Katarina Cankar
- Wageningen University and Research, Wageningen Plant Research, BU Bioscience, Droevendaalsesteeg 1, 6708PB Wageningen, the Netherlands
| | - Dirk Bosch
- Wageningen University and Research, Wageningen Plant Research, BU Bioscience, Droevendaalsesteeg 1, 6708PB Wageningen, the Netherlands
| | - Maria Alexandra Brito
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal; Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
| | - Cláudia Nunes Dos Santos
- Instituto de Biologia Experimental e Tecnológica (iBET), Av. República, Qta. Marquês, 2780-157 Oeiras, Portugal; iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Lisboa, Portugal.
| |
Collapse
|
6
|
Gellen G, Klement E, Biwott K, Schlosser G, Kalló G, Csősz É, Medzihradszky KF, Bacso Z. Cross-Linking Mass Spectrometry on P-Glycoprotein. Int J Mol Sci 2023; 24:10627. [PMID: 37445813 DOI: 10.3390/ijms241310627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/21/2023] [Accepted: 06/22/2023] [Indexed: 07/15/2023] Open
Abstract
The ABC transporter P-glycoprotein (Pgp) has been found to be involved in multidrug resistance in tumor cells. Lipids and cholesterol have a pivotal role in Pgp's conformations; however, it is often difficult to investigate it with conventional structural biology techniques. Here, we applied robust approaches coupled with cross-linking mass spectrometry (XL-MS), where the natural lipid environment remains quasi-intact. Two experimental approaches were carried out using different cross-linkers (i) on living cells, followed by membrane preparation and immunoprecipitation enrichment of Pgp, and (ii) on-bead, subsequent to membrane preparation and immunoprecipitation. Pgp-containing complexes were enriched employing extracellular monoclonal anti-Pgp antibodies on magnetic beads, followed by on-bead enzymatic digestion. The LC-MS/MS results revealed mono-links on Pgp's solvent-accessible residues, while intraprotein cross-links confirmed a complex interplay between extracellular, transmembrane, and intracellular segments of the protein, of which several have been reported to be connected to cholesterol. Harnessing the MS results and those of molecular docking, we suggest an epitope for the 15D3 cholesterol-dependent mouse monoclonal antibody. Additionally, enriched neighbors of Pgp prove the strong connection of Pgp to the cytoskeleton and other cholesterol-regulated proteins. These findings suggest that XL-MS may be utilized for protein structure and network analyses in such convoluted systems as membrane proteins.
Collapse
Affiliation(s)
- Gabriella Gellen
- MTA-ELTE Lendület Ion Mobility Mass Spectrometry Research Group, Department of Analytical Chemistry, Institute of Chemistry, ELTE Eötvös Loránd University, H-1117 Budapest, Hungary
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem tér 1., H-4032 Debrecen, Hungary
- Doctoral School of Molecular Cell and Immune Biology, University of Debrecen, Egyetem tér 1., H-4032 Debrecen, Hungary
| | - Eva Klement
- Single Cell Omics Advanced Core Facility, HCEMM, H-6728 Szeged, Hungary
- Laboratory of Proteomics Research, BRC, H-6726 Szeged, Hungary
| | - Kipchumba Biwott
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem tér 1., H-4032 Debrecen, Hungary
- Doctoral School of Molecular Cell and Immune Biology, University of Debrecen, Egyetem tér 1., H-4032 Debrecen, Hungary
| | - Gitta Schlosser
- MTA-ELTE Lendület Ion Mobility Mass Spectrometry Research Group, Department of Analytical Chemistry, Institute of Chemistry, ELTE Eötvös Loránd University, H-1117 Budapest, Hungary
| | - Gergő Kalló
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Egyetem tér 1., H-4032 Debrecen, Hungary
- Proteomics Core Facility, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Egyetem tér 1., H-4032 Debrecen, Hungary
| | - Éva Csősz
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Egyetem tér 1., H-4032 Debrecen, Hungary
- Proteomics Core Facility, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Egyetem tér 1., H-4032 Debrecen, Hungary
| | | | - Zsolt Bacso
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem tér 1., H-4032 Debrecen, Hungary
- Doctoral School of Molecular Cell and Immune Biology, University of Debrecen, Egyetem tér 1., H-4032 Debrecen, Hungary
- Faculty of Pharmacology, University of Debrecen, Egyetem tér 1., H-4032 Debrecen, Hungary
| |
Collapse
|
7
|
Chai AB, Callaghan R, Gelissen IC. Regulation of P-Glycoprotein in the Brain. Int J Mol Sci 2022; 23:ijms232314667. [PMID: 36498995 PMCID: PMC9740459 DOI: 10.3390/ijms232314667] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/18/2022] [Accepted: 11/22/2022] [Indexed: 11/27/2022] Open
Abstract
Maintenance of the tightly regulated homeostatic environment of the brain is facilitated by the blood-brain barrier (BBB). P-glycoprotein (P-gp), an ATP-binding cassette transporter, is expressed on the luminal surface of the endothelial cells in the BBB, and actively exports a wide variety of substrates to limit exposure of the vulnerable brain environment to waste buildup and neurotoxic compounds. Downregulation of P-gp expression and activity at the BBB have been reported with ageing and in neurodegenerative diseases. Upregulation of P-gp at the BBB contributes to poor therapeutic outcomes due to altered pharmacokinetics of CNS-acting drugs. The regulation of P-gp is highly complex, but unravelling the mechanisms involved may help the development of novel and nuanced strategies to modulate P-gp expression for therapeutic benefit. This review summarises the current understanding of P-gp regulation in the brain, encompassing the transcriptional, post-transcriptional and post-translational mechanisms that have been identified to affect P-gp expression and transport activity.
Collapse
Affiliation(s)
- Amanda B. Chai
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
| | - Richard Callaghan
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Ingrid C. Gelissen
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
- Correspondence: ; Tel.: +61-2-8627-0357
| |
Collapse
|
8
|
The Impact of P-Glycoprotein on Opioid Analgesics: What's the Real Meaning in Pain Management and Palliative Care? Int J Mol Sci 2022; 23:ijms232214125. [PMID: 36430602 PMCID: PMC9695906 DOI: 10.3390/ijms232214125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/01/2022] [Accepted: 11/11/2022] [Indexed: 11/18/2022] Open
Abstract
Opioids are widely used in cancer and non-cancer pain management. However, many transporters at the blood-brain barrier (BBB), such as P-glycoprotein (P-gp, ABCB1/MDR1), may impair their delivery to the brain, thus leading to opioid tolerance. Nonetheless, opioids may regulate P-gp expression, thus altering the transport of other compounds, namely chemotherapeutic agents, resulting in pharmacoresistance. Other kinds of painkillers (e.g., acetaminophen, dexamethasone) and adjuvant drugs used for neuropathic pain may act as P-gp substrates and modulate its expression, thus making pain management challenging. Inflammatory conditions are also believed to upregulate P-gp. The role of P-gp in drug-drug interactions is currently under investigation, since many P-gp substrates may also act as substrates for the cytochrome P450 enzymes, which metabolize a wide range of xenobiotics and endobiotics. Genetic variability of the ABCB1/MDR1 gene may be accountable for inter-individual variation in opioid-induced analgesia. P-gp also plays a role in the management of opioid-induced adverse effects, such as constipation. Peripherally acting mu-opioid receptors antagonists (PAMORAs), such as naloxegol and naldemedine, are substrates of P-gp, which prevent their penetration in the central nervous system. In our review, we explore the interactions between P-gp and opioidergic drugs, with their implications in clinical practice.
Collapse
|
9
|
Eng ME, Imperio GE, Bloise E, Matthews SG. ATP-binding cassette (ABC) drug transporters in the developing blood-brain barrier: role in fetal brain protection. Cell Mol Life Sci 2022; 79:415. [PMID: 35821142 PMCID: PMC11071850 DOI: 10.1007/s00018-022-04432-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 05/27/2022] [Accepted: 06/15/2022] [Indexed: 12/19/2022]
Abstract
The blood-brain barrier (BBB) provides essential neuroprotection from environmental toxins and xenobiotics, through high expression of drug efflux transporters in endothelial cells of the cerebral capillaries. However, xenobiotic exposure, stress, and inflammatory stimuli have the potential to disrupt BBB permeability in fetal and post-natal life. Understanding the role and ability of the BBB in protecting the developing brain, particularly with respect to drug/toxin transport, is key to promoting long-term brain health. Drug transporters, particularly P-gp and BCRP are expressed in early gestation at the developing BBB and have a crucial role in developmental homeostasis and fetal brain protection. We have highlighted several factors that modulate drug transporters at the developing BBB, including synthetic glucocorticoid (sGC), cytokines, maternal infection, and growth factors. Some factors have the potential to increase expression and function of drug transporters and increase brain protection (e.g., sGC, transforming growth factor [TGF]-β). However, others inhibit drug transporters expression and function at the BBB, increasing brain exposure to xenobiotics (e.g., tumor necrosis factor [TNF], interleukin [IL]-6), negatively impacting brain development. This has implications for pregnant women and neonates, who represent a vulnerable population and may be exposed to drugs and environmental toxins, many of which are P-gp and BCRP substrates. Thus, alterations in regulated transport across the developing BBB may induce long-term changes in brain health and compromise pregnancy outcome. Furthermore, a large portion of neonatal adverse drug reactions are attributed to agents that target or access the nervous system, such as stimulants (e.g., caffeine), anesthetics (e.g., midazolam), analgesics (e.g., morphine) and antiretrovirals (e.g., Zidovudine); thus, understanding brain protection is key for the development of strategies to protect the fetal and neonatal brain.
Collapse
Affiliation(s)
- Margaret E Eng
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Medical Sciences Bldg. Rm. 3207. 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
| | | | - Enrrico Bloise
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Medical Sciences Bldg. Rm. 3207. 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Canada
- Department of Morphology, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Stephen G Matthews
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Medical Sciences Bldg. Rm. 3207. 1 King's College Circle, Toronto, ON, M5S 1A8, Canada.
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Canada.
- Department of Obstetrics and Gynecology, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada.
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada.
| |
Collapse
|
10
|
Low JY, Laiho M. Caveolae-Associated Molecules, Tumor Stroma, and Cancer Drug Resistance: Current Findings and Future Perspectives. Cancers (Basel) 2022; 14:cancers14030589. [PMID: 35158857 PMCID: PMC8833326 DOI: 10.3390/cancers14030589] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/21/2022] [Accepted: 01/22/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Cell membranes contain small invaginations called caveolae. They are a specialized lipid domain and orchestrate cellular signaling events, mechanoprotection, and lipid homeostasis. Formation of the caveolae depends on two classes of proteins, the caveolins and cavins, which form large complexes that allow their self-assembly into caveolae. Loss of either of these two proteins leads to distortion of the caveolae structure and disruption of many physiological processes that affect diseases of the muscle, metabolic states governing lipids, and the glucose balance as well as cancers. In cancers, the expression of caveolins and cavins is heterogenous, and they undergo alterations both in the tumors and the surrounding tumor microenvironment stromal cells. Remarkably, their expression and function has been associated with resistance to many cancer drugs. Here, we summarize the current knowledge of the resistance mechanisms and how this knowledge could be applied into the clinic in future. Abstract The discovery of small, “cave-like” invaginations at the plasma membrane, called caveola, has opened up a new and exciting research area in health and diseases revolving around this cellular ultrastructure. Caveolae are rich in cholesterol and orchestrate cellular signaling events. Within caveola, the caveola-associated proteins, caveolins and cavins, are critical components for the formation of these lipid rafts, their dynamics, and cellular pathophysiology. Their alterations underlie human diseases such as lipodystrophy, muscular dystrophy, cardiovascular disease, and diabetes. The expression of caveolins and cavins is modulated in tumors and in tumor stroma, and their alterations are connected with cancer progression and treatment resistance. To date, although substantial breakthroughs in cancer drug development have been made, drug resistance remains a problem leading to treatment failures and challenging translation and bench-to-bedside research. Here, we summarize the current progress in understanding cancer drug resistance in the context of caveola-associated molecules and tumor stroma and discuss how we can potentially design therapeutic avenues to target these molecules in order to overcome treatment resistance.
Collapse
Affiliation(s)
- Jin-Yih Low
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA;
- Correspondence: ; Tel.: +1-410-502-9748; Fax: +1-410-502-2821
| | - Marikki Laiho
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA;
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
11
|
Abstract
The accumulation of neurotoxic amyloid-beta (Aβ) in the brain is one of the characteristic hallmarks of Alzheimer's disease (AD). Aβ-peptide brain homeostasis is governed by its production and various clearance mechanisms. The blood-brain barrier provides a large surface area for influx and efflux mechanisms into and out of the brain. Different transporters and receptors have been implicated to play crucial roles in Aβ clearance from brain. Besides Aβ transport, the blood-brain barrier tightly regulates the brain's microenvironment; however, vascular alterations have been shown in patients with AD. Here, we summarize how the blood-brain barrier changes during aging and in disease and focus on recent findings of how the ABC transporter P-glycoprotein (ABCB1/P-gp) and the receptor low-density lipoprotein receptor-related protein 1 (LRP1) play a role in Aβ clearance from brain.
Collapse
|
12
|
Wang LM, Wang YT, Yang WX. Engineered nanomaterials induce alterations in biological barriers: focus on paracellular permeability. Nanomedicine (Lond) 2021; 16:2725-2741. [PMID: 34870452 DOI: 10.2217/nnm-2021-0165] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Engineered nanoparticles (ENPs) are widely used in medical diagnosis and treatment, as food additives and as energy materials. ENPs may exert adverse or beneficial effects on the human body, which may be linked to interactions with biological barriers. In this review, the authors summarize the influences of four typical metal/metal oxide nanomaterials (Ag, TiO2, Au, ZnO nanoparticles) on the paracellular permeability of biological barriers. Disruptions on tight junctions, adhesion junctions, gap junctions and desmosomes via complex signaling pathways, such as the MAPK, PKC and ROCK signaling pathways, affect paracellular permeability. Reactive oxygen species and cytokines underlie the mechanism of ENP-triggered alterations in paracellular permeability. This review provides the information necessary for the cautious application of nanoparticles in medicine and life sciences in the future.
Collapse
Affiliation(s)
- Lan-Min Wang
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, 310058, PR China
| | - Yu-Ting Wang
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, 310058, PR China
| | - Wan-Xi Yang
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, 310058, PR China
| |
Collapse
|
13
|
Gericke B, Borsdorf S, Wienböker I, Noack A, Noack S, Löscher W. Similarities and differences in the localization, trafficking, and function of P-glycoprotein in MDR1-EGFP-transduced rat versus human brain capillary endothelial cell lines. Fluids Barriers CNS 2021; 18:36. [PMID: 34344390 PMCID: PMC8330100 DOI: 10.1186/s12987-021-00266-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 06/28/2021] [Indexed: 12/24/2022] Open
Abstract
Background In vitro models based on brain capillary endothelial cells (BCECs) are among the most versatile tools in blood–brain barrier research for testing drug penetration into the brain and how this is affected by efflux transporters such as P-glycoprotein (Pgp). However, compared to freshly isolated brain capillaries or primary BCECs, the expression of Pgp in immortalized BCEC lines is markedly lower, which prompted us previously to transduce the widely used human BCEC line hCMEC/D3 with a doxycycline-inducible MDR1-EGFP fusion plasmid. The EGFP-labeled Pgp in these cells allows studying the localization and trafficking of the transporter and how these processes are affected by drug exposure. Here we used this strategy for the rat BCEC line RBE4 and performed a face-to-face comparison of RBE4 and hCMEC/D3 wild-type (WT) and MDR1-EGFP transduced cells. Methods MDR1-EGFP-transduced variants were derived from WT cells by lentiviral transduction, using an MDR1-linker-EGFP vector. Localization, trafficking, and function of Pgp were compared in WT and MDR1-EGFP transduced cell lines. Primary cultures of rat BCECs and freshly isolated rat brain capillaries were used for comparison. Results All cells exhibited typical BCEC morphology. However, significant differences were observed in the localization of Pgp in that RBE4-MDR1-EGFP cells expressed Pgp primarily at the plasma membrane, whereas in hCMEC/D3 cells, the Pgp-EGFP fusion protein was visible both at the plasma membrane and in endolysosomal vesicles. Exposure to doxorubicin increased the number of Pgp-EGFP-positive endolysosomes, indicating a lysosomotropic effect. Furthermore, lysosomal trapping of doxorubicin was observed, likely contributing to the protection of the cell nucleus from damage. In cocultures of WT and MDR1-EGFP transduced cells, intercellular Pgp-EGFP trafficking was observed in RBE4 cells as previously reported for hCMEC/D3 cells. Compared to WT cells, the MDR1-EGFP transduced cells exhibited a significantly higher expression and function of Pgp. However, the junctional tightness of WT and MDR1-EGFP transduced RBE4 and hCMEC/D3 cells was markedly lower than that of primary BCECs, excluding the use of the cell lines for studying vectorial drug transport. Conclusions The present data indicate that MDR1-EGFP transduced RBE4 cells are an interesting tool to study the biogenesis of lysosomes and Pgp-mediated lysosomal drug trapping in response to chemotherapeutic agents and other compounds at the level of the blood–brain barrier. Supplementary Information The online version contains supplementary material available at 10.1186/s12987-021-00266-z.
Collapse
Affiliation(s)
- Birthe Gericke
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Bünteweg 17, 30559, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany
| | - Saskia Borsdorf
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Bünteweg 17, 30559, Hannover, Germany
| | - Inka Wienböker
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Bünteweg 17, 30559, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany
| | - Andreas Noack
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Bünteweg 17, 30559, Hannover, Germany
| | - Sandra Noack
- Department of Trauma Surgery, Hannover Medical School, Hannover, Germany
| | - Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Bünteweg 17, 30559, Hannover, Germany. .,Center for Systems Neuroscience, Hannover, Germany.
| |
Collapse
|
14
|
Abstract
From the viewpoint of drug discovery, it is an important issue to elucidate the drug permeability at the human central nervous system (CNS) barriers and the molecular mechanisms in the cells forming CNS barriers especially during CNS diseases. I introduced quantitative proteomics techniques into the blood-brain barrier (BBB) study, then quantitatively investigated the transport system at the human BBB and clarified the quantitative differences in protein expression levels and functions of transporters and receptors between animals and humans, or in vitro and in vivo. Based on the difference in the absolute expression level of transporters between in vitro and in vivo, I demonstrated that the drug efflux activity of P-glycoprotein (P-gp) at in vivo BBB can be accurately reconstructed from the in vitro system, not only in mouse models but also monkeys similar to humans and pathological conditions. Furthermore, I discovered Claudin-11 as another tight junction molecule expressed at the CNS barriers, and clarified that it contributes to the disruption of the CNS barriers in multiple sclerosis. Furthermore, it was also elucidated that the P-gp dysfunction causes excessive brain entry of glucocorticoid which causes a nerve damage in cerebral infarct, and it can be suppressed by targeting Abl/Src kinases. These suggest that targeting the tight junctions and transporters, which are important molecules at the CNS barriers, would potentially lead to the treatment of CNS diseases. In this review, I would like to introduce a new CNS barrier study opened by quantitative proteomics research.
Collapse
Affiliation(s)
- Yasuo Uchida
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences, Tohoku University
| |
Collapse
|
15
|
Ding Y, Zhong Y, Baldeshwiler A, Abner EL, Bauer B, Hartz AMS. Protecting P-glycoprotein at the blood-brain barrier from degradation in an Alzheimer's disease mouse model. Fluids Barriers CNS 2021; 18:10. [PMID: 33676539 PMCID: PMC7937299 DOI: 10.1186/s12987-021-00245-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Accepted: 02/25/2021] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND Failure to clear Aβ from the brain is partly 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 AD, P-gp levels are reduced, which contributes to impaired Aβ brain clearance. However, the mechanism responsible for decreased P-gp levels is poorly understood and there are no strategies available to protect P-gp. We previously demonstrated in isolated brain capillaries ex vivo that human Aβ40 (hAβ40) triggers P-gp degradation by activating the ubiquitin-proteasome pathway. In this pathway, hAβ40 initiates P-gp ubiquitination, leading to internalization and proteasomal degradation of P-gp, which then results in decreased P-gp protein expression and transport activity levels. Here, we extend this line of research and present results from an in vivo study using a transgenic mouse model of AD (human amyloid precursor protein (hAPP)-overexpressing mice; Tg2576). METHODS In our study, hAPP mice were treated with vehicle, nocodazole (NCZ, microtubule inhibitor to block P-gp internalization), or a combination of NCZ and the P-gp inhibitor cyclosporin A (CSA). We determined P-gp protein expression and transport activity levels in isolated mouse brain capillaries and Aβ levels in plasma and brain tissue. RESULTS Treating hAPP mice with 5 mg/kg NCZ for 14 days increased P-gp levels to levels found in WT mice. Consistent with this, P-gp-mediated hAβ42 transport in brain capillaries was increased in NCZ-treated hAPP mice compared to untreated hAPP mice. Importantly, NCZ treatment significantly lowered hAβ40 and hAβ42 brain levels in hAPP mice, whereas hAβ40 and hAβ42 levels in plasma remained unchanged. CONCLUSIONS These findings provide in vivo evidence that microtubule inhibition maintains P-gp protein expression and transport activity levels, which in turn helps to lower hAβ brain levels in hAPP mice. Thus, protecting P-gp at the blood-brain barrier may provide a novel therapeutic strategy for AD and other Aβ-based pathologies.
Collapse
Affiliation(s)
- Yujie Ding
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, 40536, USA
| | - Yu Zhong
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, 40536, USA
| | - Andrea Baldeshwiler
- Department of Pharmacy Practice and Pharmaceutical Sciences, College of Pharmacy, University of Minnesota, Duluth, Minnesota, 55812, USA
| | - Erin L Abner
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, 40536, USA
- Department of Epidemiology, College of Public Health, University of Kentucky, Lexington, KY, 40536, USA
| | - Björn Bauer
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, 40536, USA
| | - Anika M S Hartz
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, 40536, USA.
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, 40536, USA.
- University of Kentucky Sanders-Brown Center on Aging, 800 S Limestone, Lexington, KY, 40536, USA.
| |
Collapse
|
16
|
Molecular Regulation of Canalicular ABC Transporters. Int J Mol Sci 2021; 22:ijms22042113. [PMID: 33672718 PMCID: PMC7924332 DOI: 10.3390/ijms22042113] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/15/2021] [Accepted: 02/18/2021] [Indexed: 12/17/2022] Open
Abstract
The ATP-binding cassette (ABC) transporters expressed at the canalicular membrane of hepatocytes mediate the secretion of several compounds into the bile canaliculi and therefore play a key role in bile secretion. Among these transporters, ABCB11 secretes bile acids, ABCB4 translocates phosphatidylcholine and ABCG5/G8 is responsible for cholesterol secretion, while ABCB1 and ABCC2 transport a variety of drugs and other compounds. The dysfunction of these transporters leads to severe, rare, evolutionary biliary diseases. The development of new therapies for patients with these diseases requires a deep understanding of the biology of these transporters. In this review, we report the current knowledge regarding the regulation of canalicular ABC transporters' folding, trafficking, membrane stability and function, and we highlight the role of molecular partners in these regulating mechanisms.
Collapse
|
17
|
Eustaquio Do Imperio G, Lye P, Bloise E, Matthews SG. Function of Multidrug Resistance Transporters is Disrupted by Infection Mimics in Human Brain Endothelial Cells. Tissue Barriers 2021; 9:1860616. [PMID: 33427563 PMCID: PMC8078541 DOI: 10.1080/21688370.2020.1860616] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
P-glycoprotein (P-gp/ABCB1) and breast cancer resistance protein (BCRP/ABCG2) modulate the distribution of drugs and toxins across the blood-brain barrier (BBB). Animal studies reported that infection-induced disruption of these transporters in the developing BBB impairs fetal brain protection. However, the impact of infection mimics on P-gp/BCRP function in human brain endothelium is less well understood. We hypothesized that Toll-like receptor ligands mimicking bacterial and viral infection would modify the expression and function of P-gp and BCRP in human brain endothelial cells (BECs). Human cerebral microvascular endothelial cells (hCMEC/D3) were challenged with bacterial [Lipopolysaccharide (LPS)] and viral-mimics [polyinosinic:polycytidylic acid (PolyI:C) or single-stranded RNA (ssRNA)], or pro-inflammatory cytokines interleukin (IL)-6, tumor necrosis factor (TNF)-α and interferon gamma (IFN)-ɣ. P-gp and BCRP function was assessed after 4 or 24 h, using Calcein-AM and Chlorin-6 assays, respectively. Western blot and qPCR quantified P-gp/ABCB1 and BCRP/ABCG2 expression following treatments. Infection mimics are potent modulators of drug transporters in human BECs in vitro. LPS and PolyI:C increased, while ssRNA exposure reduced P-gp activity. In contrast, LPS and PolyI:C decreased, while ssRNA increased BCRP activity (P < .05). There was little correlation between drug transporter function, gene expression and total protein level. Altered plasma membrane BCRP may suggest modified intracellular trafficking induced by infection in human BECs. Bacterial and viral infection mimics modify P-gp and BCRP transport function in human BECs, in vitro. This knowledge may contribute and have important implications for human brain protection and possible altered biodistribution of drugs and xenobiotics in the brain following exposure to TLR agonists.
Collapse
Affiliation(s)
| | - Phetcharawan Lye
- Sinai Health System, Lunenfeld-Tanenbaum Research Institute, Toronto, Ontario, Canada.,Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Enrrico Bloise
- Sinai Health System, Lunenfeld-Tanenbaum Research Institute, Toronto, Ontario, Canada.,Department of Physiology, University of Toronto, Toronto, Ontario, Canada.,Department of Morphology, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Stephen G Matthews
- Sinai Health System, Lunenfeld-Tanenbaum Research Institute, Toronto, Ontario, Canada.,Department of Physiology, University of Toronto, Toronto, Ontario, Canada.,Department of Obstetrics and Gynecology and Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
18
|
de Trizio I, Errede M, d'Amati A, Girolamo F, Virgintino D. Expression of P-gp in Glioblastoma: What we can Learn from Brain Development. Curr Pharm Des 2020; 26:1428-1437. [PMID: 32186270 DOI: 10.2174/1381612826666200318130625] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 02/18/2020] [Indexed: 02/06/2023]
Abstract
P-Glycoprotein (P-gp) is a 170-kDa transmembrane glycoprotein that works as an efflux pump and confers multidrug resistance (MDR) in normal tissues and tumors, including nervous tissues and brain tumors. In the developing telencephalon, the endothelial expression of P-gp, and the subcellular localization of the transporter at the luminal endothelial cell (EC) plasma membrane are early hallmarks of blood-brain barrier (BBB) differentiation and suggest a functional BBB activity that may complement the placental barrier function and the expression of P-gp at the blood-placental interface. In early fetal ages, P-gp has also been immunolocalized on radial glia cells (RGCs), located in the proliferative ventricular zone (VZ) of the dorsal telencephalon and now considered to be neural progenitor cells (NPCs). RG-like NPCs have been found in many regions of the developing brain and have been suggested to give rise to neural stem cells (NSCs) of adult subventricular (SVZ) neurogenic niches. The P-gp immunosignal, associated with RG-like NPCs during cortical histogenesis, progressively decreases in parallel with the last waves of neuroblast migrations, while 'outer' RGCs and the deriving astrocytes do not stain for the efflux transporter. These data suggest that in human glioblastoma (GBM), P-gp expressed by ECs may be a negligible component of tumor MDR. Instead, tumor perivascular astrocytes may dedifferentiate and resume a progenitor-like P-gp activity, becoming MDR cells and contribute, together with perivascular P-gpexpressing glioma stem-like cells (GSCs), to the MDR profile of GBM vessels. In conclusion, the analysis of Pgp immunolocalization during brain development may contribute to identify the multiple cellular sources in the GBM vessels that may be involved in P-gp-mediated chemoresistance and can be responsible for GBM therapy failure and tumor recurrence.
Collapse
Affiliation(s)
- Ignazio de Trizio
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Human Anatomy and Histology Unit, University of Bari, School of Medicine, Bari, Italy.,Department of Neurosurgery, Neurocenter of Southern Switzerland, Regional Hospital Lugano, Lugano, Switzerland
| | - Mariella Errede
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Human Anatomy and Histology Unit, University of Bari, School of Medicine, Bari, Italy
| | - Antonio d'Amati
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Human Anatomy and Histology Unit, University of Bari, School of Medicine, Bari, Italy
| | - Francesco Girolamo
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Human Anatomy and Histology Unit, University of Bari, School of Medicine, Bari, Italy
| | - Daniela Virgintino
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Human Anatomy and Histology Unit, University of Bari, School of Medicine, Bari, Italy
| |
Collapse
|
19
|
VEGF/VEGFR-2 system exerts neuroprotection against Phoneutria nigriventer spider envenomation through PI3K-AKT-dependent pathway. Toxicon 2020; 185:76-90. [PMID: 32649934 DOI: 10.1016/j.toxicon.2020.06.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 06/23/2020] [Accepted: 06/23/2020] [Indexed: 01/19/2023]
Abstract
This study was undertaken to elucidate why VEGF/VEGFR-2 is elevated in the hippocampus of rats injected with Phoneutria nigriventer spider venom (PNV). PNV delays Na+ channels inactivation; blocks Ca2+ and K+ channels, increases glutamate release, causes blood-brain breakdown (BBBb), brain edema and severe excitotoxicity. Analytical FT-IR spectroscopy showed profound alteration in molecular biochemical state, with evidences for VEGFR-2 (KDR/Flk-1) signaling mediation. By blocking VEGF/VEGFR-2 binding via pre-treatment with itraconazole we demonstrated that animals' condition was deteriorated soon at 1-2 h post-PNV exposure concurrently with decreased expression of VEGF, BBB-associated proteins, ZO-1, β-catenin, laminin, P-gp (P-glycoprotein), Neu-N (neuron's viability marker) and MAPKphosphorylated-p38, while phosphorylated-ERK and Src pathways were increased. At 5 h and coinciding with incipient signs of animals' recuperation, the proteins associated with protection (HIF-1α, VEGF, VEGFR-1, VEGFR-2, Neu-N, occludin, β-catenin, laminin, P-gp efflux protein, phosphorylated-p38) increased thus indicating p38 pathway activation together with paracellular route strengthening. However, the BBB transcellular trafficking and caspase-3 increased (pro-apoptotic pathway activation). At 24 h, the transcellular route reestablished physiological state but the pro-survival pathway PI3K/(p-Akt) dropped in animals underwent VEGF/VEGFR-2 binding inhibition, whereas it was significantly activated at matched interval in PNV group without prior itraconazole; these results demonstrate impaired VEGF' survival effects at 24 h. The inhibition of VEGF/VEGFR-2 binding identified 5 h as turning point at which multi-level dynamic interplay was elicited to reverse hippocampal damage. Collectively, the data confirmed VEGFR-2 signaling via serine-threonine kinase Akt as neuroprotective pathway against PNV-induced damage. Further studies are needed to elucidate mechanisms underlying PNV effects.
Collapse
|
20
|
Novel Intrinsic Mechanisms of Active Drug Extrusion at the Blood-Brain Barrier: Potential Targets for Enhancing Drug Delivery to the Brain? Pharmaceutics 2020; 12:pharmaceutics12100966. [PMID: 33066604 PMCID: PMC7602420 DOI: 10.3390/pharmaceutics12100966] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/07/2020] [Accepted: 10/08/2020] [Indexed: 12/13/2022] Open
Abstract
The blood-brain barrier (BBB) limits the pharmacotherapy of several brain disorders. In addition to the structural and metabolic characteristics of the BBB, the ATP-driven, drug efflux transporter P-glycoprotein (Pgp) is a selective gatekeeper of the BBB; thus, it is a primary hindrance to drug delivery into the brain. Here, we review the complex regulation of Pgp expression and functional activity at the BBB with an emphasis on recent studies from our laboratory. In addition to traditional processes such as transcriptional regulation and posttranscriptional or posttranslational modification of Pgp expression and functionality, novel mechanisms such as intra- and intercellular Pgp trafficking and intracellular Pgp-mediated lysosomal sequestration in BBB endothelial cells with subsequent disposal by blood neutrophils are discussed. These intrinsic mechanisms of active drug extrusion at the BBB are potential therapeutic targets that could be used to modulate P-glycoprotein activity in the treatment of brain diseases and enhance drug delivery to the brain.
Collapse
|
21
|
Direct and indirect cholesterol effects on membrane proteins with special focus on potassium channels. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1865:158706. [DOI: 10.1016/j.bbalip.2020.158706] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 03/19/2020] [Accepted: 03/30/2020] [Indexed: 12/16/2022]
|
22
|
Hoshi Y, Uchida Y, Tachikawa M, Ohtsuki S, Couraud PO, Suzuki T, Terasaki T. Oxidative stress-induced activation of Abl and Src kinases rapidly induces P-glycoprotein internalization via phosphorylation of caveolin-1 on tyrosine-14, decreasing cortisol efflux at the blood-brain barrier. J Cereb Blood Flow Metab 2020; 40:420-436. [PMID: 30621530 PMCID: PMC7370610 DOI: 10.1177/0271678x18822801] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Exposure of the brain to high levels of glucocorticoids during ischemia-reperfusion induces neuronal cell death. Oxidative stress alters blood-brain barrier (BBB) function during ischemia-reperfusion, and so we hypothesized that it might impair P-glycoprotein (P-gp)-mediated efflux transport of glucocorticoids at the BBB. Therefore, the purpose of this study was to clarify the molecular mechanism of this putative decrease of P-gp-mediated efflux function. First, we established that H2O2 treatment of a human in vitro BBB model (hCMEC/D3) reduced both P-gp efflux transport activity and protein expression on the plasma membrane within 20 min. These results suggested that the rapid decrease of efflux function might be due to internalization of P-gp. Furthermore, H2O2 treatment markedly increased tyrosine-14-phosphorylated caveolin-1, which is involved in P-gp internalization. A brain perfusion study in rats showed that cortisol efflux at the BBB was markedly decreased by H2O2 administration, and inhibitors of Abl kinase and Src kinase, which phosphorylate tyrosine-14 in caveolin-1, suppressed this decrease. Overall, these findings support the idea that oxidative stress-induced activation of Abl kinase and Src kinase induces internalization of P-gp via the phosphorylation of tyrosine-14 in caveolin-1, leading to a rapid decrease of P-gp-mediated cortisol efflux at the BBB.
Collapse
Affiliation(s)
- Yutaro Hoshi
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Yasuo Uchida
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Masanori Tachikawa
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Sumio Ohtsuki
- Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | | | - Takashi Suzuki
- Department of Pathology and Histotechnology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tetsuya Terasaki
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| |
Collapse
|
23
|
Fan Y, Si W, Ji W, Wang Z, Gao Z, Tian R, Song W, Zhang H, Niu R, Zhang F. Rack1 mediates Src binding to drug transporter P-glycoprotein and modulates its activity through regulating Caveolin-1 phosphorylation in breast cancer cells. Cell Death Dis 2019; 10:394. [PMID: 31113938 PMCID: PMC6529477 DOI: 10.1038/s41419-019-1633-y] [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: 01/23/2019] [Revised: 05/02/2019] [Accepted: 05/06/2019] [Indexed: 12/24/2022]
Abstract
The failure of chemotherapy and the emergence of multidrug resistance (MDR) are the major obstacles for effective therapy in locally advanced and metastatic breast cancer. Overexpression of the drug transporter P-glycoprotein (P-gp) in cancer cells is one of the main causes of MDR due to its ability to efflux anticancer drugs out of cells. Although the signaling node that regulates the expression of P-gp has been intensively investigated; the regulatory mechanism underlying P-gp transport activity remains obscure. Herein, we reported that Rack1 and tyrosine kinase Src confer drug resistance through modulating the transport function of P-gp without altering its protein level. We provide evidences that Rack1 and Src regulate P-gp activity by modulating caveolin-1 (Cav1) phosphorylation. Importantly, Rack1 acts as a signaling hub and mediates Src binding to P-gp, thereby facilitating the phosphorylation of Cav1 by Src and abolishing the inhibitory effect of Cav1 on P-gp. Taken together, our results demonstrate the pivotal roles of Rack1 and Src in modulating P-gp activity in drug-resistant cells. Our findings also provide novel insights into the mechanism regulating P-gp transport activity. Rack1 may represent a new target for the development of effective therapies for reversing drug resistance.
Collapse
Affiliation(s)
- Yanling Fan
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
| | - Weiyao Si
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
| | - Wei Ji
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
| | - Zhiyong Wang
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
| | - Zicong Gao
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
| | - Ran Tian
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
| | - Weijie Song
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
| | - He Zhang
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
| | - Ruifang Niu
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China. .,Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China. .,Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China. .,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China.
| | - Fei Zhang
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China. .,Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China. .,Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China. .,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China.
| |
Collapse
|
24
|
Tome ME, Jarvis CK, Schaefer CP, Jacobs LM, Herndon JM, Hunn KC, Arkwright NB, Kellohen KL, Mierau PC, Davis TP. Acute pain alters P-glycoprotein-containing protein complexes in rat cerebral microvessels: Implications for P-glycoprotein trafficking. J Cereb Blood Flow Metab 2018; 38:2209-2222. [PMID: 30346224 PMCID: PMC6282220 DOI: 10.1177/0271678x18803623] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
P-glycoprotein (PgP) is the major drug efflux pump in human cerebral microvessels. PgP prevents pathogens, toxins and therapeutic drugs from entering the CNS. Understanding the molecular regulation of PgP activity will suggest novel mechanisms to improve CNS drug delivery. Previously, we found that during peripheral inflammatory pain (PIP) (3 h after λ carrageenan injection in the rat paw), PgP traffics to the cortical microvessel endothelial cell plasma membrane concomitant with increased PgP activity. In the current study, we measured the changes in composition of PgP-containing protein complexes after PIP in rat microvessel isolates. We found that a portion of the PgP is contained in a multi-protein complex that also contains the caveolar proteins CAV1, SDPR, PTRF and PRKCDBP. With PIP, total CAV1 bound to PgP was unchanged; however, phosphorylated CAV1 (Y14P-CAV1) in the complex increased. There were few PgP/CAV1 complexes relative to total PgP and CAV1 in the microvessels suggesting CAV1 bound to PgP is unlikely to affect total PgP activity. However, both PgP and CAV1 trafficked away from the nucleus in response to PIP. These data suggest that P-CAV1 bound to PgP potentially regulates PgP trafficking and contributes to the acute PgP activity increase after a PIP stimulus.
Collapse
Affiliation(s)
- Margaret E Tome
- Department of Pharmacology, University of Arizona, Tucson, AZ, USA
| | - Chelsea K Jarvis
- Department of Pharmacology, University of Arizona, Tucson, AZ, USA
| | | | - Leigh M Jacobs
- Department of Pharmacology, University of Arizona, Tucson, AZ, USA
| | - Joseph M Herndon
- Department of Pharmacology, University of Arizona, Tucson, AZ, USA
| | - Kristen C Hunn
- Department of Pharmacology, University of Arizona, Tucson, AZ, USA
| | | | | | - Peyton C Mierau
- Department of Pharmacology, University of Arizona, Tucson, AZ, USA
| | - Thomas P Davis
- Department of Pharmacology, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
25
|
Stavrovskaya AA, Rybalkina EY. Recent Advances in the Studies of Molecular Mechanisms Regulating Multidrug Resistance in Cancer Cells. BIOCHEMISTRY (MOSCOW) 2018; 83:779-786. [PMID: 30200862 DOI: 10.1134/s0006297918070015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Here we present new approaches to better understanding multidrug resistance (MDR) development in cancer cells, such as identification of components of a complex process of MDR evolution. Recent advances in the studies of MDR are discussed: 1) chemotherapy agents might be involved in the selection of cancer stem cells resulting in the elevated drug resistance and enhanced tumorigenicity; 2) cell-cell interactions have a great effect on the MDR emergence and evolution; 3) mechanotransduction is an important signaling mechanism in cell-cell interactions; 4) proteins of the ABC transporter family which are often involved in MDR might be transferred between cells via microvesicles (epigenetic MDR regulation); 5) proteins providing cell-to-cell transfer of functional P-glycoprotein (MDR1 protein) via microvesicles have been investigated; 6) P-glycoprotein may serve to regulate apoptosis, as well as transcription and translation of target genes/proteins. Although proving once again that MDR is a complex multi-faceted process, these data open new approaches to overcoming it.
Collapse
Affiliation(s)
- A A Stavrovskaya
- Blokhin Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, 115478, Russia.
| | - E Yu Rybalkina
- Blokhin Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, 115478, Russia
| |
Collapse
|
26
|
Magnesium Reduces Blood-Brain Barrier Permeability and Regulates Amyloid-β Transcytosis. Mol Neurobiol 2018; 55:7118-7131. [PMID: 29383689 DOI: 10.1007/s12035-018-0896-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 01/09/2018] [Indexed: 10/18/2022]
Abstract
Poor Mg status is a risk factor for Alzheimer's disease (AD), and the underlying mechanisms remain elusive. Here, we provided the first evidence that elevated Mg levels significantly reduced the blood-brain barrier (BBB) permeability and regulated its function in vitro. Transient receptor potential melastatin 7 (TRPM7) and magnesium transporter subtype 1 (MagT1) were two major cellular receptors mediating entry of extracellular Mg2+ into the cells. Elevated Mg levels also induced an accelerated clearance of amyloid-β peptide (Aβ) from the brain to the blood side via BBB transcytosis through low-density lipoprotein receptor-related protein (LRP) and phosphatidylinositol binding clathrin assembly protein (PICALM), while reduced the influx of Aβ from the blood to the brain side involving receptor for advanced glycation end products (RAGE) and caveolae. Mg enhanced BBB barrier properties and overall expression of LRP1 and PICALM whereas reduced that of RAGE and caveolin-1. Apical-to-basolateral and vice versa steady-state Aβ flux achieved an equilibrium of 18 and 0.27 fmol/min/cm2, respectively, about 30 min after the initial addition of physiological levels of free Aβ. Knockdown of caveolin-1 or disruption of caveolae membrane microdomains reduced RAGE-mediated influx significantly, but not LRP1-mediated efflux of Aβ. Stimulating endothelial cells with vascular endothelial growth factor (VEGF) enhanced caveolin-1 phosphorylation and RAGE expression. Co-immunoprecipitation demonstrated that RAGE, but not LRP1, was physically associated with caveolin-1. Thus, Mg can reduce BBB permeability and promote BBB clearance of Aβ from the brain by increasing the expression of LRP1 and PICALM while reducing the level of RAGE and caveolin-1.
Collapse
|
27
|
Pokharel D, Roseblade A, Oenarto V, Lu JF, Bebawy M. Proteins regulating the intercellular transfer and function of P-glycoprotein in multidrug-resistant cancer. Ecancermedicalscience 2017; 11:768. [PMID: 29062386 PMCID: PMC5636210 DOI: 10.3332/ecancer.2017.768] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Indexed: 12/15/2022] Open
Abstract
Chemotherapy is an essential part of anticancer treatment. However, the overexpression of P-glycoprotein (P-gp) and the subsequent emergence of multidrug resistance (MDR) hampers successful treatment clinically. P-gp is a multidrug efflux transporter that functions to protect cells from xenobiotics by exporting them out from the plasma membrane to the extracellular space. P-gp inhibitors have been developed in an attempt to overcome P-gp-mediated MDR; however, lack of specificity and dose limiting toxicity have limited their effectiveness clinically. Recent studies report on accessory proteins that either directly or indirectly regulate P-gp expression and function and which are necessary for the establishment of the functional phenotype in cancer cells. This review discusses the role of these proteins, some of which have been recently proposed to comprise an interactive complex, and discusses their contribution towards MDR. We also discuss the role of other pathways and proteins in regulating P-gp expression in cells. The potential for these proteins as novel therapeutic targets provides new opportunities to circumvent MDR clinically.
Collapse
Affiliation(s)
- Deep Pokharel
- Discipline of Pharmacy, The Graduate School of Health, The University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Ariane Roseblade
- Discipline of Pharmacy, The Graduate School of Health, The University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Vici Oenarto
- Discipline of Pharmacy, The Graduate School of Health, The University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Jamie F Lu
- Discipline of Pharmacy, The Graduate School of Health, The University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Mary Bebawy
- Discipline of Pharmacy, The Graduate School of Health, The University of Technology Sydney, Sydney, NSW 2007, Australia.,Laboratory of Cancer Cell Biology and Therapeutics, The University of Technology Sydney, Sydney, NSW 2007, Australia
| |
Collapse
|
28
|
Tomono T, Yano K, Ogihara T. Snail-Induced Epithelial-to-Mesenchymal Transition Enhances P-gp-Mediated Multidrug Resistance in HCC827 Cells. J Pharm Sci 2017; 106:2642-2649. [DOI: 10.1016/j.xphs.2017.03.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 03/03/2017] [Accepted: 03/09/2017] [Indexed: 12/12/2022]
|
29
|
Gomes MJ, Kennedy PJ, Martins S, Sarmento B. Delivery of siRNA silencing P-gp in peptide-functionalized nanoparticles causes efflux modulation at the blood–brain barrier. Nanomedicine (Lond) 2017; 12:1385-1399. [DOI: 10.2217/nnm-2017-0023] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: Explore the use of transferrin-receptor peptide-functionalized nanoparticles (NPs) targeting blood–brain barrier (BBB) as siRNA carriers to silence P-glycoprotein (P-gp). Materials & methods: Permeability experiments were assessed through a developed BBB cell-based model; P-gp mRNA expression was evaluated in vitro; rhodamine 123 permeability was assessed after cell monolayer treatment with siRNA NPs. Results: Beyond their ability to improve siRNA permeability through the BBB by twofold, 96-h post-transfection, functionalized polymeric NPs successfully reduced P-gp mRNA expression up to 52%, compared with nonfunctionalized systems. Subsequently, the permeability of rhodamine 123 through the human BBB model increased up to 27%. Conclusion: Developed BBB-targeted NPs induced P-gp downregulation and consequent increase on P-gp substrate permeability, revealing their ability to modulate drug efflux at the BBB.
Collapse
Affiliation(s)
- Maria João Gomes
- i3S, Instituto de Investigação e Inovação em Saúde, Rua Alfredo Allen, 208, 4200–135 Porto, Portugal
- INEB, Instituto de Engenharia Biomédica, Biocarrier Group, Rua Alfredo Allen, 208, 4200–135 Porto, Portugal
- ICBAS, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050–313 Porto, Portugal
| | - Patrick J Kennedy
- i3S, Instituto de Investigação e Inovação em Saúde, Rua Alfredo Allen, 208, 4200–135 Porto, Portugal
- INEB, Instituto de Engenharia Biomédica, Biocarrier Group, Rua Alfredo Allen, 208, 4200–135 Porto, Portugal
- ICBAS, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050–313 Porto, Portugal
- IPATIMUP, Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Rua Alfredo Allen, 208, 4200–393 Porto, Portugal
| | - Susana Martins
- Department of Physics, Chemistry & Pharmacy, University of Southern Denmark, Campusvej 55, DK-5230 Odense, Denmark
| | - Bruno Sarmento
- i3S, Instituto de Investigação e Inovação em Saúde, Rua Alfredo Allen, 208, 4200–135 Porto, Portugal
- INEB, Instituto de Engenharia Biomédica, Biocarrier Group, Rua Alfredo Allen, 208, 4200–135 Porto, Portugal
- CESPU, Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, Rua Central de Gandra, 1317, 4585–116 Gandra, Portugal
| |
Collapse
|
30
|
Reversal effects of local anesthetics on P-glycoprotein-mediated cancer multidrug resistance. Anticancer Drugs 2017; 28:243-249. [DOI: 10.1097/cad.0000000000000455] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
31
|
Malhaire H, Gimel JC, Roger E, Benoît JP, Lagarce F. How to design the surface of peptide-loaded nanoparticles for efficient oral bioavailability? Adv Drug Deliv Rev 2016; 106:320-336. [PMID: 27058155 DOI: 10.1016/j.addr.2016.03.011] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Revised: 03/17/2016] [Accepted: 03/28/2016] [Indexed: 01/22/2023]
Abstract
The oral administration of proteins is a current challenge to be faced in the field of therapeutics. There is currently much interest in nanocarriers since they can enhance oral bioavailability. For lack of a clear definition, the key characteristics of nanoparticles have been highlighted. Specific surface area is one of these characteristics and represents a huge source of energy that can be used to control the biological fate of the carrier. The review discusses nanocarrier stability, mucus interaction and absorption through the intestinal epithelium. The protein corona, which has raised interest over the last decade, is also discussed. The universal ideal surface is a myth and over-coated carriers are not a solution either. Besides, common excipients can be useful on several targets. The suitable design should rather take into account the composition, structure and behavior of unmodified nanomaterials.
Collapse
|
32
|
Tome ME, Herndon JM, Schaefer CP, Jacobs LM, Zhang Y, Jarvis CK, Davis TP. P-glycoprotein traffics from the nucleus to the plasma membrane in rat brain endothelium during inflammatory pain. J Cereb Blood Flow Metab 2016; 36:1913-1928. [PMID: 27466374 PMCID: PMC5094312 DOI: 10.1177/0271678x16661728] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 06/28/2016] [Indexed: 02/01/2023]
Abstract
P-glycoprotein (PgP), a drug efflux pump in blood-brain barrier endothelial cells, is a major clinical obstacle for effective central nervous system drug delivery. Identifying PgP regulatory pathways that can be exploited clinically is critical for improving central nervous system drug delivery. We previously found that PgP activity increases in rat brain microvessels concomitant with decreased central nervous system drug delivery in response to acute peripheral inflammatory pain. In the current study, we tested the hypothesis that PgP traffics to the luminal plasma membrane of the microvessel endothelial cells from intracellular stores during peripheral inflammatory pain. Using immunofluorescence microscopy, we detected PgP in endothelial cell nuclei and in the luminal plasma membrane in control animals. Following peripheral inflammatory pain, luminal PgP staining increased while staining in the nucleus decreased. Biochemical analysis of nuclear PgP content confirmed our visual observations. Peripheral inflammatory pain also increased endothelial cell luminal staining of polymerase 1 and transcript release factor/cavin1 and serum deprivation response protein/cavin2, two caveolar scaffold proteins, without changing caveolin1 or protein kinase C delta binding protein/cavin3 location. Our data (a) indicate that PgP traffics from stores in the nucleus to the endothelial cell luminal membrane in response to peripheral inflammatory pain; (b) provide an explanation for our previous observation that peripheral inflammatory pain inhibits central nervous system drug uptake; and (c) suggest a novel regulatory mechanism for PgP activity in rat brain.
Collapse
Affiliation(s)
- Margaret E Tome
- Department of Pharmacology, University of Arizona, Tucson, USA
| | | | | | - Leigh M Jacobs
- Department of Pharmacology, University of Arizona, Tucson, USA
| | - Yifeng Zhang
- Department of Pharmacology, University of Arizona, Tucson, USA
| | | | - Thomas P Davis
- Department of Pharmacology, University of Arizona, Tucson, USA
| |
Collapse
|
33
|
Wu M, Li T, Chen L, Peng S, Liao W, Bai R, Zhao X, Yang H, Wu C, Zeng H, Liu Y. Essential oils from Inula japonica and Angelicae dahuricae enhance sensitivity of MCF-7/ADR breast cancer cells to doxorubicin via multiple mechanisms. JOURNAL OF ETHNOPHARMACOLOGY 2016; 180:18-27. [PMID: 26795076 DOI: 10.1016/j.jep.2016.01.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2015] [Revised: 11/26/2015] [Accepted: 01/18/2016] [Indexed: 06/05/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Angelicae dahurica (Hoffm.) Benth. & Hook.f.ex Franch. & Sav combined with Pueraria and Gastrodia elata Bl. combined with Inula japonica Thunb. are widely used in herb-pairs of traditional chinese medicine. Previous studies have shown that Angelicae dahuricae essential oil (ADO) enhanced puerarin internalization into ABCB1-overexpressed Caco-2 cells. These findings suggest the possibility that essential oils may enhance the absorption via certain mechanisms related to ABCB1 and reverse multidrug resistance (MDR). AIM OF THE STUDY ADO and essential oils from Inula japonica (IJO) may reverse ABCB1-mediated MDR, but this ability has not been investigated in detail in the well-established cancer cell lines. In this study, the underlying molecular mechanisms were further investigated to examine how IJO and ADO reverse MDR in the resistant human breast cancer cell line of MCF-7/ADR. Also this work may help uncover the conceivable compatibility mechanisms of above herb-pairs involved in ABCB1. MATERIALS AND METHODS The MDR human breast cancer MCF-7/ADR cells were treated with IJO, its sesquiterpene component isoalantolactone (ISO) or ADOat non- cytotoxic concentrations. The MDR ability was examined by measuring the sensitivity to doxorubicin (DOX), DOX accumulation and efflux, ABCB1 ATPase activity, ABCB1 expression, membrane fluidity, and stability and localization of lipid rafts and caveolae. Finally, the molecular modeling was performed to postulate how ISO interacts with ABCB1. RESULTS Treating MCF-7/ADR cells with IJ oil, ISO or AD oil reversed MDR 2- to 3-fold, without affecting the sensitivity of the non-MDR parental cell line. Mechanistic studies showed that these oils down-regulated mRNA and protein expression of ABCB1, and reduced the stability of lipid rafts in the cell membrane, which has previously been shown to reduce ABCB1-mediated transport. On the other hand, IJO, ISO and ADO did not inhibit ABCB1 ATPase activity, and fluorescence polarization experiments showed that low concentrations of the oils did not appear to alter membrane fluidity, unlike some MDR-reversing agents, ISO showed a higher docking score than verapamil but lower than dofequidar and tariquidar. CONCLUSIONS Our results suggest that IJO, ISO and ADO could reverse MDR by down-regulating ABCB1 expression and reducing lipid raft stability. These findings may be useful for developing safer and effective MDR reversal agents and also help find out the compatibility mechanisms.
Collapse
Affiliation(s)
- Min Wu
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, PR China; Department of Pharmacy, Chengdu Medical College, Chengdu 610081, Sichuan, PR China.
| | - Tingting Li
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, PR China.
| | - Lilan Chen
- Department of Pharmacy, Chengdu Medical College, Chengdu 610081, Sichuan, PR China.
| | - Sugang Peng
- Department of Pharmacy, Chengdu Medical College, Chengdu 610081, Sichuan, PR China.
| | - Wei Liao
- Department of Pharmacy, Chengdu Medical College, Chengdu 610081, Sichuan, PR China.
| | - Ruolan Bai
- Department of Pharmacy, Chengdu Medical College, Chengdu 610081, Sichuan, PR China.
| | - Xue Zhao
- Department of Pharmacy, Chengdu Medical College, Chengdu 610081, Sichuan, PR China.
| | - Hong Yang
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, PR China; Center for Information in Biomedicine, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China.
| | - Chunhui Wu
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, PR China; Center for Information in Biomedicine, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China.
| | - Hongjuan Zeng
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, PR China; Center for Information in Biomedicine, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China.
| | - Yiyao Liu
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, PR China; Center for Information in Biomedicine, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China.
| |
Collapse
|
34
|
Lee CY, Lai TY, Tsai MK, Ou-Yang P, Tsai CY, Wu SW, Hsu LC, Chen JS. The influence of a caveolin-1 mutant on the function of P-glycoprotein. Sci Rep 2016; 6:20486. [PMID: 26843476 PMCID: PMC4740904 DOI: 10.1038/srep20486] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 01/05/2016] [Indexed: 12/02/2022] Open
Abstract
The genetic heterogeneity in cancer cells has an increased chance in the acquisition of new mutant such as drug-resistant phenotype in cancer cells. The phenotype of drug resistance in cancer cells could be evaluated by the number or function of drug transporters on cell membranes, which would lead to decreased intracellular anti-cancer drugs concentration. Caveolae are flask-shaped invaginations on cell membrane that function in membrane trafficking, endocytosis, and as a compartment where receptors and signaling proteins are concentrated. Caveolin-1 (CAV1) is the principal structural protein of caveolae and closely correlates with multidrug resistance in cancer cells. In a systematic study of the ubiquitin-modified proteome, lysine 176 of CAV1 was identified as a potential post-translational modification site for ubiquitination. In this article, we identified a mutation at lysine 176 to arginine (K176R) on CAV1 would interfere with the biogenesis of caveolae and broke the interaction of CAV1 with P-glycoprotein. Functional assays further revealed that K176R mutant of CAV1 in cancer cells increased the transport activity of P-glycoprotein and decreased the killing ability of anti-cancer drugs in non-small-cell lung cancer cell lines.
Collapse
Affiliation(s)
- Chih-Yuan Lee
- Department of Surgery, No. 7 Chung San South Road, Taipei 10002, Taiwan.,Institute of Molecular Medicine, College of Medicine, National Taiwan University, No. 1 Jen-Ai Road, Sec. 1, Taipei 10002, Taiwan
| | - Ting-Yu Lai
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, No. 1 Jen-Ai Road, Sec. 1, Taipei 10002, Taiwan
| | - Meng-Kun Tsai
- Department of Surgery, No. 7 Chung San South Road, Taipei 10002, Taiwan
| | - Pu Ou-Yang
- Department of Medical Research, National Taiwan University Hospital, No. 7 Chung San South Road, Taipei 10002, Taiwan
| | - Ching-Yi Tsai
- Department of Medical Research, National Taiwan University Hospital, No. 7 Chung San South Road, Taipei 10002, Taiwan
| | - Shu-Wei Wu
- Department of Surgery, No. 7 Chung San South Road, Taipei 10002, Taiwan
| | - Li-Chung Hsu
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, No. 1 Jen-Ai Road, Sec. 1, Taipei 10002, Taiwan
| | - Jin-Shing Chen
- Department of Surgery, No. 7 Chung San South Road, Taipei 10002, Taiwan
| |
Collapse
|
35
|
Zhang S, Cao W, Yue M, Zheng N, Hu T, Yang S, Dong Z, Lu S, Mo S. Caveolin-1 affects tumor drug resistance in esophageal squamous cell carcinoma by regulating expressions of P-gp and MRP1. Tumour Biol 2016; 37:9189-96. [PMID: 26768616 DOI: 10.1007/s13277-015-4778-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Accepted: 12/29/2015] [Indexed: 12/15/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is the most common cancer in China, and multidrug resistance (MDR) remains one of the biggest problems in ESCC chemotherapy. In this study, we aimed to investigate the mechanism of Caveolin-1, an integral membrane protein, on regulating ESCC MDR. First, immunohistochemistry was used to check the protein expression of Caveolin-1, MDR-related protein of P-glycoprotein (P-gp), and multidrug resistance protein 1 (MRP1) in 84 pathologically characterized ESCC tissues, matched adjacent tumor, and adjacent normal-looking tissues. The results showed that Caveolin-1 expression level was elevated in ESCC tissues than that of matched adjacent tumor and adjacent normal-looking tissues (P < 0.05), and the expression of Caveolin-1 has close correlation with P-gp and MRP1 during tumor genesis of ESCC (P = 0.034, P = 0.009, respectively). Then, Caveolin-1 overexpression and knockdown were used to investigate its effect on expressions of P-gp and MRP1 in ESCC cell line Ec9706. The messenger RNA (mRNA) and protein expression levels of P-gp and MRP1 were checked by real-time quantitative reverse transcription-PCR (qRT-PCR) and Western blot (WB). The results showed that Caveolin-1 overexpression significantly promotes the mRNA and protein expression of MRP1 (P < 0.05), while almost has no effect on the mRNA and protein expression of P-gp (P > 0.05); Cavoelin-1 knockdown inhibits the mRNA and protein expressions of both P-gp and MRP1 (P < 0.05). The similar result was found in another ESCC cell line Eca109. So, it is concluded that Caveolin-1 affects ESCC MDR by regulating the expressions of P-gp and MRP1; therefore, it can be taken as a significant marker and target in tumor therapy.
Collapse
Affiliation(s)
- Song Zhang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Wenbo Cao
- Department of Basic Science of Oncology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, 450001, China
| | - Mingjin Yue
- Henan Tianxing Education and Media Company, Limited, Zhengzhou, Henan Province, 450002, China
| | - Naigang Zheng
- Department of Basic Science of Oncology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, 450001, China
| | - Tao Hu
- Department of Basic Science of Oncology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, 450001, China
| | - Shengli Yang
- Department of Basic Science of Oncology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, 450001, China
- Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, Henan Province, 450001, China
| | - Ziming Dong
- Department of Basic Science of Oncology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, 450001, China
- Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, Henan Province, 450001, China
| | - Shixin Lu
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Saijun Mo
- Department of Basic Science of Oncology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, 450001, China.
- Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, Henan Province, 450001, China.
| |
Collapse
|
36
|
Qosa H, Miller DS, Pasinelli P, Trotti D. Regulation of ABC efflux transporters at blood-brain barrier in health and neurological disorders. Brain Res 2015; 1628:298-316. [PMID: 26187753 PMCID: PMC4681613 DOI: 10.1016/j.brainres.2015.07.005] [Citation(s) in RCA: 165] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2015] [Revised: 06/28/2015] [Accepted: 07/02/2015] [Indexed: 01/16/2023]
Abstract
The strength of the blood-brain barrier (BBB) in providing protection to the central nervous system from exposure to circulating chemicals is maintained by tight junctions between endothelial cells and by a broad range of transporter proteins that regulate exchange between CNS and blood. The most important transporters that restrict the permeability of large number of toxins as well as therapeutic agents are the ABC transporters. Among them, P-gp, BCRP, MRP1 and MRP2 are the utmost studied. These efflux transporters are neuroprotective, limiting the brain entry of neurotoxins; however, they could also restrict the entry of many therapeutics and contribute to CNS pharmacoresistance. Characterization of several regulatory pathways that govern expression and activity of ABC efflux transporters in the endothelium of brain capillaries have led to an emerging consensus that these processes are complex and contain several cellular and molecular elements. Alterations in ABC efflux transporters expression and/or activity occur in several neurological diseases. Here, we review the signaling pathways that regulate expression and transport activity of P-gp, BCRP, MRP1 and MRP2 as well as how their expression/activity changes in neurological diseases. This article is part of a Special Issue entitled SI: Neuroprotection.
Collapse
Affiliation(s)
- Hisham Qosa
- Weinberg Unit for ALS Research, Farber Institute for Neuroscience, Department of Neuroscience, Thomas Jefferson University, 900 Walnut street, Philadelphia, PA 19107, USA.
| | - David S Miller
- Laboratory of Signal Transduction, NIH/NIEHS, Research Triangle Park, NC 27709, USA
| | - Piera Pasinelli
- Weinberg Unit for ALS Research, Farber Institute for Neuroscience, Department of Neuroscience, Thomas Jefferson University, 900 Walnut street, Philadelphia, PA 19107, USA
| | - Davide Trotti
- Weinberg Unit for ALS Research, Farber Institute for Neuroscience, Department of Neuroscience, Thomas Jefferson University, 900 Walnut street, Philadelphia, PA 19107, USA.
| |
Collapse
|
37
|
Evidence for P-Glycoprotein Involvement in Cell Volume Regulation Using Coulter Sizing in Flow Cytometry. Int J Mol Sci 2015; 16:14318-37. [PMID: 26114386 PMCID: PMC4519844 DOI: 10.3390/ijms160714318] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 05/06/2015] [Accepted: 05/14/2015] [Indexed: 12/13/2022] Open
Abstract
The regulation of cell volume is an essential function that is coupled to a variety of physiological processes such as receptor recycling, excitability and contraction, cell proliferation, migration, and programmed cell death. Under stress, cells undergo emergency swelling and respond to such a phenomenon with a regulatory volume decrease (RVD) where they release cellular ions, and other osmolytes as well as a concomitant loss of water. The link between P-glycoprotein, a transmembrane transporter, and cell volume regulation is controversial, and changes in cells volume are measured using microscopy or electrophysiology. For instance, by using the patch-clamp method, our team demonstrated that chloride currents activated in the RVD were more intense and rapid in a breast cancer cell line overexpressing the P-glycoprotein (P-gp). The Cell Lab Quanta SC is a flow cytometry system that simultaneously measures electronic volume, side scatter and three fluorescent colors; altogether this provides unsurpassed population resolution and accurate cell counting. Therefore, here we propose a novel method to follow cellular volume. By using the Coulter-type channel of the cytometer Cell Lab Quanta SC MPL (multi-platform loading), we demonstrated a role for the P-gp during different osmotic treatments, but also a differential activity of the P-gp through the cell cycle. Altogether, our data strongly suggests a role of P-gp in cell volume regulation.
Collapse
|
38
|
Tome ME, Schaefer CP, Jacobs LM, Zhang Y, Herndon JM, Matty FO, Davis TP. Identification of P-glycoprotein co-fractionating proteins and specific binding partners in rat brain microvessels. J Neurochem 2015; 134:200-10. [PMID: 25832806 DOI: 10.1111/jnc.13106] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 02/26/2015] [Accepted: 03/23/2015] [Indexed: 11/27/2022]
Abstract
Drug delivery to the brain for the treatment of pathologies with a CNS component is a significant clinical challenge. P-glycoprotein (PgP), a drug efflux pump in the endothelial cell membrane, is a major factor in preventing therapeutics from crossing the blood-brain barrier (BBB). Identifying PgP regulatory mechanisms is key to developing agents to modulate PgP activity. Previously, we found that PgP trafficking was altered concomitant with increased PgP activity and disassembly of high molecular weight PgP-containing complexes during acute peripheral inflammatory pain. These data suggest that PgP activity is post-translationally regulated at the BBB. The goal of the current study was to identify proteins that co-localize with PgP in rat brain microvessel endothelial cell membrane microdomains and use the data to suggest potential regulatory mechanisms. Using new density gradients of microvessel homogenates, we identified two unique pools (1,2) of PgP in membrane fractions. Caveolar constituents, caveolin1, cavin1, and cavin2, co-localized with PgP in these fractions indicating the two pools contained caveolae. A chaperone (Hsc71), protein disulfide isomerase and endosomal/lysosomal sorting proteins (Rab5, Rab11a) also co-fractionated with PgP in the gradients. These data suggest signaling pathways with a potential role in post-translational regulation of PgP activity at the BBB.
Collapse
Affiliation(s)
- Margaret E Tome
- Department of Medical Pharmacology, University of Arizona, Tucson, Arizona, USA
| | - Charles P Schaefer
- Department of Medical Pharmacology, University of Arizona, Tucson, Arizona, USA
| | - Leigh M Jacobs
- Department of Medical Pharmacology, University of Arizona, Tucson, Arizona, USA
| | - Yifeng Zhang
- Department of Medical Pharmacology, University of Arizona, Tucson, Arizona, USA
| | - Joseph M Herndon
- Department of Medical Pharmacology, University of Arizona, Tucson, Arizona, USA
| | - Fabian O Matty
- Department of Medical Pharmacology, University of Arizona, Tucson, Arizona, USA
| | - Thomas P Davis
- Department of Medical Pharmacology, University of Arizona, Tucson, Arizona, USA
| |
Collapse
|
39
|
Sanon VP, Sawaki D, Mjaatvedt CH, Jourdan‐Le Saux C. Myocardial Tissue Caveolae. Compr Physiol 2015; 5:871-86. [DOI: 10.1002/cphy.c140050] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
40
|
Zhang Y, Qu X, Teng Y, Li Z, Xu L, Liu J, Ma Y, Fan Y, Li C, Liu S, Wang Z, Hu X, Zhang J, Liu Y. Cbl-b inhibits P-gp transporter function by preventing its translocation into caveolae in multiple drug-resistant gastric and breast cancers. Oncotarget 2015; 6:6737-48. [PMID: 25788263 PMCID: PMC4466646 DOI: 10.18632/oncotarget.3253] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Accepted: 01/29/2015] [Indexed: 12/25/2022] Open
Abstract
The transport function of P-glycoprotein (P-gp) requires its efficient localization to caveolae, a subset of lipid rafts, and disruption of caveolae suppresses P-gp transport function. However, the regulatory molecules involved in the translocation of P-gp into caveolae remain unknown. In the present study, we showed that c-Src dependent Caveolin-1 phosphorylation promoted the translocation of P-gp into caveolae, resulting in multidrug resistance in adriamycin resistant gastric cancer SGC7901/Adr and breast cancer MCF-7/Adr cells. In a negative feedback loop, the translocation of Cbl-b from the nucleus to the cytoplasm prevented the localization of P-gp to caveolae resulting in the reversal of MDR through the ubiquitination and degradation of c-Src. Clinical data showed a significant positive relationship between Cbl-b expression and survival in P-gp positive breast cancer patients who received anthracycline-based chemotherapy. Our findings identified a new regulatory mechanism of P-gp transport function in multiple drug-resistant gastric and breast cancers.
Collapse
Affiliation(s)
- Ye Zhang
- 1 Department of Medical Oncology, the First Hospital of China Medical University, Shenyang 110001, China
| | - Xiujuan Qu
- 1 Department of Medical Oncology, the First Hospital of China Medical University, Shenyang 110001, China
| | - Yuee Teng
- 1 Department of Medical Oncology, the First Hospital of China Medical University, Shenyang 110001, China
| | - Zhi Li
- 1 Department of Medical Oncology, the First Hospital of China Medical University, Shenyang 110001, China
| | - Ling Xu
- 1 Department of Medical Oncology, the First Hospital of China Medical University, Shenyang 110001, China
| | - Jing Liu
- 1 Department of Medical Oncology, the First Hospital of China Medical University, Shenyang 110001, China
| | - Yanju Ma
- 1 Department of Medical Oncology, the First Hospital of China Medical University, Shenyang 110001, China
| | - Yibo Fan
- 1 Department of Medical Oncology, the First Hospital of China Medical University, Shenyang 110001, China
| | - Ce Li
- 1 Department of Medical Oncology, the First Hospital of China Medical University, Shenyang 110001, China
| | - Shizhou Liu
- 1 Department of Medical Oncology, the First Hospital of China Medical University, Shenyang 110001, China
| | - Zhenning Wang
- 2 Department of Surgical Oncology and General Surgery, the First Hospital of China Medical University, Shenyang 110001, China
| | - Xuejun Hu
- 3 Department of Medical Respiratory, the First Hospital of China Medical University, Shenyang 110001, China
| | - Jingdong Zhang
- 1 Department of Medical Oncology, the First Hospital of China Medical University, Shenyang 110001, China
| | - Yunpeng Liu
- 1 Department of Medical Oncology, the First Hospital of China Medical University, Shenyang 110001, China
| |
Collapse
|
41
|
Miller DS. Regulation of ABC transporters blood-brain barrier: the good, the bad, and the ugly. Adv Cancer Res 2015; 125:43-70. [PMID: 25640266 DOI: 10.1016/bs.acr.2014.10.002] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The brain capillary endothelial cells that constitute the blood-brain barrier express multiple ABC transport proteins on the luminal, blood-facing, plasma membrane. These transporters function as ATP-driven efflux pumps for xenobiotics and endogenous metabolites. High expression of these ABC transporters at the barrier is a major obstacle to the delivery of therapeutics, including chemotherapeutics, to the CNS. Here, I review the signals that alter ABC transporter expression and transport function with an emphasis on P-glycoprotein, Mrp2, and breast cancer resistance protein (BCRP), the efflux transporters for which we have the most detailed picture of regulation. Recent work shows that transporter protein expression can be upregulated in response to inflammatory and oxidative stress, therapeutic drugs, diet, and persistent environmental pollutants; as a consequence, drug delivery to the brain is reduced (potentially bad and ugly). In contrast, basal transport activity of P-glycoprotein and BCRP can be reduced through complex signaling pathways that involve events in and on the brain capillary endothelial cells. Targeting these signaling events provides opportunities to rapidly and reversibly increase brain accumulation of drugs that are substrates for the transporters (potentially good). The clinical usefulness of targeting signaling to reduce efflux transporter activity and improve drug delivery to the CNS remains to be established.
Collapse
Affiliation(s)
- David S Miller
- Laboratory of Toxicology and Pharmacology, National Institute of Environmental Health Sciences, National Institutes of Health, North Carolina, USA.
| |
Collapse
|
42
|
P-glycoprotein trafficking as a therapeutic target to optimize CNS drug delivery. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2014; 71:25-44. [PMID: 25307213 DOI: 10.1016/bs.apha.2014.06.009] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The primary function of the blood-brain barrier (BBB)/neurovascular unit is to protect the central nervous system (CNS) from potentially harmful xenobiotic substances and maintain CNS homeostasis. Restricted access to the CNS is maintained via a combination of tight junction proteins as well as a variety of efflux and influx transporters that limits the transcellular and paracellular movement of solutes. Of the transporters identified at the BBB, P-glycoprotein (P-gp) has emerged as the transporter that is the greatest obstacle to effective CNS drug delivery. In this chapter, we provide data to support intracellular protein trafficking of P-gp within cerebral capillary microvessels as a potential target for improved drug delivery. We show that pain-induced changes in P-gp trafficking are associated with changes in P-gp's association with caveolin-1, a key scaffolding/trafficking protein that colocalizes with P-gp at the luminal membrane of brain microvessels. Changes in colocalization with the phosphorylated and nonphosphorylated forms of caveolin-1, by pain, are accompanied by dynamic changes in the distribution, relocalization, and activation of P-gp "pools" between microvascular endothelial cell subcellular compartments. Since redox-sensitive processes may be involved in signaling disassembly of higher-order structures of P-gp, we feel that manipulating redox signaling, via specific protein targeting at the BBB, may protect disulfide bond integrity of P-gp reservoirs and control trafficking to the membrane surface, providing improved CNS drug delivery. The advantage of therapeutic drug "relocalization" of a protein is that the physiological impact can be modified, temporarily or long term, despite pathology-induced changes in gene transcription.
Collapse
|
43
|
ABC Transporter Regulation by Signaling at the Blood–Brain Barrier. PHARMACOLOGY OF THE BLOOD BRAIN BARRIER: TARGETING CNS DISORDERS 2014; 71:1-24. [DOI: 10.1016/bs.apha.2014.06.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
44
|
Tachikawa M, Uchida Y, Ohtsuki S, Terasaki T. Recent Progress in Blood–Brain Barrier and Blood–CSF Barrier Transport Research: Pharmaceutical Relevance for Drug Delivery to the Brain. DRUG DELIVERY TO THE BRAIN 2014. [DOI: 10.1007/978-1-4614-9105-7_2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
45
|
Beloqui A, Solins MN, Gascn AR, del Pozo-Rodrguez A, des Rieux A, Prat V. Mechanism of transport of saquinavir-loaded nanostructured lipid carriers across the intestinal barrier. J Control Release 2013; 166:115-23. [DOI: 10.1016/j.jconrel.2012.12.021] [Citation(s) in RCA: 159] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2012] [Revised: 12/11/2012] [Accepted: 12/16/2012] [Indexed: 11/30/2022]
|
46
|
McCaffrey G, Davis TP. Physiology and pathophysiology of the blood-brain barrier: P-glycoprotein and occludin trafficking as therapeutic targets to optimize central nervous system drug delivery. J Investig Med 2012; 60:1131-40. [PMID: 23138008 PMCID: PMC3851303 DOI: 10.2310/jim.0b013e318276de79] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The blood-brain barrier (BBB) is a physical and metabolic barrier that separates the central nervous system from the peripheral circulation. Central nervous system drug delivery across the BBB is challenging, primarily because of the physical restriction of paracellular diffusion between the endothelial cells that comprise the microvessels of the BBB and the activity of efflux transporters that quickly expel back into the capillary lumen a wide variety of xenobiotics. Therapeutic manipulation of protein trafficking is emerging as a novel means of modulating protein function, and in this minireview, the targeting of the trafficking of 2 key BBB proteins, P-glycoprotein and occludin, is presented as a novel, reversible means of optimizing central nervous system drug delivery.
Collapse
Affiliation(s)
- Gwen McCaffrey
- Department of Medical Pharmacology, University of Arizona College of Medicine, Tucson, AZ 85745, USA.
| | | |
Collapse
|
47
|
McCaffrey G, Staatz WD, Sanchez-Covarrubias L, Finch JD, Demarco K, Laracuente ML, Ronaldson PT, Davis TP. P-glycoprotein trafficking at the blood-brain barrier altered by peripheral inflammatory hyperalgesia. J Neurochem 2012; 122:962-75. [PMID: 22716933 DOI: 10.1111/j.1471-4159.2012.07831.x] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
P-glycoprotein (ABCB1/MDR1, EC 3.6.3.44), the major efflux transporter at the blood-brain barrier (BBB), is a formidable obstacle to CNS pharmacotherapy. Understanding the mechanism(s) for increased P-glycoprotein activity at the BBB during peripheral inflammatory pain is critical in the development of novel strategies to overcome the significant decreases in CNS analgesic drug delivery. In this study, we employed the λ-carrageenan pain model (using female Sprague-Dawley rats), combined with confocal microscopy and subcellular fractionation of cerebral microvessels, to determine if increased P-glycoprotein function, following the onset of peripheral inflammatory pain, is associated with a change in P-glycoprotein trafficking which leads to pain-induced effects on analgesic drug delivery. Injection of λ-carrageenan into the rat hind paw induced a localized, inflammatory pain (hyperalgesia) and simultaneously, at the BBB, a rapid change in colocalization of P-glycoprotein with caveolin-1, a key scaffolding/trafficking protein. Subcellular fractionation of isolated cerebral microvessels revealed that the bulk of P-glycoprotein constitutively traffics to membrane domains containing high molecular weight, disulfide-bonded P-glycoprotein-containing structures that cofractionate with membrane domains enriched with monomeric and high molecular weight, disulfide-bonded, caveolin-1-containing structures. Peripheral inflammatory pain promoted a dynamic redistribution between membrane domains of P-glycoprotein and caveolin-1. Disassembly of high molecular weight P-glycoprotein-containing structures within microvascular endothelial luminal membrane domains was accompanied by an increase in ATPase activity, suggesting a potential for functionally active P-glycoprotein. These results are the first observation that peripheral inflammatory pain leads to specific structural changes in P-glycoprotein responsible for controlling analgesic drug delivery to the CNS.
Collapse
Affiliation(s)
- Gwen McCaffrey
- Department of Medical Pharmacology, University of Arizona College of Medicine, Tucson, AZ 85745, USA.
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Minami T, Kijima T, Otani Y, Kohmo S, Takahashi R, Nagatomo I, Hirata H, Suzuki M, Inoue K, Takeda Y, Kida H, Tachibana I, Kumanogoh A. HER2 as therapeutic target for overcoming ATP-binding cassette transporter-mediated chemoresistance in small cell lung cancer. Mol Cancer Ther 2012; 11:830-41. [PMID: 22389470 DOI: 10.1158/1535-7163.mct-11-0884] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Small cell lung cancer (SCLC) easily acquires multidrug resistance after successful initial therapy. Overexpression of ATP-binding cassette (ABC) transporters is important for the multidrug resistance. Among them, ABCB1 and ABCG2 are known to be upregulated in chemoresistant SCLC cells. We found that human epidermal growth factor receptor 2 (HER2) expressions are also upregulated in chemoresistant SBC-3/ETP, SBC-3/SN-38, and SBC-3/CDDP cells, compared with chemosensitive SBC-3 cells. Lapatinib, a tyrosine kinase inhibitor of HER2, could not suppress proliferation of these HER2-positive SCLC cells alone but successfully restored chemosensitivity to etoposide and SN-38 with a clinically applicable concentration. The reversal effect of lapatinib was thought to be caused by inhibition of drug efflux pump functions of ABC transporters, although lapatinib itself has been reported to be a substrate for them. Moreover, knocking down of HER2 by an short interfering RNA weakened the effect of lapatinib on ABCB1, indicating the involvement of HER2 in the inhibitory mechanisms. Notably, we showed that caveolin-1 and Src play key roles in modulating ABCB1 function via HER2 inactivation. In SBC-3/ETP cells, dephosphorylation of HER2 by lapatinib activates Src and successively leads to increased caveolin-1 phosphorylation. Through this process, caveolin-1 dissociates from HER2 and strengthens association with ABCB1, and finally impairs the pump functions. Furthermore, we showed that treatment by lapatinib in combination with etoposide or irinotecan significantly suppresses the growth of subcutaneous SBC-3/ETP and SBC-3/SN-38 tumors in mice, respectively. Collectively, these results indicate that combination therapy with lapatinib and cytotoxic agents could conquer ABC transporter-mediated chemoresistance especially in HER2-positive SCLC.
Collapse
Affiliation(s)
- Toshiyuki Minami
- Department of Respiratory Medicine, Allergy and Rheumatic Diseases, Osaka University Graduate School of Medicine, Osaka University, Osaka, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Brun E, Carrière M, Mabondzo A. In vitro evidence of dysregulation of blood-brain barrier function after acute and repeated/long-term exposure to TiO(2) nanoparticles. Biomaterials 2011; 33:886-96. [PMID: 22027597 DOI: 10.1016/j.biomaterials.2011.10.025] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2011] [Accepted: 10/10/2011] [Indexed: 12/13/2022]
Abstract
The effects of titanium dioxide nanoparticles (TiO(2) NPs) on blood-brain barrier (BBB) function are unknown. Here, we report such evidence of adverse effects after in vitro exposure of a rat primary cell-based BBB model to NPs. BBB integrity was studied by measuring the flux of sucrose through the monolayer. P-glycoprotein (P-gp) activity was assessed by measuring the passage of vinblastine. Transcription profiles of P-gp and other ABC transporters as well as of cytokines were investigated by real-time PCR. Electron microscopy and particle-induced X-ray emission measurements were performed. We compared several exposure modalities, from early to chronic, mimicking a brain-to-blood transport or a systemic contamination. In the first case, BBB integrity was preserved, but P-gp activity of endothelial cells (BECs) was reduced. In the second case, BBB integrity and P-gp function were impaired from 5 μg/mL for 24 h and expression of tight junction proteins and efflux transporters was modulated. An inflammatory response had repercussions on ABC transporter expression of glial cells. We demonstrate that NPs accumulated in BECs and crossed the cell monolayer. These findings suggest that there is an immunoregulatory loop between inflammatory components, BECs and glial cells in the dysfunction of the BBB during exposure to TiO(2) NPs.
Collapse
Affiliation(s)
- Emilie Brun
- CEA, Direction des Sciences du Vivant, iBiTec-S, Service de Pharmacologie et d'Immunoanalyse, F91191 Gif-sur-Yvette, France
| | | | | |
Collapse
|
50
|
Mo S, Yang S, Cui Z. New glimpses of caveolin-1 functions in embryonic development and human diseases. FRONTIERS IN BIOLOGY 2011; 6:367. [PMID: 32215005 PMCID: PMC7089126 DOI: 10.1007/s11515-011-1132-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/14/2010] [Accepted: 12/30/2010] [Indexed: 11/17/2022]
Abstract
Caveolin-1 (Cav-1) isoforms, including Cav-1α and Cav-1β, were identified as integral membrane proteins and the major components of caveolae. Cav-1 proteins are highly conserved during evolution from {itCaenorhabditis elegans} to human and are capable of interacting with many signaling molecules through their caveolin scaffolding domains to regulate the activities of multiple signaling pathways. Thus, Cav-1 plays crucial roles in the regulation of cellular proliferation, differentiation and apoptosis in a cell-specific and contextual manner. In addition, Cav-1 is essential for embryonic development of vertebrates owing to its regulation of BMP, Wnt, TGF-β and other key signaling molecules. Moreover, Cav-1 is mainly expressed in terminally differentiated cells and its abnormal expression is often associated with human diseases, such as tumor progression, cardiovascular diseases, fibrosis, lung regeneration, and diseases related to virus. In this review, we will further discuss the potential of Cav-1 as a target for disease therapy and multiple drug resistance.
Collapse
Affiliation(s)
- Saijun Mo
- Department of Basic Oncology, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001 China
| | - Shengli Yang
- Department of Basic Oncology, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001 China
| | - Zongbin Cui
- Key Laboratory of Biodiversity and Conservation of Aquatic Organism, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072 China
| |
Collapse
|