1
|
Okyar A, Ozturk Civelek D, Akyel YK, Surme S, Pala Kara Z, Kavakli IH. The role of the circadian timing system on drug metabolism and detoxification: an update. Expert Opin Drug Metab Toxicol 2024; 20:503-517. [PMID: 38753451 DOI: 10.1080/17425255.2024.2356167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 05/13/2024] [Indexed: 05/18/2024]
Abstract
INTRODUCTION The 24-hour variations in drug absorption, distribution, metabolism, and elimination, collectively known as pharmacokinetics, are fundamentally influenced by rhythmic physiological processes regulated by the molecular clock. Recent advances have elucidated the intricacies of the circadian timing system and the molecular interplay between biological clocks, enzymes and transporters in preclinical level. AREA COVERED Circadian rhythm of the drug metabolizing enzymes and carrier efflux functions possess a major role for drug metabolism and detoxification. The efflux and metabolism function of intestines and liver seems important. The investigations revealed that the ABC and SLC transporter families, along with cytochrome p-450 systems in the intestine, liver, and kidney, play a dominant role in the circadian detoxification of drugs. Additionally, the circadian control of efflux by the blood-brain barrier is also discussed. EXPERT OPINION The influence of the circadian timing system on drug pharmacokinetics significantly impacts the efficacy, adverse effects, and toxicity profiles of various drugs. Moreover, the emergence of sex-related circadian changes in the metabolism and detoxification processes has underscored the importance of considering gender-specific differences in drug tolerability and pharmacology. A better understanding of coupling between central clock and circadian metabolism/transport contributes to the development of more rational drug utilization and the implementation of chronotherapy applications.
Collapse
Affiliation(s)
- Alper Okyar
- Department of Pharmacology, Istanbul University Faculty of Pharmacy, Istanbul, Turkiye
| | - Dilek Ozturk Civelek
- Department of Pharmacology, Faculty of Pharmacy, Bezmialem Vakif University, Istanbul, Turkiye
| | - Yasemin Kubra Akyel
- Department of Medical Pharmacology, School of Medicine, Istanbul Medipol University, Istanbul, Turkey
- Department of Biochemistry and Biophysics, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Saliha Surme
- Molecular Biology and Genetics, Koc University, Istanbul, Türkiye
- Chemical and Biological Engineering, Koc University, Istanbul, Türkiye
| | - Zeliha Pala Kara
- Department of Pharmacology, Istanbul University Faculty of Pharmacy, Istanbul, Turkiye
| | - I Halil Kavakli
- Molecular Biology and Genetics, Koc University, Istanbul, Türkiye
- Chemical and Biological Engineering, Koc University, Istanbul, Türkiye
| |
Collapse
|
2
|
Huttunen KM. Improving drug delivery to the brain: the prodrug approach. Expert Opin Drug Deliv 2024; 21:683-693. [PMID: 38738934 DOI: 10.1080/17425247.2024.2355180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 05/10/2024] [Indexed: 05/14/2024]
Abstract
INTRODUCTION The prodrug approach has been thought to be a simple solution to improve brain drug delivery for decades. Nevertheless, it still comes as a surprise that there is relatively little success in the field. The best example anti-parkinsonian drug levodopa has been serendipitously discovered to be a transporter-utilizing brain-delivered prodrug rather than a rationally developed one. AREAS COVERED The lack of success can mainly be explained by the insufficient understanding of the role of membrane proteins that can facilitate drug delivery at dynamic barriers, such as the blood-brain barrier (BBB), but also by the sparse knowledge of prodrug bioconverting enzymes in the brain. This review summarizes the current status of the prodrug attempts that have been developed in the past to improve brain drug delivery. EXPERT OPINION With the expandingly improved analytical and computational technologies, it is anticipated that enhanced brain drug delivery will be eventually achieved for most of the central nervous system (CNS) acting drugs. However, this requires that carrier-mediated (pro)drug delivery methods are implemented in the very early phases of the drug development processes and not as a last step to survive a problematic investigational drug candidate.
Collapse
Affiliation(s)
- Kristiina M Huttunen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
3
|
Avilez-Avilez JJ, Medina-Flores MF, Gómez-Gonzalez B. Sleep loss impairs blood-brain barrier function: Cellular and molecular mechanisms. VITAMINS AND HORMONES 2024; 126:77-96. [PMID: 39029977 DOI: 10.1016/bs.vh.2024.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/21/2024]
Abstract
Sleep is a physiological process that preserves the integrity of the neuro-immune-endocrine network to maintain homeostasis. Sleep regulates the production and secretion of hormones, neurotransmitters, cytokines and other inflammatory mediators, both at the central nervous system (CNS) and at the periphery. Sleep promotes the removal of potentially toxic metabolites out of the brain through specialized systems such as the glymphatic system, as well as the expression of specific transporters in the blood-brain barrier. The blood-brain barrier maintains CNS homeostasis by selectively transporting metabolic substrates and nutrients into the brain, by regulating the efflux of metabolic waste products, and maintaining bidirectional communication between the periphery and the CNS. All those processes are disrupted during sleep loss. Brain endothelial cells express the blood-brain barrier phenotype, which arises after cell-to-cell interactions with mural cells, like pericytes, and after the release of soluble factors by astroglial endfeet. Astroglia, pericytes and brain endothelial cells respond differently to sleep loss; evidence has shown that sleep loss induces a chronic low-grade inflammatory state at the CNS, which is associated with blood-brain barrier dysfunction. In animal models, blood-brain barrier dysfunction is characterized by increased blood-brain barrier permeability, decreased tight junction protein expression and pericyte detachment from the capillary wall. Blood-brain barrier dysfunction may promote defects in brain clearance of potentially neurotoxic metabolites and byproducts of neural physiology, which may eventually contribute to neurodegenerative diseases. This chapter aims to describe the cellular and molecular mechanisms by which sleep loss modifies the function of the blood-brain barrier.
Collapse
Affiliation(s)
- Jessica Janeth Avilez-Avilez
- Graduate Program in Experimental Biology, Universidad Autónoma Metropolitana, Mexico City, Mexico; Area of Neurosciences, Department of Biology of Reproduction, Universidad Autónoma Metropolitana, Mexico City, Mexico
| | - María Fernanda Medina-Flores
- Graduate Program in Experimental Biology, Universidad Autónoma Metropolitana, Mexico City, Mexico; Area of Neurosciences, Department of Biology of Reproduction, Universidad Autónoma Metropolitana, Mexico City, Mexico
| | - Beatriz Gómez-Gonzalez
- Area of Neurosciences, Department of Biology of Reproduction, Universidad Autónoma Metropolitana, Mexico City, Mexico.
| |
Collapse
|
4
|
Abstract
The blood-brain barrier (BBB) is a critical interface separating the central nervous system from the peripheral circulation, ensuring brain homeostasis and function. Recent research has unveiled a profound connection between the BBB and circadian rhythms, the endogenous oscillations synchronizing biological processes with the 24-hour light-dark cycle. This review explores the significance of circadian rhythms in the context of BBB functions, with an emphasis on substrate passage through the BBB. Our discussion includes efflux transporters and the molecular timing mechanisms that regulate their activities. A significant focus of this review is the potential implications of chronotherapy, leveraging our knowledge of circadian rhythms for improving drug delivery to the brain. Understanding the temporal changes in BBB can lead to optimized timing of drug administration, to enhance therapeutic efficacy for neurological disorders while reducing side effects. By elucidating the interplay between circadian rhythms and drug transport across the BBB, this review offers insights into innovative therapeutic interventions.
Collapse
Affiliation(s)
- Mari Kim
- Cell Biology Department, Emory University, Atlanta, GA, USA
| | | | | |
Collapse
|
5
|
Fong H, Zhou B, Feng H, Luo C, Bai B, Zhang J, Wang Y. Recapitulation of Structure-Function-Regulation of Blood-Brain Barrier under (Patho)Physiological Conditions. Cells 2024; 13:260. [PMID: 38334652 PMCID: PMC10854731 DOI: 10.3390/cells13030260] [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: 10/30/2023] [Revised: 01/12/2024] [Accepted: 01/19/2024] [Indexed: 02/10/2024] Open
Abstract
The blood-brain barrier (BBB) is a remarkable and intricate barrier that controls the exchange of molecules between the bloodstream and the brain. Its role in maintaining the stability of the central nervous system cannot be overstated. Over the years, advancements in neuroscience and technology have enabled us to delve into the cellular and molecular components of the BBB, as well as its regulation. Yet, there is a scarcity of comprehensive reviews that follow a logical framework of structure-function-regulation, particularly focusing on the nuances of BBB regulation under both normal and pathological conditions. This review sets out to address this gap by taking a historical perspective on the discovery of the BBB and highlighting the major observations that led to its recognition as a distinct brain barrier. It explores the intricate cellular elements contributing to the formation of the BBB, including endothelial cells, pericytes, astrocytes, and neurons, emphasizing their collective role in upholding the integrity and functionality of the BBB. Furthermore, the review delves into the dynamic regulation of the BBB in physiological states, encompassing neural, humoral, and auto-regulatory mechanisms. By shedding light on these regulatory processes, a deeper understanding of the BBB's response to various physiological cues emerges. This review also investigates the disruption of the BBB integrity under diverse pathological conditions, such as ischemia, infection, and toxin exposure. It elucidates the underlying mechanisms that contribute to BBB dysfunction and explores potential therapeutic strategies that aim to restore the BBB integrity and function. Overall, this recapitulation provides valuable insights into the structure, functions, and regulation of the BBB. By integrating historical perspectives, cellular elements, regulatory mechanisms, and pathological implications, this review contributes to a more comprehensive understanding of the BBB and paves the way for future research and therapeutic interventions.
Collapse
Affiliation(s)
- Hin Fong
- Faculty of Medicine, International School, Jinan University, Guangzhou 510632, China; (H.F.); (C.L.); (B.B.)
| | - Botao Zhou
- Department of Physiology, Basic Medical and Public Health School, Jinan University, Guangzhou 510632, China;
| | - Haixiao Feng
- Gies College of Business, University of Illinois Urbana-Champaign, Urbana-Champaign, IL 61801, USA;
| | - Chuoying Luo
- Faculty of Medicine, International School, Jinan University, Guangzhou 510632, China; (H.F.); (C.L.); (B.B.)
| | - Boren Bai
- Faculty of Medicine, International School, Jinan University, Guangzhou 510632, China; (H.F.); (C.L.); (B.B.)
| | - John Zhang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92350, USA;
- Department of Neurosurgery, Loma Linda University, Loma Linda, CA 92350, USA
| | - Yuechun Wang
- Department of Physiology, Basic Medical and Public Health School, Jinan University, Guangzhou 510632, China;
| |
Collapse
|
6
|
Salmina AB, Alexandrova OP, Averchuk AS, Korsakova SA, Saridis MR, Illarioshkin SN, Yurchenko SO. Current progress and challenges in the development of brain tissue models: How to grow up the changeable brain in vitro? J Tissue Eng 2024; 15:20417314241235527. [PMID: 38516227 PMCID: PMC10956167 DOI: 10.1177/20417314241235527] [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: 09/25/2023] [Accepted: 02/12/2024] [Indexed: 03/23/2024] Open
Abstract
In vitro modeling of brain tissue is a promising but not yet resolved problem in modern neurobiology and neuropharmacology. Complexity of the brain structure and diversity of cell-to-cell communication in (patho)physiological conditions make this task almost unachievable. However, establishment of novel in vitro brain models would ultimately lead to better understanding of development-associated or experience-driven brain plasticity, designing efficient approaches to restore aberrant brain functioning. The main goal of this review is to summarize the available data on methodological approaches that are currently in use, and to identify the most prospective trends in development of neurovascular unit, blood-brain barrier, blood-cerebrospinal fluid barrier, and neurogenic niche in vitro models. The manuscript focuses on the regulation of adult neurogenesis, cerebral microcirculation and fluids dynamics that should be reproduced in the in vitro 4D models to mimic brain development and its alterations in brain pathology. We discuss approaches that are critical for studying brain plasticity, deciphering the individual person-specific trajectory of brain development and aging, and testing new drug candidates in the in vitro models.
Collapse
Affiliation(s)
- Alla B Salmina
- Brain Science Institute, Research Center of Neurology, Moscow, Russia
- Bauman Moscow State Technical University, Moscow, Russia
| | - Olga P Alexandrova
- Brain Science Institute, Research Center of Neurology, Moscow, Russia
- Bauman Moscow State Technical University, Moscow, Russia
| | - Anton S Averchuk
- Brain Science Institute, Research Center of Neurology, Moscow, Russia
- Bauman Moscow State Technical University, Moscow, Russia
| | | | | | | | | |
Collapse
|
7
|
Järvinen J, Montaser AB, Adla SK, Leppänen J, Lehtonen M, Vellonen KS, Laitinen T, Jalkanen A, Elmquist WF, Timonen J, Huttunen KM, Rautio J. Altering distribution profile of palbociclib by its prodrugs. Eur J Pharm Sci 2024; 192:106637. [PMID: 37967656 DOI: 10.1016/j.ejps.2023.106637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 11/10/2023] [Accepted: 11/11/2023] [Indexed: 11/17/2023]
Abstract
Palbociclib, a cyclin-dependent kinase (CDK) 4/6 inhibitor, is currently used clinically for treating hormone receptor-positive and human epidermal growth factor receptor 2 negative breast cancer. Additionally, it has the potential to be utilized in the treatment of various tumors, including malignant glioblastoma. Previous research has indicated that palbociclib is a substrate for two efflux transporters, P-glycoprotein (P-gp; MDR1) and breast cancer-resistant protein (BCRP), which restrict the brain exposure of palbociclib. In the present study, our objective was to alter the brain distribution pattern of palbociclib by creating and assessing two novel prodrugs through in vitro, in situ, and in vivo evaluations. To this end, we synthesized two prodrugs of palbociclib by attaching it to the tyrosine promoiety at the para- (PD1) and meta-(PD2) position via a carbamate bond. We hypothesized that the prodrugs could bypass efflux transporter-mediated drug resistance by leveraging the l-type amino acid transporter (LAT1) to facilitate their transport across the blood-brain barrier (BBB) and into cancer cells, such as glioma cells that express LAT1. The compounds PD1 and PD2 did not show selective binding and had limited inhibitory effects on LAT1 in three cell lines (MCF-7, U87-MG, HEK-hLAT1). However, PD1 and PD2 demonstrated the ability to evade efflux mechanisms, and their in vitro uptake profiles were comparable to that of palbociclib, indicating their potential for effective cellular transport. In in situ and in vivo studies, brain uptake was not significantly improved compared to palbociclib, but the pharmacokinetic profiles showed encouraging enhancements. PD1 exhibited a higher AUCbrain/plasma ratio, suggesting safer dosing, while PD2 showed favorable long-acting pharmacokinetics. Although our prodrug design did not significantly improve palbociclib brain delivery due to the potential size limitation of the prodrugs, the study provides valuable insights for future prodrug development and drug delivery strategies targeting specific transporters.
Collapse
Affiliation(s)
- Juulia Järvinen
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland.
| | - Ahmed B Montaser
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Santosh Kumar Adla
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Jukka Leppänen
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Marko Lehtonen
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Kati-Sisko Vellonen
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Tuomo Laitinen
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Aaro Jalkanen
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - William F Elmquist
- Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, MN, United States
| | - Juri Timonen
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland; Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5E, P.O. Box 56, 00014 Helsinki, Finland
| | - Kristiina M Huttunen
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Jarkko Rautio
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| |
Collapse
|
8
|
Eide PK, Lashkarivand A, Pripp AH, Valnes LM, Hovd M, Ringstad G, Blennow K, Zetterberg H. Mechanisms behind changes of neurodegeneration biomarkers in plasma induced by sleep deprivation. Brain Commun 2023; 5:fcad343. [PMID: 38130841 PMCID: PMC10733810 DOI: 10.1093/braincomms/fcad343] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 11/08/2023] [Accepted: 12/11/2023] [Indexed: 12/23/2023] Open
Abstract
Acute sleep deprivation has been shown to affect cerebrospinal fluid and plasma concentrations of biomarkers associated with neurodegeneration, though the mechanistic underpinnings remain unknown. This study compared individuals who, for one night, were either subject to total sleep deprivation or free sleep, (i) examining plasma concentrations of neurodegeneration biomarkers the morning after sleep deprivation or free sleep and (ii) determining how overnight changes in biomarkers plasma concentrations correlate with indices of meningeal lymphatic and glymphatic clearance functions. Plasma concentrations of amyloid-β 40 and 42, phosphorylated tau peptide 181, glial fibrillary acid protein and neurofilament light were measured longitudinally in subjects who from Day 1 to Day 2 either underwent total sleep deprivation (n = 7) or were allowed free sleep (n = 21). The magnetic resonance imaging contrast agent gadobutrol was injected intrathecally, serving as a cerebrospinal fluid tracer. Population pharmacokinetic model parameters of gadobutrol cerebrospinal fluid-to-blood clearance were utilized as a proxy of meningeal lymphatic clearance capacity and intrathecal contrast-enhanced magnetic resonance imaging as a proxy of glymphatic function. After one night of acute sleep deprivation, the plasma concentrations of amyloid-β 40 and 42 were reduced, but not the ratio, and concentrations of the other biomarkers were unchanged. The overnight change in amyloid-β 40 and 42 plasma concentrations in the sleep group correlated significantly with indices of meningeal lymphatic clearance capacity, while this was not seen for the other neurodegeneration biomarkers. However, overnight change in plasma concentrations of amyloid-β 40 and 42 did not correlate with the glymphatic marker. On the other hand, the overnight change in plasma concentration of phosphorylated tau peptide 181 correlated significantly with the marker of glymphatic function in the sleep deprivation group but not in the sleep group. The present data add to the evidence of the role of sleep and sleep deprivation on plasma neurodegeneration concentrations; however, the various neurodegeneration biomarkers respond differently with different mechanisms behind sleep-induced alterations in amyloid-β and tau plasma concentrations. Clearance capacity of meningeal lymphatics seems more important for sleep-induced changes in amyloid-β 40 and 42 plasma concentrations, while glymphatic function seems most important for change in plasma concentration of phosphorylated tau peptide 181 during sleep deprivation. Altogether, the present data highlight diverse mechanisms behind sleep-induced effects on concentrations of plasma neurodegeneration biomarkers.
Collapse
Affiliation(s)
- Per Kristian Eide
- Department of Neurosurgery, Oslo University Hospital—Rikshospitalet, N-0424 Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, N-0316 Oslo, Norway
| | - Aslan Lashkarivand
- Department of Neurosurgery, Oslo University Hospital—Rikshospitalet, N-0424 Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, N-0316 Oslo, Norway
| | - Are Hugo Pripp
- Oslo Centre of Biostatistics and Epidemiology, Research Support Services, Oslo University Hospital, N-0424 Oslo, Norway
- Faculty of Health Sciences, Oslo Metropolitan University, N-0130 Oslo, Norway
| | - Lars Magnus Valnes
- Department of Neurosurgery, Oslo University Hospital—Rikshospitalet, N-0424 Oslo, Norway
| | - Markus Hovd
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, N-0316 Oslo, Norway
- Department of Transplantation Medicine, Oslo University Hospital, N-0424 Oslo, Norway
| | - Geir Ringstad
- Department of Radiology, Oslo University Hospital—Rikshospitalet, N-0424 Oslo, Norway
- Department of Geriatrics and Internal medicine, Sorlandet Hospital, N-4836 Arendal, Norway
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, S-405 30 Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, S-405 30 Gothenburg, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, S-405 30 Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, S-405 30 Gothenburg, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London WC1E 6BT, UK
- UK Dementia Research Institute at UCL, London WC1E 6BT, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong 999077, China
- Department of Medicine, UW School of Medicine and Public Health, Madison, WI 53726, USA
| |
Collapse
|
9
|
Wevers NR, De Vries HE. Microfluidic models of the neurovascular unit: a translational view. Fluids Barriers CNS 2023; 20:86. [PMID: 38008744 PMCID: PMC10680291 DOI: 10.1186/s12987-023-00490-9] [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: 07/14/2023] [Accepted: 11/15/2023] [Indexed: 11/28/2023] Open
Abstract
The vasculature of the brain consists of specialized endothelial cells that form a blood-brain barrier (BBB). This barrier, in conjunction with supporting cell types, forms the neurovascular unit (NVU). The NVU restricts the passage of certain substances from the bloodstream while selectively permitting essential nutrients and molecules to enter the brain. This protective role is crucial for optimal brain function, but presents a significant obstacle in treating neurological conditions, necessitating chemical modifications or advanced drug delivery methods for most drugs to cross the NVU. A deeper understanding of NVU in health and disease will aid in the identification of new therapeutic targets and drug delivery strategies for improved treatment of neurological disorders.To achieve this goal, we need models that reflect the human BBB and NVU in health and disease. Although animal models of the brain's vasculature have proven valuable, they are often of limited translational relevance due to interspecies differences or inability to faithfully mimic human disease conditions. For this reason, human in vitro models are essential to improve our understanding of the brain's vasculature under healthy and diseased conditions. This review delves into the advancements in in vitro modeling of the BBB and NVU, with a particular focus on microfluidic models. After providing a historical overview of the field, we shift our focus to recent developments, offering insights into the latest achievements and their associated constraints. We briefly examine the importance of chip materials and methods to facilitate fluid flow, emphasizing their critical roles in achieving the necessary throughput for the integration of microfluidic models into routine experimentation. Subsequently, we highlight the recent strides made in enhancing the biological complexity of microfluidic NVU models and propose recommendations for elevating the biological relevance of future iterations.Importantly, the NVU is an intricate structure and it is improbable that any model will fully encompass all its aspects. Fit-for-purpose models offer a valuable compromise between physiological relevance and ease-of-use and hold the future of NVU modeling: as simple as possible, as complex as needed.
Collapse
Affiliation(s)
- Nienke R Wevers
- MIMETAS BV, De Limes 7, Oegstgeest, 2342 DH, The Netherlands.
| | - Helga E De Vries
- Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam Neuroscience - Neuroinfection and Neuroinflammation, De Boelelaan 1117, Amsterdam, the Netherlands
| |
Collapse
|
10
|
Schulz JA, Hartz AMS, Bauer B. ABCB1 and ABCG2 Regulation at the Blood-Brain Barrier: Potential New Targets to Improve Brain Drug Delivery. Pharmacol Rev 2023; 75:815-853. [PMID: 36973040 PMCID: PMC10441638 DOI: 10.1124/pharmrev.120.000025] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 03/10/2023] [Accepted: 03/10/2023] [Indexed: 03/29/2023] Open
Abstract
The drug efflux transporters ABCB1 and ABCG2 at the blood-brain barrier limit the delivery of drugs into the brain. Strategies to overcome ABCB1/ABCG2 have been largely unsuccessful, which poses a tremendous clinical problem to successfully treat central nervous system (CNS) diseases. Understanding basic transporter biology, including intracellular regulation mechanisms that control these transporters, is critical to solving this clinical problem.In this comprehensive review, we summarize current knowledge on signaling pathways that regulate ABCB1/ABCG2 at the blood-brain barrier. In Section I, we give a historical overview on blood-brain barrier research and introduce the role that ABCB1 and ABCG2 play in this context. In Section II, we summarize the most important strategies that have been tested to overcome the ABCB1/ABCG2 efflux system at the blood-brain barrier. In Section III, the main component of this review, we provide detailed information on the signaling pathways that have been identified to control ABCB1/ABCG2 at the blood-brain barrier and their potential clinical relevance. This is followed by Section IV, where we explain the clinical implications of ABCB1/ABCG2 regulation in the context of CNS disease. Lastly, in Section V, we conclude by highlighting examples of how transporter regulation could be targeted for therapeutic purposes in the clinic. SIGNIFICANCE STATEMENT: The ABCB1/ABCG2 drug efflux system at the blood-brain barrier poses a significant problem to successful drug delivery to the brain. The article reviews signaling pathways that regulate blood-brain barrier ABCB1/ABCG2 and could potentially be targeted for therapeutic purposes.
Collapse
Affiliation(s)
- Julia A Schulz
- Department of Pharmaceutical Sciences, College of Pharmacy (J.A.S., B.B.), Sanders-Brown Center on Aging and Department of Pharmacology and Nutritional Sciences, College of Medicine (A.M.S.H.), University of Kentucky, Lexington, Kentucky
| | - Anika M S Hartz
- Department of Pharmaceutical Sciences, College of Pharmacy (J.A.S., B.B.), Sanders-Brown Center on Aging and Department of Pharmacology and Nutritional Sciences, College of Medicine (A.M.S.H.), University of Kentucky, Lexington, Kentucky
| | - Björn Bauer
- Department of Pharmaceutical Sciences, College of Pharmacy (J.A.S., B.B.), Sanders-Brown Center on Aging and Department of Pharmacology and Nutritional Sciences, College of Medicine (A.M.S.H.), University of Kentucky, Lexington, Kentucky
| |
Collapse
|
11
|
Liu H, Barthélemy NR, Ovod V, Bollinger JG, He Y, Chahin SL, Androff B, Bateman RJ, Lucey BP. Acute sleep loss decreases CSF-to-blood clearance of Alzheimer's disease biomarkers. Alzheimers Dement 2023; 19:3055-3064. [PMID: 36695437 PMCID: PMC10366339 DOI: 10.1002/alz.12930] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 11/22/2022] [Accepted: 12/20/2022] [Indexed: 01/26/2023]
Abstract
INTRODUCTION Sleep deprivation increases cerebrospinal fluid (CSF) amyloid beta (Aβ) and tau levels; however, sleep's effect on Aβ and tau in plasma is unknown. METHODS In a cross-over design, CSF Aβ and tau concentrations were measured in five cognitively normal individuals who had blood and CSF collected every 2 hours for 36 hours during sleep-deprived and normal sleep control conditions. RESULTS Aβ40, Aβ42, unphosphorylated tau threonine181 (T181), unphosphorylated tau threonine-217 (T217), and phosphorylated T181 (pT181) concentrations increased ∼35% to 55% in CSF and decreased ∼5% to 15% in plasma during sleep deprivation. CSF/plasma ratios of all Alzheimer's disease (AD) biomarkers increased during sleep deprivation while the CSF/plasma albumin ratio, a measure of blood-CSF barrier permeability, decreased. CSF and plasma Aβ42/40, pT181/T181, and pT181/Aβ42 ratios were stable longitudinally in both groups. DISCUSSION These findings show that sleep loss alters some plasma AD biomarkers by lowering brain clearance mechanisms and needs to be taken into account when interpreting individual plasma AD biomarkers but not ratios.
Collapse
Affiliation(s)
- Haiyan Liu
- Department of Neurology, Washington University School of Medicine, St Louis, MO
| | - Nicolas R. Barthélemy
- Department of Neurology, Washington University School of Medicine, St Louis, MO
- Tracy Family SILQ Center, Washington University School of Medicine, St Louis, MO
| | - Vitaliy Ovod
- Department of Neurology, Washington University School of Medicine, St Louis, MO
- Tracy Family SILQ Center, Washington University School of Medicine, St Louis, MO
| | - James G. Bollinger
- Department of Neurology, Washington University School of Medicine, St Louis, MO
- Tracy Family SILQ Center, Washington University School of Medicine, St Louis, MO
| | - Yingxin He
- Department of Neurology, Washington University School of Medicine, St Louis, MO
- Tracy Family SILQ Center, Washington University School of Medicine, St Louis, MO
| | - Samir L. Chahin
- Department of Neurology, Washington University School of Medicine, St Louis, MO
- Tracy Family SILQ Center, Washington University School of Medicine, St Louis, MO
| | - Brendan Androff
- Department of Neurology, Washington University School of Medicine, St Louis, MO
- Tracy Family SILQ Center, Washington University School of Medicine, St Louis, MO
| | - Randall J. Bateman
- Department of Neurology, Washington University School of Medicine, St Louis, MO
- Tracy Family SILQ Center, Washington University School of Medicine, St Louis, MO
| | - Brendan P. Lucey
- Department of Neurology, Washington University School of Medicine, St Louis, MO
- Tracy Family SILQ Center, Washington University School of Medicine, St Louis, MO
- Center On Biological Rhythms and Sleep, Washington University School of Medicine, St Louis, MO
| |
Collapse
|
12
|
Berezin CT, Bergum N, Torres Lopez GM, Vigh J. Morphine pharmacokinetics and opioid transporter expression at the blood-retina barrier of male and female mice. Front Pharmacol 2023; 14:1206104. [PMID: 37388441 PMCID: PMC10301758 DOI: 10.3389/fphar.2023.1206104] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 06/01/2023] [Indexed: 07/01/2023] Open
Abstract
Opioids are effective analgesics for treating moderate to severe pain, however, their use must be weighed against their dangerous side effects. Investigations into opioid pharmacokinetics provide crucial information regarding both on- and off-target drug effects. Our recent work showed that morphine deposits and accumulates in the mouse retina at higher concentrations than in the brain upon chronic systemic exposure. We also found reduced retinal expression of P-glycoprotein (P-gp), a major opioid extruder at the blood-brain barrier (BBB). Here, we systematically interrogated the expression of three putative opioid transporters at the blood-retina barrier (BRB): P-gp, breast cancer resistance protein (Bcrp) and multidrug resistance protein 2 (Mrp2). Using immunohistochemistry, we found robust expression of P-gp and Bcrp, but not Mrp2, at the inner BRB of the mouse retina. Previous studies have suggested that P-gp expression may be regulated by sex hormones. However, upon acute morphine treatment we found no sex differences in morphine deposition levels in the retina or brain, nor on transporter expression in the retinas of males and females with a high or low estrogen:progesterone ratio. Importantly, we found that P-gp, but not Bcrp, expression significantly correlated with morphine concentration in the retina, suggesting P-gp is the predominant opioid transporter at the BRB. In addition, fluorescence extravasation studies revealed that chronic morphine treatment did not alter the permeability of either the BBB or BRB. Together, these data suggest that reduced P-gp expression mediates retinal morphine accumulation upon systemic delivery, and in turn, potential effects on circadian photoentrainment.
Collapse
Affiliation(s)
- Casey-Tyler Berezin
- Cell and Molecular Biology Graduate Program, Colorado State University, Fort Collins, CO, United States
| | - Nikolas Bergum
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Glenda M. Torres Lopez
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Jozsef Vigh
- Cell and Molecular Biology Graduate Program, Colorado State University, Fort Collins, CO, United States
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| |
Collapse
|
13
|
Skapetze L, Owino S, Lo EH, Arai K, Merrow M, Harrington M. Rhythms in barriers and fluids: Circadian clock regulation in the aging neurovascular unit. Neurobiol Dis 2023; 181:106120. [PMID: 37044366 DOI: 10.1016/j.nbd.2023.106120] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 03/27/2023] [Accepted: 04/07/2023] [Indexed: 04/14/2023] Open
Abstract
The neurovascular unit is where two very distinct physiological systems meet: The central nervous system (CNS) and the blood. The permeability of the barriers separating these systems is regulated by time, including both the 24 h circadian clock and the longer processes of aging. An endogenous circadian rhythm regulates the transport of molecules across the blood-brain barrier and the circulation of the cerebrospinal fluid and the glymphatic system. These fluid dynamics change with time of day, and with age, and especially in the context of neurodegeneration. Factors may differ depending on brain region, as can be highlighted by consideration of circadian regulation of the neurovascular niche in white matter. As an example of a potential target for clinical applications, we highlight chaperone-mediated autophagy as one mechanism at the intersection of circadian dysregulation, aging and neurodegenerative disease. In this review we emphasize key areas for future research.
Collapse
Affiliation(s)
- Lea Skapetze
- Institute of Medical Psychology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Sharon Owino
- Neuroscience Program, Smith College, Northampton, MA 01060, United States of America
| | - Eng H Lo
- Neuroprotection Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ken Arai
- Neuroprotection Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Martha Merrow
- Institute of Medical Psychology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Mary Harrington
- Neuroscience Program, Smith College, Northampton, MA 01060, United States of America.
| |
Collapse
|
14
|
Ogata S, Ito S, Masuda T, Ohtsuki S. Diurnal Changes in Protein Expression at the Blood-Brain Barrier in Mice. Biol Pharm Bull 2022; 45:751-756. [PMID: 35650102 DOI: 10.1248/bpb.b22-00016] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Circadian rhythms influence the transport function of the blood-brain barrier (BBB) and peripheral organs. However, the influence of circadian rhythms on protein expression in the BBB remains to be completely elucidated. Therefore, we aimed to investigate diurnal changes in protein expression in the mouse BBB using quantitative proteomics. Quantitative proteomics showed that the expression of 67, 10, and 20 proteins in the isolated mouse brain capillary fraction changed significantly at zeitgeber time (ZT) 6, 12, and 18, respectively, compared to ZT0. Among them, the levels of 44 proteins were significantly increased at ZT6 and then returned to the same level as ZT0 at ZT12 and ZT18. Gene ontology analysis indicated that the proteins significantly increased at ZT6 were majorly related to translation. The brain capillary endothelial cell-selective proteins sepiapterin reductase and vascular endothelial growth factor receptor 2 showed diurnal variation. In contrast, the expression of ABC transporters, SLC transporters, and receptors associated with receptor-mediated transcytosis, and tight junction proteins did not change within a day. The present findings demonstrated that protein expression related to transport function and physical barrier at the BBB was maintained throughout the day, although the proteins involved in some biological processes exhibited diurnal variation at the BBB.
Collapse
Affiliation(s)
- Seiryo Ogata
- Department of Pharmaceutical Microbiology, Graduate School of Pharmaceutical Sciences, Kumamoto University
| | - Shingo Ito
- Department of Pharmaceutical Microbiology, Graduate School of Pharmaceutical Sciences, Kumamoto University.,Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University
| | - Takeshi Masuda
- Department of Pharmaceutical Microbiology, Graduate School of Pharmaceutical Sciences, Kumamoto University.,Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University
| | - Sumio Ohtsuki
- Department of Pharmaceutical Microbiology, Graduate School of Pharmaceutical Sciences, Kumamoto University.,Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University
| |
Collapse
|
15
|
Huttunen KM, Terasaki T, Urtti A, Montaser AB, Uchida Y. Pharmacoproteomics of Brain Barrier Transporters and Substrate Design for the Brain Targeted Drug Delivery. Pharm Res 2022; 39:1363-1392. [PMID: 35257288 PMCID: PMC9246989 DOI: 10.1007/s11095-022-03193-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 02/08/2022] [Indexed: 12/12/2022]
Abstract
One of the major reasons why central nervous system (CNS)-drug development has been challenging in the past, is the barriers that prevent substances entering from the blood circulation into the brain. These barriers include the blood-brain barrier (BBB), blood-spinal cord barrier (BSCB), blood-cerebrospinal fluid barrier (BCSFB), and blood-arachnoid barrier (BAB), and they differ from each other in their transporter protein expression and function as well as among the species. The quantitative expression profiles of the transporters in the CNS-barriers have been recently revealed, and in this review, it is described how they affect the pharmacokinetics of compounds and how these expression differences can be taken into account in the prediction of brain drug disposition in humans, an approach called pharmacoproteomics. In recent years, also structural biology and computational resources have progressed remarkably, enabling a detailed understanding of the dynamic processes of transporters. Molecular dynamics simulations (MDS) are currently used commonly to reveal the conformational changes of the transporters and to find the interactions between the substrates and the protein during the binding, translocation in the transporter cavity, and release of the substrate on the other side of the membrane. The computational advancements have also aided in the rational design of transporter-utilizing compounds, including prodrugs that can be actively transported without losing potency towards the pharmacological target. In this review, the state-of-art of these approaches will be also discussed to give insights into the transporter-mediated drug delivery to the CNS.
Collapse
Affiliation(s)
- Kristiina M Huttunen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland.
| | - Tetsuya Terasaki
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland.
| | - Arto Urtti
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Ahmed B Montaser
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Yasuo Uchida
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai, 980-8578, Japan
| |
Collapse
|
16
|
Lu D, Wang Z, Wu B. Pharmacokinetics-based Chronotherapy. Curr Drug Metab 2022; 23:2-7. [PMID: 34994324 DOI: 10.2174/1389200223666220106124218] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/06/2021] [Accepted: 12/06/2021] [Indexed: 11/22/2022]
Abstract
Dosing time-dependency of pharmacokinetics (or chronopharmacokinetics) has been long recognized. Studies in recent years have revealed that diurnal rhythmicity in expression of drug-metabolizing enzymes and transporters (DMETs) are key factors determining chronopharmacokinetics. In this article, we briefly summarize current knowledge with respect to circadian mechanisms of DMETs and discuss how rhythmic DMETs are translated to drug chronoeffects. More importantly, we present our perspectives on pharmacokinetics-based chronotherapy.
Collapse
Affiliation(s)
- Danyi Lu
- Institute of Molecular Rhythm and Metabolism, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhigang Wang
- Department of Intensive Care Unit, First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Baojian Wu
- Institute of Molecular Rhythm and Metabolism, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
17
|
Walker WH, Sprowls SA, Bumgarner JR, Liu JA, Meléndez-Fernández OH, Walton JC, Lockman PR, DeVries AC, Nelson RJ. Circadian Influences on Chemotherapy Efficacy in a Mouse Model of Brain Metastases of Breast Cancer. Front Oncol 2021; 11:752331. [PMID: 34956876 PMCID: PMC8695439 DOI: 10.3389/fonc.2021.752331] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 11/19/2021] [Indexed: 11/30/2022] Open
Abstract
Chemotherapy is more effective in the treatment of peripheral tumors than brain metastases, likely reflecting the reduced ability of chemotherapy to cross the blood-brain barrier (BBB) and blood-tumor barrier at efficacious concentrations. Recent studies demonstrate circadian regulation of the BBB. Thus, we predicted that optimally timed chemotherapy would increase anti-tumor efficacy in a model of brain metastases of breast cancer (BMBC). First, we characterized novel daily alterations in BBB permeability to a commonly used chemotherapeutic, 14C-paclitaxel, within BMBC following injections given at four time points across the day. Peak and trough 14C-paclitaxel concentrations within BMBC occurred during the mid-dark phase and at the beginning of the light phase, respectively. Notably, chemotherapy injections during the dark phase increased cell death within BMBC and delayed onset of neurological symptoms relative to injections during the light phase. These data provide strong evidence for the beneficial effects of chrono-chemotherapy for the treatment of BMBC.
Collapse
Affiliation(s)
- William H. Walker
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States
| | - Samuel A. Sprowls
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, WV, United States
| | - Jacob R. Bumgarner
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States
| | - Jennifer A. Liu
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States
| | | | - James C. Walton
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States
| | - Paul R. Lockman
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, WV, United States
- WVU Cancer Institute, West Virginia University, Morgantown, WV, United States
| | - A. Courtney DeVries
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States
- WVU Cancer Institute, West Virginia University, Morgantown, WV, United States
- Department of Medicine, Division of Oncology/Hematology, West Virginia University, Morgantown, WV, United States
| | - Randy J. Nelson
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States
| |
Collapse
|
18
|
Silva S, Bicker J, Falcão A, Fortuna A. Antidepressants and Circadian Rhythm: Exploring Their Bidirectional Interaction for the Treatment of Depression. Pharmaceutics 2021; 13:1975. [PMID: 34834391 PMCID: PMC8624696 DOI: 10.3390/pharmaceutics13111975] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/13/2021] [Accepted: 11/16/2021] [Indexed: 01/07/2023] Open
Abstract
Scientific evidence that circadian rhythms affect pharmacokinetics and pharmacodynamics has highlighted the importance of drug dosing-time. Circadian oscillations alter drug absorption, distribution, metabolism, and excretion (ADME) as well as intracellular signaling systems, target molecules (e.g., receptors, transporters, and enzymes), and gene transcription. Although several antidepressant drugs are clinically available, less than 50% of depressed patients respond to first-line pharmacological treatments. Chronotherapeutic approaches to enhance the effectiveness of antidepressants are not completely known. Even so, experimental results found until this day suggest a positive influence of drug dosing-time on the efficacy of depression therapy. On the other hand, antidepressants have also demonstrated to modulate circadian rhythmicity and sleep-wake cycles. This review aims to evidence the potential of chronotherapy to improve the efficacy and/or safety of antidepressants. It includes pre-clinical and clinical studies that demonstrate the relevance of determining the most appropriate time of administration for antidepressant drugs. In parallel, their positive influence on the resynchronization of disrupted circadian rhythms is also herein discussed. It is expected that this review will promote the investigation of chronotherapy for the treatment of depression, contribute to a better understanding of the relationship between antidepressants and circadian rhythms, and consequently promote the development of new therapeutics.
Collapse
Affiliation(s)
- Soraia Silva
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal; (S.S.); (A.F.); (A.F.)
- CIBIT—Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal
| | - Joana Bicker
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal; (S.S.); (A.F.); (A.F.)
- CIBIT—Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal
| | - Amílcar Falcão
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal; (S.S.); (A.F.); (A.F.)
- CIBIT—Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal
| | - Ana Fortuna
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal; (S.S.); (A.F.); (A.F.)
- CIBIT—Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal
| |
Collapse
|
19
|
Toyohara J, Sakata M, Ishibashi K, Mossel P, Imai M, Wagatsuma K, Tago T, Imabayashi E, Colabufo NA, Luurtsema G, Ishii K. First clinical assessment of [ 18F]MC225, a novel fluorine-18 labelled PET tracer for measuring functional P-glycoprotein at the blood-brain barrier. Ann Nucl Med 2021; 35:1240-1252. [PMID: 34368924 DOI: 10.1007/s12149-021-01666-9] [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: 05/25/2021] [Accepted: 08/02/2021] [Indexed: 12/01/2022]
Abstract
OBJECTIVE 5-(1-(2-[18F]fluoroethoxy))-[3-(6,7-dimethoxy-3,4-dihydro-1H-isoquinolin-2-yl)-propyl]-5,6,7,8-tetrahydronaphthalen ([18F]MC225) is a selective substrate for P-glycoprotein (P-gp), possessing suitable properties for measuring overexpression of P-gp in the brain. This is the first-in-human study to examine safety, radiation dosimetry and P-gp function at the blood-brain barrier (BBB) of [18F]MC225 in healthy subjects. METHODS [18F]MC225 biodistribution and dosimetry were determined in 3 healthy male subjects, using serial 2 h and intermittent 4 and 6 h whole-body PET scans acquired after [18F]MC225 injection. Dynamic [18F]MC225 brain PET (90 min) was obtained in 5 healthy male subjects. Arterial blood was sampled at various time intervals during scanning and the fraction of unchanged [18F]MC225 in plasma was determined. T1-weighted MRI was performed for anatomical coregistration. Total distribution volume (VT) was estimated using 1- and 2-tissue-compartment models (1-TCM and 2-TCM, respectively). VT was also estimated using the Logan graphical method (Logan plot) (t* = 20 min). Surrogate parameters without blood sampling (area-under the curve [AUC] of regional time-activity curves [TACs] and negative slope of calculated TACs) were compared with the VT values. RESULTS No serious adverse events occurred throughout the study period. Although biodistribution implied hepatobiliary excretion, secretion of radioactivity from liver to small intestine through the gallbladder was very slow. Total renal excreted radioactivity recovered during 6 h after injection was < 2%ID. Absorbed dose was the highest in the pancreas (mean ± SD, 203 ± 45 μGy/MBq) followed by the liver (83 ± 11 μGy/MBq). Mean effective dose with and without urination was 17 ± 1 μSv/MBq. [18F]MC225 readily entered the brain, distributing homogeneously in grey matter regions. 2-TCM provided lower Akaike information criterion scores than did 1-TCM. VT estimated by Logan plot was well correlated with that of 2-TCM (r2 > 0.9). AUCs of TACs were positively correlated with VT (2-TCM) values (r2: AUC0-60 min = 0.61, AUC0-30 min = 0.62, AUC30-60 min = 0.59, p < 0.0001). Negative slope of SUV TACs was negatively correlated with VT (2-TCM) values (r2 = 0.53, p < 0.0001). CONCLUSIONS This initial evaluation indicated that [18F]MC225 is a suitable and safe PET tracer for measuring P-gp function at the BBB.
Collapse
Affiliation(s)
- Jun Toyohara
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan.
| | - Muneyuki Sakata
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan
| | - Kenji Ishibashi
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan
| | - Pascalle Mossel
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Masamichi Imai
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan
| | - Kei Wagatsuma
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan
- School of Allied Health Science, Kitasato University, 1-15-1 Kitasato, Sagamihara, Kanagawa, 252-0373, Japan
| | - Tetsuro Tago
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan
| | - Etsuko Imabayashi
- Department of Diagnostic Radiology, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan
| | - Nicola A Colabufo
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari, via Orabona 4, 70125, Bari, Italy
| | - Gert Luurtsema
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Kenji Ishii
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan
| |
Collapse
|
20
|
Tuura RO, Volk C, Callaghan F, Jaramillo V, Huber R. Sleep-related and diurnal effects on brain diffusivity and cerebrospinal fluid flow. Neuroimage 2021; 241:118420. [PMID: 34302966 DOI: 10.1016/j.neuroimage.2021.118420] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 06/25/2021] [Accepted: 07/20/2021] [Indexed: 10/20/2022] Open
Abstract
The question of how waste products are cleared from the brain, and the role which sleep plays in this process, is critical for our understanding of a range of physical and mental illnesses. In rodents, both circadian and sleep-related processes appear to facilitate clearance of waste products. The purpose of this study was to investigate whether overnight changes in diffusivity, brain volumes, and cerebrospinal fluid flow measured with MRI are associated with sleep parameters from overnight high-density sleep EEG, and circadian markers. In healthy adults investigated with MRI before and after sleep EEG, we observed an increase in water diffusivity overnight, which was positively related to the proportion of total sleep time spent in rapid eye movement (REM) sleep, and negatively associated with the fraction of sleep time spent in non rapid eye movement (NREM) sleep. Diffusivity was also associated with the sleep midpoint, a circadian marker. CSF flow increased overnight; this increase was unrelated to sleep or diffusivity measures but was associated with circadian markers. These results provide evidence for both sleep related and diurnal effects on water compartmentalisation within the brain.
Collapse
Affiliation(s)
- Ruth O'Gorman Tuura
- Center for MR Research, University Children's Hospital, University of Zürich, Switzerland; Children's Research Center, University Children's Hospital, University of Zürich, Switzerland.
| | - Carina Volk
- Center for MR Research, University Children's Hospital, University of Zürich, Switzerland; Children's Research Center, University Children's Hospital, University of Zürich, Switzerland
| | - Fraser Callaghan
- Center for MR Research, University Children's Hospital, University of Zürich, Switzerland; Children's Research Center, University Children's Hospital, University of Zürich, Switzerland
| | - Valeria Jaramillo
- Children's Research Center, University Children's Hospital, University of Zürich, Switzerland; Child Development Center and Pediatric Sleep Center, University Children's Hospital, Zürich, Switzerland
| | - Reto Huber
- Children's Research Center, University Children's Hospital, University of Zürich, Switzerland; Child Development Center and Pediatric Sleep Center, University Children's Hospital, Zürich, Switzerland; Department of Child and Adolescent Psychiatry and Psychotherapy, Psychiatric Hospital, University of Zurich, Zurich, Switzerland
| |
Collapse
|
21
|
Semyachkina-Glushkovskaya O, Mamedova A, Vinnik V, Klimova M, Saranceva E, Ageev V, Yu T, Zhu D, Penzel T, Kurths J. Brain Mechanisms of COVID-19-Sleep Disorders. Int J Mol Sci 2021; 22:6917. [PMID: 34203143 PMCID: PMC8268116 DOI: 10.3390/ijms22136917] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/15/2021] [Accepted: 06/17/2021] [Indexed: 02/06/2023] Open
Abstract
2020 and 2021 have been unprecedented years due to the rapid spread of the modified severe acute respiratory syndrome coronavirus around the world. The coronavirus disease 2019 (COVID-19) causes atypical infiltrated pneumonia with many neurological symptoms, and major sleep changes. The exposure of people to stress, such as social confinement and changes in daily routines, is accompanied by various sleep disturbances, known as 'coronasomnia' phenomenon. Sleep disorders induce neuroinflammation, which promotes the blood-brain barrier (BBB) disruption and entry of antigens and inflammatory factors into the brain. Here, we review findings and trends in sleep research in 2020-2021, demonstrating how COVID-19 and sleep disorders can induce BBB leakage via neuroinflammation, which might contribute to the 'coronasomnia' phenomenon. The new studies suggest that the control of sleep hygiene and quality should be incorporated into the rehabilitation of COVID-19 patients. We also discuss perspective strategies for the prevention of COVID-19-related BBB disorders. We demonstrate that sleep might be a novel biomarker of BBB leakage, and the analysis of sleep EEG patterns can be a breakthrough non-invasive technology for diagnosis of the COVID-19-caused BBB disruption.
Collapse
Affiliation(s)
- Oxana Semyachkina-Glushkovskaya
- Institute of Physics, Humboldt University, Newtonstrasse 15, 12489 Berlin, Germany;
- Department of Biology, Saratov State University, Atrakhanskaya Str. 83, 410012 Saratov, Russia; (A.M.); (V.V.); (M.K.); (E.S.); (V.A.)
| | - Aysel Mamedova
- Department of Biology, Saratov State University, Atrakhanskaya Str. 83, 410012 Saratov, Russia; (A.M.); (V.V.); (M.K.); (E.S.); (V.A.)
| | - Valeria Vinnik
- Department of Biology, Saratov State University, Atrakhanskaya Str. 83, 410012 Saratov, Russia; (A.M.); (V.V.); (M.K.); (E.S.); (V.A.)
| | - Maria Klimova
- Department of Biology, Saratov State University, Atrakhanskaya Str. 83, 410012 Saratov, Russia; (A.M.); (V.V.); (M.K.); (E.S.); (V.A.)
| | - Elena Saranceva
- Department of Biology, Saratov State University, Atrakhanskaya Str. 83, 410012 Saratov, Russia; (A.M.); (V.V.); (M.K.); (E.S.); (V.A.)
| | - Vasily Ageev
- Department of Biology, Saratov State University, Atrakhanskaya Str. 83, 410012 Saratov, Russia; (A.M.); (V.V.); (M.K.); (E.S.); (V.A.)
| | - Tingting Yu
- Wuhan National Laboratory for Optoelectronics, Britton Chance Center for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan 430074, China; (T.Y.); (D.Z.)
- MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Dan Zhu
- Wuhan National Laboratory for Optoelectronics, Britton Chance Center for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan 430074, China; (T.Y.); (D.Z.)
- MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Thomas Penzel
- Department of Biology, Saratov State University, Atrakhanskaya Str. 83, 410012 Saratov, Russia; (A.M.); (V.V.); (M.K.); (E.S.); (V.A.)
- Sleep Medicine Center, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Jürgen Kurths
- Institute of Physics, Humboldt University, Newtonstrasse 15, 12489 Berlin, Germany;
- Department of Biology, Saratov State University, Atrakhanskaya Str. 83, 410012 Saratov, Russia; (A.M.); (V.V.); (M.K.); (E.S.); (V.A.)
- Potsdam Institute for Climate Impact Research, Telegrafenberg A31, 14473 Potsdam, Germany
| |
Collapse
|
22
|
Demarez C, De Assis LVM, Krohn M, Ramella N, Schwaninger M, Oster H, Astiz M. The trophoblast clock controls transport across placenta in mice. Development 2021; 148:256558. [PMID: 33913482 DOI: 10.1242/dev.197673] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 03/22/2021] [Indexed: 12/20/2022]
Abstract
In mammals, 24-h rhythms of physiology and behavior are organized by a body-wide network of clock genes and proteins. Despite the well-known function of the adult circadian system, the roles of maternal, fetal and placental clocks during pregnancy are poorly defined. In the mature mouse placenta, the labyrinth zone (LZ) is of fetal origin and key for selective nutrient and waste exchange. Recently, clock gene expression has been detected in LZ and other fetal tissues; however, there is no evidence of a placental function controlled by the LZ clock. Here, we demonstrate that specifically the trophoblast layer of the LZ harbors an already functional clock by late gestation, able to regulate in a circadian manner the expression and activity of the xenobiotic efflux pump, ATP-binding cassette sub-family B member 1 (ABCB1), likely gating the fetal exposure to drugs from the maternal circulation to certain times of the day. As more than 300 endogenous and exogenous compounds are substrates of ABCB1, our results might have implications in choosing the maternal treatment time when aiming either maximal/minimal drug availability to the fetus/mother.
Collapse
Affiliation(s)
- Cécile Demarez
- Institute of Neurobiology, Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck. Marie-Curie-Straße, 23562 Lübeck, Germany
| | | | - Markus Krohn
- Institute for Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck. Marie-Curie-Straße, 23562 Lübeck, Germany
| | - Nahuel Ramella
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Calles 60 y 120, 1900 La Plata, Argentina
| | - Markus Schwaninger
- Institute for Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck. Marie-Curie-Straße, 23562 Lübeck, Germany
| | - Henrik Oster
- Institute of Neurobiology, Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck. Marie-Curie-Straße, 23562 Lübeck, Germany
| | - Mariana Astiz
- Institute of Neurobiology, Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck. Marie-Curie-Straße, 23562 Lübeck, Germany
| |
Collapse
|
23
|
Zhang SL, Lahens NF, Yue Z, Arnold DM, Pakstis PP, Schwarz JE, Sehgal A. A circadian clock regulates efflux by the blood-brain barrier in mice and human cells. Nat Commun 2021; 12:617. [PMID: 33504784 PMCID: PMC7841146 DOI: 10.1038/s41467-020-20795-9] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 12/18/2020] [Indexed: 01/30/2023] Open
Abstract
The blood-brain barrier (BBB) is critical for neural function. We report here circadian regulation of the BBB in mammals. Efflux of xenobiotics by the BBB oscillates in mice, with highest levels during the active phase and lowest during the resting phase. This oscillation is abrogated in circadian clock mutants. To elucidate mechanisms of circadian regulation, we profiled the transcriptome of brain endothelial cells; interestingly, we detected limited circadian regulation of transcription, with no evident oscillations in efflux transporters. We recapitulated the cycling of xenobiotic efflux using a human microvascular endothelial cell line to find that the molecular clock drives cycling of intracellular magnesium through transcriptional regulation of TRPM7, which appears to contribute to the rhythm in efflux. Our findings suggest that considering circadian regulation may be important when therapeutically targeting efflux transporter substrates to the CNS.
Collapse
Affiliation(s)
- Shirley L Zhang
- Chronobiology and Sleep Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
- Howard Hughes Medical Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| | - Nicholas F Lahens
- Institute for Translational Medicine and Therapeutics (ITMAT), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhifeng Yue
- Chronobiology and Sleep Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Howard Hughes Medical Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Denice M Arnold
- Chronobiology and Sleep Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Peter P Pakstis
- Chronobiology and Sleep Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Jessica E Schwarz
- Chronobiology and Sleep Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Amita Sehgal
- Chronobiology and Sleep Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
- Howard Hughes Medical Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
24
|
Segarra M, Aburto MR, Acker-Palmer A. Blood-Brain Barrier Dynamics to Maintain Brain Homeostasis. Trends Neurosci 2021; 44:393-405. [PMID: 33423792 DOI: 10.1016/j.tins.2020.12.002] [Citation(s) in RCA: 152] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 10/03/2020] [Accepted: 12/07/2020] [Indexed: 01/18/2023]
Abstract
The blood-brain barrier (BBB) is a dynamic platform for exchange of substances between the blood and the brain parenchyma, and it is an essential functional gatekeeper for the central nervous system (CNS). While it is widely recognized that BBB disruption is a hallmark of several neurovascular pathologies, an aspect of the BBB that has received somewhat less attention is the dynamic modulation of BBB tightness to maintain brain homeostasis in response to extrinsic environmental factors and physiological changes. In this review, we summarize how BBB integrity adjusts in critical stages along the life span, as well as how BBB permeability can be altered by common stressors derived from nutritional habits, environmental factors and psychological stress.
Collapse
Affiliation(s)
- Marta Segarra
- Neuro and Vascular Guidance, Buchmann Institute for Molecular Life Sciences (BMLS) and Institute of Cell Biology and Neuroscience, Max-von-Laue-Strasse 15, D-60438, Frankfurt am Main, Germany; Cardio-Pulmonary Institute (CPI), Max-von-Laue-Strasse 15, D-60438, Frankfurt am Main, Germany.
| | - Maria R Aburto
- Neuro and Vascular Guidance, Buchmann Institute for Molecular Life Sciences (BMLS) and Institute of Cell Biology and Neuroscience, Max-von-Laue-Strasse 15, D-60438, Frankfurt am Main, Germany
| | - Amparo Acker-Palmer
- Neuro and Vascular Guidance, Buchmann Institute for Molecular Life Sciences (BMLS) and Institute of Cell Biology and Neuroscience, Max-von-Laue-Strasse 15, D-60438, Frankfurt am Main, Germany; Cardio-Pulmonary Institute (CPI), Max-von-Laue-Strasse 15, D-60438, Frankfurt am Main, Germany; Max Planck Institute for Brain Research, Max-von-Laue-Strasse 4, 60438 Frankfurt am Main, Germany.
| |
Collapse
|
25
|
Pácha J, Balounová K, Soták M. Circadian regulation of transporter expression and implications for drug disposition. Expert Opin Drug Metab Toxicol 2020; 17:425-439. [PMID: 33353445 DOI: 10.1080/17425255.2021.1868438] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Introduction: Solute Carrier (SLC) and ATP-binding cassette (ABC) transporters expressed in the intestine, liver, and kidney determine the absorption, distribution, and excretion of drugs. In addition, most molecular and cellular processes show circadian rhythmicity controlled by circadian clocks that leads to diurnal variations in the pharmacokinetics and pharmacodynamics of many drugs and affects their therapeutic efficacy and toxicity.Area covered: This review provides an overview of the current knowledge on the circadian rhythmicity of drug transporters and the molecular mechanisms of their circadian control. Evidence for coupling drug transporters to circadian oscillators and the plausible candidates conveying circadian clock signals to target drug transporters, particularly transcription factors operating as the output of clock genes, is discussed.Expert opinion: The circadian machinery has been demonstrated to interact with the uptake and efflux of various drug transporters. The evidence supports the concept that diurnal changes that affect drug transporters may influence the pharmacokinetics of the drugs. However, more systematic studies are required to better define the timing of pharmacologically important drug transporter regulation and determine tissue- and sex-dependent differences. Finally, the transfer of knowledge based on the results and conclusions obtained primarily from animal models will require careful validation before it is applied to humans.
Collapse
Affiliation(s)
- Jiří Pácha
- Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Kateřina Balounová
- Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic.,Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Matúš Soták
- Department of Molecular and Clinical Medicine, Wallenberg Laboratory, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden.,Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
26
|
Profaci CP, Munji RN, Pulido RS, Daneman R. The blood-brain barrier in health and disease: Important unanswered questions. J Exp Med 2020; 217:151582. [PMID: 32211826 PMCID: PMC7144528 DOI: 10.1084/jem.20190062] [Citation(s) in RCA: 426] [Impact Index Per Article: 85.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 09/21/2019] [Accepted: 11/21/2019] [Indexed: 12/11/2022] Open
Abstract
The blood vessels vascularizing the central nervous system exhibit a series of distinct properties that tightly control the movement of ions, molecules, and cells between the blood and the parenchyma. This "blood-brain barrier" is initiated during angiogenesis via signals from the surrounding neural environment, and its integrity remains vital for homeostasis and neural protection throughout life. Blood-brain barrier dysfunction contributes to pathology in a range of neurological conditions including multiple sclerosis, stroke, and epilepsy, and has also been implicated in neurodegenerative diseases such as Alzheimer's disease. This review will discuss current knowledge and key unanswered questions regarding the blood-brain barrier in health and disease.
Collapse
Affiliation(s)
- Caterina P Profaci
- Department of Neurosciences, University of California, San Diego, San Diego, CA.,Department of Pharmacology, University of California, San Diego, San Diego, CA
| | - Roeben N Munji
- Department of Neurosciences, University of California, San Diego, San Diego, CA.,Department of Pharmacology, University of California, San Diego, San Diego, CA
| | - Robert S Pulido
- Department of Neurosciences, University of California, San Diego, San Diego, CA.,Department of Pharmacology, University of California, San Diego, San Diego, CA
| | - Richard Daneman
- Department of Neurosciences, University of California, San Diego, San Diego, CA.,Department of Pharmacology, University of California, San Diego, San Diego, CA
| |
Collapse
|
27
|
Lu D, Zhao M, Chen M, Wu B. Circadian Clock-Controlled Drug Metabolism: Implications for Chronotherapeutics. Drug Metab Dispos 2020; 48:395-406. [PMID: 32114506 DOI: 10.1124/dmd.120.090472] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 02/18/2020] [Indexed: 12/20/2022] Open
Abstract
Dependence of drug metabolism on dosing time has long been recognized. However, only recently are the underlying mechanisms for circadian drug metabolism being clarified. Diurnal rhythmicity in expression of drug-metabolizing enzymes is believed to be a key factor determining circadian metabolism. Supporting the notion that biological rhythms are generated and maintained by the circadian clock, a number of diurnal enzymes are under the control of the circadian clock. In general, circadian clock genes generate and regulate diurnal rhythmicity in drug-metabolizing enzymes via transcriptional actions on one or two of three cis-elements (i.e., E-box, D-box, and Rev-erb response element or RAR-related orphan receptor response element). Additionally, cycling or clock-controlled nuclear receptors such as hepatocyte nuclear factor 4α and peroxisome proliferator-activated receptor γ are contributors to diurnal enzyme expression. These newly discovered mechanisms for each of the rhythmic enzymes are reviewed in this article. We also discuss how the rhythms of enzymes are translated to circadian pharmacokinetics and drug chronotoxicity, which has direct implications for chronotherapeutics. Our discussion is also extended to two diurnal transporters (P-glycoprotein and multidrug resistance-associated protein 2) that have an important role in drug absorption. Although the experimental evidence is lacking in metabolism-based chronoefficacy, circadian genes (e.g., Rev-erbα) as drug targets are shown to account for diurnal variability in drug efficacy. SIGNIFICANCE STATEMENT: Significant progress has been made in understanding the molecular mechanisms for generation of diurnal rhythmicity in drug-metabolizing enzymes. In this article, we review the newly discovered mechanisms for each of the rhythmic enzymes and discuss how the rhythms of enzymes are translated to circadian pharmacokinetics and drug chronotoxicity, which has direct implications for chronotherapeutics.
Collapse
Affiliation(s)
- Danyi Lu
- Research Center for Biopharmaceutics and Pharmacokinetics, College of Pharmacy, Jinan University, Guangzhou, China (D.L., M.Z., M.C., B.W.) and International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou, China (B.W.)
| | - Mengjing Zhao
- Research Center for Biopharmaceutics and Pharmacokinetics, College of Pharmacy, Jinan University, Guangzhou, China (D.L., M.Z., M.C., B.W.) and International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou, China (B.W.)
| | - Min Chen
- Research Center for Biopharmaceutics and Pharmacokinetics, College of Pharmacy, Jinan University, Guangzhou, China (D.L., M.Z., M.C., B.W.) and International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou, China (B.W.)
| | - Baojian Wu
- Research Center for Biopharmaceutics and Pharmacokinetics, College of Pharmacy, Jinan University, Guangzhou, China (D.L., M.Z., M.C., B.W.) and International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou, China (B.W.)
| |
Collapse
|
28
|
Bicker J, Alves G, Falcão A, Fortuna A. Timing in drug absorption and disposition: The past, present, and future of chronopharmacokinetics. Br J Pharmacol 2020; 177:2215-2239. [PMID: 32056195 DOI: 10.1111/bph.15017] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Revised: 01/05/2020] [Accepted: 01/27/2020] [Indexed: 12/20/2022] Open
Abstract
The importance of drug dosing time in pharmacokinetics, pharmacodynamics, and toxicity is receiving increasing attention from the scientific community. In spite of mounting evidence that circadian oscillations affect drug absorption, distribution, metabolism, and excretion (ADME), there remain many unanswered questions in this field and, occasionally, conflicting experimental results. Such data arise not only from translational difficulties caused by interspecies differences but also from variability in study design and a lack of understanding of how the circadian clock affects physiological factors that strongly influence ADME, namely, the expression and activity of drug transporters. Hence, the main goal of this review is to provide an updated analysis of the role of the circadian rhythm in drug absorption, distribution across blood-tissue barriers, metabolism in hepatic and extra-hepatic tissues, and hepatobiliary and renal excretion. It is expected that the research suggestions proposed here will contribute to a tissue-targeted and time-targeted pharmacotherapy.
Collapse
Affiliation(s)
- Joana Bicker
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal.,CIBIT/ICNAS-Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
| | - Gilberto Alves
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Amílcar Falcão
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal.,CIBIT/ICNAS-Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
| | - Ana Fortuna
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal.,CIBIT/ICNAS-Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
29
|
Loryan I, Hammarlund-Udenaes M, Syvänen S. Brain Distribution of Drugs: Pharmacokinetic Considerations. Handb Exp Pharmacol 2020; 273:121-150. [PMID: 33258066 DOI: 10.1007/164_2020_405] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
It is crucial to understand the basic principles of drug transport, from the site of delivery to the site of action within the CNS, in order to evaluate the possible utility of a new drug candidate for CNS action, or possible CNS side effects of non-CNS targeting drugs. This includes pharmacokinetic aspects of drug concentration-time profiles in plasma and brain, blood-brain barrier transport and drug distribution within the brain parenchyma as well as elimination processes from the brain. Knowledge of anatomical and physiological aspects connected with drug delivery is crucial in this context. The chapter is intended for professionals working in the field of CNS drug development and summarizes key pharmacokinetic principles and state-of-the-art experimental methodologies to assess brain drug disposition. Key parameters, describing the extent of unbound (free) drug across brain barriers, in particular blood-brain and blood-cerebrospinal fluid barriers, are presented along with their application in drug development. Special emphasis is given to brain intracellular pharmacokinetics and its role in evaluating target engagement. Fundamental neuropharmacokinetic differences between small molecular drugs and biologicals are discussed and critical knowledge gaps are outlined.
Collapse
Affiliation(s)
- Irena Loryan
- Translational PKPD Group, Department of Pharmacy, Uppsala University, Uppsala, Sweden.
| | | | - Stina Syvänen
- Department of Public Health and Caring Sciences/Geriatrics, Uppsala University, Rudbeck Laboratory, Uppsala, Sweden
| |
Collapse
|
30
|
Imaging the effect of the circadian light-dark cycle on the glymphatic system in awake rats. Proc Natl Acad Sci U S A 2019; 117:668-676. [PMID: 31848247 PMCID: PMC6955326 DOI: 10.1073/pnas.1914017117] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Homeostasis and the daily rhythms in brain function and temperature are coupled to the circadian light–dark cycle. MRI was used to study the redistribution of intraventricular contrast agent in awake rats during the night when they are active and during the day when at rest. Redistribution is lowest during the day and highest at night and parallels the gradients and regional variations in brain temperatures reported in the literature. The brain areas of low parenchymal redistribution are associated with high temperatures and have a high density of blood vessels that may be an essential part of the organization of the glymphatic system regulating brain temperature, blood gases, nutrients, metabolites, and waste products over the light–dark cycle. The glymphatic system functions in the removal of potentially harmful metabolites and proteins from the brain. Dynamic, contrast-enhanced MRI was used in fully awake rats to follow the redistribution of intraventricular contrast agent entrained to the light–dark cycle and its hypothetical relationship to the sleep–waking cycle, blood flow, and brain temperature in specific brain areas. Brain areas involved in circadian timing and sleep–wake rhythms showed the lowest redistribution of contrast agent during the light phase or time of inactivity and sleep in rats. Global brain redistribution of contrast agent was heterogeneous. The redistribution was highest along the dorsal cerebrum and lowest in the midbrain/pons and along the ventral surface of the brain. This heterogeneous redistribution of contrast agent paralleled the gradients and regional variations in brain temperatures reported in the literature for awake animals. Three-dimensional quantitative ultrashort time-to-echo contrast-enhanced imaging was used to reconstruct small, medium, and large arteries and veins in the rat brain and revealed areas of lowest redistribution overlapped with this macrovasculature. This study raises new questions and theoretical considerations of the impact of the light–dark cycle, brain temperature, and blood flow on the function of the glymphatic system.
Collapse
|
31
|
Abstract
Fundamental aspects of neurobiology are time-of-day regulated. Therefore, it is not surprising that neurodegenerative and psychiatric diseases are accompanied by sleep and circadian rhythm disruption. Although the direction of causation remains unclear, abnormal sleep-wake patterns often occur early in disease, exacerbate progression, and are a common primary complaint from patients. Circadian medicine incorporates knowledge of 24-hour biological rhythms to improve treatment. This article highlights how research and technologic advances in circadian biology might translate to improved patient care.
Collapse
|
32
|
Cuddapah VA, Zhang SL, Sehgal A. Regulation of the Blood-Brain Barrier by Circadian Rhythms and Sleep. Trends Neurosci 2019; 42:500-510. [PMID: 31253251 PMCID: PMC6602072 DOI: 10.1016/j.tins.2019.05.001] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 04/27/2019] [Accepted: 05/01/2019] [Indexed: 01/09/2023]
Abstract
The blood-brain barrier (BBB) is an evolutionarily conserved, structural, and functional separation between circulating blood and the central nervous system (CNS). By controlling permeability into and out of the nervous system, the BBB has a critical role in the precise regulation of neural processes. Here, we review recent studies demonstrating that permeability at the BBB is dynamically controlled by circadian rhythms and sleep. An endogenous circadian rhythm in the BBB controls transporter function, regulating permeability across the BBB. In addition, sleep promotes the clearance of metabolites along the BBB, as well as endocytosis across the BBB. Finally, we highlight the implications of this regulation for diseases, including epilepsy.
Collapse
Affiliation(s)
- Vishnu Anand Cuddapah
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Center for Sleep and Circadian Neurobiology, Chronobiology Program, and Howard Hughes Medical Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shirley L Zhang
- Center for Sleep and Circadian Neurobiology, Chronobiology Program, and Howard Hughes Medical Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Amita Sehgal
- Center for Sleep and Circadian Neurobiology, Chronobiology Program, and Howard Hughes Medical Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
33
|
Abstract
Mechanisms for elimination of metabolites from ISF include metabolism, blood-brain barrier transport and non-selective, perivascular efflux, this last being assessed by measuring the clearance of markers like inulin. Clearance describes elimination. Clearance of a metabolite generated within the brain is determined as its elimination rate divided by its concentration in interstitial fluid (ISF). However, the more frequently measured parameter is the rate constant for elimination determined as elimination rate divided by amount present, which thus depends on both the elimination processes and the distribution of the metabolite in the brain. The relative importance of the various elimination mechanisms depends on the particular metabolite. Little is known about the effects of sleep on clearance via metabolism or blood-brain barrier transport, but studies with inulin in mice comparing perivascular effluxes during sleep and wakefulness reveal a 4.2-fold increase in clearance. Amongst the important brain metabolites considered, CO2 is eliminated so rapidly across the blood-brain barrier that clearance is blood flow limited and elimination quickly balances production. Glutamate is removed from ISF primarily by uptake into astrocytes and conversion to glutamine, but also by transport across the blood-brain barrier. Both lactate and amyloid-β are eliminated by metabolism, blood-brain barrier transport and perivascular efflux and both show decreased production, decreased ISF concentration and increased perivascular clearance during sleep. Taken altogether available data indicate that sleep increases perivascular and non-perivascular clearances for amyloid-β which reduces its concentration and may have long-term consequences for the formation of plaques and cerebral arterial deposits.
Collapse
Affiliation(s)
- Stephen B Hladky
- Department of Pharmacology, University of Cambridge, Cambridge, CB2 1PD, UK.
| | - Margery A Barrand
- Department of Pharmacology, University of Cambridge, Cambridge, CB2 1PD, UK
| |
Collapse
|
34
|
De Lange E, vd Berg D, Bellanti F, Voskuyl R, Syvänen S. P-glycoprotein protein expression versus functionality at the blood-brain barrier using immunohistochemistry, microdialysis and mathematical modeling. Eur J Pharm Sci 2018; 124:61-70. [DOI: 10.1016/j.ejps.2018.08.022] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Revised: 08/14/2018] [Accepted: 08/15/2018] [Indexed: 10/28/2022]
|
35
|
Zhang SL, Yue Z, Arnold DM, Artiushin G, Sehgal A. A Circadian Clock in the Blood-Brain Barrier Regulates Xenobiotic Efflux. Cell 2018; 173:130-139.e10. [PMID: 29526461 PMCID: PMC5866247 DOI: 10.1016/j.cell.2018.02.017] [Citation(s) in RCA: 162] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Revised: 12/22/2017] [Accepted: 02/07/2018] [Indexed: 12/29/2022]
Abstract
Endogenous circadian rhythms are thought to modulate responses to external factors, but mechanisms that confer time-of-day differences in organismal responses to environmental insults/therapeutic treatments are poorly understood. Using a xenobiotic, we find that permeability of the Drosophila "blood"-brain barrier (BBB) is higher at night. The permeability rhythm is driven by circadian regulation of efflux and depends on a molecular clock in the perineurial glia of the BBB, although efflux transporters are restricted to subperineurial glia (SPG). We show that transmission of circadian signals across the layers requires cyclically expressed gap junctions. Specifically, during nighttime, gap junctions reduce intracellular magnesium ([Mg2+]i), a positive regulator of efflux, in SPG. Consistent with lower nighttime efflux, nighttime administration of the anti-epileptic phenytoin is more effective at treating a Drosophila seizure model. These findings identify a novel mechanism of circadian regulation and have therapeutic implications for drugs targeted to the central nervous system.
Collapse
Affiliation(s)
- Shirley L Zhang
- Center for Sleep and Circadian Neurobiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Chronobiology Program at Penn and Howard Hughes Medical Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Zhifeng Yue
- Chronobiology Program at Penn and Howard Hughes Medical Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Denice M Arnold
- Chronobiology Program at Penn and Howard Hughes Medical Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Gregory Artiushin
- Chronobiology Program at Penn and Howard Hughes Medical Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Amita Sehgal
- Center for Sleep and Circadian Neurobiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Chronobiology Program at Penn and Howard Hughes Medical Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
36
|
Abbott NJ, Pizzo ME, Preston JE, Janigro D, Thorne RG. The role of brain barriers in fluid movement in the CNS: is there a 'glymphatic' system? Acta Neuropathol 2018; 135:387-407. [PMID: 29428972 DOI: 10.1007/s00401-018-1812-4] [Citation(s) in RCA: 383] [Impact Index Per Article: 54.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 01/16/2018] [Accepted: 01/24/2018] [Indexed: 12/15/2022]
Abstract
Brain fluids are rigidly regulated to provide stable environments for neuronal function, e.g., low K+, Ca2+, and protein to optimise signalling and minimise neurotoxicity. At the same time, neuronal and astroglial waste must be promptly removed. The interstitial fluid (ISF) of the brain tissue and the cerebrospinal fluid (CSF) bathing the CNS are integral to this homeostasis and the idea of a glia-lymph or 'glymphatic' system for waste clearance from brain has developed over the last 5 years. This links bulk (convective) flow of CSF into brain along the outside of penetrating arteries, glia-mediated convective transport of fluid and solutes through the brain extracellular space (ECS) involving the aquaporin-4 (AQP4) water channel, and finally delivery of fluid to venules for clearance along peri-venous spaces. However, recent evidence favours important amendments to the 'glymphatic' hypothesis, particularly concerning the role of glia and transfer of solutes within the ECS. This review discusses studies which question the role of AQP4 in ISF flow and the lack of evidence for its ability to transport solutes; summarizes attributes of brain ECS that strongly favour the diffusion of small and large molecules without ISF flow; discusses work on hydraulic conductivity and the nature of the extracellular matrix which may impede fluid movement; and reconsiders the roles of the perivascular space (PVS) in CSF-ISF exchange and drainage. We also consider the extent to which CSF-ISF exchange is possible and desirable, the impact of neuropathology on fluid drainage, and why using CSF as a proxy measure of brain components or drug delivery is problematic. We propose that new work and key historical studies both support the concept of a perivascular fluid system, whereby CSF enters the brain via PVS convective flow or dispersion along larger caliber arteries/arterioles, diffusion predominantly regulates CSF/ISF exchange at the level of the neurovascular unit associated with CNS microvessels, and, finally, a mixture of CSF/ISF/waste products is normally cleared along the PVS of venules/veins as well as other pathways; such a system may or may not constitute a true 'circulation', but, at the least, suggests a comprehensive re-evaluation of the previously proposed 'glymphatic' concepts in favour of a new system better taking into account basic cerebrovascular physiology and fluid transport considerations.
Collapse
Affiliation(s)
- N Joan Abbott
- Faculty of Life Sciences and Medicine, Institute of Pharmaceutical Science, King's College London, Franklin Wilkins Building 3.82, 150 Stamford St, London, SE1 9NH, UK.
| | - Michelle E Pizzo
- Division of Pharmaceutical Sciences, University of Wisconsin-Madison School of Pharmacy, Madison, WI, USA
- Clinical Neuroengineering Training Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Jane E Preston
- Faculty of Life Sciences and Medicine, Institute of Pharmaceutical Science, King's College London, Franklin Wilkins Building 3.82, 150 Stamford St, London, SE1 9NH, UK
| | - Damir Janigro
- Flocel Inc., Cleveland, OH, USA
- Department of Physiology, Case Western Reserve University, Cleveland, OH, USA
| | - Robert G Thorne
- Division of Pharmaceutical Sciences, University of Wisconsin-Madison School of Pharmacy, Madison, WI, USA.
- Clinical Neuroengineering Training Program, University of Wisconsin-Madison, Madison, WI, USA.
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, USA.
- Cellular and Molecular Pathology Graduate Training Program, University of Wisconsin-Madison, Madison, WI, USA.
- Institute for Clinical and Translational Research, University of Wisconsin-Madison, Madison, WI, USA.
- , 5113 Rennebohm Hall, 777 Highland Avenue, Madison, WI, 53705, USA.
| |
Collapse
|
37
|
Kervezee L, Hartman R, van den Berg DJ, Meijer JH, de Lange EC. Diurnal variation in the pharmacokinetics and brain distribution of morphine and its major metabolite. Eur J Pharm Sci 2017; 109S:S132-S139. [DOI: 10.1016/j.ejps.2017.05.048] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 05/22/2017] [Indexed: 10/19/2022]
|
38
|
Ozturk N, Ozturk D, Kavakli IH, Okyar A. Molecular Aspects of Circadian Pharmacology and Relevance for Cancer Chronotherapy. Int J Mol Sci 2017; 18:E2168. [PMID: 29039812 PMCID: PMC5666849 DOI: 10.3390/ijms18102168] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 10/13/2017] [Accepted: 10/14/2017] [Indexed: 02/01/2023] Open
Abstract
The circadian timing system (CTS) controls various biological functions in mammals including xenobiotic metabolism and detoxification, immune functions, cell cycle events, apoptosis and angiogenesis. Although the importance of the CTS is well known in the pharmacology of drugs, it is less appreciated at the clinical level. Genome-wide studies highlighted that the majority of drug target genes are controlled by CTS. This suggests that chronotherapeutic approaches should be taken for many drugs to enhance their effectiveness. Currently chronotherapeutic approaches are successfully applied in the treatment of different types of cancers. The chronotherapy approach has improved the tolerability and antitumor efficacy of anticancer drugs both in experimental animals and in cancer patients. Thus, chronobiological studies have been of importance in determining the most appropriate time of administration of anticancer agents to minimize their side effects or toxicity and enhance treatment efficacy, so as to optimize the therapeutic ratio. This review focuses on the underlying mechanisms of the circadian pharmacology i.e., chronopharmacokinetics and chronopharmacodynamics of anticancer agents with the molecular aspects, and provides an overview of chronotherapy in cancer and some of the recent advances in the development of chronopharmaceutics.
Collapse
Affiliation(s)
- Narin Ozturk
- Department of Pharmacology, Faculty of Pharmacy, Istanbul University, TR-34116 Beyazit-Istanbul, Turkey.
| | - Dilek Ozturk
- Department of Pharmacology, Faculty of Pharmacy, Istanbul University, TR-34116 Beyazit-Istanbul, Turkey.
- Department of Pharmacology, Faculty of Pharmacy, Bezmialem Vakif University, TR-34093 Fatih-Istanbul, Turkey.
| | - Ibrahim Halil Kavakli
- Departments of Molecular Biology and Genetics and Chemical and Biological Engineering, Koc University, TR-34450 Sariyer-Istanbul, Turkey.
| | - Alper Okyar
- Department of Pharmacology, Faculty of Pharmacy, Istanbul University, TR-34116 Beyazit-Istanbul, Turkey.
| |
Collapse
|
39
|
Husse J, Kiehn JT, Barclay JL, Naujokat N, Meyer-Kovac J, Lehnert H, Oster H. Tissue-Specific Dissociation of Diurnal Transcriptome Rhythms During Sleep Restriction in Mice. Sleep 2017; 40:3751182. [DOI: 10.1093/sleep/zsx068] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
40
|
Liao MZ, Shen DD, Mao Q. P-gp/ABCB1 exerts differential impacts on brain and fetal exposure to norbuprenorphine-Response to 'Letter to the Editor'. Pharmacol Res 2017; 121:253. [PMID: 28411002 DOI: 10.1016/j.phrs.2017.04.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 04/06/2017] [Indexed: 11/19/2022]
Affiliation(s)
- Michael Z Liao
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, USA
| | - Danny D Shen
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, USA
| | - Qingcheng Mao
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
41
|
Dash RP, Rais R. P-gp/ABCB1 exerts differential impacts on brain and fetal exposure to norbuprenorphine-Significance of zeitgeber time on pharmacokinetics. Pharmacol Res 2017; 121:251-252. [PMID: 28400151 DOI: 10.1016/j.phrs.2017.04.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 04/05/2017] [Accepted: 04/06/2017] [Indexed: 11/29/2022]
Affiliation(s)
- Ranjeet Prasad Dash
- Drug Metabolism and Pharmacokinetics, Johns Hopkins Drug Discovery Program, Johns Hopkins University, 855 North Wolfe Street, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University, 855 North Wolfe Street, Baltimore, MD 21205, USA
| | - Rana Rais
- Drug Metabolism and Pharmacokinetics, Johns Hopkins Drug Discovery Program, Johns Hopkins University, 855 North Wolfe Street, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University, 855 North Wolfe Street, Baltimore, MD 21205, USA.
| |
Collapse
|
42
|
Yamamoto Y, Danhof M, de Lange ECM. Microdialysis: the Key to Physiologically Based Model Prediction of Human CNS Target Site Concentrations. AAPS JOURNAL 2017; 19:891-909. [DOI: 10.1208/s12248-017-0050-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 01/25/2017] [Indexed: 01/03/2023]
|
43
|
Intracerebral microdialysis in blood-brain barrier drug research with focus on nanodelivery. DRUG DISCOVERY TODAY. TECHNOLOGIES 2016; 20:13-18. [PMID: 27986218 DOI: 10.1016/j.ddtec.2016.07.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 07/13/2016] [Indexed: 01/09/2023]
Abstract
Microdialysis has contributed significantly to advance the understanding of BBB transport of drugs and to reveal key aspects of BBB transport, including quantifying active efflux and active uptake. Microdialysis studies on pharmacokinetic-pharmacodynamic relationships have given in-depth understanding of the processes involved. Recently, nanodelivery to the brain has been investigated with microdialysis, contributing to nanodelivery science by giving quantitative information on the possible success of different delivery vehicles and how they are involved in BBB transport.
Collapse
|
44
|
Savolainen H, Meerlo P, Elsinga PH, Windhorst AD, Dierckx RA, Colabufo NA, van Waarde A, Luurtsema G. P-glycoprotein Function in the Rodent Brain Displays a Daily Rhythm, a Quantitative In Vivo PET Study. AAPS JOURNAL 2016; 18:1524-1531. [DOI: 10.1208/s12248-016-9973-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 08/08/2016] [Indexed: 12/21/2022]
|
45
|
Lagerweij T, Hiddingh L, Biesmans D, Crommentuijn MH, Cloos J, Li XN, Kogiso M, Tannous BA, Vandertop WP, Noske DP, Kaspers GJ, Würdinger T, Hulleman E. A chemical screen for medulloblastoma identifies quercetin as a putative radiosensitizer. Oncotarget 2016; 7:35776-35788. [PMID: 26967057 PMCID: PMC5094961 DOI: 10.18632/oncotarget.7980] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Accepted: 02/20/2016] [Indexed: 11/29/2022] Open
Abstract
Treatment of medulloblastoma in children fails in approximately 30% of patients, and is often accompanied by severe late sequelae. Therefore, more effective drugs are needed that spare normal tissue and diminish long-term side effects. Since radiotherapy plays a pivotal role in the treatment of medulloblastoma, we set out to identify novel drugs that could potentiate the effect of ionizing radiation.Thereto, a small molecule library, consisting of 960 chemical compounds, was screened for its ability to sensitize towards irradiation. This small molecule screen identified the flavonoid quercetin as a novel radiosensitizer for the medulloblastoma cell lines DAOY, D283-med, and, to a lesser extent, D458-med at low micromolar concentrations and irradiation doses used in fractionated radiation schemes. Quercetin did not affect the proliferation of neural precursor cells or normal human fibroblasts. Importantly, in vivo experiments confirmed the radiosensitizing properties of quercetin. Administration of this flavonoid at the time of irradiation significantly prolonged survival in orthotopically xenografted mice. Together, these findings indicate that quercetin is a potent radiosensitizer for medulloblastoma cells that may be a promising lead for the treatment of medulloblastoma in patients.
Collapse
Affiliation(s)
- Tonny Lagerweij
- Department of Pediatric Oncology/Hematology, VU University Medical Center, Amsterdam, The Netherlands
- Department of Neurosurgery, VU University Medical Center, Amsterdam, The Netherlands
- Department of Neuro-oncology Research Group, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Lotte Hiddingh
- Department of Pediatric Oncology/Hematology, VU University Medical Center, Amsterdam, The Netherlands
- Department of Neurosurgery, VU University Medical Center, Amsterdam, The Netherlands
- Department of Neuro-oncology Research Group, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Dennis Biesmans
- Department of Pediatric Oncology/Hematology, VU University Medical Center, Amsterdam, The Netherlands
- Department of Neuro-oncology Research Group, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Matheus H.W. Crommentuijn
- Department of Neuro-oncology Research Group, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Jacqueline Cloos
- Department of Pediatric Oncology/Hematology, VU University Medical Center, Amsterdam, The Netherlands
| | - Xiao-Nan Li
- Department of Pediatrics, Texas Children's Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Mari Kogiso
- Department of Pediatrics, Texas Children's Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Bakhos A. Tannous
- Department of Neurology, Molecular Neurogenetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - W. Peter Vandertop
- Department of Neurosurgery, VU University Medical Center, Amsterdam, The Netherlands
| | - David P. Noske
- Department of Neurosurgery, VU University Medical Center, Amsterdam, The Netherlands
- Department of Neuro-oncology Research Group, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Gertjan J.L. Kaspers
- Department of Pediatric Oncology/Hematology, VU University Medical Center, Amsterdam, The Netherlands
| | - Tom Würdinger
- Department of Neurosurgery, VU University Medical Center, Amsterdam, The Netherlands
- Department of Neuro-oncology Research Group, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
- Department of Neurology, Molecular Neurogenetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Esther Hulleman
- Department of Pediatric Oncology/Hematology, VU University Medical Center, Amsterdam, The Netherlands
- Department of Neurosurgery, VU University Medical Center, Amsterdam, The Netherlands
- Department of Neuro-oncology Research Group, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
46
|
Simon MJ, Iliff JJ. Regulation of cerebrospinal fluid (CSF) flow in neurodegenerative, neurovascular and neuroinflammatory disease. Biochim Biophys Acta Mol Basis Dis 2015; 1862:442-51. [PMID: 26499397 DOI: 10.1016/j.bbadis.2015.10.014] [Citation(s) in RCA: 221] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 09/23/2015] [Accepted: 10/19/2015] [Indexed: 12/20/2022]
Abstract
Cerebrospinal fluid (CSF) circulation and turnover provides a sink for the elimination of solutes from the brain interstitium, serving an important homeostatic role for the function of the central nervous system. Disruption of normal CSF circulation and turnover is believed to contribute to the development of many diseases, including neurodegenerative conditions such as Alzheimer's disease, ischemic and traumatic brain injury, and neuroinflammatory conditions such as multiple sclerosis. Recent insights into CSF biology suggesting that CSF and interstitial fluid exchange along a brain-wide network of perivascular spaces termed the 'glymphatic' system suggest that CSF circulation may interact intimately with glial and vascular function to regulate basic aspects of brain function. Dysfunction within this glial vascular network, which is a feature of the aging and injured brain, is a potentially critical link between brain injury, neuroinflammation and the development of chronic neurodegeneration. Ongoing research within this field may provide a powerful new framework for understanding the common links between neurodegenerative, neurovascular and neuroinflammatory disease, in addition to providing potentially novel therapeutic targets for these conditions. This article is part of a Special Issue entitled: Neuro Inflammation edited by Helga E. de Vries and Markus Schwaninger.
Collapse
Affiliation(s)
- Matthew J Simon
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, OR, USA; Neuroscience Graduate Program, Oregon Health & Science University, Portland, OR, USA
| | - Jeffrey J Iliff
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, OR, USA; Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, USA; Neuroscience Graduate Program, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
47
|
de Lange ECM, Hammarlund-Udenaes M. Translational aspects of blood-brain barrier transport and central nervous system effects of drugs: From discovery to patients. Clin Pharmacol Ther 2015; 97:380-94. [DOI: 10.1002/cpt.76] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Revised: 01/06/2015] [Accepted: 01/06/2015] [Indexed: 02/06/2023]
Affiliation(s)
- ECM de Lange
- Leiden Academic Centre for Drug Research; Division of Pharmacology; Leiden University, Gorlaeus Laboratories; Leiden The Netherlands
| | | |
Collapse
|