1
|
Zhao N, Chung TD, Guo Z, Jamieson JJ, Liang L, Linville RM, Pessell AF, Wang L, Searson PC. The influence of physiological and pathological perturbations on blood-brain barrier function. Front Neurosci 2023; 17:1289894. [PMID: 37937070 PMCID: PMC10626523 DOI: 10.3389/fnins.2023.1289894] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 10/06/2023] [Indexed: 11/09/2023] Open
Abstract
The blood-brain barrier (BBB) is located at the interface between the vascular system and the brain parenchyma, and is responsible for communication with systemic circulation and peripheral tissues. During life, the BBB can be subjected to a wide range of perturbations or stresses that may be endogenous or exogenous, pathological or therapeutic, or intended or unintended. The risk factors for many diseases of the brain are multifactorial and involve perturbations that may occur simultaneously (e.g., two-hit model for Alzheimer's disease) and result in different outcomes. Therefore, it is important to understand the influence of individual perturbations on BBB function in isolation. Here we review the effects of eight perturbations: mechanical forces, temperature, electromagnetic radiation, hypoxia, endogenous factors, exogenous factors, chemical factors, and pathogens. While some perturbations may result in acute or chronic BBB disruption, many are also exploited for diagnostic or therapeutic purposes. The resultant outcome on BBB function depends on the dose (or magnitude) and duration of the perturbation. Homeostasis may be restored by self-repair, for example, via processes such as proliferation of affected cells or angiogenesis to create new vasculature. Transient or sustained BBB dysfunction may result in acute or pathological symptoms, for example, microhemorrhages or hypoperfusion. In more extreme cases, perturbations may lead to cytotoxicity and cell death, for example, through exposure to cytotoxic plaques.
Collapse
Affiliation(s)
- Nan Zhao
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, United States
| | - Tracy D. Chung
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, United States
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Zhaobin Guo
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, United States
| | - John J. Jamieson
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, United States
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Lily Liang
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, United States
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Raleigh M. Linville
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, United States
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Alex F. Pessell
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, United States
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Linus Wang
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, United States
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Peter C. Searson
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, United States
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
2
|
Zou G, Xia J, Luo H, Xiao D, Jin J, Miao C, Zuo X, Gao Q, Zhang Z, Xue T, You Y, Zhang Y, Zhang L, Xiong W. Combined alcohol and cannabinoid exposure leads to synergistic toxicity by affecting cerebellar Purkinje cells. Nat Metab 2022; 4:1138-1149. [PMID: 36109623 DOI: 10.1038/s42255-022-00633-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 08/03/2022] [Indexed: 11/09/2022]
Abstract
Combined use of cannabis and alcohol results in greater psychoactive toxicity than either substance alone, but the underlying central mechanisms behind this worsened outcome remain unclear. Here we show that the synergistic effect of Δ9-tetrahydrocannabinol (THC) and ethanol on motor incoordination in mice is achieved by activating presynaptic type 1 cannabinoid receptors (CB1R) and potentiating extrasynaptic glycine receptors (GlyR) within cerebellar Purkinje cells (PCs). The combination of ethanol and THC significantly reduces miniature excitatory postsynaptic current frequency in a CB1R-dependent manner, while increasing the extrasynaptic GlyR-mediated chronic chloride current, both leading to decreased PC activity. Ethanol enhances THC actions by boosting the blood-brain-barrier permeability of THC and enriching THC in the cell membrane. Di-desoxy-THC, a designed compound that specifically disrupts THC-GlyR interaction without affecting the basic functions of CB1R and GlyR, is able to restore PC function and motor coordination in mice. Our findings provide potential therapeutic strategies for overcoming the synergistic toxicity caused by combining cannabis and alcohol use.
Collapse
Affiliation(s)
- Guichang Zou
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China
| | - Jing Xia
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Heyi Luo
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Dan Xiao
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jin Jin
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Chenjian Miao
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xin Zuo
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Qianqian Gao
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, China
| | - Zhi Zhang
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Tian Xue
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yezi You
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, China
| | - Ye Zhang
- Department of Anesthesiology and Perioperative Medicine, The Second Hospital of Anhui Medical University, Hefei, China
| | - Li Zhang
- Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Wei Xiong
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China.
- Anhui Province Key Laboratory of Biomedical Aging Research, Hefei, China.
| |
Collapse
|
3
|
Abstract
Leptin for over 25 years has been a central theme in the study of appetite, obesity, and starvation. As the major site of leptin production is peripheral, and the site of action of greatest interest is the hypothalamus, how leptin accesses the central nervous system (CNS) and crosses the blood-brain barrier (BBB) has been of great interest. We review here the ongoing research that addresses fundamental questions such as the sites of leptin resistances in obesity and other conditions, the causes of resistances and their relations to one another, the three barrier sites of entry into the CNS, why recent studies using suprapharmacological doses cannot address these questions but give insight into nonsaturable entry of leptin into the CNS, and how that might be useful in using leptin therapeutically. The current status of the controversy of whether the short form of the leptin receptor acts as the BBB leptin transporter and how obesity may transform leptin transport is reviewed. Review of these and other topics summarizes in a new appreciation of what leptin may have actually evolved to do and what physiological role leptin resistance may play. © 2021 American Physiological Society. Compr Physiol 11:1-19, 2021.
Collapse
Affiliation(s)
- William A Banks
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, Washington, USA.,Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
4
|
Carrino D, Branca JJV, Becatti M, Paternostro F, Morucci G, Gulisano M, Di Cesare Mannelli L, Pacini A. Alcohol-Induced Blood-Brain Barrier Impairment: An In Vitro Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:2683. [PMID: 33799986 PMCID: PMC7967408 DOI: 10.3390/ijerph18052683] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/01/2021] [Accepted: 03/04/2021] [Indexed: 12/16/2022]
Abstract
In recent years, alcohol abuse has dramatically grown with deleterious consequence for people's health and, in turn, for health care costs. It has been demonstrated, in humans and animals, that alcohol intoxication induces neuroinflammation and neurodegeneration thus leading to brain impairments. Furthermore, it has been shown that alcohol consumption is able to impair the blood-brain barrier (BBB), but the molecular mechanisms underlining this detrimental effect have not been fully elucidated. For this reason, in this study we investigated the effects of alcohol exposure on a rat brain endothelial (RBE4) cell line, as an in vitro-validated model of brain microvascular endothelial cells. To assess whether alcohol caused a concentration-related response, the cells were treated at different times with increasing concentrations (10-1713 mM) of ethyl alcohol (EtOH). Microscopic and molecular techniques, such as cell viability assay, immunofluorescence and Western blotting, were used to examine the mechanisms involved in alcohol-induced brain endothelial cell alterations including tight junction distribution, apoptosis, and reactive oxygen species production. Our findings clearly demonstrate that alcohol causes the formation of gaps between cells by tight junction disassembly, triggered by the endoplasmic reticulum and oxidative stress, highlighted by GRP78 chaperone upregulation and increase in reactive oxygen species production, respectively. The results from this study shed light on the mechanisms underlying alcohol-induced blood-brain barrier dysfunction and a better understanding of these processes will allow us to take advantage of developing new therapeutic strategies in order to prevent the deleterious effects of alcohol.
Collapse
Affiliation(s)
- Donatello Carrino
- Department Experimental and Clinical Medicine, Anatomy and Histology Section, University of Firenze, 50134 Firenze, Italy; (D.C.); (J.J.V.B.); (F.P.); (M.G.)
| | - Jacopo Junio Valerio Branca
- Department Experimental and Clinical Medicine, Anatomy and Histology Section, University of Firenze, 50134 Firenze, Italy; (D.C.); (J.J.V.B.); (F.P.); (M.G.)
| | - Matteo Becatti
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Firenze, 50134 Firenze, Italy;
| | - Ferdinando Paternostro
- Department Experimental and Clinical Medicine, Anatomy and Histology Section, University of Firenze, 50134 Firenze, Italy; (D.C.); (J.J.V.B.); (F.P.); (M.G.)
| | - Gabriele Morucci
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy;
| | - Massimo Gulisano
- Department Experimental and Clinical Medicine, Anatomy and Histology Section, University of Firenze, 50134 Firenze, Italy; (D.C.); (J.J.V.B.); (F.P.); (M.G.)
| | - Lorenzo Di Cesare Mannelli
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Pharmacology and Toxicology Section, University of Firenze, 50139 Firenze, Italy;
| | - Alessandra Pacini
- Department Experimental and Clinical Medicine, Anatomy and Histology Section, University of Firenze, 50134 Firenze, Italy; (D.C.); (J.J.V.B.); (F.P.); (M.G.)
| |
Collapse
|
5
|
Rodriguez EA, Yamamoto BK. Toxic Effects of Methamphetamine on Perivascular Health: Co-morbid Effects of Stress and Alcohol Use Disorders. Curr Neuropharmacol 2021; 19:2092-2107. [PMID: 34344290 PMCID: PMC9185763 DOI: 10.2174/1570159x19666210803150023] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 07/09/2021] [Accepted: 07/19/2021] [Indexed: 12/04/2022] Open
Abstract
Methamphetamine (Meth) abuse presents a global problem and commonly occurs with stress and/or alcohol use disorders. Regardless, the biological causes and consequences of these comorbidities are unclear. Whereas the mechanisms of Meth, stress, and alcohol abuse have been examined individually and well-characterized, these processes overlap significantly and can impact the neural and peripheral consequences of Meth. This review focuses on the deleterious cardio- and cerebrovascular effects of Meth, stress, alcohol abuse, and their comorbid effects on the brain and periphery. Points of emphasis are on the composition of the blood-brain barrier and their effects on the heart and vasculature. The autonomic nervous system, inflammation, and oxidative stress are specifically highlighted as common mediators of the toxic consequences to vascular and perivascular health. A significant portion of the Meth abusing population also presents with stress and alcohol use disorders, prompting a need to understand the mechanisms underlying their comorbidities. Little is known about their possible convergent effects. Therefore, the purpose of this critical review is to identify shared mechanisms of Meth, chronic stress, and alcohol abuse that contributes to the dysfunction of vascular health and underscores the need for studies that directly address their interactions.
Collapse
Affiliation(s)
- Eric A. Rodriguez
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Bryan K. Yamamoto
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
6
|
Chronic alcohol exposure reduces acetylated histones in the sleep-wake regulatory brain regions to cause insomnia during withdrawal. Neuropharmacology 2020; 180:108332. [PMID: 32961200 DOI: 10.1016/j.neuropharm.2020.108332] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 09/15/2020] [Accepted: 09/17/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Alcohol use disorder (AUD) develops after chronic and heavy use of alcohol. Insomnia, a hallmark of AUD, plays a crucial role in the development of AUD. However, the causal mechanisms are unknown. Since chronic alcohol reduces acetylated histones and disrupts the epigenome, we hypothesized that chronic alcohol exposure will reduce acetylated histones in wake-promoting regions of the brain to cause insomnia during alcohol withdrawal. METHODS Adult male C57BL/6J mice, surgically instrumented for electrophysiological monitoring of sleep-wakefulness, were exposed to chronic alcohol (6.8%) consumption using Lieber-DeCarli liquid diet. Three experiments were performed. First, the effect of chronic alcohol consumption was examined on sleep-wakefulness during 7 days of withdrawal. Second, the expression of acetylated histones, H3 lysine 14 (AcH3K14), was examined in two major sleep-wake regulatory brain regions: basal forebrain (BF) and lateral hypothalamus (LH) of the brain by using western blotting. Next, blockade of histone deacetylase, via systemic administration of TSA was examined on alcohol-induced changes in sleep-wakefulness. RESULTS Alcoholic mice displayed a significant reduction in the quality and quantity of NREM sleep coupled with a significant increase in wakefulness that lasted for several days during alcohol withdrawal. In addition, alcoholic mice displayed a significant reduction in the expression of AcH3K14 in both BF and LH. Systemic administration of TSA significantly attenuated insomnia and improved the quality and quantity of sleep during alcohol withdrawal. CONCLUSIONS Based on our results, we suggest that a causal relationship exists between reduced histone acetylation and insomnia during alcohol withdrawal.
Collapse
|
7
|
Bach P, Koopmann A, Kiefer F. The Impact of Appetite-Regulating Neuropeptide Leptin on Alcohol Use, Alcohol Craving and Addictive Behavior: A Systematic Review of Preclinical and Clinical Data. Alcohol Alcohol 2020; 56:149-165. [PMID: 32490525 DOI: 10.1093/alcalc/agaa044] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 04/27/2020] [Accepted: 04/28/2020] [Indexed: 12/18/2022] Open
Abstract
AIMS The appetite regulating hormone leptin, which is mainly secreted from adipose tissue, is an important regulator of food intake and modulator of reward-driven behavior. Leptin exerts its biological actions via binding to the leptin receptor, which is expressed in the hypothalamus, but also in the hippocampus, the amygdala and the substantia nigra. In the ventral tegmental area (VTA), leptin attenuates the firing rate of dopaminergic neurons that project to the Nucleus accumbens (NAc), which serves as relay to other brain areas of the "addiction network", such as the prefrontal cortex. This suggests that leptin plays a role in the processing of rewards in the context of substance use disorders such as alcohol use disorder, especially through attenuation of dopaminergic activity in the mesolimbic reward system. This supports the plausibility of leptin's potential effects in alcohol use disorder. METHODS We searched MEDLINE from 1990 to February 2020. All abstracts were screened for relevance and we only included publications reporting original data with a full text available in English language. Studies that did not report leptin-data, reviews or case reports/series were not included. RESULTS We identified a total of N=293 studies of whom a total of N=55 preclinical and clinical studies met the specified criteria. N=40 studies investigated the effects of alcohol on leptin plasma levels, N=9 studies investigated the effects of leptin on alcohol craving and N=6 studies investigated the effects of leptin on relapse and alcohol consumption. CONCLUSIONS In this review of preclinical and clinical data, we assess the role of leptin in alcohol use and the development and maintenance of an alcohol use disorder, alcohol craving and relapse. Integrating the existing preclinical and clinical data on leptin may reveal new and innovative targets for the treatment of substance use disorders in the future.
Collapse
Affiliation(s)
- Patrick Bach
- Department of Addictive Behavior and Addiction Medicine, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, J5/68159 Mannheim, Germany.,Feuerlein Center on Translational Addiction Medicine (FCTS), University of Heidelberg, J5/68159 Mannheim, Germany
| | - Anne Koopmann
- Department of Addictive Behavior and Addiction Medicine, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, J5/68159 Mannheim, Germany.,Feuerlein Center on Translational Addiction Medicine (FCTS), University of Heidelberg, J5/68159 Mannheim, Germany
| | - Falk Kiefer
- Department of Addictive Behavior and Addiction Medicine, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, J5/68159 Mannheim, Germany.,Feuerlein Center on Translational Addiction Medicine (FCTS), University of Heidelberg, J5/68159 Mannheim, Germany
| |
Collapse
|
8
|
Kanzaki A, Tada H, Otsuka A, Nakamura T. Severe tardive dyskinesia induced by domperidone in presenile and non-dementia type 2 diabetes man with alcohol misuse showing albuminocytological dissociation and white matter hyperintensity. BMJ Case Rep 2019; 12:12/5/e228789. [DOI: 10.1136/bcr-2018-228789] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Domperidone has difficulty passing the blood–brain barrier, thus rarely causes tardive dyskinesia. Furthermore, its symptoms in adults are generally mild. Although both alcohol and diabetes are thought to increase the risk of development of tardive dyskinesia, their impact remains controversial, especially diabetes, and factors related to worsened tardive dyskinesia have not been clearly elucidated. A 59-year-old man with type 2 diabetes and history of alcohol misuse, who had been chronically prescribed domperidone at 15 mg/day, showed severe tardive dyskinesia, which was remitted within several days by stopping the drug. In our case, albuminocytological dissociation and white matter hyperintensity on MRI were confirmed, which were thought to be related to blood–brain barrier dysfunction. This present findings indicate that alcohol misuse and type 2 diabetes, as well as albuminocytological dissociation and white matter hyperintensity may result in severe tardive dyskinesia, even in individuals receiving domperidone.
Collapse
|
9
|
Abstract
Our understanding of adipose tissue as an endocrine organ has been transformed over the last 20 years. During this time, a number of adipocyte-derived factors or adipokines have been identified. This article will review evidence for how adipokines acting via the central nervous system (CNS) regulate normal physiology and disease pathology. The reported CNS-mediated effects of adipokines are varied and include the regulation of energy homeostasis, autonomic nervous system activity, the reproductive axis, neurodevelopment, cardiovascular function, and cognition. Due to the wealth of information available and the diversity of their known functions, the archetypal adipokines leptin and adiponectin will be focused on extensively. Other adipokines with established CNS actions will also be discussed. Due to the difficulties associated with studying CNS function on a molecular level in humans, the majority of our knowledge, and as such the studies described in this paper, comes from work in experimental animal models; however, where possible the relevant data from human studies are also highlighted. © 2017 American Physiological Society. Compr Physiol 7:1359-1406, 2017.
Collapse
Affiliation(s)
- Craig Beall
- Biomedical Neuroscience Research Group, Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, Devon, United Kingdom
| | - Lydia Hanna
- Biomedical Neuroscience Research Group, Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, Devon, United Kingdom
| | - Kate L J Ellacott
- Biomedical Neuroscience Research Group, Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, Devon, United Kingdom
| |
Collapse
|
10
|
Neves AR, Queiroz JF, Lima SAC, Reis S. Apo E-Functionalization of Solid Lipid Nanoparticles Enhances Brain Drug Delivery: Uptake Mechanism and Transport Pathways. Bioconjug Chem 2017; 28:995-1004. [PMID: 28355061 DOI: 10.1021/acs.bioconjchem.6b00705] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Several strategies have been implemented to enhance brain drug delivery, and herein solid lipid nanoparticles functionalized with apolipoprotein E were tested in hCMEC/D3 cell monolayers. The mean diameter of 160 nm, negative charge of -12 mV, and their lipophilic characteristics make these nanosystems suitable for brain delivery. Confocal images and flow cytometry data showed a cellular uptake increase of 1.8-fold for SLN-Palmitate-ApoE and 1.9-fold for SLN-DSPE-ApoE when compared with the non-functionalized SLNs. Clathrin-mediated endocytosis was distinguished as the preferential internalization pathway involved in cellular uptake and nanoparticles could cross the blood-brain barrier predominantly by a transcellular pathway. The understanding of the mechanisms involved in the transport of these nanosystems through the blood-brain barrier may potentiate their application on brain drug delivery.
Collapse
Affiliation(s)
- Ana Rute Neves
- UCIBIO, REQUIMTE, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto , Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Joana Fontes Queiroz
- UCIBIO, REQUIMTE, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto , Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Sofia A Costa Lima
- UCIBIO, REQUIMTE, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto , Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Salette Reis
- UCIBIO, REQUIMTE, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto , Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| |
Collapse
|
11
|
Northrop NA, Yamamoto BK. Methamphetamine effects on blood-brain barrier structure and function. Front Neurosci 2015; 9:69. [PMID: 25788874 PMCID: PMC4349189 DOI: 10.3389/fnins.2015.00069] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 02/17/2015] [Indexed: 01/28/2023] Open
Abstract
Methamphetamine (Meth) is a widely abuse psychostimulant. Traditionally, studies have focused on the neurotoxic effects of Meth on monoaminergic neurotransmitter terminals. Recently, both in vitro and in vivo studies have investigated the effects of Meth on the BBB and found that Meth produces a decrease in BBB structural proteins and an increase in BBB permeability to various molecules. Moreover, preclinical studies are validated by clinical studies in which human Meth users have increased concentrations of toxins in the brain. Therefore, this review will focus on the structural and functional disruption of the BBB caused by Meth and the mechanisms that contribute to Meth-induced BBB disruption. The review will reveal that the mechanisms by which Meth damages dopamine and serotonin terminals are similar to the mechanisms by which the blood-brain barrier (BBB) is damaged. Furthermore, this review will cover the factors that are known to potentiate the effects of Meth (McCann et al., 1998) on the BBB, such as stress and HIV, both of which are co-morbid conditions associated with Meth abuse. Overall, the goal of this review is to demonstrate that the scope of damage produced by Meth goes beyond damage to monoaminergic neurotransmitter systems to include BBB disruption as well as provide a rationale for investigating therapeutics to treat Meth-induced BBB disruption. Since a breach of the BBB can have a multitude of consequences, therapies directed toward the treatment of BBB disruption may help to ameliorate the long-term neurodegeneration and cognitive deficits produced by Meth and possibly even Meth addiction.
Collapse
Affiliation(s)
- Nicole A Northrop
- Department of Neurosciences, University of Toledo College of Medicine Toledo, OH, USA
| | - Bryan K Yamamoto
- Department of Neurosciences, University of Toledo College of Medicine Toledo, OH, USA
| |
Collapse
|
12
|
Ozcan M, Saatci T, Ayar A, Canpolat S, Kelestimur H. Leptin activates cytosolic calcium responses through protein kinase-C dependent mechanism in immortalized RFamide-related peptide-3 neurons. Brain Res 2015; 1601:8-14. [DOI: 10.1016/j.brainres.2014.12.053] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2014] [Revised: 12/20/2014] [Accepted: 12/29/2014] [Indexed: 10/24/2022]
|
13
|
Polyzos SA, Mantzoros CS. Leptin in health and disease: facts and expectations at its twentieth anniversary. Metabolism 2015; 64:5-12. [PMID: 25467841 DOI: 10.1016/j.metabol.2014.10.017] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 10/20/2014] [Accepted: 10/20/2014] [Indexed: 02/07/2023]
Affiliation(s)
- Stergios A Polyzos
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Christos S Mantzoros
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA; Boston VA Healthcare system and Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
14
|
Abstract
The blood-brain barrier (BBB) regulates the blood-to-brain passage of gastrointestinal hormones, thus informing the brain about feeding and nutritional status. Disruption of this communication results in dysregulation of feeding and body weight control. Leptin, which crosses the BBB to inform the CNS about adiposity, provides an example. Impaired leptin transport, especially coupled with central resistance, results in obesity. Various substances/conditions regulate leptin BBB transport. For example, triglycerides inhibit leptin transport. This may represent an evolutionary adaptation in that hypertriglyceridemia occurs during starvation. Inhibition of leptin, an anorectic, during starvation could have survival advantages. The large number of other substances that influence feeding is explained by the complexity of feeding. This complexity includes cognitive aspects; animals in the wild are faced with cost/benefit analyses to feed in the safest, most economical way. This cognitive aspect partially explains why so many feeding substances affect neurogenesis, neuroprotection, and cognition. The relation between triglycerides and cognition may be partially mediated through triglyceride's ability to regulate the BBB transport of cognitively active gastrointestinal hormones such as leptin, insulin, and ghrelin.
Collapse
Affiliation(s)
- William A Banks
- GRECC, Veterans Affairs Puget Sound Health Care System and Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington 98108, USA.
| |
Collapse
|
15
|
Banks WA, Gertler A, Solomon G, Niv-Spector L, Shpilman M, Yi X, Batrakova E, Vinogradov S, Kabanov AV. Principles of strategic drug delivery to the brain (SDDB): development of anorectic and orexigenic analogs of leptin. Physiol Behav 2011; 105:145-9. [PMID: 21669216 DOI: 10.1016/j.physbeh.2011.05.024] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2011] [Revised: 05/19/2011] [Accepted: 05/23/2011] [Indexed: 11/19/2022]
Abstract
The blood-brain barrier (BBB) presents a tremendous challenge for the delivery of drugs to the central nervous system (CNS). This includes drugs that target brain receptors for the treatment of obesity and anorexia. Strategic drug delivery to brain (SDDB) is an approach that considers in depth the relations among the BBB, the candidate therapeutic, the CNS target, and the disease state to be treated. Here, we illustrate principles of SDDB with two different approaches to developing drugs based on leptin. In normal body weight humans and in non-obese rodents, leptin is readily transported across the BBB and into the CNS where it inhibits feeding and enhances thermogenesis. However, in obesity, the transport of leptin across the BBB is impaired, resulting in a resistance to leptin. As a result, it is difficult to treat obesity with leptin or its analogs that depend on the leptin transporter for access to the CNS. To treat obesity, we developed a leptin agonist modified by the addition of pluronic block copolymers (P85-leptin). P85-leptin retains biological activity and is capable of crossing the BBB by a mechanism that is not dependent on the leptin transporter. As such, P85-leptin is able to cross the BBB of obese mice at a rate similar to that of native leptin in lean mice. To treat anorexia, we developed a leptin antagonist modified by pegylation (PEG-MLA) that acts primarily by blocking the BBB transporter for endogenous, circulating leptin. This prevents blood-borne, endogenous leptin from entering the CNS, essentially mimicking the leptin resistance seen in obesity, and resulting in a significant increase in adiposity. These examples illustrate two strategies in which an understanding of the interactions among the BBB, CNS targets, and candidate therapeutics under physiologic and diseased conditions can be used to develop drugs effective for the treatment of brain disease.
Collapse
Affiliation(s)
- W A Banks
- Geriatrics Research Education and Clinical Center - Veterans Affairs Puget Sound Health Care System and Division of Gerontology and Geriatric Medicine, Department of Internal Medicine, University of Washington, Seattle, WA, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Bhaskar S, Tian F, Stoeger T, Kreyling W, de la Fuente JM, Grazú V, Borm P, Estrada G, Ntziachristos V, Razansky D. Multifunctional Nanocarriers for diagnostics, drug delivery and targeted treatment across blood-brain barrier: perspectives on tracking and neuroimaging. Part Fibre Toxicol 2010; 7:3. [PMID: 20199661 PMCID: PMC2847536 DOI: 10.1186/1743-8977-7-3] [Citation(s) in RCA: 287] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2009] [Accepted: 03/03/2010] [Indexed: 01/03/2023] Open
Abstract
Nanotechnology has brought a variety of new possibilities into biological discovery and clinical practice. In particular, nano-scaled carriers have revolutionalized drug delivery, allowing for therapeutic agents to be selectively targeted on an organ, tissue and cell specific level, also minimizing exposure of healthy tissue to drugs. In this review we discuss and analyze three issues, which are considered to be at the core of nano-scaled drug delivery systems, namely functionalization of nanocarriers, delivery to target organs and in vivo imaging. The latest developments on highly specific conjugation strategies that are used to attach biomolecules to the surface of nanoparticles (NP) are first reviewed. Besides drug carrying capabilities, the functionalization of nanocarriers also facilitate their transport to primary target organs. We highlight the leading advantage of nanocarriers, i.e. their ability to cross the blood-brain barrier (BBB), a tightly packed layer of endothelial cells surrounding the brain that prevents high-molecular weight molecules from entering the brain. The BBB has several transport molecules such as growth factors, insulin and transferrin that can potentially increase the efficiency and kinetics of brain-targeting nanocarriers. Potential treatments for common neurological disorders, such as stroke, tumours and Alzheimer's, are therefore a much sought-after application of nanomedicine. Likewise any other drug delivery system, a number of parameters need to be registered once functionalized NPs are administered, for instance their efficiency in organ-selective targeting, bioaccumulation and excretion. Finally, direct in vivo imaging of nanomaterials is an exciting recent field that can provide real-time tracking of those nanocarriers. We review a range of systems suitable for in vivo imaging and monitoring of drug delivery, with an emphasis on most recently introduced molecular imaging modalities based on optical and hybrid contrast, such as fluorescent protein tomography and multispectral optoacoustic tomography. Overall, great potential is foreseen for nanocarriers in medical diagnostics, therapeutics and molecular targeting. A proposed roadmap for ongoing and future research directions is therefore discussed in detail with emphasis on the development of novel approaches for functionalization, targeting and imaging of nano-based drug delivery systems, a cutting-edge technology poised to change the ways medicine is administered.
Collapse
Affiliation(s)
- Sonu Bhaskar
- Instituto Universitario de Nanociencia de Aragón (INA), Universidad de Zaragoza, Zaragoza, Spain
- Zaragoza University Hospital-Miguel Servet, and Instituto Aragonés de Ciencias de la Salud (I+CS), Zaragoza, Spain
| | - Furong Tian
- Comprehensive Pneumology Centre, Institute of Lung Biology and Disease, Helmholtz Zentrum München, Neuherberg, Germany
| | - Tobias Stoeger
- Comprehensive Pneumology Centre, Institute of Lung Biology and Disease, Helmholtz Zentrum München, Neuherberg, Germany
| | - Wolfgang Kreyling
- Comprehensive Pneumology Centre, Institute of Lung Biology and Disease, Helmholtz Zentrum München, Neuherberg, Germany
| | - Jesús M de la Fuente
- Instituto Universitario de Nanociencia de Aragón (INA), Universidad de Zaragoza, Zaragoza, Spain
| | - Valeria Grazú
- Instituto Universitario de Nanociencia de Aragón (INA), Universidad de Zaragoza, Zaragoza, Spain
| | - Paul Borm
- Centre of Expertise in Life Sciences, Zuyd University, Heerlen, the Netherlands
| | - Giovani Estrada
- Institute of Bioinformatics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Vasilis Ntziachristos
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München, and Technische Universität München, Germany
| | - Daniel Razansky
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München, and Technische Universität München, Germany
| |
Collapse
|
17
|
Banks WA. Mouse models of neurological disorders: a view from the blood-brain barrier. Biochim Biophys Acta Mol Basis Dis 2009; 1802:881-8. [PMID: 19879356 DOI: 10.1016/j.bbadis.2009.10.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2009] [Revised: 10/20/2009] [Accepted: 10/23/2009] [Indexed: 12/16/2022]
Abstract
The number of disease models that involve an aspect of blood-brain barrier (BBB) dysregulation have increased tremendously. The main factors contributing to this expansion have been an increased number of diseases in which the BBB is known to be involved, an increase in the known functions of the BBB, and an increase in the number of models and tools with which those diverse functions can be studied. In many cases, the BBB may be a target of disease; current thinking would include hypertensive encephalopathy and perhaps stroke in this category. Another category are those diseases in which special attributes of the BBB may predispose to disease; for example, the ability of a pathogen to cross the BBB often depends on the pathogen's ability to invoke transcytotic pathways in the brain endothelial or choroid plexus cell. Of special interest are those diseases in which the BBB may be the primary seat of disease or play a major role in the onset or progression of the disease. An increasing number of diseases are so categorized in which BBB dysfunction or dysregulation plays a major role; this review highlights such roles for the BBB including those proposed for Alzheimer's disease and obesity.
Collapse
Affiliation(s)
- William A Banks
- GRECC, Veterans Affairs Medical Center-St. Louis and Saint Louis University School of Medicine, Division of Geriatrics, Department of Internal Medicine, 915 N. Grand Blvd, St. Louis, MO 63106, USA.
| |
Collapse
|
18
|
Abstract
Interleukin15 (IL 15) is a proinflammatory cytokine with elevated concentrations in autoimmune diseases involving the periphery (e.g. rheumatoid arthritis) and CNS (e.g. multiple sclerosis). Its interactions with the blood-brain barrier (BBB) were studied in normal and lipopolysaccharide (LPS)-treated mice. (125)I-IL15 remained intact for at least 10 min after i.v. injection and reached CNS parenchyma with regional differences between brain and spinal cord. Both in vivo and in situ brain perfusion of (125)I-IL15 showed that its permeation of the BBB was non-saturable. LPS induced a significant increase of IL15 uptake by the brain and spinal cord, partly related to a higher general permeability of the BBB. The results suggest that the BBB is an interface for blood-borne IL15 to interact with the CNS in the basal state and during inflammation.
Collapse
Affiliation(s)
- Weihong Pan
- Pennington Biomedical Research Center, Baton Rouge, Louisiana 70808, USA.
| | | | | | | |
Collapse
|