1
|
van Setten A, Uleman JF, Melis RJF, Lawlor B, Riksen NP, Claassen JAHR, de Heus RAA. No association between markers of systemic inflammation and endothelial dysfunction with Alzheimer's disease progression: a longitudinal study. GeroScience 2024:10.1007/s11357-024-01294-x. [PMID: 39085534 DOI: 10.1007/s11357-024-01294-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 07/18/2024] [Indexed: 08/02/2024] Open
Abstract
INTRODUCTION Systemic inflammation and endothelial dysfunction are potentially modifiable factors implicated in Alzheimer's disease (AD), which offer potential therapeutic targets to slow disease progression. METHODS We investigated the relationship between baseline circulating levels of inflammatory (TNF-α, IL-1ß) and endothelial cell markers (VCAM-1, ICAM-1, E-selectin) and 18-month cognitive decline (ADAS-cog12) in 266 mild-to-moderate AD patients from the NILVAD study. We employed individual growth models to examine associations, potential mediation, and interaction effects while adjusting for confounders. RESULTS The average increase in ADAS-cog12 scores over all patients was 8.1 points in 18 months. No significant association was found between the markers and the rate of cognitive decline. Mediation analysis revealed no mediating role for endothelial cell markers, and interaction effects were not observed. DISCUSSION Our results do not support the role of systemic inflammation or endothelial dysfunction in progression in persons with AD.
Collapse
Affiliation(s)
- Arne van Setten
- Department of Geriatric Medicine, Radboudumc Alzheimer Center, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jeroen F Uleman
- Copenhagen Health Complexity Center, Department of Public Health, University of Copenhagen, Oster Farimagsgade 5, 1353, Copenhagen K, Denmark.
| | - René J F Melis
- Department of Geriatric Medicine, Radboudumc Alzheimer Center, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Brian Lawlor
- Global Brain Health Institute, Trinity College, Dublin, Ireland
| | - Niels P Riksen
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jurgen A H R Claassen
- Department of Geriatric Medicine, Radboudumc Alzheimer Center, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Rianne A A de Heus
- Department of Geriatric Medicine, Radboudumc Alzheimer Center, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Primary and Community Care, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
2
|
Rhea EM, Banks WA. Insulin and the blood-brain barrier. VITAMINS AND HORMONES 2024; 126:169-190. [PMID: 39029972 DOI: 10.1016/bs.vh.2024.02.002] [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
The blood-brain barrier (BBB) predominantly regulates insulin transport into and levels within the brain. The BBB is also an important site of insulin binding and mediator of insulin receptor (INSR) signaling. The insulin transporter is separate from the INSR, highlighting the important, unique role of each protein in this structure. After a brief introduction on the structure of insulin and the INSR, we discuss the importance of insulin interactions at the BBB, the properties of the insulin transporter and the role of the BBB insulin transporter in various physiological conditions. We go on to further describe insulin BBB signaling and the impact not only within brain endothelial cells but also the cascade into other cell types within the brain. We close with future considerations to advance our knowledge about the importance of insulin at the BBB.
Collapse
Affiliation(s)
- Elizabeth M Rhea
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States; Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA, United States.
| | - William A Banks
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States; Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA, United States
| |
Collapse
|
3
|
Banks WA, Hansen KM, Erickson MA, Crews FT. High-mobility group box 1 (HMGB1) crosses the BBB bidirectionally. Brain Behav Immun 2023; 111:386-394. [PMID: 37146655 DOI: 10.1016/j.bbi.2023.04.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 04/25/2023] [Accepted: 04/30/2023] [Indexed: 05/07/2023] Open
Abstract
High-mobility group box 1 (HMGB1) is a ubiquitous protein that regulates transcription in the nucleus, and is an endogenous damage-associated molecular pattern molecule that activates the innate immune system. HMGB1 activates the TLR4 and RAGE recepto, inducing downstream signals reminiscent of cytokines that have been found to cross the blood-brain barrier (BBB). Blood HMGB1 increases in stroke, sepsis, senescence, alcohol binge drinking and other conditions. Here, we examined the ability of HMGB1 radioactively labeled with iodine (I-HMGB1) to cross the BBB. We found that I-HMGB1 readily entered into mouse brain from the circulation with a unidirectional influx rate of 0.654 μl/g-min. All brain regions tested took up I-HMGB1; uptake was greatest by the olfactory bulb and least in the striatum. Transport was not reliably inhibited by unlabeled HMGB1 nor by inhibitors of TLR4, TLR2, RAGE, or CXCR4. Uptake was enhanced by co-injection of wheatgerm agglutinin, suggestive of involvement of absorptive transcytosis as a mechanism of transport. Induction of inflammation/neuroinflammation with lipopolysaccharide is known to increase blood HMGB1; we report here that brain transport is also increased by LPS-induced inflammation. Finally, we found that I-HMGB1 was also transported in the brain-to-blood direction, with both unlabeled HMGB1 or lipopolysaccharide increasing the transport rate. These results show that HMGB1 can bidirectionally cross the BBB and that those transport rates are enhanced by inflammation. Such transport provides a mechanism by which HMGB1 levels would impact neuroimmune signaling in both the brain and periphery.
Collapse
Affiliation(s)
- William A Banks
- Geriatric Research Educational and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, US State; Division of Gerontology and Geriatric Medicine, University of Washington School of Medicine, Seattle, WA, US State.
| | - Kim M Hansen
- Geriatric Research Educational and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, US State; Division of Gerontology and Geriatric Medicine, University of Washington School of Medicine, Seattle, WA, US State
| | - Michelle A Erickson
- Geriatric Research Educational and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, US State; Division of Gerontology and Geriatric Medicine, University of Washington School of Medicine, Seattle, WA, US State
| | - Fulton T Crews
- Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, US State
| |
Collapse
|
4
|
Abdelsalam M, Ahmed M, Osaid Z, Hamoudi R, Harati R. Insights into Exosome Transport through the Blood-Brain Barrier and the Potential Therapeutical Applications in Brain Diseases. Pharmaceuticals (Basel) 2023; 16:571. [PMID: 37111328 PMCID: PMC10144189 DOI: 10.3390/ph16040571] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 03/31/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
Drug delivery to the central nervous system (CNS) is limited due to the presence of the blood-brain barrier (BBB), a selective physiological barrier located at the brain microvessels that regulates the flow of cells, molecules and ions between the blood and the brain. Exosomes are nanosized extracellular vesicles expressed by all cell types and that function as cargos, allowing for communication between the cells. The exosomes were shown to cross or regulate the BBB in healthy and disease conditions. However, the mechanistic pathways by which exosomes cross the BBB have not been fully elucidated yet. In this review, we explore the transport mechanisms of exosomes through the BBB. A large body of evidence suggests that exosome transport through the BBB occurs primarily through transcytosis. The transcytosis mechanisms are influenced by several regulators. Inflammation and metastasis also enhance exosome trafficking across the BBB. We also shed light on the therapeutical applications of exosomes for treating brain diseases. Further investigations are essential to provide clearer insights related to trafficking of exosomes across the BBB and disease treatment.
Collapse
Affiliation(s)
- Manal Abdelsalam
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (M.A.); (M.A.); (Z.O.)
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
| | - Munazza Ahmed
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (M.A.); (M.A.); (Z.O.)
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
| | - Zaynab Osaid
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (M.A.); (M.A.); (Z.O.)
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
| | - Rifat Hamoudi
- Clinical Sciences Department, College of Medicine, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates;
- Division of Surgery and Interventional Science, University College London, London W1W 7EJ, UK
| | - Rania Harati
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (M.A.); (M.A.); (Z.O.)
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
| |
Collapse
|
5
|
Shpakov AO, Zorina II, Derkach KV. Hot Spots for the Use of Intranasal Insulin: Cerebral Ischemia, Brain Injury, Diabetes Mellitus, Endocrine Disorders and Postoperative Delirium. Int J Mol Sci 2023; 24:3278. [PMID: 36834685 PMCID: PMC9962062 DOI: 10.3390/ijms24043278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/27/2023] [Accepted: 01/31/2023] [Indexed: 02/11/2023] Open
Abstract
A decrease in the activity of the insulin signaling system of the brain, due to both central insulin resistance and insulin deficiency, leads to neurodegeneration and impaired regulation of appetite, metabolism, endocrine functions. This is due to the neuroprotective properties of brain insulin and its leading role in maintaining glucose homeostasis in the brain, as well as in the regulation of the brain signaling network responsible for the functioning of the nervous, endocrine, and other systems. One of the approaches to restore the activity of the insulin system of the brain is the use of intranasally administered insulin (INI). Currently, INI is being considered as a promising drug to treat Alzheimer's disease and mild cognitive impairment. The clinical application of INI is being developed for the treatment of other neurodegenerative diseases and improve cognitive abilities in stress, overwork, and depression. At the same time, much attention has recently been paid to the prospects of using INI for the treatment of cerebral ischemia, traumatic brain injuries, and postoperative delirium (after anesthesia), as well as diabetes mellitus and its complications, including dysfunctions in the gonadal and thyroid axes. This review is devoted to the prospects and current trends in the use of INI for the treatment of these diseases, which, although differing in etiology and pathogenesis, are characterized by impaired insulin signaling in the brain.
Collapse
Affiliation(s)
- Alexander O. Shpakov
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 194223 St. Petersburg, Russia
| | | | | |
Collapse
|
6
|
Banks WA, Noonan C, Rhea EM. Evidence for an alternative insulin transporter at the blood-brain barrier. AGING PATHOBIOLOGY AND THERAPEUTICS 2022; 4:100-108. [PMID: 36644126 PMCID: PMC9837797 DOI: 10.31491/apt.2022.12.100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Accumulating evidence suggests there is an alternative insulin transporter besides the insulin receptor at the blood-brain barrier (BBB), responsible for shuttling insulin from the circulation into the brain. In this review, we summarize key features of the BBB and what makes it unique compared to other capillary beds; summarize what we know about insulin BBB transport; provide an extensive list of diseases, physiological states, and serum factors tested in modifying insulin BBB transport; and lastly, highlight potential alternative transport systems that may be involved in or have already been tested in mediating insulin BBB transport. Identifying the transport system for insulin at the BBB would aide in controlling central nervous system (CNS) insulin levels in multiple diseases and conditions including Alzheimer's disease (AD) and obesity, where availability of insulin to the CNS is limited.
Collapse
Affiliation(s)
- William A. Banks
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington, Seattle, WA 98195, USA
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Cassidy Noonan
- Research and Development, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
- University of Washington, Seattle, WA 98195, USA
| | - Elizabeth M. Rhea
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington, Seattle, WA 98195, USA
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
| |
Collapse
|
7
|
Rhea EM, Banks WA, Raber J. Insulin Resistance in Peripheral Tissues and the Brain: A Tale of Two Sites. Biomedicines 2022; 10:1582. [PMID: 35884888 PMCID: PMC9312939 DOI: 10.3390/biomedicines10071582] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/27/2022] [Accepted: 06/29/2022] [Indexed: 12/12/2022] Open
Abstract
The concept of insulin resistance has been around since a few decades after the discovery of insulin itself. To allude to the classic Charles Dicken's novel published 62 years before the discovery of insulin, in some ways, this is the best of times, as the concept of insulin resistance has expanded to include the brain, with the realization that insulin has a life beyond the regulation of glucose. In other ways, it is the worst of times as insulin resistance is implicated in devastating diseases, including diabetes mellitus, obesity, and Alzheimer's disease (AD) that affect the brain. Peripheral insulin resistance affects nearly a quarter of the United States population in adults over age 20. More recently, it has been implicated in AD, with the degree of brain insulin resistance correlating with cognitive decline. This has led to the investigation of brain or central nervous system (CNS) insulin resistance and the question of the relation between CNS and peripheral insulin resistance. While both may involve dysregulated insulin signaling, the two conditions are not identical and not always interlinked. In this review, we compare and contrast the similarities and differences between peripheral and CNS insulin resistance. We also discuss how an apolipoprotein involved in insulin signaling and related to AD, apolipoprotein E (apoE), has distinct pools in the periphery and CNS and can indirectly affect each system. As these systems are both separated but also linked via the blood-brain barrier (BBB), we discuss the role of the BBB in mediating some of the connections between insulin resistance in the brain and in the peripheral tissues.
Collapse
Affiliation(s)
- Elizabeth M. Rhea
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington, Seattle, WA 98195, USA; (E.M.R.); (W.A.B.)
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
| | - William A. Banks
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington, Seattle, WA 98195, USA; (E.M.R.); (W.A.B.)
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA
- Departments of Neurology and Radiation Medicine, Division of Neuroscience, ONPRC, Oregon Health & Science University, Portland, OR 97239, USA
| |
Collapse
|
8
|
Caioni G, Cimini A, Benedetti E. Food Contamination: An Unexplored Possible Link between Dietary Habits and Parkinson’s Disease. Nutrients 2022; 14:nu14071467. [PMID: 35406080 PMCID: PMC9003245 DOI: 10.3390/nu14071467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/29/2022] [Accepted: 03/29/2022] [Indexed: 12/10/2022] Open
Abstract
Importance of a healthy lifestyle in maintaining the population’s well-being and health, especially in terms of balanced nutrition, is well known. Food choice of and dieting habits could impact disease management, which is especially true for Parkinson’s disease (PD). However, nowadays, it is not that simple to maintain a balance in nutrition, and the idea of a healthy diet tends to fade as the consequence of a western lifestyle. This should not only be dealt with in the context of food choice, but also from an environmental point of view. What we put into our bodies is strictly related to the quality of ecosystems we live in. For these reasons, attention should be directed to all the pollutants, which in many cases, we unknowingly ingest. It will be necessary to explore the interaction between food and environment, since human activity also influences the raw materials destined for consumption. This awareness can be achieved by means of an innovative scientific approach, which involves the use of new models, in order to overcome the traditional scientific investigations included in the study of Parkinson’s disease.
Collapse
Affiliation(s)
- Giulia Caioni
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (G.C.); (A.C.)
| | - Annamaria Cimini
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (G.C.); (A.C.)
- Department of Biology, Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA 19122, USA
| | - Elisabetta Benedetti
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (G.C.); (A.C.)
- Correspondence: ; Tel.: +39-086-243-3267
| |
Collapse
|
9
|
Galea I. The blood-brain barrier in systemic infection and inflammation. Cell Mol Immunol 2021; 18:2489-2501. [PMID: 34594000 PMCID: PMC8481764 DOI: 10.1038/s41423-021-00757-x] [Citation(s) in RCA: 225] [Impact Index Per Article: 75.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 08/04/2021] [Indexed: 02/08/2023] Open
Abstract
The vascular blood-brain barrier is a highly regulated interface between the blood and brain. Its primary function is to protect central neurons while signaling the presence of systemic inflammation and infection to the brain to enable a protective sickness behavior response. With increasing degrees and duration of systemic inflammation, the vascular blood-brain barrier becomes more permeable to solutes, undergoes an increase in lymphocyte trafficking, and is infiltrated by innate immune cells; endothelial cell damage may occasionally occur. Perturbation of neuronal function results in the clinical features of encephalopathy. Here, the molecular and cellular anatomy of the vascular blood-brain barrier is reviewed, first in a healthy context and second in a systemic inflammatory context. Distinct from the molecular and cellular mediators of the blood-brain barrier's response to inflammation, several moderators influence the direction and magnitude at genetic, system, cellular and molecular levels. These include sex, genetic background, age, pre-existing brain pathology, systemic comorbidity, and gut dysbiosis. Further progress is required to define and measure mediators and moderators of the blood-brain barrier's response to systemic inflammation in order to explain the heterogeneity observed in animal and human studies.
Collapse
Affiliation(s)
- Ian Galea
- grid.5491.90000 0004 1936 9297Clinical Neurosciences, Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD UK
| |
Collapse
|
10
|
The Blood-Brain Barrier, Oxidative Stress, and Insulin Resistance. Antioxidants (Basel) 2021; 10:antiox10111695. [PMID: 34829566 PMCID: PMC8615183 DOI: 10.3390/antiox10111695] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 10/22/2021] [Accepted: 10/25/2021] [Indexed: 12/12/2022] Open
Abstract
The blood–brain barrier (BBB) is a network of specialized endothelial cells that regulates substrate entry into the central nervous system (CNS). Acting as the interface between the periphery and the CNS, the BBB must be equipped to defend against oxidative stress and other free radicals generated in the periphery to protect the CNS. There are unique features of brain endothelial cells that increase the susceptibility of these cells to oxidative stress. Insulin signaling can be impacted by varying levels of oxidative stress, with low levels of oxidative stress being necessary for signaling and higher levels being detrimental. Insulin must cross the BBB in order to access the CNS, levels of which are important in peripheral metabolism as well as cognition. Any alterations in BBB transport due to oxidative stress at the BBB could have downstream disease implications. In this review, we cover the interactions of oxidative stress at the BBB, how insulin signaling is related to oxidative stress, and the impact of the BBB in two diseases greatly affected by oxidative stress and insulin resistance: diabetes mellitus and Alzheimer’s disease.
Collapse
|
11
|
Rhea EM, Banks WA. A historical perspective on the interactions of insulin at the blood-brain barrier. J Neuroendocrinol 2021; 33:e12929. [PMID: 33433042 PMCID: PMC8052275 DOI: 10.1111/jne.12929] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/18/2020] [Accepted: 12/02/2020] [Indexed: 11/30/2022]
Abstract
Subsequent to the discovery of insulin in 1921, the role of insulin in the brain has been investigated throughly. The ability of insulin to act within the brain to regulate peripheral glucose levels helped evolve the research surrounding the ability of insulin to be transported into the brain. Investigations aiming to determine the transport of insulin into the brain from the circulation soon followed. Once it was established that insulin could enter the brain, the ability of insulin to bind brain microvessels and regulators of this process were determined. As technology advanced, quantitative measurements to specify the transport rate of insulin across the blood-brain barrier (BBB) and the impact of physiological conditions and diseases were the logical next steps. Lastly, with the advent of genetic mouse models and high-specificity antagonists, the specific role of the insulin receptor in mediating insulin transport could begin to be explored. In this review, we summarise the main findings throughout the decades regarding the interactions of insulin at the BBB.
Collapse
Affiliation(s)
- Elizabeth M. Rhea
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA 98159
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, 1660 S Columbian Way, Seattle, Washington, USA 98108
- Corresponding author: Elizabeth M. Rhea;
| | - William A. Banks
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA 98159
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, 1660 S Columbian Way, Seattle, Washington, USA 98108
| |
Collapse
|
12
|
Reddiar SB, Jin L, Wai DCC, Csoti A, Panyi G, Norton RS, Nicolazzo JA. Lipopolysaccharide influences the plasma and brain pharmacokinetics of subcutaneously-administered HsTX1[R14A], a K V1.3-blocking peptide. Toxicon 2021; 195:29-36. [PMID: 33689790 DOI: 10.1016/j.toxicon.2021.03.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 02/22/2021] [Accepted: 03/03/2021] [Indexed: 12/01/2022]
Abstract
KV1.3 is a voltage-gated potassium channel that is upregulated in neuroinflammatory conditions, such as Alzheimer's disease and Parkinson's disease. HsTX1[R14A] is a potent and selective peptide blocker of KV1.3 with the potential to block microglial KV1.3, but its brain uptake is expected to be limited owing to the restrictive nature of the blood-brain barrier. To assess its peripheral and brain exposure, a LC-MS/MS assay was developed to quantify HsTX1[R14A] concentrations in mouse plasma and brain homogenate that was reliable and reproducible in the range of 6.7-66.7 nM (r2 = 0.9765) and 15-150 pmol/g (r2 = 0.9984), respectively. To assess if neuroinflammation affected HsTX1[R14A] disposition, C57BL/6 mice were administered HsTX1[R14A] subcutaneously (2 mg/kg) 24 h after an intraperitoneal dose of Escherichia coli lipopolysaccharide (LPS), which is commonly used to induce neuroinflammation; brain and plasma concentrations of HsTX1[R14A] were then quantified over 120 min. LPS treatment significantly retarded the decline in HsTX1[R14A] plasma concentrations, presumably as a result of reducing renal clearance, and led to substantial brain uptake of HsTX1[R14A], presumably through disruption of brain inter-endothelial tight junctions. This study suggests that HsTX1[R14A] may reach microglia in sufficient concentrations to block KV1.3 in neuroinflammatory conditions, and therefore has the potential to reduce neurodegenerative diseases.
Collapse
Affiliation(s)
- Sanjeevini Babu Reddiar
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Liang Jin
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Dorothy C C Wai
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Agota Csoti
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Hungary
| | - Gyorgy Panyi
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Hungary
| | - Raymond S Norton
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia; ARC Centre for Fragment-Based Design, Monash University, Parkville, Victoria, Australia
| | - Joseph A Nicolazzo
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia.
| |
Collapse
|
13
|
Simon F, Guyot L, Garcia J, Vilchez G, Bardel C, Chenel M, Tod M, Payen L. Impact of interleukin‐6 on drug transporters and permeability in the hCMEC/D3 blood–brain barrier model. Fundam Clin Pharmacol 2020; 35:397-409. [DOI: 10.1111/fcp.12596] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 07/27/2020] [Accepted: 07/30/2020] [Indexed: 12/17/2022]
Affiliation(s)
- Florian Simon
- EA3738 Faculté de médecine de Lyon‐Sud Université de Lyon 1 165 chemin du Grand Revoyet, Faculté de médecine et maïeutique Oullins France69921France
- Laboratoire de Biochimie‐Toxicologie Centre Hospitalier Lyon‐Sud Hospices civils de Lyon 165 chemin du Grand Revoyet Pierre‐Bénite France69310France
- Institut de Recherches Internationales Servier Direction of Clinical PK and Pharmacometrics 50 rue Carnot Suresnes92150France
| | - Laetitia Guyot
- Laboratoire de Biochimie‐Toxicologie Centre Hospitalier Lyon‐Sud Hospices civils de Lyon 165 chemin du Grand Revoyet Pierre‐Bénite France69310France
| | - Jessica Garcia
- Laboratoire de Biochimie‐Toxicologie Centre Hospitalier Lyon‐Sud Hospices civils de Lyon 165 chemin du Grand Revoyet Pierre‐Bénite France69310France
| | - Gaelle Vilchez
- Hospices Civils de Lyon Department of Biostatistics 162 avenue Lacassagne Lyon69424France
| | - Claire Bardel
- Hospices Civils de Lyon Department of Biostatistics 162 avenue Lacassagne Lyon69424France
| | - Marylore Chenel
- Institut de Recherches Internationales Servier Direction of Clinical PK and Pharmacometrics 50 rue Carnot Suresnes92150France
| | - Michel Tod
- EA3738 Faculté de médecine de Lyon‐Sud Université de Lyon 1 165 chemin du Grand Revoyet, Faculté de médecine et maïeutique Oullins France69921France
| | - Léa Payen
- Laboratoire de Biochimie‐Toxicologie Centre Hospitalier Lyon‐Sud Hospices civils de Lyon 165 chemin du Grand Revoyet Pierre‐Bénite France69310France
| |
Collapse
|
14
|
Galindo DC, Banks WA, Rhea EM. The impact of acute rosiglitazone on insulin pharmacokinetics at the blood-brain barrier. Endocrinol Diabetes Metab 2020; 3:e00149. [PMID: 32704569 PMCID: PMC7375048 DOI: 10.1002/edm2.149] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 05/02/2020] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION CNS insulin levels are decreased and insulin receptor signalling is dampened in Alzheimer's disease (AD). Increasing CNS insulin levels through a variety of methods has been shown to improve memory. Indeed, medications routinely used to improve insulin resistance in type 2 diabetes are now being repurposed for memory enhancement. CNS insulin is primarily derived from the circulation, by an active transport system at the blood-brain barrier (BBB). The goal of this study was to determine whether rosiglitazone (RSG), a drug used to improve insulin sensitivity in type 2 diabetes, could enhance insulin transport at the BBB, as a potential therapeutic for improving memory. METHODS Using radioactively labelled insulin and the multiple-time regression analysis technique, we measured the rate of insulin BBB transport and level of vascular binding in mice pretreated with vehicle or 10 µg RSG in the presence or absence of an insulin receptor inhibitor. RESULTS Although we found acute RSG administration does not affect insulin transport at the BBB, it does restore BBB vascular binding of insulin in an insulin receptor-resistant state. CONCLUSIONS Acute RSG treatment does not alter insulin BBB transport in healthy mice but can restore insulin receptor binding at the BBB in an insulin-resistant state.
Collapse
Affiliation(s)
| | - William A. Banks
- Department of MedicineUniversity of WashingtonSeattleWAUSA
- Research and DevelopmentVeterans Affairs Puget Sound Healthcare SystemSeattleWAUSA
| | - Elizabeth M. Rhea
- Department of MedicineUniversity of WashingtonSeattleWAUSA
- Research and DevelopmentVeterans Affairs Puget Sound Healthcare SystemSeattleWAUSA
| |
Collapse
|
15
|
Transport of Extracellular Vesicles across the Blood-Brain Barrier: Brain Pharmacokinetics and Effects of Inflammation. Int J Mol Sci 2020; 21:ijms21124407. [PMID: 32575812 PMCID: PMC7352415 DOI: 10.3390/ijms21124407] [Citation(s) in RCA: 270] [Impact Index Per Article: 67.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 06/08/2020] [Accepted: 06/10/2020] [Indexed: 01/08/2023] Open
Abstract
Extracellular vesicles can cross the blood–brain barrier (BBB), but little is known about passage. Here, we used multiple-time regression analysis to examine the ability of 10 exosome populations derived from mouse, human, cancerous, and non-cancerous cell lines to cross the BBB. All crossed the BBB, but rates varied over 10-fold. Lipopolysaccharide (LPS), an activator of the innate immune system, enhanced uptake independently of BBB disruption for six exosomes and decreased uptake for one. Wheatgerm agglutinin (WGA) modulated transport of five exosome populations, suggesting passage by adsorptive transcytosis. Mannose 6-phosphate inhibited uptake of J774A.1, demonstrating that its BBB transporter is the mannose 6-phosphate receptor. Uptake rates, patterns, and effects of LPS or WGA were not predicted by exosome source (mouse vs. human) or cancer status of the cell lines. The cell surface proteins CD46, AVβ6, AVβ3, and ICAM-1 were variably expressed but not predictive of transport rate nor responses to LPS or WGA. A brain-to-blood efflux mechanism variably affected CNS retention and explains how CNS-derived exosomes enter blood. In summary, all exosomes tested here readily crossed the BBB, but at varying rates and by a variety of vesicular-mediated mechanisms involving specific transporters, adsorptive transcytosis, and a brain-to-blood efflux system.
Collapse
|
16
|
Logsdon AF, Schindler AG, Meabon JS, Yagi M, Herbert MJ, Banks WA, Raskind MA, Marshall DA, Keene CD, Perl DP, Peskind ER, Cook DG. Nitric oxide synthase mediates cerebellar dysfunction in mice exposed to repetitive blast-induced mild traumatic brain injury. Sci Rep 2020; 10:9420. [PMID: 32523011 PMCID: PMC7287110 DOI: 10.1038/s41598-020-66113-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 03/16/2020] [Indexed: 02/02/2023] Open
Abstract
We investigated the role of nitric oxide synthase (NOS) in mediating blood-brain barrier (BBB) disruption and peripheral immune cell infiltration in the cerebellum following blast exposure. Repetitive, but not single blast exposure, induced delayed-onset BBB disruption (72 hours post-blast) in cerebellum. The NOS inhibitor N(G)-nitro-L-arginine methyl ester (L-NAME) administered after blast blocked BBB disruption and prevented CD4+ T-cell infiltration into cerebellum. L-NAME also blocked blast-induced increases in intercellular adhesion molecule-1 (ICAM-1), a molecule that plays a critical role in regulating blood-to-brain immune cell trafficking. Blocking NOS-mediated BBB dysfunction during this acute/subacute post-blast interval (24-71 hours after the last blast) also prevented sensorimotor impairment on a rotarod task 30 days later, long after L-NAME cleared the body. In postmortem brains from Veterans/military Servicemembers with blast-related TBI, we found marked Purkinje cell dendritic arbor structural abnormalities, which were comparable to neuropathologic findings in the blast-exposed mice. Taken collectively, these results indicate that blast provokes delayed-onset of NOS-dependent pathogenic cascades that can later emerge as behavioral dysfunction. These results also further implicate the cerebellum as a brain region vulnerable to blast-induced mTBI.
Collapse
Affiliation(s)
- Aric F. Logsdon
- 0000 0004 0420 6540grid.413919.7Geriatric Research Education and Clinical Center (GRECC), Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108 USA ,0000000122986657grid.34477.33Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195 USA
| | - Abigail G. Schindler
- 0000 0004 0420 6540grid.413919.7Geriatric Research Education and Clinical Center (GRECC), Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108 USA ,0000000122986657grid.34477.33Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA 98195 USA
| | - James S. Meabon
- 0000 0004 0420 6540grid.413919.7VA Northwest Mental Illness Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108 USA ,0000000122986657grid.34477.33Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA 98195 USA
| | - Mayumi Yagi
- 0000 0004 0420 6540grid.413919.7Geriatric Research Education and Clinical Center (GRECC), Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108 USA
| | - Melanie J. Herbert
- 0000 0004 0420 6540grid.413919.7Geriatric Research Education and Clinical Center (GRECC), Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108 USA
| | - William A. Banks
- 0000 0004 0420 6540grid.413919.7Geriatric Research Education and Clinical Center (GRECC), Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108 USA ,0000000122986657grid.34477.33Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195 USA
| | - Murray A. Raskind
- 0000 0004 0420 6540grid.413919.7VA Northwest Mental Illness Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108 USA ,0000000122986657grid.34477.33Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA 98195 USA
| | - Desiree A. Marshall
- 0000000122986657grid.34477.33Department of Pathology, University of Washington, Seattle, WA 98195 USA
| | - C. Dirk Keene
- 0000000122986657grid.34477.33Department of Pathology, University of Washington, Seattle, WA 98195 USA
| | - Daniel P. Perl
- 0000 0001 0421 5525grid.265436.0Department of Pathology, Center for Neuroscience and Regenerative Medicine, School of Medicine, Uniformed Services University, Bethesda, MD 20814 USA
| | - Elaine R. Peskind
- 0000 0004 0420 6540grid.413919.7VA Northwest Mental Illness Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108 USA ,0000000122986657grid.34477.33Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA 98195 USA
| | - David G. Cook
- 0000 0004 0420 6540grid.413919.7Geriatric Research Education and Clinical Center (GRECC), Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108 USA ,0000000122986657grid.34477.33Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195 USA
| |
Collapse
|
17
|
Rhea EM, Raber J, Banks WA. ApoE and cerebral insulin: Trafficking, receptors, and resistance. Neurobiol Dis 2020; 137:104755. [PMID: 31978603 PMCID: PMC7050417 DOI: 10.1016/j.nbd.2020.104755] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 01/13/2020] [Accepted: 01/20/2020] [Indexed: 12/16/2022] Open
Abstract
Central nervous system (CNS) insulin resistance is associated with Alzheimer's disease (AD). In addition, the apolipoprotein E4 (apoE4) isoform is a risk factor for AD. The connection between these two factors in relation to AD is being actively explored. We summarize this literature with a focus on the transport of insulin and apoE across the blood-brain barrier (BBB) and into the CNS, the impact of apoE and insulin on the BBB, and the interactions between apoE, insulin, and the insulin receptor once present in the CNS. We highlight how CNS insulin resistance is apparent in AD and potential ways to overcome this resistance by repurposing currently approved drugs, with apoE genotype taken into consideration as the treatment response following most interventions is apoE isoform-dependent. This review is part of a special issue focusing on apoE in AD and neurodegeneration.
Collapse
Affiliation(s)
- Elizabeth M Rhea
- Research and Development, Veterans Affairs Puget Sound Healthcare System, Seattle, WA 98108, United States of America; Department of Medicine, University of Washington, Seattle, WA 98195, United States of America.
| | - Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, United States of America; Departments of Neurology and Radiation Medicine, Division of Neuroscience, ONPRC, Oregon Health & Science University, Portland, OR 97239, United States of America
| | - William A Banks
- Research and Development, Veterans Affairs Puget Sound Healthcare System, Seattle, WA 98108, United States of America; Department of Medicine, University of Washington, Seattle, WA 98195, United States of America
| |
Collapse
|
18
|
Erickson MA, Wilson ML, Banks WA. In vitro modeling of blood-brain barrier and interface functions in neuroimmune communication. Fluids Barriers CNS 2020; 17:26. [PMID: 32228633 PMCID: PMC7106666 DOI: 10.1186/s12987-020-00187-3] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 03/18/2020] [Indexed: 02/07/2023] Open
Abstract
Neuroimmune communication contributes to both baseline and adaptive physiological functions, as well as disease states. The vascular blood-brain barrier (BBB) and associated cells of the neurovascular unit (NVU) serve as an important interface for immune communication between the brain and periphery through the blood. Immune functions and interactions of the BBB and NVU in this context can be categorized into at least five neuroimmune axes, which include (1) immune modulation of BBB impermeability, (2) immune regulation of BBB transporters, secretions, and other functions, (3) BBB uptake and transport of immunoactive substances, (4) immune cell trafficking, and (5) BBB secretions of immunoactive substances. These axes may act separately or in concert to mediate various aspects of immune signaling at the BBB. Much of what we understand about immune axes has been from work conducted using in vitro BBB models, and recent advances in BBB and NVU modeling highlight the potential of these newer models for improving our understanding of how the brain and immune system communicate. In this review, we discuss how conventional in vitro models of the BBB have improved our understanding of the 5 neuroimmune axes. We further evaluate the existing literature on neuroimmune functions of novel in vitro BBB models, such as those derived from human induced pluripotent stem cells (iPSCs) and discuss their utility in evaluating aspects of neuroimmune communication.
Collapse
Affiliation(s)
- Michelle A Erickson
- Geriatric Research Education and Clinical Center, VA Puget Sound Healthcare System, Seattle, WA, 98108, USA.,Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA, 98104, USA
| | - Miranda L Wilson
- Geriatric Research Education and Clinical Center, VA Puget Sound Healthcare System, Seattle, WA, 98108, USA
| | - William A Banks
- Geriatric Research Education and Clinical Center, VA Puget Sound Healthcare System, Seattle, WA, 98108, USA. .,Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA, 98104, USA.
| |
Collapse
|
19
|
Gazerani P. Probiotics for Parkinson's Disease. Int J Mol Sci 2019; 20:E4121. [PMID: 31450864 PMCID: PMC6747430 DOI: 10.3390/ijms20174121] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 08/14/2019] [Accepted: 08/21/2019] [Indexed: 02/08/2023] Open
Abstract
Parkinson's disease (PD) is a complex neurological disorder classically characterized by impairments in motor system function associated with loss of dopaminergic neurons in the substantia nigra. After almost 200 years since the first description of PD by James Parkinson, unraveling the complexity of PD continues to evolve. It is now recognized that an interplay between genetic and environmental factors influences a diverse range of cellular processes, reflecting on other clinical features including non-motor symptoms. This has consequently highlighted the extensive value of early clinical diagnosis to reduce difficulties of later stage management of PD. Advancement in understanding of PD has made remarkable progress in introducing new tools and strategies such as stem cell therapy and deep brain stimulation. A link between alterations in gut microbiota and PD has also opened a new line. Evidence exists of a bidirectional pathway between the gastrointestinal tract and the central nervous system. Probiotics, prebiotics and synbiotics are being examined that might influence gut-brain axis by altering gut microbiota composition, enteric nervous system, and CNS. This review provides status on use of probiotics for PD. Limitations and future directions will also be addressed to promote further research considering use of probiotics for PD.
Collapse
Affiliation(s)
- Parisa Gazerani
- Biomedicine: Department of Health Science and Technology, Faculty of Medicine, Aalborg University,Frederik Bajers Vej 3B, 9220 Aalborg East, Denmark.
| |
Collapse
|
20
|
Rhea EM, Banks WA. Role of the Blood-Brain Barrier in Central Nervous System Insulin Resistance. Front Neurosci 2019; 13:521. [PMID: 31213970 PMCID: PMC6558081 DOI: 10.3389/fnins.2019.00521] [Citation(s) in RCA: 141] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 05/06/2019] [Indexed: 01/01/2023] Open
Abstract
The blood-brain barrier (BBB) mediates the communication between the periphery and the central nervous system (CNS). Recently, CNS insulin resistance has been elucidated to play a role in neurodegenerative disease. This has stimulated a wealth of information on the molecular impact of insulin in the brain, particularly in the improvement of cognition. Since the BBB regulates the transport of insulin into the brain and thus, helps to regulate CNS levels, alterations in the BBB response to insulin could impact CNS insulin resistance. In this review, we summarize the effect of insulin on some of the cell types that make up the BBB, including endothelial cells, neurons, astrocytes, and pericytes. We broadly discuss how these changes in specific cell types could ultimately impact the BBB. We also summarize how insulin can regulate levels of the pathological hallmarks of Alzheimer's disease, including amyloid beta (Aβ) and tau within each cell type. Finally, we suggest interventional approaches to overcome detrimental effects on the BBB in regards to changes in insulin transport.
Collapse
Affiliation(s)
- Elizabeth M Rhea
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States.,Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA, United States
| | - William A Banks
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States.,Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA, United States
| |
Collapse
|
21
|
Protective effect of surface-modified berberine nanoparticles against LPS-induced neurodegenerative changes: a preclinical study. Drug Deliv Transl Res 2019; 9:906-919. [PMID: 30868509 DOI: 10.1007/s13346-019-00626-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
22
|
Logsdon AF, Meabon JS, Cline MM, Bullock KM, Raskind MA, Peskind ER, Banks WA, Cook DG. Blast exposure elicits blood-brain barrier disruption and repair mediated by tight junction integrity and nitric oxide dependent processes. Sci Rep 2018; 8:11344. [PMID: 30054495 PMCID: PMC6063850 DOI: 10.1038/s41598-018-29341-6] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 06/25/2018] [Indexed: 12/14/2022] Open
Abstract
Mild blast-induced traumatic brain injury (TBI) is associated with blood-brain barrier (BBB) disruption. However, the mechanisms whereby blast disrupts BBB integrity are not well understood. To address this issue BBB permeability to peripherally injected 14C-sucrose and 99mTc-albumin was quantified in ten brain regions at time points ranging from 0.25 to 72 hours. In mice, repetitive (2X) blast provoked BBB permeability to 14C-sucrose that persisted in specific brain regions from 0.25 to 72 hours. However, 99mTc-albumin revealed biphasic BBB disruption (open-closed-open) over the same interval, which was most pronounced in frontal cortex and hippocampus. This indicates that blast initiates interacting BBB disruption and reparative processes in specific brain regions. Further investigation of delayed (72 hour) BBB disruption revealed that claudin-5 (CLD5) expression was disrupted specifically in the hippocampus, but not in dorsal striatum, a brain region that showed no blast-induced BBB permeability to sucrose or albumin. In addition, we found that delayed BBB permeability and disrupted CLD5 expression were blocked by the nitric oxide synthase inhibitor N(G)-nitro-L-arginine methyl ester (L-NAME). These data argue that latent nitric oxide-dependent signaling pathways initiate processes that result in delayed BBB disruption, which are manifested in a brain-region specific manner.
Collapse
Affiliation(s)
- Aric F Logsdon
- Geriatric Research Education and Clinical Center (GRECC), Veterans Affairs Puget Sound Health Care System, Seattle, WA, 98108, USA.,Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA, 98195, USA
| | - James S Meabon
- Veterans Affairs Northwest Network, Mental Illness Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, 98108, USA.,Department of Psychiatry and Behavioral Science, University of Washington School of Medicine, Seattle, WA, 98195, USA
| | - Marcella M Cline
- Geriatric Research Education and Clinical Center (GRECC), Veterans Affairs Puget Sound Health Care System, Seattle, WA, 98108, USA.,Department of Molecular and Cellular Biology, University of Washington, Seattle, WA, 98195, USA
| | - Kristin M Bullock
- Geriatric Research Education and Clinical Center (GRECC), Veterans Affairs Puget Sound Health Care System, Seattle, WA, 98108, USA
| | - Murray A Raskind
- Veterans Affairs Northwest Network, Mental Illness Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, 98108, USA.,Department of Psychiatry and Behavioral Science, University of Washington School of Medicine, Seattle, WA, 98195, USA
| | - Elaine R Peskind
- Veterans Affairs Northwest Network, Mental Illness Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, 98108, USA.,Department of Psychiatry and Behavioral Science, University of Washington School of Medicine, Seattle, WA, 98195, USA
| | - William A Banks
- Geriatric Research Education and Clinical Center (GRECC), Veterans Affairs Puget Sound Health Care System, Seattle, WA, 98108, USA.,Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA, 98195, USA
| | - David G Cook
- Geriatric Research Education and Clinical Center (GRECC), Veterans Affairs Puget Sound Health Care System, Seattle, WA, 98108, USA. .,Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA, 98195, USA.
| |
Collapse
|
23
|
|
24
|
Ghatak S, Trudler D, Dolatabadi N, Ambasudhan R. Parkinson's disease: what the model systems have taught us so far. J Genet 2018; 97:729-751. [PMID: 30027906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Parkinson's disease (PD) is a debilitating neurodegenerative disorder, for which people above the age of 60 show an increased risk. Although there has been great advancement in understanding the disease-related abnormalities in brain circuitry and development of symptomatic treatments, a cure for PD remains elusive. The discovery of PD associated gene mutations and environmental toxins have yielded animal models of the disease. These models could recapitulate several key aspects of PD, and provide more insights into the disease pathogenesis. They have also revealed novel aspects of the disease mechanism including noncell autonomous events and spreading of pathogenic protein species across the brain. Nevertheless, none of these models so far can comprehensively represent all aspects of the human disease. While the field is still searching for the perfect model system, recent developments in stem cell biology have provided a new dimension to modelling PD, especially doing it in a patient-specific manner. In the current review, we attempt to summarize the key findings in the areas discussed above, and highlight how the core PD pathology distinguishes itself from other neurodegenerative disorders while also resembling them in many aspects.
Collapse
Affiliation(s)
- Swagata Ghatak
- Neurodegenerative Disease Center, Scintillon Institute, San Diego, CA 92121, USA.
| | | | | | | |
Collapse
|
25
|
Lipopolysaccharide Induces Subacute Cerebral Microhemorrhages with Involvement of Nitric Oxide Synthase in Rats. J Stroke Cerebrovasc Dis 2018; 27:1905-1913. [DOI: 10.1016/j.jstrokecerebrovasdis.2018.02.044] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Revised: 02/09/2018] [Accepted: 02/17/2018] [Indexed: 11/22/2022] Open
|
26
|
Erickson MA, Banks WA. Neuroimmune Axes of the Blood-Brain Barriers and Blood-Brain Interfaces: Bases for Physiological Regulation, Disease States, and Pharmacological Interventions. Pharmacol Rev 2018; 70:278-314. [PMID: 29496890 PMCID: PMC5833009 DOI: 10.1124/pr.117.014647] [Citation(s) in RCA: 205] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Central nervous system (CNS) barriers predominantly mediate the immune-privileged status of the brain, and are also important regulators of neuroimmune communication. It is increasingly appreciated that communication between the brain and immune system contributes to physiologic processes, adaptive responses, and disease states. In this review, we discuss the highly specialized features of brain barriers that regulate neuroimmune communication in health and disease. In section I, we discuss the concept of immune privilege, provide working definitions of brain barriers, and outline the historical work that contributed to the understanding of CNS barrier functions. In section II, we discuss the unique anatomic, cellular, and molecular characteristics of the vascular blood-brain barrier (BBB), blood-cerebrospinal fluid barrier, and tanycytic barriers that confer their functions as neuroimmune interfaces. In section III, we consider BBB-mediated neuroimmune functions and interactions categorized as five neuroimmune axes: disruption, responses to immune stimuli, uptake and transport of immunoactive substances, immune cell trafficking, and secretions of immunoactive substances. In section IV, we discuss neuroimmune functions of CNS barriers in physiologic and disease states, as well as pharmacological interventions for CNS diseases. Throughout this review, we highlight many recent advances that have contributed to the modern understanding of CNS barriers and their interface functions.
Collapse
Affiliation(s)
- Michelle A Erickson
- Geriatric Research and Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, Washington; and Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, Washington
| | - William A Banks
- Geriatric Research and Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, Washington; and Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, Washington
| |
Collapse
|
27
|
Perez-Pardo P, Kliest T, Dodiya HB, Broersen LM, Garssen J, Keshavarzian A, Kraneveld AD. The gut-brain axis in Parkinson's disease: Possibilities for food-based therapies. Eur J Pharmacol 2017; 817:86-95. [DOI: 10.1016/j.ejphar.2017.05.042] [Citation(s) in RCA: 121] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 03/31/2017] [Accepted: 05/22/2017] [Indexed: 12/18/2022]
|
28
|
Perez-Pardo P, Hartog M, Garssen J, Kraneveld AD. Microbes Tickling Your Tummy: the Importance of the Gut-Brain Axis in Parkinson's Disease. Curr Behav Neurosci Rep 2017; 4:361-368. [PMID: 29201595 PMCID: PMC5694504 DOI: 10.1007/s40473-017-0129-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Purpose of Review Patients suffering from Parkinson's disease (PD) are known to experience gastrointestinal dysfunction that might precede the onset of motor symptoms by several years. Evidence suggests an important role of the gut-brain axis in PD pathogenesis. These interactions might be essentially influenced by the gut microbiota. Here, we review recent findings supporting that changes in the gut microbiota composition might be a trigger for inflammation contributing to neurodegeneration in PD. Recent Findings Recent research revealed that PD patients exhibit a pro-inflammatory microbiota profile in their intestinal tract that might increase gut permeability, allowing leakage of bacterial products and inflammatory mediators from the intestines. Evidence in literature indicates that alpha-synuclein deposition might start in the enteric nervous system by pro-inflammatory immune activity and then propagates to the CNS. Alternatively, the peripheral inflammatory response could impact the brain through systemic mechanisms. Summary A better understanding of the gut-brain interactions and the role of the intestinal microbiota in the regulation of immune responses might bring new insights in PD pathological progression and might lead to novel diagnostics and therapeutic approaches.
Collapse
Affiliation(s)
- Paula Perez-Pardo
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Mitch Hartog
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
- Nutricia Research, Utrecht, The Netherlands
| | - Aletta D. Kraneveld
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
- Institute for Risk Assessment Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
29
|
Can Co-Activation of Nrf2 and Neurotrophic Signaling Pathway Slow Alzheimer's Disease? Int J Mol Sci 2017; 18:ijms18061168. [PMID: 28561773 PMCID: PMC5485992 DOI: 10.3390/ijms18061168] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 05/22/2017] [Accepted: 05/27/2017] [Indexed: 12/28/2022] Open
Abstract
Alzheimer’s disease (AD) is a multifaceted disease that is hard to treat by single-modal treatment. AD starts with amyloid peptides, mitochondrial dysfunction, and oxidative stress and later is accompanied with chronic endoplasmic reticulum (ER) stress and autophagy dysfunction, resulting in more complicated pathogenesis. Currently, few treatments can modify the complicated pathogenic progress of AD. Compared to the treatment with exogenous antioxidants, the activation of global antioxidant defense system via Nrf2 looks more promising in attenuating oxidative stress in AD brains. Accompanying the activation of the Nrf2-mediated antioxidant defense system that reduce the AD-causative factor, oxidative stress, it is also necessary to activate the neurotrophic signaling pathway that replaces damaged organelles and molecules with new ones. Thus, the dual actions to activate both the Nrf2 antioxidant system and neurotrophic signaling pathway are expected to provide a better strategy to modify AD pathogenesis. Here, we review the current understanding of AD pathogenesis and neuronal defense systems and discuss a possible way to co-activate the Nrf2 antioxidant system and neurotrophic signaling pathway with the hope of helping to find a better strategy to slow AD.
Collapse
|
30
|
De Geyter D, De Smedt A, Stoop W, De Keyser J, Kooijman R. Central IGF-I Receptors in the Brain are Instrumental to Neuroprotection by Systemically Injected IGF-I in a Rat Model for Ischemic Stroke. CNS Neurosci Ther 2016; 22:611-6. [PMID: 27080541 PMCID: PMC6492886 DOI: 10.1111/cns.12550] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 03/08/2016] [Accepted: 03/20/2016] [Indexed: 01/07/2023] Open
Abstract
AIM Insulin-like growth factor I (IGF-I) is a neuroprotective agent in animal models of ischemic stroke. The purpose of this study was to determine whether systemically injected IGF-I exerts its neuroprotective action by binding to IGF-I receptors in the brain after crossing the blood-brain barrier, or via peripheral effects. METHODS To differentiate the central effects of IGF-I from systemic effects, ischemic stroke was induced in conscious male Wistar Kyoto rats by the injection of endothelin-1 adjacent to the middle cerebral artery in the right hemisphere, while either the IGF-I receptor antagonist JB-1 or vehicle was introduced into the right lateral ventricle. RESULTS Intravenous injection of recombinant human (rh)IGF-I resulted in 50% reduction in infarct size, which was counteracted by the central administration of JB-1. Furthermore, rhIGF-I was detected in both the ischemic and nonischemic hemisphere. CONCLUSIONS Systemically injected rhIGF-I passes the blood-brain barrier and protects neurons via IGF-I receptors in the brain in rats with an ischemic stroke.
Collapse
Affiliation(s)
- Deborah De Geyter
- Center for Neurosciences (C4N)Vrije Universiteit Brussel (VUB)BrusselsBelgium
| | - Ann De Smedt
- Center for Neurosciences (C4N)Vrije Universiteit Brussel (VUB)BrusselsBelgium
- Department of NeurologyUniversitair Ziekenhuis BrusselBrusselsBelgium
| | - Wendy Stoop
- Center for Neurosciences (C4N)Vrije Universiteit Brussel (VUB)BrusselsBelgium
| | - Jacques De Keyser
- Center for Neurosciences (C4N)Vrije Universiteit Brussel (VUB)BrusselsBelgium
- Department of NeurologyUniversitair Ziekenhuis BrusselBrusselsBelgium
- Department of NeurologyUniversity Medical Center GroningenGroningenThe Netherlands
| | - Ron Kooijman
- Center for Neurosciences (C4N)Vrije Universiteit Brussel (VUB)BrusselsBelgium
| |
Collapse
|
31
|
Rani V, Deshmukh R, Jaswal P, Kumar P, Bariwal J. Alzheimer's disease: Is this a brain specific diabetic condition? Physiol Behav 2016; 164:259-67. [PMID: 27235734 DOI: 10.1016/j.physbeh.2016.05.041] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2015] [Revised: 05/16/2016] [Accepted: 05/22/2016] [Indexed: 01/13/2023]
Abstract
Alzheimer's disease (AD) and type 2 diabetes (T2DM) are the two major health issues affecting millions of elderly people worldwide, with major impacts in the patient's daily life. Numerous studies have demonstrated that patients with diabetes have an increased risk of developing AD compared with healthy individuals. The principal biological mechanisms that associate with the progression of diabetes and AD are not completely understood. Impaired insulin signaling, uncontrolled glucose metabolism, oxidative stress, abnormal protein processing, and the stimulation of inflammatory pathways are common features to both AD and T2DM. In recent years brain specific abnormalities in insulin and insulin like growth factor (IGF) signaling considered as a major trigger involved in the etiopathogenesis of AD, showing T2DM like milieu. This review summarizes the pathways that might link diabetes and AD and the effect of diminished insulin.
Collapse
Affiliation(s)
- Vanita Rani
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga 142001, Punjab, India
| | - Rahul Deshmukh
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga 142001, Punjab, India.
| | - Priya Jaswal
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga 142001, Punjab, India
| | - Puneet Kumar
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga 142001, Punjab, India
| | - Jitender Bariwal
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga 142001, Punjab, India
| |
Collapse
|
32
|
Urayama A, Grubb JH, Sly WS, Banks WA. Pharmacologic manipulation of lysosomal enzyme transport across the blood-brain barrier. J Cereb Blood Flow Metab 2016; 36:476-86. [PMID: 26661222 PMCID: PMC4794098 DOI: 10.1177/0271678x15614589] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 09/24/2015] [Indexed: 12/22/2022]
Abstract
The adult blood-brain barrier, unlike the neonatal blood-brain barrier, does not transport lysosomal enzymes into brain, making enzyme replacement therapy ineffective in treating the central nervous system symptoms of lysosomal storage diseases. However, enzyme transport can be re-induced with alpha-adrenergics. Here, we examined agents that are known to alter the blood-brain barrier transport of large molecules or to induce lysosomal enzyme transport across the blood-brain barrier ((±)epinephrine, insulin, retinoic acid, and lipopolysaccharide) in 2-week-old and adult mice. In 2-week-old adolescent mice, all these pharmacologic agents increased brain and heart uptake of phosphorylated human β-glucuronidase. In 8-week-old adult mice, manipulations with (±)epinephrine, insulin, and retinoic acid were significantly effective on uptake by brain and heart. The increased uptake of phosphorylated human β-glucuronidase was inhibited by mannose 6-phosphate for the agents (±)epinephrine and retinoic acid and by L-NG-nitroarginine methyl ester for the agent lipopolysaccharide in neonatal and adult mice. An in situ brain perfusion study revealed that retinoic acid directly modulated the transport of phosphorylated human β-glucuronidase across the blood-brain barrier. The present study indicates that there are multiple opportunities to at least transiently induce phosphorylated human β-glucuronidase transport at the adult blood-brain barrier.
Collapse
Affiliation(s)
- Akihiko Urayama
- Department of Neurology, University of Texas Medical School at Houston, Houston, TX, USA
| | - Jeffrey H Grubb
- Lysosomal Research, Ultragenyx Pharmaceutical Inc., Novato, CA, USA Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - William S Sly
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - William A Banks
- Geriatric Research, Education, and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA; Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| |
Collapse
|
33
|
Banks WA, Gray AM, Erickson MA, Salameh TS, Damodarasamy M, Sheibani N, Meabon JS, Wing EE, Morofuji Y, Cook DG, Reed MJ. Lipopolysaccharide-induced blood-brain barrier disruption: roles of cyclooxygenase, oxidative stress, neuroinflammation, and elements of the neurovascular unit. J Neuroinflammation 2015; 12:223. [PMID: 26608623 PMCID: PMC4660627 DOI: 10.1186/s12974-015-0434-1] [Citation(s) in RCA: 404] [Impact Index Per Article: 44.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 11/14/2015] [Indexed: 01/31/2023] Open
Abstract
Background Disruption of the blood-brain barrier (BBB) occurs in many diseases and is often mediated by inflammatory and neuroimmune mechanisms. Inflammation is well established as a cause of BBB disruption, but many mechanistic questions remain. Methods We used lipopolysaccharide (LPS) to induce inflammation and BBB disruption in mice. BBB disruption was measured using 14C-sucrose and radioactively labeled albumin. Brain cytokine responses were measured using multiplex technology and dependence on cyclooxygenase (COX) and oxidative stress determined by treatments with indomethacin and N-acetylcysteine. Astrocyte and microglia/macrophage responses were measured using brain immunohistochemistry. In vitro studies used Transwell cultures of primary brain endothelial cells co- or tri-cultured with astrocytes and pericytes to measure effects of LPS on transendothelial electrical resistance (TEER), cellular distribution of tight junction proteins, and permeability to 14C-sucrose and radioactive albumin. Results In comparison to LPS-induced weight loss, the BBB was relatively resistant to LPS-induced disruption. Disruption occurred only with the highest dose of LPS and was most evident in the frontal cortex, thalamus, pons-medulla, and cerebellum with no disruption in the hypothalamus. The in vitro and in vivo patterns of LPS-induced disruption as measured with 14C-sucrose, radioactive albumin, and TEER suggested involvement of both paracellular and transcytotic pathways. Disruption as measured with albumin and 14C-sucrose, but not TEER, was blocked by indomethacin. N-acetylcysteine did not affect disruption. In vivo, the measures of neuroinflammation induced by LPS were mainly not reversed by indomethacin. In vitro, the effects on LPS and indomethacin were not altered when brain endothelial cells (BECs) were cultured with astrocytes or pericytes. Conclusions The BBB is relatively resistant to LPS-induced disruption with some brain regions more vulnerable than others. LPS-induced disruption appears is to be dependent on COX but not on oxidative stress. Based on in vivo and in vitro measures of neuroinflammation, it appears that astrocytes, microglia/macrophages, and pericytes play little role in the LPS-mediated disruption of the BBB.
Collapse
Affiliation(s)
- William A Banks
- Geriatric Research Education and Clinical Center-VA Puget Sound Health Care System, Seattle, WA, USA. .,Division of Gerontology and Geriatric Medicine, Department of Internal Medicine, University of Washington School of Medicine, Seattle, WA, USA.
| | - Alicia M Gray
- University of Washington School of Medicine, Seattle, WA, USA.
| | - Michelle A Erickson
- Geriatric Research Education and Clinical Center-VA Puget Sound Health Care System, Seattle, WA, USA. .,Division of Gerontology and Geriatric Medicine, Department of Internal Medicine, University of Washington School of Medicine, Seattle, WA, USA.
| | - Therese S Salameh
- Geriatric Research Education and Clinical Center-VA Puget Sound Health Care System, Seattle, WA, USA.
| | - Mamatha Damodarasamy
- Division of Gerontology and Geriatric Medicine, Department of Internal Medicine, University of Washington School of Medicine, Seattle, WA, USA.
| | - Nader Sheibani
- Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
| | - James S Meabon
- Mental Health Research Education and Clinical Center-VA Puget Sound Health Care System, Seattle, WA, USA.
| | - Emily E Wing
- Geriatric Research Education and Clinical Center-VA Puget Sound Health Care System, Seattle, WA, USA.
| | - Yoichi Morofuji
- Geriatric Research Education and Clinical Center-VA Puget Sound Health Care System, Seattle, WA, USA. .,Division of Gerontology and Geriatric Medicine, Department of Internal Medicine, University of Washington School of Medicine, Seattle, WA, USA. .,Department of Neurosurgery, University of Nagasaki, Nagasaki, Japan.
| | - David G Cook
- Geriatric Research Education and Clinical Center-VA Puget Sound Health Care System, Seattle, WA, USA.
| | - May J Reed
- Division of Gerontology and Geriatric Medicine, Department of Internal Medicine, University of Washington School of Medicine, Seattle, WA, USA.
| |
Collapse
|
34
|
Liu LB, Liu XB, Ma J, Liu YH, Li ZQ, Ma T, Zhao XH, Xi Z, Xue YX. Bradykinin increased the permeability of BTB via NOS/NO/ZONAB-mediating down-regulation of claudin-5 and occludin. Biochem Biophys Res Commun 2015; 464:118-25. [PMID: 26106824 DOI: 10.1016/j.bbrc.2015.06.082] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 06/11/2015] [Indexed: 12/12/2022]
Abstract
After demonstrating bradykinin (BK) could increase the permeability of blood-tumor barrier (BTB) via opening the tight junction (TJ), and that the possible mechanism is unclear, we demonstrated that BK could increase the expressions of eNOS and nNOS and promote ZONAB translocation into nucleus. NOS inhibitors l-NAME and 7-NI could effectively block the effect of BK on increasing BTB permeability, decreasing the expressions of claudin-5 and occludin and promoting the translocation of ZONAB. Overexpression of ZONAB could significantly enhance BK-mediating BTB permeability. Meanwhile, chromatin immunoprecipitation verified ZONAB interacted with the promoter of claudin-5 and occludin respectively. This study indicated NOS/NO/ZONAB pathway might be involved in BK's increasing the permeability of BTB.
Collapse
Affiliation(s)
- Li-bo Liu
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang 110122, PR China
| | - Xiao-bai Liu
- The 96th Class, 7-Year Program, China Medical University, Shenyang 110122, PR China
| | - Jun Ma
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang 110122, PR China
| | - Yun-hui Liu
- Department of Neurosurgery, Shengjing Hospital, China Medical University, Shenyang 110004, PR China
| | - Zhi-qing Li
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang 110122, PR China
| | - Teng Ma
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang 110122, PR China
| | - Xi-he Zhao
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang 110122, PR China
| | - Zhuo Xi
- Department of Neurosurgery, Shengjing Hospital, China Medical University, Shenyang 110004, PR China
| | - Yi-xue Xue
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang 110122, PR China.
| |
Collapse
|
35
|
Doll DN, Hu H, Sun J, Lewis SE, Simpkins JW, Ren X. Mitochondrial crisis in cerebrovascular endothelial cells opens the blood-brain barrier. Stroke 2015; 46:1681-9. [PMID: 25922503 DOI: 10.1161/strokeaha.115.009099] [Citation(s) in RCA: 138] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 04/03/2015] [Indexed: 12/25/2022]
Abstract
BACKGROUND AND PURPOSE The blood-brain barrier (BBB) is a selectively permeable cerebrovascular endothelial barrier that maintains homeostasis between the periphery and the central nervous system. BBB disruption is a consequence of ischemic stroke and BBB permeability can be altered by infection/inflammation, but the complex cellular and molecular changes that result in this BBB alteration need to be elucidated to determine mechanisms. METHODS Infection mimic (lipopolysaccharide) challenge on infarct volume, BBB permeability, infiltrated neutrophils, and functional outcomes after murine transient middle cerebral artery occlusion in vivo; mitochondrial evaluation of cerebrovascular endothelial cells challenged by lipopolysaccharide in vitro; pharmacological inhibition of mitochondria on BBB permeability in vitro and in vivo; the effects of mitochondrial inhibitor on BBB permeability, infarct volume, and functional outcomes after transient middle cerebral artery occlusion. RESULTS We report here that lipopolysaccharide worsens ischemic stroke outcome and increases BBB permeability after transient middle cerebral artery occlusion in mice. Furthermore, we elucidate a novel mechanism that compromised mitochondrial function accounts for increased BBB permeability as evidenced by: lipopolysaccharide-induced reductions in oxidative phosphorylation and subunit expression of respiratory chain complexes in cerebrovascular endothelial cells, a compromised BBB permeability induced by pharmacological inhibition of mitochondrial function in cerebrovascular endothelial cells in vitro and in an in vivo animal model, and worsened stroke outcomes in transient middle cerebral artery occlusion mice after inhibition of mitochondrial function. CONCLUSIONS We concluded that mitochondria are key players in BBB permeability. These novel findings suggest a potential new therapeutic strategy for ischemic stroke by endothelial cell mitochondrial regulation.
Collapse
Affiliation(s)
- Danielle N Doll
- From the Department of Neurobiology and Anatomy (D.N.D.), Experimental Stroke Core, Center for Basic and Translational Stroke Research (H.H., S.E.L., J.W.S., X.R.), and Department of Physiology and Pharmacology (H.H., J.S., S.E.L., J.W.S., X.R.), West Virginia University, Morgantown
| | - Heng Hu
- From the Department of Neurobiology and Anatomy (D.N.D.), Experimental Stroke Core, Center for Basic and Translational Stroke Research (H.H., S.E.L., J.W.S., X.R.), and Department of Physiology and Pharmacology (H.H., J.S., S.E.L., J.W.S., X.R.), West Virginia University, Morgantown
| | - Jiahong Sun
- From the Department of Neurobiology and Anatomy (D.N.D.), Experimental Stroke Core, Center for Basic and Translational Stroke Research (H.H., S.E.L., J.W.S., X.R.), and Department of Physiology and Pharmacology (H.H., J.S., S.E.L., J.W.S., X.R.), West Virginia University, Morgantown
| | - Sara E Lewis
- From the Department of Neurobiology and Anatomy (D.N.D.), Experimental Stroke Core, Center for Basic and Translational Stroke Research (H.H., S.E.L., J.W.S., X.R.), and Department of Physiology and Pharmacology (H.H., J.S., S.E.L., J.W.S., X.R.), West Virginia University, Morgantown
| | - James W Simpkins
- From the Department of Neurobiology and Anatomy (D.N.D.), Experimental Stroke Core, Center for Basic and Translational Stroke Research (H.H., S.E.L., J.W.S., X.R.), and Department of Physiology and Pharmacology (H.H., J.S., S.E.L., J.W.S., X.R.), West Virginia University, Morgantown
| | - Xuefang Ren
- From the Department of Neurobiology and Anatomy (D.N.D.), Experimental Stroke Core, Center for Basic and Translational Stroke Research (H.H., S.E.L., J.W.S., X.R.), and Department of Physiology and Pharmacology (H.H., J.S., S.E.L., J.W.S., X.R.), West Virginia University, Morgantown.
| |
Collapse
|
36
|
Banks WA. The blood-brain barrier in neuroimmunology: Tales of separation and assimilation. Brain Behav Immun 2015; 44:1-8. [PMID: 25172555 PMCID: PMC4275374 DOI: 10.1016/j.bbi.2014.08.007] [Citation(s) in RCA: 166] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Revised: 08/20/2014] [Accepted: 08/20/2014] [Indexed: 01/25/2023] Open
Abstract
Neuroimmunology is concerned with the relations between the central nervous and immune systems and with the mechanisms that drive those relations. The blood-brain barrier (BBB) employs mechanisms that both separate and connect these two systems. In fact, the relative immune privilege of the central nervous system (CNS) is largely attributable to the BBB's ability to prevent the unregulated exchange of immune cells and their secretions between the CNS and blood. Having separated the two systems, the BBB then participates in mechanisms that allow them to influence, communicate, and interact with one another. Likewise, the BBB itself is influenced by immune events that are occurring in the periphery and in the CNS so that these three components (the BBB, the immune system, and the CNS) form neuroimmune axes that adapt to physiological and pathological conditions. To date, four major themes have emerged by which the BBB participates in these neuroimmune axes. The first of these four, the formation of the barrier, acts to separate the immune and central nervous systems. The other three themes provide mechanisms for re-establishing communication: response of the BBB to immunomodulatory molecules (e.g., prostaglandins, cytokines, chemokines, nitric oxide) secreted by immune and CNS cells; the controlled, regulated exchange of chemokines, cytokines, and immune cells between the CNS and the blood (i.e., transport across the BBB); the secretion of immunomodulatory molecules by the BBB, often in a polarized fashion. Taken together, these mechanisms reveal the BBB to be a dynamic, interactive, and adaptable interface between the immune system and the CNS, separating them on the one hand and fostering their interactions on the other hand, adjusting to physiological changes, while being a target for disease processes. This review examines specific examples by which the BBB plays an interactive, defining role in neuroimmunology.
Collapse
Affiliation(s)
- W A Banks
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care Center, Seattle, WA, United States; Division of Gerontology and Geriatric Medicine, University of Washington School of Medicine, Seattle, WA, United States.
| |
Collapse
|
37
|
Triggering of protection mechanism against Phoneutria nigriventer spider venom in the brain. PLoS One 2014; 9:e107292. [PMID: 25211468 PMCID: PMC4161398 DOI: 10.1371/journal.pone.0107292] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 08/11/2014] [Indexed: 01/03/2023] Open
Abstract
Severe accidents caused by the "armed" spider Phoneutria nigriventer cause neurotoxic manifestations in victims. In experiments with rats, P. nigriventer venom (PNV) temporarily disrupts the properties of the BBB by affecting both the transcellular and the paracellular route. However, it is unclear how cells and/or proteins participate in the transient opening of the BBB. The present study demonstrates that PNV is a substrate for the multidrug resistance protein-1 (MRP1) in cultured astrocyte and endothelial cells (HUVEC) and increases mrp1 and cx43 and down-regulates glut1 mRNA transcripts in cultured astrocytes. The inhibition of nNOS by 7-nitroindazole suggests that NO derived from nNOS mediates some of these effects by either accentuating or opposing the effects of PNV. In vivo, MRP1, GLUT1 and Cx43 protein expression is increased differentially in the hippocampus and cerebellum, indicating region-related modulation of effects. PNV contains a plethora of Ca(2+), K(+) and Na(+) channel-acting neurotoxins that interfere with glutamate handling. It is suggested that the findings of the present study are the result of a complex interaction of signaling pathways, one of which is the NO, which regulates BBB-associated proteins in response to PNV interference on ions physiology. The present study provides additional insight into PNV-induced BBB dysfunction and shows that a protective mechanism is activated against the venom. The data shows that PNV has qualities for potential use in drug permeability studies across the BBB.
Collapse
|
38
|
Dohgu S, Banks WA. Brain pericytes increase the lipopolysaccharide-enhanced transcytosis of HIV-1 free virus across the in vitro blood-brain barrier: evidence for cytokine-mediated pericyte-endothelial cell crosstalk. Fluids Barriers CNS 2013; 10:23. [PMID: 23816186 PMCID: PMC3710206 DOI: 10.1186/2045-8118-10-23] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Accepted: 06/23/2013] [Indexed: 08/26/2023] Open
Abstract
Background Human immunodeficiency virus-1 (HIV-1) enters the brain by crossing the blood–brain barrier (BBB) as both free virus and within infected immune cells. Previous work showed that activation of the innate immune system with lipopolysaccharide (LPS) enhances free virus transport both in vivo and across monolayer monocultures of brain microvascular endothelial cells (BMECs) in vitro. Methods Here, we used monocultures and co-cultures of brain pericytes and brain endothelial cells to examine the crosstalk between these cell types in mediating the LPS-enhanced permeation of radioactively-labeled HIV-1 (I-HIV) across BMEC monolayers. Results We found that brain pericytes when co-cultured with BMEC monolayers magnified the LPS-enhanced transport of I-HIV without altering transendothelial electrical resistance, indicating that pericytes affected the transcytotic component of HIV-1 permeation. As LPS crosses the BBB poorly if at all, and since pericytes are on the abluminal side of the BBB, we postulated that luminal LPS acts indirectly on pericytes through abluminal secretions from BMECs. Consistent with this, we found that the pattern of secretion of cytokines by pericytes directly exposed to LPS was different than when the pericytes were exposed to the abluminal fluid from LPS-treated BMEC monolayers. Conclusion These results are evidence for a cellular crosstalk in which LPS acts at the luminal surface of the brain endothelial cell, inducing abluminal secretions that stimulate pericytes to release substances that enhance the permeability of the BMEC monolayer to HIV.
Collapse
Affiliation(s)
- Shinya Dohgu
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan.
| | | |
Collapse
|
39
|
Banks WA, Owen JB, Erickson MA. Insulin in the brain: there and back again. Pharmacol Ther 2012; 136:82-93. [PMID: 22820012 PMCID: PMC4134675 DOI: 10.1016/j.pharmthera.2012.07.006] [Citation(s) in RCA: 395] [Impact Index Per Article: 32.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Accepted: 07/03/2012] [Indexed: 12/12/2022]
Abstract
Insulin performs unique functions within the CNS. Produced nearly exclusively by the pancreas, insulin crosses the blood-brain barrier (BBB) using a saturable transporter, affecting feeding and cognition through CNS mechanisms largely independent of glucose utilization. Whereas peripheral insulin acts primarily as a metabolic regulatory hormone, CNS insulin has an array of effects on brain that may more closely resemble the actions of the ancestral insulin molecule. Brain endothelial cells (BECs), the cells that form the vascular BBB and contain the transporter that translocates insulin from blood to brain, are themselves regulated by insulin. The insulin transporter is altered by physiological and pathological factors including hyperglycemia and the diabetic state. The latter can lead to BBB disruption. Pericytes, pluripotent cells in intimate contact with the BECs, protect the integrity of the BBB and its ability to transport insulin. Most of insulin's known actions within the CNS are mediated through two canonical pathways, the phosphoinositide-3 kinase (PI3)/Akt and Ras/mitogen activated kinase (MAPK) cascades. Resistance to insulin action within the CNS, sometimes referred to as diabetes mellitus type III, is associated with peripheral insulin resistance, but it is possible that variable hormonal resistance syndromes exist so that resistance at one tissue bed may be independent of that at others. CNS insulin resistance is associated with Alzheimer's disease, depression, and impaired baroreceptor gain in pregnancy. These aspects of CNS insulin action and the control of its entry by the BBB are likely only a small part of the story of insulin within the brain.
Collapse
Affiliation(s)
- William A Banks
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care Center, Seattle, WA, USA.
| | | | | |
Collapse
|
40
|
Hsuchou H, Kastin AJ, Mishra PK, Pan W. C-reactive protein increases BBB permeability: implications for obesity and neuroinflammation. Cell Physiol Biochem 2012; 30:1109-19. [PMID: 23018453 DOI: 10.1159/000343302] [Citation(s) in RCA: 122] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/14/2012] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND/AIMS Acute phase C-reactive protein (CRP), elevated in obesity and inflammation, is a major binding protein for leptin. It is thought that CRP contributes to leptin resistance by preventing leptin from crossing the blood-brain barrier (BBB). Here we determined how CRP interacts with the BBB and whether it deters leptin from reaching CNS targets. METHODS BBB permeability, compartmental distribution, tracer stability, and expression of tight junction protein and inflammatory marker were determined. RESULTS CRP was stable in blood, but did not permeate the BBB in trace amounts. However, it increased paracellular permeability at a higher dose. Agouti viable (A(vy)) mice with adult-onset obesity show higher CRP entry into the brain. CRP did not permeate hCMEC/D3 cells nor change zona occludin-1 or cyclooxygenase-2 expression. An intermediate dose of CRP had no effect on leptin transport across the BBB after co-treatment. Thus, acute interactions between CRP and leptin at the BBB level were negligible and did not explain the leptin resistance seen in obesity. CONCLUSIONS The interactions of CRP and the BBB are a two-phase process, with increased paracellular permeability at a high dose that enables its entry into the CNS and serves to induce reactive gliosis and impair CNS function.
Collapse
Affiliation(s)
- Hung Hsuchou
- Blood-Brain Barrier Group, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA
| | | | | | | |
Collapse
|
41
|
Erickson MA, Dohi K, Banks WA. Neuroinflammation: a common pathway in CNS diseases as mediated at the blood-brain barrier. Neuroimmunomodulation 2012; 19:121-30. [PMID: 22248728 PMCID: PMC3707010 DOI: 10.1159/000330247] [Citation(s) in RCA: 167] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2011] [Accepted: 06/22/2011] [Indexed: 01/16/2023] Open
Abstract
The blood-brain barrier (BBB) is not simply a physical barrier but a regulatory interface between the central nervous system (CNS) and immune system. The BBB both affects and is affected by the immune system and connects at many levels with the CNS, including the following: (1) the BBB transports cytokines and secretes various substances with neuroinflammatory properties; (2) transporters are altered in disease states including traumatic injury, Alzheimer's disease and inflammatory processes; (3) cytokines and other immune secretions from the cells comprising the BBB are both constitutive and inducible; (4) immune cells are transported across the BBB by the highly regulated process termed diapedesis, which involves communication and interactions between the brain endothelial cells and the immune cells; (5) the neuroimmune system has various effects on the BBB, including modulation of important transport systems and in extreme pathological conditions even disruption of the BBB, and (6) the brain-to-blood efflux transporter P-glycoprotein is altered in inflammatory conditions, thus affecting drug delivery to the brain. In summary, the BBB is an interactive interface that regulates and defines many of the ways that the CNS and the immune system communicate with one another.
Collapse
Affiliation(s)
- Michelle A. Erickson
- Geriatrics Research Education and Clinical Center, Puget Sound Health Care System, Seattle, Wash., USA
- Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, St. Louis, Mo., USA
| | - Kenji Dohi
- Geriatrics Research Education and Clinical Center, Puget Sound Health Care System, Seattle, Wash., USA
- Division of Gerontology and Geriatric Medicine, Department of Internal Medicine, University of Washington School of Medicine, Seattle, Wash., USA
- Department of Emergency and Critical Care Medicine, Showa University School of Medicine, Tokyo, Japan
| | - William A. Banks
- Geriatrics Research Education and Clinical Center, Puget Sound Health Care System, Seattle, Wash., USA
- Division of Gerontology and Geriatric Medicine, Department of Internal Medicine, University of Washington School of Medicine, Seattle, Wash., USA
| |
Collapse
|
42
|
Shiraishi JI, Tanizawa H, Fujita M, Kawakami SI, Bungo T. Localization of hypothalamic insulin receptor in neonatal chicks: Evidence for insulinergic system control of feeding behavior. Neurosci Lett 2011; 491:177-80. [DOI: 10.1016/j.neulet.2011.01.031] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2010] [Revised: 12/28/2010] [Accepted: 01/12/2011] [Indexed: 12/21/2022]
|
43
|
Banks WA, Morley JE, Lynch JL, Lynch KM, Mooradian AD. Insulin detemir is not transported across the blood-brain barrier. Peptides 2010; 31:2284-8. [PMID: 20868713 PMCID: PMC3006236 DOI: 10.1016/j.peptides.2010.09.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2010] [Revised: 09/09/2010] [Accepted: 09/10/2010] [Indexed: 11/19/2022]
Abstract
Insulin detemir has a different profile of action on the central nervous system (CNS) than human insulin. It has been hypothesized that this is caused by an altered ability of insulin detemir to cross the blood-brain barrier (BBB). Here, we measured the permeability of the BBB to insulin detemir. We labeled insulin detemir with radioactive iodine (I-Det) and examined its ability to cross the BBB of the mouse. Permeation was assessed after intravenous injection and by brain perfusion in the presence or absence of excess insulin detemir. The ability of insulin detemir to inhibit human insulin transport across the BBB was also assessed. I-Det did not cross the BBB either after intravenous injection or when studied by brain perfusion, a method which removes or reduces the influence of circulating proteins. Unlabeled detemir was about 10 times less potent than human insulin at inhibiting the transport of radioactive human insulin across the BBB. The altered CNS profile of insulin detemir may be caused by its poor access to CNS receptors and by a block of human insulin from crossing the BBB.
Collapse
Affiliation(s)
- William A Banks
- GRECC, Veterans Affairs Puget Sound Health Care System, United States.
| | | | | | | | | |
Collapse
|
44
|
Impact of p-glycoprotein inhibition and lipopolysaccharide administration on blood-brain barrier transport of colistin in mice. Antimicrob Agents Chemother 2010; 55:502-7. [PMID: 21115788 DOI: 10.1128/aac.01273-10] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
The aim of this study was to investigate the factors limiting the blood-brain barrier (BBB) transport of colistin in healthy mice and to assess the impact of systemic inflammation on the transport of this antibiotic across the BBB. Colistin sulfate (40 mg/kg) was administered subcutaneously to Swiss outbred mice as single and multiple doses to determine any relationship between brain uptake and plasma concentrations of colistin. To assess the effect of P-glycoprotein (P-gp) on BBB transport, colistin sulfate (5 mg/kg) was concomitantly administered intravenously with PSC833 or GF120918 (10 mg/kg). Systemic inflammation was induced by three intraperitoneal injections of lipopolysaccharide (LPS; 3 mg/kg), and BBB transport of colistin was subsequently measured following subcutaneous administration and by an in situ brain perfusion. The brain uptake of colistin was low following single and multiple subcutaneous doses, with brain-to-plasma concentration ratios ranging between 0.021 and 0.037, and this was not significantly enhanced by coadministration of GF120918 or PSC833 (P > 0.05). LPS significantly increased the brain uptake of subcutaneously administered colistin with area under the brain concentration time curve (AUC(brain)) values of 11.7 ± 2.7 μg·h/g and 4.0 ± 0.3 μg·h/g for LPS- and saline-treated mice, respectively (mean ± standard deviation). Similarly, in situ perfusion of colistin led to higher antibiotic brain concentrations in LPS-treated animals than in saline-treated animals, with colistin brain-to-perfusate concentration ratios of 0.019 ± 0.001 and 0.014 ± 0.001, respectively. This study demonstrates that the BBB transport of colistin is negligible in healthy mice; however, brain concentrations of colistin can be significantly enhanced during systemic inflammation, as might be observed in infected patients.
Collapse
|
45
|
Oshima S, Nemoto E, Kuramochi M, Saitoh Y, Kobayashi D. Penetration of oseltamivir and its active metabolite into the brain after lipopolysaccharide-induced inflammation in mice. J Pharm Pharmacol 2010. [DOI: 10.1211/jpp.61.10.0018] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Abstract
Objectives
Oseltamivir phosphate is used for the treatment of influenza virus infections. Recently, oral intake has been associated with abnormal behaviour. The present study examined the brain penetration of oseltamivir phosphate and oseltamivir carboxylate, its active metabolite, during inflammation.
Methods
Male C57BL/6 mice were given three i.p. injections of lipopolysaccharide (LPS) or saline. We studied the concentration of Evans blue (a marker of blood–brain barrier function) and oseltamivir phosphate and its active metabolite in the brain and plasma.
Key findings
The brain-to-plasma ratio of Evans blue compared with saline-treated control mice increased significantly with LPS dose. LPS-induced inflammation increased the permeation of drugs through the blood-brain barrier. The concentration of oseltamivir phosphate in both brain and plasma was 2-fold higher in mice treated with LPS than in control mice. Although the plasma concentration of the active carboxylate was not significantly altered by inflammation, the brain concentration was increased 2.7-fold in mice treated with LPS compared with control mice.
Conclusions
Administration of oseltamivir phosphate in the presence of inflammation increased the brain concentration of both parent drug and active metabolite, which may explain the central nervous system side-effects observed with this agent.
Collapse
Affiliation(s)
- Shigeru Oshima
- Faculty of Pharmaceutical Sciences, Josai University, Sakado, Saitama, Japan
| | - Eiichi Nemoto
- Faculty of Pharmaceutical Sciences, Josai University, Sakado, Saitama, Japan
| | - Mika Kuramochi
- Faculty of Pharmaceutical Sciences, Josai University, Sakado, Saitama, Japan
| | - Yukiya Saitoh
- Faculty of Pharmaceutical Sciences, Josai University, Sakado, Saitama, Japan
| | - Daisuke Kobayashi
- Faculty of Pharmaceutical Sciences, Josai University, Sakado, Saitama, Japan
| |
Collapse
|
46
|
Minimal penetration of lipopolysaccharide across the murine blood-brain barrier. Brain Behav Immun 2010; 24:102-9. [PMID: 19735725 PMCID: PMC2789209 DOI: 10.1016/j.bbi.2009.09.001] [Citation(s) in RCA: 265] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2009] [Revised: 09/01/2009] [Accepted: 09/02/2009] [Indexed: 11/21/2022] Open
Abstract
LPS given peripherally or into the brain induces a neuroinflammatory response. How peripheral LPS induces its effects on brain is not clear, but one mechanism is that LPS crosses the blood-brain barrier (BBB). Alternatively, LPS acts outside the BBB by stimulating afferent nerves, acting at circumventricular organs, and altering BBB permeabilities and functions. Here, we labeled LPS with radioactive iodine (I-LPS) and coinjected it with radioactively labeled albumin (I-Alb) which acted as a vascular space marker. Measurable amounts of I-LPS associated with the BBB, most reversibly bound to brain endothelia. Brain endothelia also sequestered small amounts of I-LPS and about 0.025% of an intravenously injected dose of I-LPS crossed the BBB to enter the CNS. Disruption of the BBB with repeated injections of LPS did not enhance I-LPS uptake. Based on dose-response curves in the literature of the amounts of LPS needed to stimulate brain neuroimmune events, it is unlikely that enough peripherally administered LPS enters the CNS to invoke those events except possibly at the highest doses used and for the most sensitive brain functions. I-LPS injected into the lateral ventricle of the brain entered the circulation with the reabsorption of cerebrospinal fluid (bulk flow) as previously described. In conclusion, brain uptake of circulating I-LPS is so low that most effects of peripherally administered LPS are likely mediated through LPS receptors located outside the BBB.
Collapse
|
47
|
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
|
48
|
Banks WA, Erickson MA. The blood-brain barrier and immune function and dysfunction. Neurobiol Dis 2009; 37:26-32. [PMID: 19664708 DOI: 10.1016/j.nbd.2009.07.031] [Citation(s) in RCA: 366] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2009] [Revised: 07/20/2009] [Accepted: 07/25/2009] [Indexed: 12/20/2022] Open
Abstract
The blood-brain barrier (BBB) is the monocellular interface that divides the peripheral circulation from direct contact with the central nervous system (CNS). This interface consists of several parallel barriers that include most notably the capillary bed of the CNS and the choroid plexus. These barriers at one level create the dichotomy between the circulating factors of the immune system and the components of the CNS only to regulate interactions between the immune and central nervous systems at other levels. The BBB is thus an integral part of the neuroimmune axis. Here, we will consider four aspects of BBB-neuroimmune interactions: BBB disruption as mediated by LPS and cytokines, cytokine transport across the BBB, immune cell trafficking, and effects of lipopolysaccharide (LPS) on various functions of the BBB.
Collapse
Affiliation(s)
- William A Banks
- Geriatrics Research Educational and Clinical Center, Veterans Affairs Medical Center-St. Louis, USA.
| | | |
Collapse
|
49
|
Jaeger LB, Dohgu S, Sultana R, Lynch JL, Owen JB, Erickson MA, Shah GN, Price TO, Fleegal-Demotta MA, Butterfiled DA, Banks WA. Lipopolysaccharide alters the blood-brain barrier transport of amyloid beta protein: a mechanism for inflammation in the progression of Alzheimer's disease. Brain Behav Immun 2009; 23:507-17. [PMID: 19486646 PMCID: PMC2783557 DOI: 10.1016/j.bbi.2009.01.017] [Citation(s) in RCA: 203] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2008] [Revised: 01/20/2009] [Accepted: 01/28/2009] [Indexed: 01/05/2023] Open
Abstract
Alzheimer's disease (AD) brains are characterized by accumulation of amyloid beta protein (Abeta) and neuroinflammation. Increased blood-to-brain influx and decreased brain-to-blood efflux across the blood-brain barrier (BBB) have been proposed as mechanisms for Abeta accumulation. Epidemiological studies suggest that the nonsteroidal anti-inflammatory drug (NSAID) indomethacin slows the progression of AD. We hypothesized that inflammation alters BBB handling of Abeta. Mice treated with lipopolysaccharide (LPS) had increased brain influx and decreased brain efflux of Abeta, recapitulating the findings in AD. Neither influx nor efflux was mediated by LPS acting directly on BBB cells. Increased influx was mediated by a blood-borne factor, indomethacin-independent, blocked by the triglyceride triolein, and not related to expression of the blood-to-brain transporter of Abeta, RAGE. Serum levels of IL-6, IL-10, IL-13, and MCP-1 mirrored changes in Abeta influx. Decreased efflux was blocked by indomethacin and accompanied by decreased protein expression of the brain-to-blood transporter of Abeta, LRP-1. LPS paradoxically increased expression of neuronal LRP-1, a major source of Abeta. Thus, inflammation potentially increases brain levels of Abeta by three mechanisms: increased influx, decreased efflux, and increased neuronal production.
Collapse
Affiliation(s)
- Laura B. Jaeger
- Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, St. Louis, Missouri 63104, USA
- Geriatric Research Education and Clinical Center (GGREC), VA Medical Center, St. Louis, Missouri 63106, USA
| | - Shinya Dohgu
- Geriatric Research Education and Clinical Center (GGREC), VA Medical Center, St. Louis, Missouri 63106, USA
- Department of Internal Medicine, Division of Geriatric Medicine, St. Louis University School of Medicine, St. Louis, Missouri 63104, USA
| | - R. Sultana
- Department of Chemistry, Center of Membrane Sciences and Sander-Brown Center on Aging, University of Kentucky, Lexington, KY 40506 USA
| | - Jessica L. Lynch
- Geriatric Research Education and Clinical Center (GGREC), VA Medical Center, St. Louis, Missouri 63106, USA
- Department of Internal Medicine, Division of Geriatric Medicine, St. Louis University School of Medicine, St. Louis, Missouri 63104, USA
| | - Joshua B. Owen
- Department of Chemistry, Center of Membrane Sciences and Sander-Brown Center on Aging, University of Kentucky, Lexington, KY 40506 USA
| | - Michelle A. Erickson
- Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, St. Louis, Missouri 63104, USA
- Geriatric Research Education and Clinical Center (GGREC), VA Medical Center, St. Louis, Missouri 63106, USA
| | - Gul N. Shah
- Geriatric Research Education and Clinical Center (GGREC), VA Medical Center, St. Louis, Missouri 63106, USA
- Department of Internal Medicine, Division of Geriatric Medicine, St. Louis University School of Medicine, St. Louis, Missouri 63104, USA
| | - Tulin O. Price
- Geriatric Research Education and Clinical Center (GGREC), VA Medical Center, St. Louis, Missouri 63106, USA
- Department of Internal Medicine, Division of Geriatric Medicine, St. Louis University School of Medicine, St. Louis, Missouri 63104, USA
| | - Melissa A. Fleegal-Demotta
- Geriatric Research Education and Clinical Center (GGREC), VA Medical Center, St. Louis, Missouri 63106, USA
- Department of Internal Medicine, Division of Geriatric Medicine, St. Louis University School of Medicine, St. Louis, Missouri 63104, USA
| | - D. Allan Butterfiled
- Department of Chemistry, Center of Membrane Sciences and Sander-Brown Center on Aging, University of Kentucky, Lexington, KY 40506 USA
| | - William A. Banks
- Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, St. Louis, Missouri 63104, USA
- Geriatric Research Education and Clinical Center (GGREC), VA Medical Center, St. Louis, Missouri 63106, USA
- Department of Internal Medicine, Division of Geriatric Medicine, St. Louis University School of Medicine, St. Louis, Missouri 63104, USA
| |
Collapse
|
50
|
Salkeni MA, Lynch JL, Otamis-Price T, Banks WA. Lipopolysaccharide impairs blood-brain barrier P-glycoprotein function in mice through prostaglandin- and nitric oxide-independent pathways. J Neuroimmune Pharmacol 2008; 4:276-82. [PMID: 19039663 DOI: 10.1007/s11481-008-9138-y] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2008] [Accepted: 11/06/2008] [Indexed: 12/14/2022]
Abstract
P-glycoprotein (P-gp) is a brain-to-blood efflux system that controls the ability of many drugs and endogenous substances to access the brain. In vitro work has shown that inflammatory states mediated through lipopolysaccharide (LPS) and tumor necrosis factor-alpha first impair and then stimulate P-gp activity. Here, we determined whether LPS can affect P-gp function in vivo. Mice treated with a single intraperitoneal injection of LPS (3 mg/kg) showed an inhibition of P-gp function. As assessed by brain perfusion, inhibition began 18 h after LPS administration and lasted until 36 h after administration. P-gp protein was increased by 44%, consistent with P-gp inhibition occurring through post-translational mechanisms. Unlike other effects of LPS on blood-brain barrier function, neither nitric oxide nor prostaglandin inhibition had an effect. We conclude that induction of proinflammatory states as exemplified by LPS treatment can inhibit P-gp function in vivo at the blood-brain barrier.
Collapse
Affiliation(s)
- Mohamad A Salkeni
- Department of Internal Medicine, Division of Geriatrics, Saint Louis University School of Medicine, St Louis, USA
| | | | | | | |
Collapse
|