Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Int J Mol Sci ; 25(12)2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38928312

ABSTRACT

Serum amyloid A (SAA) proteins are highly conserved lipoproteins that are notoriously involved in the acute phase response and systemic amyloidosis, but their biological functions are incompletely understood. Recent work has shown that SAA proteins can enter the brain by crossing the intact blood-brain barrier (BBB), and that they can impair BBB functions. Once in the central nervous system (CNS), SAA proteins can have both protective and harmful effects, which have important implications for CNS disease. In this review of the thematic series on SAA, we discuss the existing literature that relates SAA to neuroinflammation and CNS disease, and the possible roles of the BBB in these relations.


Subject(s)
Blood-Brain Barrier , Serum Amyloid A Protein , Blood-Brain Barrier/metabolism , Humans , Serum Amyloid A Protein/metabolism , Animals , Central Nervous System Diseases/metabolism
2.
J Pharmacol Exp Ther ; 384(1): 205-223, 2023 01.
Article in English | MEDLINE | ID: mdl-36310035

ABSTRACT

One important function of the vascular blood-brain barrier (BBB) is to facilitate neuroimmune communication. The BBB fulfills this function, in part, through its ability to transport cytokines and chemokines. C-C motif chemokine receptor 2 (CCL2) (MCP-1) and C-C motif chemokine receptor 5 (CCL5) (RANTES) are proinflammatory chemokines that mediate neuroimmune responses to acute insults and aspects of brain injury and neurodegenerative diseases; however, a blood-to-brain transport system has not been evaluated for either chemokine in vivo. Therefore, we determined whether CCL2 and CCL5 in blood can cross the intact BBB and enter the brain. Using CD-1 mice, we found that 125I-labeled CCL2 and CCL5 crossed the BBB and entered the brain parenchyma. We next aimed to identify the mechanisms of 125I-CCL2 and 125I-CCL5 transport in an in situ brain perfusion model. We found that both heparin and eprodisate inhibited brain uptake of 125I-CCL2 and 125I-CCL5 in situ, whereas antagonists of their receptors, CCR2 or CCR5, respectively, did not, suggesting that heparan sulfates at the endothelial surface mediate BBB transport. Finally, we showed that CCL2 and CCL5 transport across the BBB increased following a single injection of 0.3 mg/kg lipopolysaccharide. These data demonstrate that CCL2 and CCL5 in the brain can derive, in part, from the circulation, especially during systemic inflammation. Further, binding to the BBB-associated heparan sulfate is a mechanism by which both chemokines can cross the intact BBB, highlighting a novel therapeutic target for treating neuroinflammation. SIGNIFICANCE STATEMENT: Our work demonstrates that C-C motif chemokine ligand 2 (CCL2) and C-C motif chemokine ligand 5 (CCL5) can cross the intact blood-brain barrier and that transport is robustly increased during inflammation. These data suggest that circulating CCL2 and CCL5 can contribute to brain levels of each chemokine. We further show that the transport of both chemokines is inhibited by heparin and eprodisate, suggesting that CCL2/CCL5-heparan sulfate interactions could be therapeutically targeted to limit accumulation of these chemokines in the brain.


Subject(s)
Blood-Brain Barrier , Heparin , Mice , Animals , Blood-Brain Barrier/metabolism , Heparin/pharmacology , Ligands , Chemokines/metabolism , Inflammation/drug therapy , Receptors, Chemokine , Heparitin Sulfate
3.
Brain Behav Immun ; 111: 386-394, 2023 07.
Article in English | MEDLINE | ID: mdl-37146655

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.


Subject(s)
Blood-Brain Barrier , HMGB1 Protein , Animals , Mice , Blood-Brain Barrier/metabolism , HMGB1 Protein/metabolism , Inflammation , Lipopolysaccharides , Toll-Like Receptor 4/metabolism
4.
Brain Behav Immun ; 109: 251-268, 2023 03.
Article in English | MEDLINE | ID: mdl-36682515

ABSTRACT

COVID-19 and especially Long COVID are associated with severe CNS symptoms and may place persons at risk to develop long-term cognitive impairments. Here, we show that two non-infective models of SARS-CoV-2 can cross the blood-brain barrier (BBB) and induce neuroinflammation, a major mechanism underpinning CNS and cognitive impairments, even in the absence of productive infection. The viral models cross the BBB by the mechanism of adsorptive transcytosis with the sugar N-acetylglucosamine being key. The delta and omicron variants cross the BB B faster than the other variants of concern, with peripheral tissue uptake rates also differing for the variants. Neuroinflammation induced by icv injection of S1 protein was greatly enhanced in young and especially in aged SAMP8 mice, a model of Alzheimer's disease, whereas sex and obesity had little effect.


Subject(s)
Alzheimer Disease , COVID-19 , Humans , Mice , Animals , Blood-Brain Barrier/metabolism , Alzheimer Disease/metabolism , SARS-CoV-2 , COVID-19/complications , Neuroinflammatory Diseases , Post-Acute COVID-19 Syndrome
5.
Int J Mol Sci ; 24(2)2023 Jan 13.
Article in English | MEDLINE | ID: mdl-36675154

ABSTRACT

The blood-brain barrier (BBB) is an interface primarily comprised of brain endothelial cells (BECs), separating the central nervous system (CNS) from the systemic circulation while carefully regulating the transport of molecules and inflammatory cells, and maintaining the required steady-state environment. Inflammation modulates many BBB functions, but the ultrastructural cytoarchitectural changes of the BBB with inflammation are understudied. Inflammation was induced in male 8-10-week-old CD-1 mice with intraperitoneal lipopolysaccharide (LPS), using a regimen (3 mg/kg at 0, 6, and 24 h) that caused robust BBB disruption but had minimal lethality at the study timepoint of 28 h. Perfusion-fixed brains were collected and the frontal cortical layer III regions were analyzed using a transmission electron microscopy (TEM). The LPS-treated mice had pronounced ultrastructural remodeling changes in BECs that included plasma membrane ruffling, increased numbers of extracellular microvesicles, small exosome formation, aberrant BEC mitochondria, increased BEC transcytosis, while tight junctions appeared to be unaltered. Aberrant pericytes were contracted with rounded nuclei and a loss of their elongated cytoplasmic processes. Surveilling microglial cells were attracted to the neurovascular unit (NVU) of BECs, and astrocyte detachment and separation were associated with the formation of a perivascular space and pericapillary edema. The LPS treatment resulted in numerous ultrastructural aberrant remodeling changes to the neurovascular unit's BECs, microglia, pericytes, and astrocytes. In summary, a disturbance of the NVU morphology is a consequence of LPS treatment.


Subject(s)
Blood-Brain Barrier , Lipopolysaccharides , Male , Mice , Animals , Blood-Brain Barrier/metabolism , Lipopolysaccharides/adverse effects , Neuroinflammatory Diseases , Endothelial Cells/metabolism , Astrocytes/metabolism , Inflammation/chemically induced , Inflammation/metabolism
6.
Int J Mol Sci ; 24(2)2023 Jan 13.
Article in English | MEDLINE | ID: mdl-36675130

ABSTRACT

Ozone (O3) is an air pollutant that primarily damages the lungs, but growing evidence supports the idea that O3 also harms the brain; acute exposure to O3 has been linked to central nervous system (CNS) symptoms such as depressed mood and sickness behaviors. However, the mechanisms by which O3 inhalation causes neurobehavioral changes are limited. One hypothesis is that factors in the circulation bridge communication between the lungs and brain following O3 exposure. In this study, our goals were to characterize neurobehavioral endpoints of O3 exposure as they relate to markers of systemic and pulmonary inflammation, with a particular focus on serum amyloid A (SAA) and kynurenine as candidate mediators of O3 behavioral effects. We evaluated O3-induced dose-, time- and sex-dependent changes in pulmonary inflammation, circulating SAA and kynurenine and its metabolic enzymes, and sickness and depressive-like behaviors in Balb/c and CD-1 mice. We found that 3 parts per million (ppm) O3, but not 2 or 1 ppm O3, increased circulating SAA and lung inflammation, which were resolved by 48 h and was worse in females. We also found that indoleamine 2,3-dioxygenase (Ido1) mRNA expression was increased in the brain and spleen 24 h after 3 ppm O3 and that kynurenine was increased in blood. Sickness and depressive-like behaviors were observed at all O3 doses (1-3 ppm), suggesting that behavioral responses to O3 can occur independently of increased SAA or neutrophils in the lungs. Using SAA knockout mice, we found that SAA did not contribute to O3-induced pulmonary damage or inflammation, systemic increases in kynurenine post-O3, or depressive-like behavior but did contribute to weight loss. Together, these findings indicate that acute O3 exposure induces transient symptoms of sickness and depressive-like behaviors that may occur in the presence or absence of overt pulmonary neutrophilia and systemic increases of SAA. SAA does not appear to contribute to pulmonary inflammation induced by O3, although it may contribute to other aspects of sickness behavior, as reflected by a modest effect on weight loss.


Subject(s)
Ozone , Pneumonia , Female , Mice , Animals , Ozone/toxicity , Serum Amyloid A Protein/metabolism , Kynurenine/metabolism , Lung/metabolism , Pneumonia/metabolism , Phenotype
7.
Lung ; 200(2): 269-275, 2022 04.
Article in English | MEDLINE | ID: mdl-35199228

ABSTRACT

PURPOSE: Anesthetics are required for procedures that deliver drugs/biologics, infectious/inflammatory agents, and toxicants directly to the lungs. However, the possible confounding effects of anesthesia on lung inflammation and injury are underreported. Here, we evaluated the effects of two commonly used anesthetic regimens on lung inflammatory responses to ozone in mice. METHODS: We tested the effects of brief isoflurane (Iso) or ketamine/xylazine/atipamezole (K/X/A) anesthesia prior to ozone exposure (4 h, 3 ppm) on lung inflammatory responses in mice. Anesthesia regimens modeled those used for non-surgical intratracheal instillations and were administered 1-2 h or 24 h prior to initiating ozone exposure. RESULTS: We found that Iso given 1-2 h prior to ozone inhibited inflammatory responses in the lung, and this effect was absent when Iso was given 23-24 h prior to ozone. In contrast, K/X/A given 1-2 h prior to ozone increased lung and systemic inflammation. CONCLUSION: Our results highlight the need to comprehensively evaluate anesthesia as an experimental variable in the assessment of lung inflammation in response to ozone and other inflammatory stimuli.


Subject(s)
Anesthesia , Ozone , Pneumonia , Animals , Humans , Inflammation/chemically induced , Lung , Mice , Ozone/toxicity , Pneumonia/chemically induced
8.
Int J Mol Sci ; 23(4)2022 Feb 21.
Article in English | MEDLINE | ID: mdl-35216491

ABSTRACT

Systemic inflammation has been implicated in the progression of Alzheimer's disease (AD); however, less is understood about how existing AD pathology contributes to adverse outcomes following acute inflammatory insults. In the present study, our goal was to determine how AD-associated amyloid beta (Aß) pathology influences the acute neuroinflammatory and behavioral responses to a moderate systemic inflammatory insult. We treated 16-18-month-old female Tg2576 (Tg) mice, which overproduce human Aß and develop plaques, and age-matched wild-type (WT) littermate mice with an intraperitoneal injection of 0.33 mg/kg lipopolysaccharide (LPS) or saline. Mice were then evaluated over the next 28 h for sickness/depressive-like behaviors (food intake, weight loss, locomotion, and sucrose preference), systemic inflammation (serum amyloid A, SAA), blood-brain barrier (BBB) disruption, astrogliosis (glial fibrillary acidic protein/GFAP), Aß, and cytokine levels in the brain. We found that LPS caused a larger reduction in body weight in Tg vs. WT mice, but that other behavioral responses to LPS did not differ by genotype. BBB disruption was not apparent in either genotype following LPS. Concentrations of the systemic inflammatory marker, SAA, in the blood and brain were significantly increased with LPS but did not significantly differ by genotype. GFAP was increased in Tg mice vs. WT but was not significantly affected by LPS in either genotype. Finally, LPS-induced increases of eight cytokines (IL-1ß, IL-6, IL-12 (p40), IL-10, IL-17A, MIP-1α/CCL3, MIP-1ß/CCL4, and RANTES/CCL5) were found to be significantly higher in Tg mice vs. WT. In summary, our data show that Aß pathology exacerbates the neuroinflammatory response to LPS and identifies cytokines that are selectively regulated by Aß. The association of worse neuroinflammation with greater weight loss in Tg mice suggests that Aß pathology could contribute to poor outcomes following a systemic inflammatory insult.


Subject(s)
Amyloid beta-Peptides/metabolism , Cytokines/metabolism , Hippocampus/metabolism , Lipopolysaccharides/metabolism , Mice, Transgenic/metabolism , Weight Loss/physiology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/metabolism , Animals , Disease Models, Animal , Female , Gliosis/metabolism , Gliosis/pathology , Hippocampus/pathology , Inflammation/metabolism , Mice , Microglia/metabolism , Microglia/pathology , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology
9.
Pharmacol Rev ; 70(2): 278-314, 2018 04.
Article in English | MEDLINE | ID: mdl-29496890

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.


Subject(s)
Blood-Brain Barrier/physiology , Central Nervous System Diseases/drug therapy , Neuroimmunomodulation/physiology , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/immunology , Central Nervous System Diseases/immunology , Drug Delivery Systems , Humans , Membrane Transport Proteins/metabolism , Neuroimmunomodulation/drug effects , Transcytosis/immunology , Transcytosis/physiology
10.
Int J Mol Sci ; 22(5)2021 Mar 06.
Article in English | MEDLINE | ID: mdl-33800954

ABSTRACT

Emerging data indicate that neurological complications occur as a consequence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. The blood-brain barrier (BBB) is a critical interface that regulates entry of circulating molecules into the CNS, and is regulated by signals that arise from the brain and blood compartments. In this review, we discuss mechanisms by which SARS-CoV-2 interactions with the BBB may contribute to neurological dysfunction associated with coronavirus disease of 2019 (COVID-19), which is caused by SARS-CoV-2. We consider aspects of peripheral disease, such as hypoxia and systemic inflammatory response syndrome/cytokine storm, as well as CNS infection and mechanisms of viral entry into the brain. We also discuss the contribution of risk factors for developing severe COVID-19 to BBB dysfunction that could increase viral entry or otherwise damage the brain.


Subject(s)
Blood-Brain Barrier/physiopathology , Blood-Brain Barrier/virology , COVID-19/virology , SARS-CoV-2/metabolism , Animals , Blood-Brain Barrier/metabolism , Brain/virology , COVID-19/epidemiology , Central Nervous System Diseases/etiology , Central Nervous System Diseases/virology , Comorbidity , Humans , SARS-CoV-2/chemistry , Viral Tropism
11.
Int J Mol Sci ; 20(7)2019 Apr 02.
Article in English | MEDLINE | ID: mdl-30986918

ABSTRACT

Age is associated with altered immune functions that may affect the brain. Brain barriers, including the blood-brain barrier (BBB) and blood-CSF barrier (BCSFB), are important interfaces for neuroimmune communication, and are affected by aging. In this review, we explore novel mechanisms by which the aging immune system alters central nervous system functions and neuroimmune responses, with a focus on brain barriers. Specific emphasis will be on recent works that have identified novel mechanisms by which BBB/BCSFB functions change with age, interactions of the BBB with age-associated immune factors, and contributions of the BBB to age-associated neurological disorders. Understanding how age alters BBB functions and responses to pathological insults could provide important insight on the role of the BBB in the progression of cognitive decline and neurodegenerative disease.


Subject(s)
Aging/immunology , Blood-Brain Barrier/immunology , Blood-Brain Barrier/physiology , Immune System/physiology , Animals , Humans , Microbiota , Nervous System Diseases/pathology
12.
J Neuroinflammation ; 15(1): 25, 2018 Jan 27.
Article in English | MEDLINE | ID: mdl-29373982

ABSTRACT

BACKGROUND: Secoisolariciresinol diglucoside (SDG), the main lignan in flaxseed, is known for its beneficial effects in inflammation, oxidative stress, heart disease, tumor progression, atherosclerosis, and diabetes. SDG might be an attractive natural compound that protects against neuroinflammation. Yet, there are no comprehensive studies to date investigating the effects of SDG on brain endothelium using relevant in vivo and in vitro models. METHODS: We evaluated the effects of orally administered SDG on neuroinflammatory responses using in vivo imaging of the brain microvasculature during systemic inflammation and aseptic encephalitis. In parallel, the anti-inflammatory actions of SDG on brain endothelium and monocytes were evaluated in vitro blood-brain barrier (BBB) model. Multiple group comparisons were performed by one-way analysis of variance with Dunnet's post hoc tests. RESULTS: We found that SDG diminished leukocyte adhesion to and migration across the BBB in vivo in the setting of aseptic encephalitis (intracerebral TNFα injection) and prevented enhanced BBB permeability during systemic inflammatory response (LPS injection). In vitro SDG pretreatment of primary human brain microvascular endothelial cells (BMVEC) or human monocytes diminished adhesion and migration of monocytes across brain endothelial monolayers in conditions mimicking CNS inflammatory responses. Consistent with our in vivo observations, SDG decreased expression of the adhesion molecule, VCAM1, induced by TNFα, or IL-1ß in BMVEC. SDG diminished expression of the active form of VLA-4 integrin (promoting leukocyte adhesion and migration) and prevented the cytoskeleton changes in primary human monocytes activated by relevant inflammatory stimuli. CONCLUSION: This study indicates that SDG directly inhibits BBB interactions with inflammatory cells and reduces the inflammatory state of leukocytes. Though more work is needed to determine the mechanism by which SDG mediates these effects, the ability of SDG to exert a multi-functional response reducing oxidative stress, inflammation, and BBB permeability makes it an exciting potential therapeutic for neuroinflammatory diseases. SDG can serve as an anti-inflammatory and barrier-protective agent in neuroinflammation.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Blood-Brain Barrier/drug effects , Butylene Glycols/pharmacology , Endothelium, Vascular/drug effects , Glucosides/pharmacology , Inflammation Mediators/antagonists & inhibitors , Microvessels/drug effects , Animals , Blood-Brain Barrier/metabolism , Cell Movement/drug effects , Cell Movement/physiology , Dose-Response Relationship, Drug , Endothelium, Vascular/metabolism , Humans , Inflammation Mediators/metabolism , Male , Mice , Microvessels/metabolism
13.
Am J Pathol ; 187(1): 91-109, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27993242

ABSTRACT

Mounting evidence implicates antiretroviral (ARV) drugs as potential contributors to the persistence and evolution of clinical and pathological presentation of HIV-associated neurocognitive disorders in the post-ARV era. Based on their ability to induce endoplasmic reticulum (ER) stress in various cell types, we hypothesized that ARV-mediated ER stress in the central nervous system resulted in chronic dysregulation of the unfolded protein response and altered amyloid precursor protein (APP) processing. We used in vitro and in vivo models to show that HIV protease inhibitor (PI) class ARVs induced neuronal damage and ER stress, leading to PKR-like ER kinase-dependent phosphorylation of the eukaryotic translation initiation factor 2α and enhanced translation of ß-site APP cleaving enzyme-1 (BACE1). In addition, PIs induced ß-amyloid production, indicative of increased BACE1-mediated APP processing, in rodent neuroglial cultures and human APP-expressing Chinese hamster ovary cells. Inhibition of BACE1 activity protected against neuronal damage. Finally, ARVs administered to mice and SIV-infected macaques resulted in neuronal damage and BACE1 up-regulation in the central nervous system. These findings implicate a subset of PIs as potential mediators of neurodegeneration in HIV-associated neurocognitive disorders.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/metabolism , HIV Protease Inhibitors/pharmacology , Protein Biosynthesis/drug effects , Protein Processing, Post-Translational/drug effects , Up-Regulation/drug effects , Animals , Axons/drug effects , Axons/metabolism , Axons/pathology , Cells, Cultured , Macaca , Male , Mice, Inbred C57BL , Microtubule-Associated Proteins/metabolism , Neuroglia/drug effects , Neuroglia/metabolism , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Protein Stability/drug effects , Rats , Ritonavir/pharmacology , Unfolded Protein Response/drug effects , eIF-2 Kinase/metabolism
14.
FASEB J ; 31(9): 3950-3965, 2017 09.
Article in English | MEDLINE | ID: mdl-28533327

ABSTRACT

Accumulating evidence suggests that O3 exposure may contribute to CNS dysfunction. Here, we posit that inflammatory and acute-phase proteins in the circulation increase after O3 exposure and systemically convey signals of O3 exposure to the CNS. To model acute O3 exposure, female Balb/c mice were exposed to 3 ppm O3 or forced air for 2 h and were studied after 6 or 24 h. Of 23 cytokines and chemokines, only KC/CXCL1 was increased in blood 6 h after O3 exposure. The acute-phase protein serum amyloid A (A-SAA) was significantly increased by 24 h, whereas C-reactive protein was unchanged. A-SAA in blood correlated with total leukocytes, macrophages, and neutrophils in bronchoalveolar lavage from O3-exposed mice. A-SAA mRNA and protein were increased in the liver. We found that both isoforms of A-SAA completely crossed the intact blood-brain barrier, although the rate of SAA2.1 influx was approximately 5 times faster than that of SAA1.1. Finally, A-SAA protein, but not mRNA, was increased in the CNS 24 h post-O3 exposure. Our findings suggest that A-SAA is functionally linked to pulmonary inflammation in our O3 exposure model and that A-SAA could be an important systemic signal of O3 exposure to the CNS.-Erickson, M. A., Jude, J., Zhao, H., Rhea, E. M., Salameh, T. S., Jester, W., Pu, S., Harrowitz, J., Nguyen, N., Banks, W. A., Panettieri, R. A., Jr., Jordan-Sciutto, K. L. Serum amyloid A: an ozone-induced circulating factor with potentially important functions in the lung-brain axis.


Subject(s)
Brain Diseases/chemically induced , Inflammation/chemically induced , Lung Diseases/chemically induced , Ozone/toxicity , Serum Amyloid A Protein/metabolism , Acute-Phase Proteins/genetics , Acute-Phase Proteins/metabolism , Animals , Brain Diseases/metabolism , Cytokines/blood , Cytokines/genetics , Cytokines/metabolism , Female , Inflammation/blood , Inflammation/metabolism , Liver/metabolism , Mice , Mice, Inbred BALB C
15.
Brain Behav Immun ; 74: 3-6, 2018 11.
Article in English | MEDLINE | ID: mdl-30172947

ABSTRACT

This year's 2018 Named Series on blood-brain interfaces highlights the importance of brain barriers as mediators of neuroimmune communication and regulators of neurological function. The term "brain interfaces" reflects our growing understanding that brain barriers such as the blood-brain barrier (BBB) and blood-CSF barrier (BCSFB) are not only gatekeepers, but facilitators of bidirectional communication between the brain and periphery. There is also an emerging appreciation that CNS sites that are exposed to blood-borne immune molecules and cells, such as the leptomeninges and circumventricular organs, may also be considered brain interfaces with important homeostatic and pathological functions. The work featured in this Series covers novel aspects of brain interface functions that focus on mechanisms regulating barrier integrity and transporter activities, downstream consequences of neurovascular injury, peripheral organ infection/injury, and clearance of pathogenic proteins. Results of these studies have emphasized new mechanisms by which brain interface dysfunction could contribute to neuroinflammation and CNS damage in eclampsia, fetal and adult hypoxic/ischemic injury, traumatic brain injury, Helicobacter infections, acute lung injury, multiple sclerosis, and Alzheimer's disease. This body of work emphasizes that brain interfaces may themselves be important therapeutic targets for a variety of CNS diseases that are associated with immune dyshomeostasis. Future works are warranted to further investigate brain interface functions in health and disease.


Subject(s)
Blood-Brain Barrier/metabolism , Neuroimmunomodulation/physiology , Animals , Blood-Brain Barrier/physiology , Brain/blood supply , Brain/metabolism , Central Nervous System Diseases/metabolism , Humans
17.
J Neuroinflammation ; 12: 223, 2015 Nov 25.
Article in English | MEDLINE | ID: mdl-26608623

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 (14)C-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 (14)C-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 (14)C-sucrose, radioactive albumin, and TEER suggested involvement of both paracellular and transcytotic pathways. Disruption as measured with albumin and (14)C-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.


Subject(s)
Blood-Brain Barrier/metabolism , Endothelial Cells/metabolism , Inflammation Mediators/metabolism , Lipopolysaccharides/toxicity , Oxidative Stress/physiology , Prostaglandin-Endoperoxide Synthases/physiology , Animals , Astrocytes/drug effects , Astrocytes/immunology , Astrocytes/metabolism , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/immunology , Cell Line, Transformed , Coculture Techniques , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Endothelial Cells/immunology , Inflammation/chemically induced , Inflammation/immunology , Inflammation/metabolism , Inflammation Mediators/immunology , Male , Mice , Oxidative Stress/drug effects
18.
J Pharmacol Exp Ther ; 349(3): 497-507, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24706984

ABSTRACT

Increased blood levels of the eotaxin chemokine C-C motif ligand 11 (CCL11) in aging were recently shown to negatively regulate adult hippocampal neurogenesis. How circulating CCL11 could affect the central nervous system (CNS) is not clear, but one possibility is that it can cross the blood-brain barrier (BBB). Here, we show that CCL11 undergoes bidirectional transport across the BBB. Transport of CCL11 from blood into whole brain (influx) showed biphasic kinetics, with a slow phase preceding a rapid phase of uptake. We found that the slow phase was explained by binding of CCL11 to cellular components in blood, whereas the rapid uptake phase was mediated by direct interactions with the BBB. CCL11, even at high doses, did not cause BBB disruption. All brain regions except striatum showed a delayed rapid-uptake phase. Striatum had only an early rapid-uptake phase, which was the fastest of any brain region. We also observed a slow but saturable transport system for CCL11 from brain to blood. C-C motif ligand 3 (CCR3), an important receptor for CCL11, did not facilitate CCL11 transport across the BBB, although high concentrations of a CCR3 inhibitor increased brain uptake without causing BBB disruption. Our results indicate that CCL11 in the circulation can access many regions of the brain outside of the neurogenic niche via transport across the BBB. This suggests that blood-borne CCL11 may have important physiologic functions in the CNS and implicates the BBB as an important regulator of physiologic versus pathologic effects of this chemokine.


Subject(s)
Blood-Brain Barrier/metabolism , Chemokine CCL11/blood , Animals , Biological Transport , Brain/blood supply , Brain/metabolism , Cells, Cultured , Chemokine CCL11/administration & dosage , Chemokine CCL11/pharmacokinetics , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Enzyme-Linked Immunosorbent Assay , Injections, Intravenous , Iodine Radioisotopes , Male , Mice , Primary Cell Culture , Regression Analysis , Tissue Distribution
20.
Exp Biol Med (Maywood) ; 248(5): 399-411, 2023 05.
Article in English | MEDLINE | ID: mdl-37012666

ABSTRACT

The blood-brain barrier (BBB) is a critical physiochemical interface that regulates communication between the brain and blood. It is comprised of brain endothelial cells which regulate the BBB's barrier and interface properties and is surrounded by supportive brain cell types including pericytes and astrocytes. Recent reports have suggested that the BBB undergoes dysfunction during normative aging and in disease. In this review, we consider the effect of cellular senescence, one of the nine hallmarks of aging, on the BBB. We first characterize known normative age-related changes at the BBB, and then evaluate changes in neurodegenerative diseases, with an emphasis on if/how cellular senescence is influencing these changes. We then discuss what insight has been gained from in vitro and in vivo studies of cellular senescence at the BBB. Finally, we evaluate mechanisms by which cellular senescence in peripheral pathologies can indirectly or directly affect BBB function.


Subject(s)
Blood-Brain Barrier , Endothelial Cells , Cellular Senescence , Brain
SELECTION OF CITATIONS
SEARCH DETAIL