Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
J Nutr ; 153(6): 1771-1782, 2023 06.
Article in English | MEDLINE | ID: mdl-37044210

ABSTRACT

BACKGROUND: Although reported dietary supplement use is common in older adults, evaluations of dietary supplement use over the past 10 y are lacking. OBJECTIVE: This analysis determined changes in reported dietary supplement use in cognitively normal older adults (aged ≥ 55 y) using the National Alzheimer's Coordinating Center data from 2015 to 2019 using a serial cross-sectional study design. METHODS: The first available visit for cognitively normal participants aged ≥ 55 y from 2015 to 2019 with a complete medication form was used, resulting in 9357 participants. Associations between visit year categories and reported use of dietary supplement categories/individual supplements were tested using categorical statistics. To determine whether the probabilities of reported supplement use changed in 2019 compared with those of 2015, z-scores and two-sided P values were used. Weighted analyses were used to confirm analytical findings. RESULTS: When comparing 2015 and 2019, the reported use of any dietary supplement decreased from 77.7% in 2015 to 71.0% in 2019 (P < 0.0001); any vitamin from 72.5% to 65.5% (P < 0.0001); any mineral from 39.2% to 30.4% (P < 0.0001); "other" nonvitamin/nonmineral supplements from 34.4% to 26.9% (P < 0.0001), calcium from 31.2% to 21.7% (P < 0.0001), multivitamins from 48.4% to 38.4% (P < 0.0001), potassium from 5.6% to 3.5% (P = 0.001), vitamin C from 13.0% to 9.2% (P = 0.0002), chondroitin from 6.0% to 4.1% (P = 0.006), glucosamine from 11.1% to 6.5% (P < 0.0001), and all omega fatty acids from 25.2% to 17.0% (P < 0.0001). Reported use increased for vitamin B7/biotin from 3.1% in 2015 to 5.8% in 2019 (P = 0.0003), melatonin from 3.1% to 5.8% (P = 0.0002), and turmeric from 1.2% to 4.7% (P < 0.0001). CONCLUSION: Although the reported use of many major dietary supplement categories and individual supplements significantly decreased in older adults from 2015 to 2019, biotin, turmeric, and melatonin significantly increased. Because biotin may interfere with some laboratory tests, this may have important public health implications.


Subject(s)
Alzheimer Disease , Melatonin , Humans , Aged , Biotin , Cross-Sectional Studies , Dietary Supplements , Vitamins
2.
J Pain ; 24(1): 24-37, 2023 01.
Article in English | MEDLINE | ID: mdl-36152760

ABSTRACT

For individuals experiencing pain, the decision to engage in clinical trials may be influenced by a number of factors including current and past care, illness severity, physical functioning, financial stress, and caregiver support. Co-occurring depression and anxiety may add to these challenges. The aim of this scoping review was to describe perspectives about clinical trial participation, including recruitment and retention among individuals with pain and pain comorbidities, including depression and/or anxiety. We searched PubMed, CINAHL, PsycINFO, and Cochrane CENTRAL databases. Study features, sample demographics, perspectives, barriers and/or motivations were collected and described. A total of 35 assessments were included in this scoping review with 24 focused on individuals with pain (24/35, 68.6%), 9 on individuals with depression and/or anxiety (9/35, 25.7%), and 2 on individuals with pain and co-occurring depression/anxiety (2/35, 5.7%). Barriers among participants with pain and those with depression included: research team's communication of information, fear of interventional risks, distrust (only among respondents with pain), too many procedures, fear of inadequate treatment, disease-life stressors, and embarrassment with study procedures (more commonly reported in participants with depression). Facilitators in both groups included: altruism and supportive staff, better access to care, and the ability to have outcome feedback (more commonly among individuals with depression). Individuals with pain and depression experience challenges that affect trial recruitment and retention. Engaging individuals with pain within research planning may assist in addressing these barriers and the needs of individuals affected by pain and/or depression. PERSPECTIVE: This review highlights the need to address barriers and facilitators to participation in clinical trials, including the need for an assessment of perspectives from underserved or marginalized populations.


Subject(s)
Anxiety , Depression , Humans , Depression/epidemiology , Depression/therapy , Anxiety/epidemiology , Anxiety/therapy , Anxiety Disorders , Pain/epidemiology
3.
Neurochem Res ; 47(3): 768-780, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34846597

ABSTRACT

Somatostatin receptor subtype 4 (SSTR4) is expressed in BV2 microglia, suggesting that SSTR4 agonists may impact microglia function. This study assessed the high-affinity SSTR4 agonist SM-I-26 (SMI) (0 nM, 10 nM, 1000 nM) against lipopolysaccharide (LPS)-induced inflammation (0, 10 or 100 ng/ml) over 6 or 24 h in BV2 microglia. Cell viability, nitrite output and mRNA expression changes of genes associated with our target (Sstr4), inflammation (Tnf-α, Il-6, Il-1ß, inos), anti-inflammatory and anti-oxidant actions (Il-10, Catalase), and mediators of Aß binding/phagocytosis (Msr1, Cd33, Trem1, Trem2) were measured. At 6 h SMI showed no effect across all conditions. At 24 h SMI (10 and 1000 nM) upregulated Sstr4 expression under inflammatory and non-inflammatory conditions. At 24 h SMI downregulated expression of the inflammatory cytokines Tnf-α (1000 nM within all LPS concentrations) and Il-6 (10 nM within 0 and 10 ng/ml LPS). At 24 h 10 nM SMI upregulated Il-10, while 1000 nM upregulated Catalase under inflammatory and non-inflammatory conditions. At 24 h Msr1 and Cd33 were upregulated by 1000 nM SMI under non-inflammatory conditions, while Trem1 was downregulated by 10 and 1000 nM SMI under mildly inflammatory and non-inflammatory conditions. These results show that SMI had concentration and time-dependent effects on mRNA expression of genes associated with different states of microglial activation. The SMI reduced Tnf-α and Il-6 inflammatory gene expression, and increased Il-10 anti-inflammatory gene expression, identifies anti-inflammatory actions of SSTR4 agonists extend to microglia.


Subject(s)
Lipopolysaccharides , Microglia , Cytokines/metabolism , Gene Expression , Humans , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Microglia/metabolism , Receptors, Somatostatin/genetics , Receptors, Somatostatin/metabolism
4.
RSC Med Chem ; 12(8): 1352-1365, 2021 Aug 18.
Article in English | MEDLINE | ID: mdl-34458738

ABSTRACT

Somatostatin receptor-4 (SST4) is highly expressed in brain regions affiliated with learning and memory. SST4 agonist treatment may act to mitigate Alzheimer's disease (AD) pathology. An integrated approach to SST4 agonist lead optimization is presented herein. High affinity and selective agonists with biological efficacy were identified through iterative cycles of a structure-based design strategy encompassing computational methods, chemistry, and preclinical pharmacology. 1,2,4-Triazole derivatives of our previously reported hit (4) showed enhanced SST4 binding affinity, activity, and selectivity. Thirty-five compounds showed low nanomolar range SST4 binding affinity, 12 having a K i < 1 nM. These compounds showed >500-fold affinity for SST4 as compared to SST2A. SST4 activities were consistent with the respective SST4 binding affinities (EC50 < 10 nM for 34 compounds). Compound 208 (SST4 K i = 0.7 nM; EC50 = 2.5 nM; >600-fold selectivity over SST2A) display a favorable physiochemical profile, and was advanced to learning and memory behavior evaluations in the senescence accelerated mouse-prone 8 model of AD-related cognitive decline. Chronic administration enhanced learning with i.p. dosing (1 mg kg-1) compared to vehicle. Chronic administration enhanced memory with both i.p. (0.01, 0.1, 1 mg kg-1) and oral (0.01, 10 mg kg-1) dosing compared to vehicle. This study identified a novel series of SST4 agonists with high affinity, selectivity, and biological activity that may be useful in the treatment of AD.

5.
PLoS One ; 16(7): e0254242, 2021.
Article in English | MEDLINE | ID: mdl-34237100

ABSTRACT

Microglia are the resident immune cell of the brain involved in the development and progression of Alzheimer's disease (AD). Modulation of microglia activity represents a potential mechanism for treating AD. Herein, the compound NNC 26-9100 (NNC) was evaluated in toxicity, nitric oxide release, Aß1-42 uptake and cytosolic calcium assays during lipopolysaccharide (LPS)-activated conditions using mouse BV2 microglia cells. After 24 hours, LPS increased cell toxicity in the alamar blue and lactate dehydrogenase assays, increased nitrite release, and increase cytoplasmic calcium. Addition of NNC decreased the LPS-induce lactate dehydrogenase release, had no effect in the alamar blue assay, decreased nitrite release and decreased cytosolic calcium. In the absence of LPS, NNC increased uptake of FITC-tagged Aß1-42. These data demonstrate that NNC treatment decreases nitrosative stress and microglia cell damage during LPS-induced activation and enhances phagocytosis of Aß1-42 during non-inflammatory conditions. Thus, NNC 26-9100 may have beneficial effects in AD and in inflammatory diseases of the brain through enhancement of microglial Aß clearance, and cell protective effects through prevention of elevated cytosolic calcium and inhibition of nitric oxide release.


Subject(s)
Aminopyridines/pharmacology , Amyloid beta-Peptides/metabolism , Calcium/metabolism , Microglia/drug effects , Nitric Oxide/metabolism , Peptide Fragments/metabolism , Phagocytosis/drug effects , Thiourea/analogs & derivatives , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Animals , Cell Line , Inflammation/drug therapy , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Mice , Microglia/metabolism , Thiourea/pharmacology
6.
Neurochem Res ; 44(11): 2670-2680, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31630317

ABSTRACT

Alzheimer's disease (AD) is a progressive neurodegenerative disorder resulting in memory and cognitive impairment. The use of somatostatin receptor subtype-4 (SSTR4) agonists have been proposed for AD treatment. This study investigated the effects of selective SSTR4 agonist NNC 26-9100 on mRNA expression of key genes associated with AD pathology (microglia mediators of Aß phagocytosis, amyloid-beta (Aß)-degrading enzymes, anti-oxidant enzymes and pro-inflammatory cytokines) in 3xTg-AD mice. Mice were administered NNC 26-9100 (0.2 µg, i.c.v.) or vehicle control, with cortical and subcortical brain tissue collected at 6 h and 24 h post-treatment. At 6 h, NNC 26-9100 treatment decreased cortical expression of cluster of differentiation-33 (Cd33) by 25%, while increasing cortical and subcortical macrophage scavenger receptor-1 (Msr1) by 1.8 and 2.0-fold, respectively. The Cd33 downregulation and Msr1 upregulation support a state of microglia associated Aß phagocytosis. At 24 h, NNC 26-9100 treatment increased the cortical expression of Sstr4 (4.9-fold), Aß-degrading enzymes neprilysin (9.3-fold) and insulin degrading enzyme (14.8-fold), and the antioxidant catalase (3.6-fold). Similar effects at 24 h were found in subcortical tissue with NNC 26-9100 treatment, but did not reach statistical significance. No changes in pro-inflammatory cytokine expression were found. These data demonstrated NNC 26-9100 facilitates transcriptional changes in brain tissue identified with Aß phagocytosis and clearance, further supporting SSTR4 as a treatment target for AD.


Subject(s)
Brain/metabolism , Microglia/metabolism , RNA, Messenger/metabolism , Receptors, Somatostatin/metabolism , Alzheimer Disease/pathology , Aminopyridines/pharmacology , Animals , Brain/cytology , Catalase/genetics , Down-Regulation/drug effects , Insulysin/genetics , Mice, Transgenic , Neprilysin/genetics , Phagocytosis/drug effects , Phagocytosis/physiology , Scavenger Receptors, Class A/genetics , Sialic Acid Binding Ig-like Lectin 3/genetics , Thiourea/analogs & derivatives , Thiourea/pharmacology , Up-Regulation/drug effects
7.
Curr Dev Nutr ; 2(10): nzy065, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30338310

ABSTRACT

BACKGROUND: Diet-mediated alterations of critical brain nutrient transporters, major facilitator super family domain-containing 2a (Mfsd2a) and glucose transporter 1 (Glut1), have wide reaching implications in brain health and disease. OBJECTIVE: The aim of the study was to examine the impact of long-term low- and high-fat diets with lard or fish oil on critical brain nutrient transporters, Mfsd2a and Glut1. METHODS: Eight-week-old male C57BL/6 mice were fed 1 of the following 4 diets for 32 wk: 10% of kcal from lard, 10% of kcal from fish oil, 41% of kcal from lard, or 41% of kcal from fish oil. Body weight and blood chemistries delineated dietary effects. Cortical and subcortical Mfsd2a and Glut1 mRNA and protein expression were evaluated, with other supportive nutrient-sensitive targets also assessed for mRNA expression changes. RESULTS: Fish-oil diets increased cortical Mfsd2a mRNA expression compared with lard diets. Subcortical Mfsd2a mRNA expression decreased as the percentage of fat in the diet increased. There was an interaction between the type and percentage of fat with cortical and subcortical Mfsd2a and cortical Glut1 protein expression. In the lard diet groups, protein expression of cortical and subcortical Mfsd2a and cortical Glut1 significantly increased as fat percentage increased. As the fat percentage increased in the fish-oil diet groups, protein expression of cortical and subcortical Mfsd2a and cortical Glut1 did not change. When comparing the fish-oil groups with 10% lard, cortical Mfsd2a protein expression was significantly higher in the 10% and 41% fish-oil groups, whereas cortical Glut1 protein expression was significantly higher in only the 10% fish-oil group. A positive correlation between cortical peroxisome proliferator-activated receptor γ mRNA expression and Mfsd2a protein expression was shown. CONCLUSION: Corresponding to chronic dietary treatment, an interaction between the type of fat and the percentage of fat exists respective to changes in brain expression of the key nutrient transporters Mfsd2a and Glut1.

8.
Medchemcomm ; 9(12): 2083-2090, 2018 Dec 01.
Article in English | MEDLINE | ID: mdl-30746066

ABSTRACT

A series of compounds containing a 1,2,4-triazole moiety were synthesized, targeting the somatostatin receptor subtype-4 (sst4). Compounds were developed in which the Phe6/Phe7/Phe11, Trp8, and Lys9 mimetic groups were interchanged at positions 3, 4, and 5 of the 1,2,4-triazole ring. The 1,2,4-triazoles containing an 2-(imidazol-4-yl)ethyl substituent at position-3 demonstrated moderate binding affinity at sst4. 1,2,4-Triazoles containing an (indol-3-yl)methyl substituent at position-5 lacked affinity at sst4. The 1,2,4-triazoles containing an aminopropyl group at position-4 showed enhanced binding affinity compared to the 3-position. One compound with an 3-(imidazol-4-yl)propyl group at position-4 (compound 44) imparted high affinity and selectivity at sst4 (sst2A = >10 000 nM; sst4 = 19 nM), acting as an agonist (EC50 = 6.8 nM). Docking 44 into a model-built structure of sst4 pointed to differences in its binding versus the other low-affinity compounds and was also in line with one of the two previously reported binding modes. A virtual screening (VS) experiment, employing two separate docking algorithms, was able to score 44 among the top-ranked poses. In summary, compound 44 represents a novel and promising lead structure towards the development of a clinically viable sst4 agonist for the treatment of conditions ranging from Alzheimer's disease to chronic pain.

9.
J Alzheimers Dis ; 54(4): 1339-1348, 2016 10 18.
Article in English | MEDLINE | ID: mdl-27589526

ABSTRACT

Glycogen synthase kinase (GSK)-3ß is a multifunctional protein that has been implicated in the pathological characteristics of Alzheimer's disease (AD), including the heightened levels of neurofibrillary tangles, amyloid-beta (Aß), and neurodegeneration. We have previously shown that an antisense oligonucleotide directed at the Tyr 216 site on GSK-3ß (GAO) when injected centrally can decrease GSK-3ß levels, improve learning and memory, and decrease oxidative stress. In addition, we showed that GAO can cross the blood-brain barrier. Herein the impact of peripherally administered GAO in both the non-transgenic SAMP8 and transgenic Tg2576 (APPswe) models of AD were examined respective to learning and memory. Brain tissues were then evaluated for expression changes in the phosphorylated-Tyr 216 residue, which leads to GSK-3ß activation, and the phosphorylated-Ser9 residue, which reduces GSK-3ß activity. SAMP8 GAO-treated mice showed improved acquisition and retention using aversive T-maze, and improved declarative memory as measured by the novel object recognition (NOR) test. Expression of the phosphorylated-Tyr 216 was decreased and the phosphorylated-Ser9 was increased in GAO-treated SAMP8 mice. Tg2576 GAO-treated mice improved acquisition and retention in both the T-maze and NOR tests, with an increased phosphorylated-Ser9 GSK-3ß expression. Results demonstrate that peripheral administration of GAO improves learning and memory, corresponding with alterations in GSK-3ß phosphorylation state. This study supports peripherally administered GAO as a viable means to mediate GSK-3ß activity within the brain and a possible treatment for AD.


Subject(s)
Alzheimer Disease/drug therapy , Cellular Senescence/drug effects , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Maze Learning/drug effects , Memory/drug effects , Oligonucleotides, Antisense/administration & dosage , Alzheimer Disease/enzymology , Alzheimer Disease/pathology , Animals , Cellular Senescence/physiology , Disease Models, Animal , Glycogen Synthase Kinase 3 beta/metabolism , Male , Maze Learning/physiology , Memory/physiology , Mice , Mice, Transgenic
10.
Adv Pharmacol ; 71: 361-90, 2014.
Article in English | MEDLINE | ID: mdl-25307223

ABSTRACT

Steroids have a wide spectrum of impact, serving as fundamental regulators of nearly every physiological process within the human body. Therapeutic applications of steroids are equally broad, with a diverse range of medications and targets. Within the central nervous system (CNS), steroids influence development, memory, behavior, and disease outcomes. Moreover, steroids are well recognized as to their impact on the vascular endothelium. The blood-brain barrier (BBB) at the level of the brain microvascular endothelium serves as the principle interface between the peripheral circulation and the brain. Steroids have been identified to impact several critical properties of the BBB, including cellular efflux mechanisms, nutrient uptake, and tight junction integrity. Such actions not only influence brain homeostasis but also the delivery of CNS-targeted therapeutics. A greater understanding of the respective steroid-BBB interactions may shed further light on the differential treatment outcomes observed across CNS pathologies. In this chapter, we examine the current therapeutic implications of steroids respective to BBB structure and function, with emphasis on glucocorticoids and estrogens.


Subject(s)
Blood-Brain Barrier/metabolism , Estrogens/metabolism , Glucocorticoids/metabolism , Animals , Humans
11.
Brain Res ; 1520: 145-56, 2013 Jul 03.
Article in English | MEDLINE | ID: mdl-23669069

ABSTRACT

Soluble amyloid-ß peptide (Aß) oligomers have been hypothesized to be primary mediators of Alzheimer's disease progression. In this regard, reduction of soluble Aß-oligomers levels within the brain may provide a viable means in which to treat the disease. Somatostatin receptor subtype-4 (SSTR4) agonists have been proposed to reduce Aß levels in the brain via enhancement of enzymatic degradation. Herein we evaluated the effect of selective SSTR4 agonist NNC 26-9100 on the changes in learning and soluble Aß42 oligomer brain content with and without co-administration of the M13-metalloproteinase family enzyme-inhibitor phosphoramidon, using the senescence-accelerated mouse prone-8 (SAMP8) model. NNC 26-9100 treatment (0.2 µg i.c.v. in 2 µL) improved learning, which was blocked by phosphoramidon (1 and 10mM, respectively). NNC 26-9100 decreased total soluble Aß42, an effect which was blocked by phosphoramidon (10mM). Extracellular, intracellular, and membrane fractions were then isolated from cortical tissue and assessed for soluble oligomer alterations. NNC 26-9100 decreased the Aß42 trimeric (12 kDa) form within the extracellular and intracellular fractions, and produced a band-split effect of the Aß42 hexameric (25 kDa) form within the extracellular fraction. These effects were also blocked by phosphoramdon (1 and 10mM, respectively). Subsequent evaluation of NNC 26-9100 in APPswe Tg2576 transgenic mice showed a similar learning improvement and corresponding reduction in soluble Aß42 oligomers within extracellular, intracellular, and membrane fractions. These data support the hypothesis that NNC 26-9100 reduces soluble Aß42 oligomers and enhances learning through a phosphoramidon-sensitive metalloproteinase-dependent mechanism.


Subject(s)
Aminopyridines/pharmacology , Cerebral Cortex/metabolism , Metalloproteases/metabolism , Receptors, Somatostatin/agonists , Thiourea/analogs & derivatives , Alzheimer Disease/metabolism , Amyloid beta-Peptides , Animals , Blotting, Western , Cerebral Cortex/drug effects , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Male , Maze Learning/drug effects , Mice , Mice, Transgenic , Thiourea/pharmacology
12.
Eur J Pharmacol ; 683(1-3): 116-24, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22449380

ABSTRACT

Soluble amyloid ß-protein (Aß) oligomers are primary mediators of synaptic dysfunction associated with the progression of Alzheimer's disease. Such Aß oligomers exist dependent on their rates of aggregation and metabolism. Use of selective somatostatin receptor-subtype agonists have been identified as a potential means to mitigate Aß accumulation in the brain, via regulation of the enzyme neprilysin. Herein, we first evaluated the impact of the somatostatin receptor subtype-4 agonist 1-[3-[N-(5-Bromopyridin-2-yl)-N-(3,4-dichlorobenzyl)amino]propyl]-3-[3-(1H-imidazol-4-yl)propyl]thiourea (NNC 26-9100) on learning and memory in 12-month SAMP8 mice (i.c.v. injection). NNC 26-9100 (0.2 µg-dose) was shown to enhance both learning (T-maze) and memory (object recognition) compared to vehicle controls. Cortical and hippocampal tissues were evaluated subsequent to NNC 26-9100 (0.2 µg) or vehicle administration for changes in neprilysin activity, along with protein expression of amyloid-precursor protein (APP), neprilysin, and Aß1₋42 oligomers within respective cellular fractions (extracellular, intracellular and membrane). NNC 26-9100 increased neprilysin activity in cortical tissue, with an associated protein expression increase in the extracellular fraction and decreased in the intracellular fraction. A decrease in intracellular APP expression was found with treatment in both cortical and hippocampal tissues. NNC 26-9100 also significantly decreased expression of Aß1₋42 trimers within both the extracellular and intracellular cortical fractions. No expression changes were found in membrane fractions for any protein. These finding suggest the potential use of selective SSTR4 agonists to mitigate toxic oligomeric forms of Aß1₋42 in critical regions of the brain identified with learning and memory decline.


Subject(s)
Aminopyridines/pharmacology , Amyloid beta-Peptides/antagonists & inhibitors , Down-Regulation/drug effects , Extracellular Fluid/drug effects , Neurons/drug effects , Nootropic Agents/pharmacology , Peptide Fragments/antagonists & inhibitors , Receptors, Somatostatin/agonists , Thiourea/analogs & derivatives , Aminopyridines/administration & dosage , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/enzymology , Cerebral Cortex/metabolism , Dose-Response Relationship, Drug , Extracellular Fluid/enzymology , Extracellular Fluid/metabolism , Hippocampus/drug effects , Hippocampus/enzymology , Hippocampus/metabolism , Injections, Intraventricular , Male , Maze Learning/drug effects , Memory/drug effects , Mice , Mice, Inbred Strains , Molecular Weight , Neprilysin/metabolism , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Neurons/enzymology , Neurons/metabolism , Nootropic Agents/administration & dosage , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Solubility , Thiourea/administration & dosage , Thiourea/pharmacology
13.
Microvasc Res ; 81(2): 198-205, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21192956

ABSTRACT

Age and estrogen levels alter blood-brain barrier (BBB) tight junction (TJ) regulation, impacting brain homeostasis and pathological outcomes. This examination evaluated BBB TJ and estrogen receptor (ER) protein expression changes in young (8-10 week) and middle-aged (10-12 month) ovariectomized female Fisher-344 rats with chronic 17ß-estradiol or placebo treatment. Middle-aged rats showed decreased protein expression of occludin with 17ß-estradiol (55 kDa band) or placebo (45, 55, 60 kDa bands) treatment compared to respective young. In young animals, 17ß-estradiol treatment increased expression of the occludin 55 kDa band over placebo; however, this effect was lost in the middle-aged animals. In both young and middle-aged animals, expression of claudin-5 (23, 32 kDa bands) and ERα (66 kDa) increased with 17ß-estradiol treatment, while junctional adhesion molecule-A showed no change across all groups. However, ERα expression (66 kDa) was significantly reduced in the middle-aged animals compared to young placebo treated animals. Measurement of BBB TJ permeability via in situ perfusion of (14)C-sucrose showed no change with age or treatment. Our results show that increasing age and 17ß-estradiol treatment alters the expression of ERα and distinct BBB TJ protein isoforms without altering functional paracellular permeability.


Subject(s)
Aging/physiology , Blood-Brain Barrier/metabolism , Estradiol/pharmacology , Ovariectomy , Receptors, Estrogen/metabolism , Tight Junctions/metabolism , Animals , Blood-Brain Barrier/drug effects , Brain/blood supply , Brain/drug effects , Brain/metabolism , Capillary Permeability/drug effects , Capillary Permeability/physiology , Cell Adhesion Molecules/metabolism , Claudin-5 , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Female , Membrane Proteins/metabolism , Microvessels/drug effects , Microvessels/metabolism , Occludin , Perfusion , Rats , Rats, Inbred F344 , Sucrose/metabolism , Tight Junctions/drug effects
14.
Eur J Pharmacol ; 654(1): 53-9, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21185826

ABSTRACT

Selective somatostatin receptor subtype agonists have been proposed as a means to mitigate learning and memory loss associated with Alzheimer's disease. The first aim of this study evaluated blood-to-brain transport and regional brain distribution of NNC 26-9100, a selective somatostatin subtype-4 (sst4) receptor agonist. The entry rate of (131)I-NNC 26-9100 was K(i)=0.25 µl/g min, with an ~93% association with the parenchymal component. The second goal of this study was to evaluate the effect of chronic NNC 26-9100 administration (i.p.) on learning and memory, brain Aß(x-42) levels, and protein expression of sst4 receptor and amyloid precursor protein (APP) in the senescence-accelerated mouse p8 (SAMP8) model of Alzheimer's disease. Mice chronically treated with NNC 26-9100 showed improved learning (day 21) and memory (day 28) using the T-maze paradigm (20 and 200 µg). Ex vivo tissue analyses showed a decline in Aß(x-42) levels at the 20 µg dose, while no alterations were observed in sst4 receptor or APP protein expression compared to vehicle controls. These findings indicate NNC 26-9100 is taken up into key brain regions associated with learning and memory. Furthermore, chronic administration of NNC 26-9100 improved learning and memory and decreased Aß(x-42) brain levels. These results suggest sst4 receptor agonists may provide a viable therapy in the treatment of Alzheimer's disease and other forms of cognitive impairment.


Subject(s)
Aminopyridines/pharmacology , Maze Learning/drug effects , Memory/drug effects , Receptors, Somatostatin/agonists , Thiourea/analogs & derivatives , Aminopyridines/administration & dosage , Aminopyridines/pharmacokinetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Blood-Brain Barrier/metabolism , Brain/metabolism , Gene Expression Regulation/drug effects , Male , Mice , Peptide Fragments/metabolism , Receptors, Somatostatin/genetics , Thiourea/administration & dosage , Thiourea/pharmacokinetics , Thiourea/pharmacology , Time Factors , Tissue Distribution
15.
Neurobiol Dis ; 32(2): 200-19, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18790057

ABSTRACT

The blood-brain barrier (BBB) is formed by the endothelial cells of cerebral microvessels, providing a dynamic interface between the peripheral circulation and the central nervous system. The tight junctions (TJs) between the endothelial cells serve to restrict blood-borne substances from entering the brain. Under ischemic stroke conditions decreased BBB TJ integrity results in increased paracellular permeability, directly contributing to cerebral vasogenic edema, hemorrhagic transformation, and increased mortality. This loss of TJ integrity occurs in a phasic manner, which is contingent on several interdependent mechanisms (ionic dysregulation, inflammation, oxidative and nitrosative stress, enzymatic activity, and angiogenesis). Understanding the inter-relation of these mechanisms is critical for the development of new therapies. This review focuses on those aspects of ischemic stroke impacting BBB TJ integrity and the principle regulatory pathways, respective to the phases of paracellular permeability.


Subject(s)
Blood-Brain Barrier/physiopathology , Stroke/pathology , Tight Junctions/metabolism , Animals , Humans , Permeability , Stroke/physiopathology
16.
Microvasc Res ; 75(1): 91-6, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17651765

ABSTRACT

The blood-brain barrier (BBB) serves as a critical regulator of brain homeostasis. Following hypoxia (i.e. 6% oxygen/1 h) and reoxygenation (H/R), the BBB tight junctional complex is disrupted, resulting in increased BBB permeability and the development of vasogenic brain edema. In this study, we examined the effect of H/R on the in vivo rat BBB over a 36 h time course in conjunction with paracellular permeability, gray matter edema, and systemic inflammatory activity. A biphasic increase was observed in the brain uptake of (14)C-sucrose, a paracellular permeability marker; with the first increase at the 10 min reoxygenation time point, and the second increase at the 6-18 h time points. Increased brain water weight gain (edema) also showed a biphasic response with the first increase at the 10 min-1 h reoxygenation time points; and the second increase at only the 24 h time point. Analysis of serum derived cytokines (IL-1beta, TNFalpha, IL-6, IL-10, and IFNgamma) demonstrated that only IL-1beta and IL-6 were at detectable levels, but these levels were similar to controls. White blood cell counts showed significant decreases in lymphocytes (10 min-3 h), increases in monocytes (10 min-3 h and 12 h), and increases in polymorphonuclear cells (1 h and 3 h). We have shown that H/R elicits a biphasic increase in paracellular permeability and edema, which parallel to post-stroke sequelae, despite the lack of occlusion or complete depletion of oxygen.


Subject(s)
Blood-Brain Barrier/metabolism , Brain Edema/etiology , Capillary Permeability , Hypoxia/metabolism , Oxidative Stress , Animals , Brain Edema/blood , Brain Edema/metabolism , Brain Edema/pathology , Carbon Radioisotopes , Cytokines/blood , Disease Models, Animal , Female , Hypoxia/complications , Leukocyte Count , Rats , Rats, Sprague-Dawley , Sucrose/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...