Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Cell Mol Neurobiol ; 43(2): 797-811, 2023 Mar.
Article in English | MEDLINE | ID: mdl-35362880

ABSTRACT

Alzheimer's disease (AD) develops into dementia over a period of several years, during which subjective cognitive impairment (SCI) and mild cognitive impairment (MCI) can be used as intermediary diagnoses of increasing severity. Chronic neuroinflammation resulting from insufficient resolution is involved in the pathogenesis of AD and is associated with cognitive impairment. Specialized pro-resolving lipid mediators (LMs) that promote the resolution of inflammation may be valuable markers in AD diagnosis and as therapeutic targets. Liquid chromatography-tandem mass spectrometry was used to analyze pro-resolving and pro-inflammatory LMs in cerebrospinal fluid (CSF) from patients with cognitive impairment ranging from subjective impairment to a diagnosis of AD and correlated to cognition, CSF tau, and ß-amyloid. Resolvin (Rv) D4, RvD1, neuroprotectin D1 (NPD1), maresin 1 (MaR1), and RvE4 were lower in AD and/or MCI compared to SCI. The pro-inflammatory LTB4 and 15-HETE were higher in AD and MCI, respectively, while PGD2, PGE2, and PGF2a were decreased in AD, compared to SCI. RvD4 was also negatively correlated to AD tangle biomarkers, and positive correlations to cognitive test scores were observed for both pro-resolving LMs and their precursor fatty acids. In this exploratory study of the lipidome in CSF of AD, MCI, and SCI, the results indicate a shift in the LM profile from pro-resolving to pro-inflammatory in progression to AD, suggesting that it may be of use as a biomarker when followed by confirmation by replication studies.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Humans , Alzheimer Disease/pathology , Amyloid beta-Peptides , Cognition , Inflammation , Biomarkers , tau Proteins , Peptide Fragments , Disease Progression
2.
J Cell Mol Med ; 25(1): 434-447, 2021 01.
Article in English | MEDLINE | ID: mdl-33225628

ABSTRACT

Alzheimer's disease (AD) is the most common dementia, characterized by pathological accumulation of ß-amyloid (Aß) and hyperphosphorylation of tau protein, together with a damaging chronic inflammation. The lack of effective treatments urgently warrants new therapeutic strategies. Resolution of inflammation, associated with beneficial and regenerative activities, is mediated by specialized pro-resolving lipid mediators (SPMs) including maresin 1 (MaR1). Decreased levels of MaR1 have been observed in AD brains. However, the pro-resolving role of MaR1 in AD has not been fully investigated. In the present study, human monocyte-derived microglia (MdM) and a differentiated human monocyte cell line (THP-1 cells) exposed to Aß were used as models of AD neuroinflammation. We have studied the potential of MaR1 to inhibit pro-inflammatory activation of Aß and assessed its ability to stimulate phagocytosis of Aß42 . MaR1 inhibited the Aß42 -induced increase in cytokine secretion and stimulated the uptake of Aß42 in both MdM and differentiated THP-1 cells. MaR1 was also found to decrease chemokine secretion and reduce the associated increase in the activation marker CD40. Activation of kinases involved in transduction of inflammation was not affected by MaR1, but the activity of nuclear factor (NF)-κB was decreased. Our data show that MaR1 exerts effects that indicate a pro-resolving role in the context of AD and thus presents itself as a potential therapeutic target for AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Docosahexaenoic Acids/metabolism , Microglia/metabolism , Alzheimer Disease/genetics , Amyloid beta-Peptides/genetics , Blotting, Western , Cell Survival/genetics , Cell Survival/physiology , Cells, Cultured , Docosahexaenoic Acids/genetics , Humans , Immunohistochemistry , NF-kappa B/metabolism , Phagocytosis/genetics , Phagocytosis/physiology , THP-1 Cells
3.
Glia ; 68(7): 1347-1360, 2020 07.
Article in English | MEDLINE | ID: mdl-31944407

ABSTRACT

Inflammation can be resolved by pro-homeostatic lipids called specialized pro-resolving mediators (SPMs) upon activation of their receptors. Dysfunctional inflammatory resolution is now considered as a driver of chronic neuroinflammation and Alzheimer's disease (AD) pathogenesis. We have previously shown that SPM levels were reduced and also that SPM-binding receptors were increased in patients with AD compared to age-matched controls. Individuals with Down syndrome (DS) exhibit accelerated acquisition of AD neuropathology, dementia, and neuroinflammation at an earlier age than the general population. Beneficial effects of inducing resolution in DS have not been investigated previously. The effects of the SPM resolvin E1 (RvE1) in a DS mouse model (Ts65Dn) were investigated with regard to inflammation, neurodegeneration, and memory deficits. A moderate dose of RvE1 for 4 weeks in middle-aged Ts65Dn mice elicited a significant reduction in memory loss, along with reduced levels of serum pro-inflammatory cytokines, and reduced microglial activation in the hippocampus of Ts65Dn mice but had no effects in age-matched normosomic mice. There were no observable adverse side effects in Ts65Dn or in normosomic mice. These findings suggest that SPMs may represent a novel drug target for individuals with DS and others at risk of developing AD.


Subject(s)
Alzheimer Disease/drug therapy , Down Syndrome/drug therapy , Eicosapentaenoic Acid/analogs & derivatives , Hippocampus/drug effects , Memory Disorders/prevention & control , Alzheimer Disease/pathology , Animals , Down Syndrome/pathology , Eicosapentaenoic Acid/pharmacology , Hippocampus/pathology , Male , Maze Learning/physiology , Mice, Transgenic
4.
Biomacromolecules ; 18(12): 4323-4330, 2017 Dec 11.
Article in English | MEDLINE | ID: mdl-29131611

ABSTRACT

Amine functional polymers, especially cationically charged, are interesting biomacromolecules for several reasons, including easy cell membrane entrance, their ability to escape endosomes through the proton sponge effect, spontaneous complexation and delivery of drugs and siRNA, and simple functionalization in aqueous solutions. Dendrimers, a subclass of precision polymers, are monodisperse and exhibit a large and exact number of peripheral end groups in relation to their size and have shown promise in drug delivery, biomedical imaging and as antiviral agents. In this work, hydroxyl functional dendrimers of generation 1 to 5 based on 2,2-bis(methylol)propionic acid (bis-MPA) were modified to bear 6 to 96 peripheral amino groups through esterification reactions with beta-alanine. All dendrimers were isolated in high yields and with remarkable monodispersity. This was successfully accomplished utilizing the present advantages of fluoride-promoted esterification (FPE) with imidazole-activated monomers. Straightforward postfunctionalization was conducted on a second generation amino-functional dendrimer with tetraethylene glycol through NHS-amidation and carbonyl diimidazole (CDI) activation to full conversion with short reaction times. Fast biodegradation of the dendrimers through loss of peripheral beta-alanine groups was observed and generational- and dose-dependent cytotoxicity was evaluated with a set of cell lines. An increase in neurotoxicity compared to hydroxyl-functional dendrimers was shown in neuronal cells, however, the dendrimers were slightly less neurotoxic than commercially available poly(amidoamine) dendrimers (PAMAMs). Additionally, their effect on bacteria was evaluated and the second generation dendrimer was found unique inhibiting the growth of Escherichia coli at physiological conditions while being nontoxic toward human cells. Finally, these results cement a robust and sustainable synthetic route to amino-functional polyester dendrimers with interesting chemical and biological properties.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Dendrimers/chemistry , Polyesters/chemistry , Polymers/chemistry , Animals , Cations/chemistry , Cell Line , Cell Line, Tumor , Drug Delivery Systems/methods , Fluorides/chemistry , Humans , Mice , Phosphates/chemistry , Polyamines/chemistry , Polyethylene Glycols/chemistry , RAW 264.7 Cells , RNA, Small Interfering/chemistry
5.
J Lipid Res ; 56(3): 674-681, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25616438

ABSTRACT

Specialized proresolving mediators (SPMs) induce resolution of inflammation. SPMs are derivatives of n-3 and n-6 PUFAs and may mediate their beneficial effects. It is unknown whether supplementation with PUFAs influences the production of SPMs. Alzheimer's disease (AD) is associated with brain inflammation and reduced levels of SPMs. The OmegAD study is a randomized, double-blind, and placebo-controlled clinical trial on AD patients, in which placebo or a supplement of 1.7 g DHA and 0.6 g EPA was taken daily for 6 months. Plasma levels of arachidonic acid decreased, and DHA and EPA levels increased after 6 months of n-3 FA treatment. Peripheral blood mononuclear cells (PBMCs) were obtained before and after the trial. Analysis of the culture medium of PBMCs incubated with amyloid-ß 1-40 showed unchanged levels of the SPMs lipoxin A4 and resolvin D1 in the group supplemented with n-3 FAs, whereas a decrease was seen in the placebo group. The changes in SPMs showed correspondence to cognitive changes. Changes in the levels of SPMs were positively correlated to changes in transthyretin. We conclude that supplementation with n-3 PUFAs for 6 months prevented a reduction in SPMs released from PBMCs of AD patients, which was associated with changes in cognitive function.


Subject(s)
Alzheimer Disease , Cognition/drug effects , Fatty Acids, Omega-3/administration & dosage , Inflammation Mediators/blood , Leukocytes, Mononuclear/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/blood , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/pharmacology , Cells, Cultured , Double-Blind Method , Fatty Acids, Omega-6/administration & dosage , Female , Humans , Leukocytes, Mononuclear/pathology , Male , Peptide Fragments/pharmacology
6.
Alzheimers Dement ; 11(1): 40-50.e1-2, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24530025

ABSTRACT

BACKGROUND: Resolution is the final stage of the inflammatory response, when restoration of tissue occurs. Failure may lead to chronic inflammation, which is known as part of the pathology in the brain of individuals with Alzheimer's disease (AD). METHODS: Specialized pro-resolving mediators (SPMs), receptors, biosynthetic enzyme, and downstream effectors involved in resolution were analyzed in postmortem hippocampal tissue from AD patients and non-AD subjects. SPMs were analyzed in cerebrospinal fluid (CSF). RESULTS: SPMs and SPM receptors were detected in the human brain. Levels of the SPM lipoxin A4 (LXA4) were reduced in AD, both in the CSF and hippocampus. An enzyme involved in LXA4 synthesis and two SPM receptors were elevated in AD brains. LXA4 and RvD1 levels in CSF correlated with Mini-Mental State Examination (MMSE) scores. CONCLUSIONS: A resolution pathway exists in the brain and the alterations described herein strongly suggest a dysfunction of this pathway in AD. MMSE correlations suggest a connection with cognitive function in AD.


Subject(s)
Alzheimer Disease/metabolism , Hippocampus/metabolism , Inflammation Mediators/metabolism , Aged , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/enzymology , Alzheimer Disease/pathology , Amyloid beta-Peptides/cerebrospinal fluid , Biomarkers/cerebrospinal fluid , Case-Control Studies , Cognitive Dysfunction/cerebrospinal fluid , Cognitive Dysfunction/enzymology , Cognitive Dysfunction/pathology , Docosahexaenoic Acids/cerebrospinal fluid , Female , Hippocampus/enzymology , Hippocampus/pathology , Humans , Inflammation/cerebrospinal fluid , Inflammation/enzymology , Inflammation/metabolism , Inflammation/pathology , Inflammation Mediators/cerebrospinal fluid , Lipoxins/cerebrospinal fluid , Lipoxygenase/cerebrospinal fluid , Male , Middle Aged , Receptors, Formyl Peptide/analysis , Receptors, Lipoxin/analysis , tau Proteins/cerebrospinal fluid
7.
J Cell Mol Med ; 17(11): 1434-43, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24034597

ABSTRACT

Experimental neural cell therapies, including donor neural stem/progenitor cells (NPCs) have been reported to offer beneficial effects on the recovery after an injury and to counteract inflammatory and degenerative processes in the central nervous system (CNS). The interplay between donor neural cells and the host CNS still to a large degree remains unclear, in particular in human allogeneic conditions. Here, we focused our studies on the interaction of human NPCs and microglia utilizing a co-culture model. In co-cultures, both NPCs and microglia showed increased survival and proliferation compared with mono-cultures. In the presence of microglia, a larger subpopulation of NPCs expressed the progenitor cell marker nestin, whereas a smaller group of NPCs expressed the neural markers polysialylated neural cell adhesion molecule, A2B5 and glial fibrillary acidic protein compared with NPC mono-cultures. Microglia thus hindered differentiation of NPCs. The presence of human NPCs increased microglial phagocytosis of latex beads. Furthermore, we observed that the expression of CD200 molecules on NPCs and the CD200 receptor protein on microglia was enhanced in co-cultures, whereas the release of transforming growth factor-ß was increased suggesting anti-inflammatory features of the co-cultures. To conclude, the interplay between human allogeneic NPCs and microglia, significantly affected their respective proliferation and phenotype. Neural cell therapy including human donor NPCs may in addition to offering cell replacement, modulate host microglial phenotypes and functions to benefit neuroprotection and repair.


Subject(s)
Microglia/physiology , Neural Stem Cells/physiology , Allografts , Antigens, CD/metabolism , Antigens, Surface/metabolism , Cell Communication , Cell Differentiation , Cell Proliferation , Cell Survival , Cells, Cultured , Coculture Techniques , Humans , Interleukin-6/metabolism , Orexin Receptors , Phagocytosis , Phenotype , Receptors, Cell Surface/metabolism , Regenerative Medicine , Transforming Growth Factor beta1/metabolism
8.
Curr Opin Clin Nutr Metab Care ; 15(2): 134-43, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22316559

ABSTRACT

PURPOSE OF REVIEW: The omega-3 fatty acids (ω-3 FAs) docosahexaenoic acid and eicosapentaenoic acid are dietary components which have been ascribed many different health benefits. Inflammation is present in, and contributes to, pathological conditions in the central nervous system (CNS). Microglia are the primary cells with immune function in the CNS, and inflammation mediated by activated microglia is present in pathological conditions. In this review, we present and discuss findings on the modulation of microglial activities by ω-3 FAs in vivo as well as in vitro, and propose mechanisms for their effects. RECENT FINDINGS: The majority of studies show that ω-3 FAs have anti-inflammatory effects on microglia. However, phagocytosis is an activity associated with inflammation and is increased by ω-3 FAs. This can be understood in the light of recent research on the resolution of inflammation. Resolution is induced by proresolving factors, which are metabolites of ω-3 FAs. Proresolving factors are anti-inflammatory and have been shown to increase phagocytosis. Other mechanisms of the anti-inflammatory actions of ω-3 FAs involve the peroxisome proliferator-activated receptor-γ, ω-3 FA incorporation into the cell membrane, and inhibition of ion currents. SUMMARY: Immunomodulation by ω-3 FAs is mediated by several pathways that are interconnected and is a potential therapy for disorders in the CNS.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Docosahexaenoic Acids/pharmacology , Eicosapentaenoic Acid/pharmacology , Immunomodulation/drug effects , Microglia/drug effects , Animals , Central Nervous System/drug effects , Central Nervous System/metabolism , Humans , Inflammation/drug therapy , Inflammation/physiopathology , Microglia/metabolism , Models, Animal , Phagocytosis/drug effects
9.
Commun Biol ; 5(1): 245, 2022 03 21.
Article in English | MEDLINE | ID: mdl-35314851

ABSTRACT

Sustained microglial activation and increased pro-inflammatory signalling cause chronic inflammation and neuronal damage in Alzheimer's disease (AD). Resolution of inflammation follows neutralization of pathogens and is a response to limit damage and promote healing, mediated by pro-resolving lipid mediators (LMs). Since resolution is impaired in AD brains, we decided to test if intranasal administration of pro-resolving LMs in the AppNL-G-F/NL-G-F mouse model for AD could resolve inflammation and ameliorate pathology in the brain. A mixture of the pro-resolving LMs resolvin (Rv) E1, RvD1, RvD2, maresin 1 (MaR1) and neuroprotectin D1 (NPD1) was administered to stimulate their respective receptors. We examined amyloid load, cognition, neuronal network oscillations, glial activation and inflammatory factors. The treatment ameliorated memory deficits accompanied by a restoration of gamma oscillation deficits, together with a dramatic decrease in microglial activation. These findings open potential avenues for therapeutic exploration of pro-resolving LMs in AD, using a non-invasive route.


Subject(s)
Alzheimer Disease , Administration, Intranasal , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Amyloid beta-Peptides , Animals , Inflammation , Mice
10.
Curr Alzheimer Res ; 18(2): 157-170, 2021.
Article in English | MEDLINE | ID: mdl-33784960

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) develops into dementia after several years, and subjective cognitive impairment (SCI) and mild cognitive impairment (MCI) are used as intermediary diagnoses of increasing severity. Inflammation is an important part of AD pathology and provides potential novel biomarkers and treatment targets. OBJECTIVE: To identify novel potential biomarkers of AD in cerebrospinal fluid (CSF) and create a molecular pattern of inflammatory factors providing differentiation between AD and SCI. METHODS: We analyzed 43 inflammatory-related mediators in CSF samples from a cohort of SCI and AD cases vetted for confounding factors (Training cohort). Using multivariate analysis (MVA), a model for discrimination between SCI and AD was produced, which we then applied to a larger nonvetted cohort (named Test cohort). The data were analyzed for factors showing differences between diagnostic groups and factors that differed between the vetted and non-vetted cohorts. The relationship of the factors to the agreement between model and clinical diagnosis was investigated. RESULTS: A good MVA model able to discriminate AD from SCI without including tangle and plaque biomarkers was produced from the Training cohort. The model showed 50% agreement with clinical diagnosis in the Test cohort. Comparison of the cohorts indicated different patterns of factors distinguishing SCI from AD. As an example, soluble interleukin (IL)-6Rα showed lower levels in AD cases in the Training cohort, whereas placental growth factor (PlGF) and serum amyloid A (SAA) levels were higher in AD cases of the Test cohort. The levels of p-tau were also higher in the Training cohort. CONCLUSION: This study provides new knowledge regarding the involvement of inflammation in AD by indicating different patterns of factors in CSF depending on whether potential confounding comorbidities are present or not, and presents sIL-6Rα as a potential new biomarker for improved diagnosis of AD.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction/diagnosis , Comorbidity , Inflammation , Aged , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/pathology , Amyloid/blood , Female , Humans , Male , Middle Aged , Sensitivity and Specificity , tau Proteins
11.
Acta Neuropathol Commun ; 9(1): 116, 2021 06 29.
Article in English | MEDLINE | ID: mdl-34187579

ABSTRACT

Sustained brain chronic inflammation in Alzheimer's disease (AD) includes glial cell activation, an increase in cytokines and chemokines, and lipid mediators (LMs), concomitant with decreased pro-homeostatic mediators. The inflammatory response at the onset of pathology engages activation of pro-resolving, pro-homeostatic LMs followed by a gradual decrease. We used an APP knock-in (App KI) AD mouse that accumulates ß-amyloid (Aß) and presents cognitive deficits (at 2 and 6 months of age, respectively) to investigate LMs, their precursors, biosynthetic enzymes and receptors, glial activation, and inflammatory proteins in the cerebral cortex and hippocampus at 2-, 4-, 8- and 18-month-old in comparison with wild-type (WT) mice. We used LC-mass-spectrometry and MALDI molecular imaging to analyze LMs and phospholipids, and immunochemistry for proteins. Our results revealed an age-specific lipid and cytokine profile, and glial activation in the App KI mice. Despite an early onset of Aß pathology, pro-inflammatory and pro-resolving LMs were prominently increased only in the oldest age group. Furthermore, the LM biosynthetic enzymes increased, and their receptor expression decreased in the aged App KI mice. Arachidonic acid (AA)-containing phospholipid molecular species were elevated, correlating with decreased cPLA2 activity. MALDI molecular imaging depicted differential distribution of phospholipids according to genotype in hippocampal layers. Brain histology disclosed increased microglia proliferation starting from young age in the App KI mice, while astrocyte numbers were enhanced in older ages. Our results demonstrate that the brain lipidome is modified preferentially during aging as compared to amyloid pathology in the model studied here. However, alterations in phospholipids signal early pathological changes in membrane composition.


Subject(s)
Aging/pathology , Alzheimer Disease/pathology , Brain/pathology , Phospholipids/metabolism , Aging/metabolism , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Brain/metabolism , Disease Models, Animal , Gene Knock-In Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
12.
J Alzheimers Dis ; 83(3): 1291-1301, 2021.
Article in English | MEDLINE | ID: mdl-34420949

ABSTRACT

BACKGROUND: Studies have suggested a connection between a decrease in the levels of polyunsaturated fatty acids (PUFAs) and Alzheimer's disease (AD). We aimed to assess the effect of supplementation with omega-3 fatty acids (n-3 FAs) on biomarkers analyzed in the cerebrospinal fluid (CSF) of patients diagnosed with AD. OBJECTIVE: To investigate the effects of daily supplementation with 2.3 g of PUFAs in AD patients on the biomarkers in CSF described below. We also explored the possible correlation between these biomarkers and the performance in the cognitive test Mini-Mental State Examination (MMSE). METHODS: Thirty-three patients diagnosed with AD were randomized to either treatment with a daily intake of 2.3 g of n-3 FAs (n  =  18) or placebo (n  =  15). CSF samples were collected at baseline and after six months of treatment, and the following biomarkers were analyzed: Aß 38, Aß 40, Aß 42, t-tau, p-tau, neurofilament light (NfL), chitinase-3-like protein 1 (YKL-40), acetylcholinesterase (AChE), butyrylcholinesterase (BuChE), soluble IL-1 receptor type II (sIL-1RII), and IL-6. RESULTS: There were no significant differences between the groups concerning the level of the different biomarkers in the CSF at baseline. Within the treatment group, there was a small but significant increase in both YKL-40 (p = 0.04) and NfL (p = 0.03), while the other CSF biomarkers remained stable. CONCLUSION: Supplementation with n-3 FAs had a statistically significant effect on NfL and YKL-40, resulting in an increase of both biomarkers, indicating a possible increase of inflammatory response and axonal damage. This increase in biomarkers did not correlate with MMSE score.


Subject(s)
Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/diet therapy , Biomarkers , Fatty Acids, Omega-3/cerebrospinal fluid , Fatty Acids, Omega-3/therapeutic use , Administration, Oral , Aged , Alzheimer Disease/blood , Amyloid beta-Peptides/cerebrospinal fluid , Biomarkers/blood , Biomarkers/cerebrospinal fluid , Fatty Acids, Omega-3/blood , Female , Humans , Male , Mental Status and Dementia Tests/statistics & numerical data , Neurofilament Proteins/cerebrospinal fluid , tau Proteins/cerebrospinal fluid
13.
J Lipid Res ; 51(5): 1179-85, 2010 May.
Article in English | MEDLINE | ID: mdl-19965584

ABSTRACT

Omega-3 fatty acids, e.g., dokosahexaenoic acid (DHA) and eikosapentaenoic acid (EPA), ameliorate inflammatory reactions by various mechanisms, but the role of prostaglandins remains unclear. Our aim was to determine if dietary supplementation with a DHA-rich fish oil influenced the release of PGF(2alpha) from peripheral blood mononuclear cells (PBMC). In the OmegAD study, 174 Alzheimer disease patients received either 1.7 g DHA plus 0.6 g EPA or a placebo daily for six months. PBMCs from the 21 (9 on fish oil and 12 on placebo) first-randomized patients were stimulated with either lipopolysaccharide (LPS) or phytohemagglutinin (PHA) before and after 6 months. Our results showed that plasma concentrations of DHA and EPA increased significantly at 6 months in the omega-3 group. PGF(2alpha) release from LPS- (but not from PHA-) stimulated PBMC was significantly diminished in this group; no change was noted in the placebo group. PGF(2alpha) changes correlated inversely with changes in plasma DHA and EPA. Decreased IL-6 and IL-1(beta) levels correlated with decreased PGF(2alpha) levels. The stimulus-specific PGF(2alpha) release from PBMC after 6 months of oral supplementation with the DHA-rich fish oil might be one event related to reduced inflammatory reactions associated with omega-3 fatty acid intake.


Subject(s)
Dietary Supplements , Dinoprost/metabolism , Fatty Acids, Omega-3/administration & dosage , Fatty Acids, Omega-3/pharmacology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Administration, Oral , Aged , Aged, 80 and over , Alzheimer Disease/blood , Alzheimer Disease/metabolism , Culture Media, Conditioned/metabolism , Cytokines/metabolism , Dinoprost/biosynthesis , Dinoprost/blood , Fatty Acids, Omega-3/blood , Female , Humans , Lipopolysaccharides/pharmacology , Male , Middle Aged , Monocytes/drug effects , Monocytes/metabolism , Phytohemagglutinins/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism
14.
Brain Pathol ; 30(3): 614-640, 2020 05.
Article in English | MEDLINE | ID: mdl-31912564

ABSTRACT

Neuroinflammation is a key element of AD pathology and conceivably a result of a disturbed resolution. Resolution of inflammation is an active process which is strictly orchestrated following the acute inflammatory response after removal of the inflammatory stimuli. Acute inflammation is actively terminated by specialized pro-resolving mediators (SPMs) thereby promoting healing and return to homeostasis. Failed resolution may contribute to persistent neuroinflammation and aggravate AD pathology. BLT1 (leukotriene B4 receptor) and ChemR23 (chemerin receptor 23) are receptors for the SPM resolvin (Rv) E1 and are important clinical targets for ending inflammation. In AD, the levels of SPMs are decreased, and pro-inflammatory mediators are increased. In the current study, the distribution of BLT1 and ChemR23 receptors in control brains and in AD as well as correlations with AD pathology was examined for the first time. BLT1 and ChemR23 were analyzed in different regions of post-mortem human brain from cases with AD, early-onset AD and mild cognitive impairment (MCI) and healthy controls, using western blotting and immunohistochemistry. BLT1 and ChemR23 were detected in neurons and glial cells in all examined regions of the human brain, with markedly higher levels in AD than in controls. The receptor levels correlated with the density of staining for the inflammation markers HLA-DR and YKL-40 for microglia and astrocytes, respectively, and elevated staining coincided with high Braak stages in AD. The relative staining densities of these receptors were higher in the basal forebrain, cingulate gyrus and hippocampal regions compared to the cerebellum and frontal cortex (BA46). In conclusion, alterations in the expression of the resolution receptor BLT1 in AD have not been reported previously and the changes in both BLT1 and ChemR23 suggest a disturbed resolution pathway in several regions of the AD brain that may play a role in disease pathology.


Subject(s)
Alzheimer Disease/metabolism , Brain/metabolism , Receptors, Chemokine/metabolism , Receptors, Leukotriene B4/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Astrocytes/metabolism , Astrocytes/pathology , Brain/pathology , Female , Humans , Inflammation/metabolism , Inflammation/pathology , Male , Middle Aged , Neurons/metabolism , Neurons/pathology
15.
Dement Geriatr Cogn Disord ; 27(5): 481-90, 2009.
Article in English | MEDLINE | ID: mdl-19439966

ABSTRACT

BACKGROUND: omega-3 fatty acids (omega-3 FAs) found in dietary fish or fish oils are anti-inflammatory agents that may influence Alzheimer's disease (AD). OBJECTIVE: To study the effects of dietary omega-3 FA supplementation on inflammatory markers in cerebrospinal fluid (CSF) and plasma from patients with mild to moderate AD. METHODS: Thirty-five patients (70.3 +/- 8.2 years) were randomized to a daily intake of 2.3 g omega-3 FAs or placebo for 6 months. The inflammatory markers interleukin (IL)-6, tumour necrosis factor-alpha and soluble interleukin-1 receptor type II (sIL-1RII) were analysed in CSF and plasma at baseline and at 6 months. The AD markers tau-protein, hyperphosphorylated tau-protein and beta-amyloid (Abeta(1-42)) were assessed in CSF. High-sensitivity C-reactive protein was assessed in plasma. A possible relation to the APOE genotype was investigated. RESULTS: There was no significant treatment effect of omega-3 FAs on inflammatory and AD biomarkers in CSF or on inflammatory markers in plasma, nor was there any relation with APOE. A significant correlation was observed at baseline between sIL-1RII and Abeta(1-42) levels in CSF. CONCLUSIONS: Treatment of AD patients with omega-3 FAs for 6 months did not influence inflammatory or biomarkers in CSF or plasma. The correlation between sIL-1RII and Abeta(1-42) may reflect the reciprocal interactions between IL-1 and Abeta peptides.


Subject(s)
Alzheimer Disease/blood , Alzheimer Disease/cerebrospinal fluid , Fatty Acids, Omega-3/therapeutic use , Inflammation/blood , Inflammation/cerebrospinal fluid , Aged , Amyloid beta-Peptides/blood , Apolipoproteins E/genetics , Biomarkers/blood , Biomarkers/cerebrospinal fluid , C-Reactive Protein/cerebrospinal fluid , Cytokines/blood , Dietary Supplements , Double-Blind Method , Female , Genotype , Humans , Male , Neuropsychological Tests , tau Proteins/blood
16.
ACS Chem Neurosci ; 10(3): 1462-1477, 2019 03 20.
Article in English | MEDLINE | ID: mdl-30673220

ABSTRACT

A new generation of ligands designed to interact with the α-helix/ß-strand discordant region of the amyloid-ß peptide (Aß) and to counteract its oligomerization is presented. These ligands are designed to interact with and stabilize the Aß central helix (residues 13-26) in an α-helical conformation with increased interaction by combining properties of several first-generation ligands. The new peptide-like ligands aim at extended hydrophobic and polar contacts across the central part of the Aß, that is, "clamping" the target. Molecular dynamics (MD) simulations of the stability of the Aß central helix in the presence of a set of second-generation ligands were performed and revealed further stabilization of the Aß α-helical conformation, with larger number of polar and nonpolar contacts between ligand and Aß, compared to first-generation ligands. The synthesis of selected novel Aß-targeting ligands was performed in solution via an active ester coupling approach or on solid-phase using an Fmoc chemistry protocol. This included incorporation of aliphatic hydrocarbon moieties, a branched triamino acid with an aliphatic hydrocarbon tail, and an amino acid with a 4'- N, N-dimethylamino-1,8-naphthalimido group in the side chain. The ability of the ligands to reduce Aß1-42 neurotoxicity was evaluated by gamma oscillation experiments in hippocampal slice preparations. The "clamping" second-generation ligands were found to be effective antineurotoxicity agents and strongly prevented the degradation of gamma oscillations by physiological concentration of monomeric Aß1-42 at a stoichiometric ratio.


Subject(s)
Amyloid beta-Peptides/toxicity , Drug Delivery Systems/methods , Molecular Dynamics Simulation , Peptide Fragments/administration & dosage , Peptidomimetics/administration & dosage , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Animals , Cell Line, Tumor , Female , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Ligands , Male , Mice , Mice, Inbred C57BL , Organ Culture Techniques , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/metabolism , Peptide Fragments/toxicity , Peptidomimetics/metabolism
17.
J Alzheimers Dis ; 69(1): 189-197, 2019.
Article in English | MEDLINE | ID: mdl-30958356

ABSTRACT

BACKGROUND: Trials of supplementation with omega-3 fatty acids (ω3-FAs) in patients with mild cognitive impairment or Alzheimer's disease (AD) have produced inconsistent effects on cognitive decline. There is evidence of an interaction between B vitamin status and ω3-FAs in relation to brain atrophy and cognitive decline. OBJECTIVE: We investigated whether baseline levels of plasma total homocysteine (tHcy), a marker of B vitamin status, modify the effects of ω3-FAs supplementation on cognitive performance in moderate AD. METHODS: This post hoc analysis of the OmegAD trial included 171 community-based patients with AD (MMSE≥15): 88 patients received daily doses of 1.7 g docosahexaenoic acid and 0.6 g eicosapentaenoic acid for 6 months. Treatment outcome on cognition was analyzed according to baseline levels of tHcy using a general linear model and ANCOVA. RESULTS: We found significant interactions between ω3-FA supplementation and tHcy on cognition and clinical stage assessed by MMSE (p = 0.040), global CDR (p = 0.059), and CDRsob (p = 0.023), but not on ADAS-cog (p = 0.649). In patients with tHcy levels <11.7µmol/L, ω3-FA supplementation improved cognitive performance as measured by MMSE (+7.1%, 95% CI: 0.59 to 13.7%, p = 0.033) and clinical status as measured by CDRsob (-22.3%, 95% CI: -5.8 to -38.7%, p = 0.009) compared with placebo. CONCLUSION: The effect of ω3-FA supplementation on MMSE and CDR appears to be influenced by baseline tHcy, suggesting that adequate B vitamin status is required to obtain beneficial effects of ω3-FA on cognition.


Subject(s)
Alzheimer Disease/drug therapy , Cognition/drug effects , Fatty Acids, Omega-3/therapeutic use , Homocysteine/blood , Aged , Alzheimer Disease/blood , Alzheimer Disease/psychology , Dietary Supplements , Fatty Acids, Omega-3/pharmacology , Female , Humans , Male , Treatment Outcome
18.
Ther Adv Neurol Disord ; 11: 1756286418791107, 2018.
Article in English | MEDLINE | ID: mdl-30116300

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disease characterized by progressive memory loss and dementia. Accumulating evidence suggests that inflammation is involved in the pathogenesis of AD. Epidemiological studies suggest that use of anti-inflammatory drugs is associated with a lower incidence of AD. However, clinical trials with anti-inflammatory drugs have not been successful. Recent studies have shown that inflammation is resolved by a process that is mediated by a group of lipid mediators, so called specialized pro-resolving lipid mediators (SPMs). Unlike anti-inflammatory strategies, which usually involve inhibition of the synthesis of inflammatory mediators, stimulating the resolution of inflammation is aimed at ending inflammation in a similar fashion as under normal physiological conditions. We have previously shown that pathways of resolution are impaired in AD. Moreover, we found that SPMs can improve neuronal survival and increase microglial phagocytosis of amyloid beta (Aß) in in vitro studies, indicating that stimulating resolution of inflammation may be a potential therapeutic target in AD. In this review, we summarize recent findings regarding resolution of inflammation in AD. We also discuss possible strategies to stimulate the resolution of inflammation in AD, specifically focusing on signaling pathways, including SPMs, their receptors and enzymes involved in their formation.

19.
J Alzheimers Dis ; 61(2): 515-519, 2018.
Article in English | MEDLINE | ID: mdl-29154271

ABSTRACT

Low tissue levels of the major marine ω3 fatty acids (FAs) DHA and EPA are found in Alzheimer's disease (AD). We investigated if healthy proxies to AD patients have higher levels of these ω3 FAs. We observed lower levels of EPA and DHA in subcutaneous adipose tissue biopsies from 64 AD patients compared with 16 cognitively healthy proxies. No significant difference was observed when pairwise comparisons were made between a subset of 16 AD patients and their co-habiting proxies. Larger studies are needed to replicate these findings and to determine if they could depend on FA intake or differences in metabolism.


Subject(s)
Alzheimer Disease/metabolism , Fatty Acids, Omega-3/analysis , Subcutaneous Fat/chemistry , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Case-Control Studies , Dietary Supplements , Female , Humans , Male , Regression Analysis
20.
Physiol Behav ; 92(1-2): 121-8, 2007 Sep 10.
Article in English | MEDLINE | ID: mdl-17597167

ABSTRACT

There is ample evidence for the occurrence of inflammatory processes in most major neurodegenerative disorders, both in acute conditions such as traumatic brain injury and stroke, and in chronic disorders such as Alzheimer's disease, epilepsy, amyotrophic lateral sclerosis and Parkinson's disease. Studies on inflammatory factors such as pro- and antiinflammatory cytokines in experimental models of neurodegenerative disorders suggest that they are not merely bystanders, but may be involved in the neurodegenerative process. In addition, there are findings indicating that inflammatory factors may have beneficial effects on the nervous system, particularly during development of the nervous system. The challenge is to understand when, where and during which circumstances inflammation and inflammatory factors are positive or negative for neuronal survival and functioning. Some of our studies on cytokines, particularly the interleukin-1 system, are summarised and discussed in relation to neurodegeneration, cognition, and temperature changes.


Subject(s)
Body Temperature Regulation/immunology , Brain/immunology , Cytokines/immunology , Inflammation , Neurodegenerative Diseases/immunology , Animals , Central Nervous System Diseases/immunology , Central Nervous System Diseases/pathology , Cognition/physiology , Humans , Interleukin-1/immunology
SELECTION OF CITATIONS
SEARCH DETAIL