Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2023 Nov 17.
Article in English | MEDLINE | ID: mdl-38014333

ABSTRACT

Type 2 diabetes (T2D) is implicated as a risk factor for Alzheimer's disease (AD), the most common form of dementia. In this work, we investigated neuroinflammatory responses of primary neurons to potentially circulating, blood-brain barrier (BBB) permeable metabolites associated with AD, T2D, or both. We identified nine metabolites associated with protective or detrimental properties of AD and T2D in literature (lauric acid, asparagine, fructose, arachidonic acid, aminoadipic acid, sorbitol, retinol, tryptophan, niacinamide) and stimulated primary mouse neuron cultures with each metabolite before quantifying cytokine secretion via Luminex. We employed unsupervised clustering, inferential statistics, and partial least squares discriminant analysis to identify relationships between cytokine concentration and disease-associations of metabolites. We identified MCP-1, a cytokine associated with monocyte recruitment, as differentially abundant between neurons stimulated by metabolites associated with protective and detrimental properties of AD and T2D. We also identified IL-9, a cytokine that promotes mast cell growth, to be differentially associated with T2D. Indeed, cytokines, such as MCP-1 and IL-9, released from neurons in response to BBB-permeable metabolites associated with T2D may contribute to AD development by downstream effects of neuroinflammation.

2.
Cell Mol Bioeng ; 16(4): 405-421, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37811007

ABSTRACT

Introduction: Neuroinflammation and metabolic dysfunction are early alterations in Alzheimer's disease (AD) brain that are thought to contribute to disease onset and progression. Glial activation due to protein deposition results in cytokine secretion and shifts in brain metabolism, which have been observed in AD patients. However, the mechanism by which this immunometabolic feedback loop can injure neurons and cause neurodegeneration remains unclear. Methods: We used Luminex XMAP technology to quantify hippocampal cytokine concentrations in the 5xFAD mouse model of AD at milestone timepoints in disease development. We used partial least squares regression to build cytokine signatures predictive of disease progression, as compared to healthy aging in wild-type littermates. We applied the disease-defining cytokine signature to wild-type primary neuron cultures and measured downstream changes in gene expression using the NanoString nCounter system and mitochondrial function using the Seahorse Extracellular Flux live-cell analyzer. Results: We identified a pattern of up-regulated IFNγ, IP-10/CXCL10, and IL-9 as predictive of advanced disease. When healthy neurons were exposed to these cytokines in proportions found in diseased brain, gene expression of mitochondrial electron transport chain complexes, including ATP synthase, was suppressed. In live cells, basal and maximal mitochondrial respiration were impaired following cytokine stimulation. Conclusions: We identify a pattern of cytokine secretion predictive of progressing amyloid-ß pathology in the 5xFAD mouse model of AD that reduces expression of mitochondrial electron transport complexes and impairs mitochondrial respiration in healthy neurons. We establish a mechanistic link between disease-specific immune cues and impaired neuronal metabolism, potentially causing neuronal vulnerability and susceptibility to degeneration in AD. Supplementary Information: The online version contains supplementary material available at 10.1007/s12195-023-00782-y.

3.
bioRxiv ; 2023 Apr 07.
Article in English | MEDLINE | ID: mdl-37066287

ABSTRACT

Introduction: Neuroinflammation and metabolic dysfunction are early alterations in Alzheimer's disease brain that are thought to contribute to disease onset and progression. Glial activation due to protein deposition results in cytokine secretion and shifts in brain metabolism, which have been observed in Alzheimer's disease patients. However, the mechanism by which this immunometabolic feedback loop can injure neurons and cause neurodegeneration remains unclear. Methods: We used Luminex XMAP technology to quantify hippocampal cytokine concentrations in the 5xFAD mouse model of Alzheimer's disease at milestone timepoints in disease development. We used partial least squares regression to build cytokine signatures predictive of disease progression, as compared to healthy aging in wild-type littermates. We applied the disease-defining cytokine signature to wild-type primary neuron cultures and measured downstream changes in gene expression using the NanoString nCounter system and mitochondrial function using the Seahorse Extracellular Flux live-cell analyzer. Results: We identified a pattern of up-regulated IFNγ, IP-10, and IL-9 as predictive of advanced disease. When healthy neurons were exposed to these cytokines in proportions found in diseased brain, gene expression of mitochondrial electron transport chain complexes, including ATP synthase, was suppressed. In live cells, basal and maximal mitochondrial respiration were impaired following cytokine stimulation. Conclusions: An Alzheimer's disease-specific pattern of cytokine secretion reduces expression of mitochondrial electron transport complexes and impairs mitochondrial respiration in healthy neurons. We establish a mechanistic link between disease-specific immune cues and impaired neuronal metabolism, potentially causing neuronal vulnerability and susceptibility to degeneration in Alzheimer's disease.

4.
J Neurochem ; 165(4): 536-549, 2023 05.
Article in English | MEDLINE | ID: mdl-36762973

ABSTRACT

Apolipoprotein E (APOE) is a lipid transporter produced predominantly by astrocytes in the brain. The ε4 variant of APOE (APOE4) is the strongest and most common genetic risk factor for Alzheimer's disease (AD). Although the molecular mechanisms of this increased risk are unclear, APOE4 is known to alter immune signaling and lipid and glucose metabolism. Astrocytes provide various forms of support to neurons, including regulating neuronal metabolism and immune responses through cytokine signaling. Changes in astrocyte function because of APOE4 may therefore decrease neuronal support, leaving neurons more vulnerable to stress and disease insults. To determine whether APOE4 alters astrocyte neuronal support functions, we measured glycolytic and oxidative metabolism of neurons treated with conditioned media from APOE4 or APOE3 (the common, risk-neutral variant) primary astrocyte cultures. We found that APOE4 neurons treated with conditioned media from resting APOE4 astrocytes had similar metabolism to APOE3 neurons treated with media from resting APOE3 astrocytes, but treatment with astrocytic conditioned media from astrocytes challenged with amyloid-ß (Aß), a key pathological protein in AD, caused APOE4 neurons to increase their basal mitochondrial and glycolytic metabolic rates more than APOE3 neurons. These changes were not because of differences in astrocytic lactate production or glucose utilization, but instead correlated with increased glycolytic ATP production and a lack of cytokine secretion in response to Aß. Additionally, we identified that astrocytic cytokine signatures could predict basal metabolism of neurons treated with the astrocytic conditioned media. Together, these findings suggest that in the presence of Aß, APOE4 astrocytes alter immune and metabolic functions that result in a compensatory increase in neuronal metabolic stress.


Subject(s)
Alzheimer Disease , Apolipoprotein E4 , Mice , Animals , Humans , Apolipoprotein E4/genetics , Astrocytes/metabolism , Apolipoprotein E3/genetics , Apolipoprotein E3/metabolism , Culture Media, Conditioned/pharmacology , Mice, Transgenic , Cells, Cultured , Apolipoproteins E/metabolism , Amyloid beta-Peptides/metabolism , Neurons/metabolism , Alzheimer Disease/metabolism
5.
Neurobiol Aging ; 123: 154-169, 2023 03.
Article in English | MEDLINE | ID: mdl-36572594

ABSTRACT

The ε4 variant of apolipoprotein E (APOE) is the strongest and most common genetic risk factor for Alzheimer's disease (AD). While the mechanism of conveyed risk is incompletely understood, promotion of inflammation, dysregulated metabolism, and protein misfolding and aggregation are contributors to accelerating disease. Here we determined the concurrent effects of systemic metabolic changes and brain inflammation in young (3-month-old) and aged (18-month-old) male and female mice carrying the APOE4 gene. Using functional metabolic assays alongside multivariate modeling of hippocampal cytokine levels, we found that brain cytokine signatures are predictive of systemic metabolic outcomes, independent of AD proteinopathies. Male and female mice each produce different cytokine signatures as they age and as their systemic metabolic phenotype declines, and these signatures are APOE genotype dependent. Ours is the first study to identify a quantitative and predictive link between systemic metabolism and specific pathological cytokine signatures in the brain. Our results highlight the effects of APOE4 beyond the brain and suggest the potential for bi-directional influence of risk factors in the brain and periphery.


Subject(s)
Alzheimer Disease , Apolipoprotein E4 , Mice , Male , Female , Animals , Apolipoprotein E4/genetics , Apolipoprotein E4/metabolism , Cytokines/metabolism , Apolipoproteins E/genetics , Brain/metabolism , Genotype , Alzheimer Disease/metabolism , Apolipoprotein E3/genetics , Apolipoprotein E2/genetics
6.
ACS Chem Neurosci ; 13(13): 1979-1991, 2022 07 06.
Article in English | MEDLINE | ID: mdl-35713284

ABSTRACT

The aggregation of the amyloid beta (Aß) peptide is associated with Alzheimer's disease (AD) pathogenesis. Cell membrane composition, especially monosialotetrahexosylganglioside (GM1), is known to promote the formation of Aß fibrils, yet little is known about the roles of GM1 in the early steps of Aß oligomer formation. Here, by using GM1-contained liposomes as a mimic of the neuronal cell membrane, we demonstrate that GM1 is a critical trigger of Aß oligomerization and aggregation. We find that GM1 not only promotes the formation of Aß fibrils but also facilitates the maintenance of Aß42 oligomers on liposome membranes. We structurally characterize the Aß42 oligomers formed on the membrane and find that GM1 captures Aß by binding to its arginine-5 residue. To interrogate the mechanism of Aß42 oligomer toxicity, we design a new liposome-based Ca2+-encapsulation assay and provide new evidence for the Aß42 ion channel hypothesis. Finally, we determine the toxicity of Aß42 oligomers formed on membranes. Overall, by uncovering the roles of GM1 in mediating early Aß oligomer formation and maintenance, our work provides a novel direction for pharmaceutical research for AD.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Alzheimer Disease/metabolism , Amyloid/metabolism , Amyloid beta-Peptides/metabolism , G(M1) Ganglioside/chemistry , Humans , Liposomes , Peptide Fragments/metabolism
7.
Front Cell Neurosci ; 15: 645233, 2021.
Article in English | MEDLINE | ID: mdl-33815065

ABSTRACT

More than 6 million Americans are currently living with Alzheimer's disease (AD), and the incidence is growing rapidly with our aging population. Numerous therapeutics have failed to make it to the clinic, potentially due to a focus on presumptive pathogenic proteins instead of cell-type-specific signaling mechanisms. The tau propagation hypothesis that inter-neuronal tau transfer drives AD pathology has recently garnered attention, as accumulation of pathological tau in the brain has high clinical significance in correlating with progression of cognitive AD symptoms. However, studies on tau pathology in AD are classically neuron-centric and have greatly overlooked cell-type specific effects of tau internalization, degradation, and propagation. While the contribution of microglia to tau processing and propagation is beginning to be recognized and understood, astrocytes, glial cells in the brain important for maintaining neuronal metabolic, synaptic, trophic, and immune function which can produce, internalize, degrade, and propagate tau are understudied in their ability to affect AD progression through tau pathology. Here, we showcase evidence for whether tau uptake by astrocytes may be beneficial or detrimental to neuronal health and how astrocytes and their immunometabolic functions may be key targets for future successful AD therapies.

8.
Cell Metab ; 32(1): 44-55.e6, 2020 07 07.
Article in English | MEDLINE | ID: mdl-32402267

ABSTRACT

Age is a non-modifiable risk factor for the inflammation that underlies age-associated diseases; thus, anti-inflammaging drugs hold promise for increasing health span. Cytokine profiling and bioinformatic analyses showed that Th17 cytokine production differentiates CD4+ T cells from lean, normoglycemic older and younger subjects, and mimics a diabetes-associated Th17 profile. T cells from older compared to younger subjects also had defects in autophagy and mitochondrial bioenergetics that associate with redox imbalance. Metformin ameliorated the Th17 inflammaging profile by increasing autophagy and improving mitochondrial bioenergetics. By contrast, autophagy-targeting siRNA disrupted redox balance in T cells from young subjects and activated the Th17 profile by activating the Th17 master regulator, STAT3, which in turn bound IL-17A and F promoters. Mitophagy-targeting siRNA failed to activate the Th17 profile. We conclude that metformin improves autophagy and mitochondrial function largely in parallel to ameliorate a newly defined inflammaging profile that echoes inflammation in diabetes.


Subject(s)
Aging/drug effects , Autophagy/drug effects , Hypoglycemic Agents/pharmacology , Inflammation/metabolism , Metformin/pharmacology , Mitochondria/drug effects , Adult , Aging/metabolism , Humans , Middle Aged , Mitochondria/metabolism
9.
Sci Rep ; 9(1): 14413, 2019 10 08.
Article in English | MEDLINE | ID: mdl-31595002

ABSTRACT

Tristetraprolin (TTP), encoded by the Zfp36 gene, is a zinc-finger protein that regulates RNA stability primarily through association with 3' untranslated regions (3' UTRs) of target mRNAs. While TTP is expressed abundantly in the intestines, its function in intestinal epithelial cells (IECs) is unknown. Here we used a cre-lox system to remove Zfp36 in the mouse epithelium to uncover a role for TTP in IECs and to identify target genes in these cells. While TTP was largely dispensable for establishment and maintenance of the colonic epithelium, we found an expansion of the proliferative zone and an increase in goblet cell numbers in the colon crypts of Zfp36ΔIEC mice. Furthermore, through RNA-sequencing of transcripts isolated from the colons of Zfp36fl/fl and Zfp36ΔIEC mice, we found that expression of inducible nitric oxide synthase (iNos or Nos2) was elevated in TTP-knockout IECs. We demonstrate that TTP interacts with AU-rich elements in the Nos2 3' UTR and suppresses Nos2 expression. In comparison to control Zfp36fl/fl mice, Zfp36ΔIEC mice were less susceptible to dextran sodium sulfate (DSS)-induced acute colitis. Together, these results demonstrate that TTP in IECs targets Nos2 expression and aggravates acute colitis.


Subject(s)
Colitis/genetics , Colon/metabolism , Nitric Oxide Synthase Type II/genetics , Tristetraprolin/genetics , 3' Untranslated Regions/genetics , Animals , Colitis/chemically induced , Colitis/pathology , Colon/pathology , Dextran Sulfate/toxicity , Disease Models, Animal , Gene Expression Regulation, Enzymologic/genetics , Gene Knockout Techniques , Heterogeneous Nuclear Ribonucleoprotein D0 , High-Throughput Nucleotide Sequencing , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Intestines/enzymology , Intestines/pathology , Mice , RNA Stability/genetics , RNA-Binding Proteins/genetics
10.
Biol Sex Differ ; 10(1): 31, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31262355

ABSTRACT

Obesity is a global epidemic that greatly increases risk for developing cardiovascular disease and type II diabetes. Sex differences in the obese phenotype are well established in experimental animal models and clinical populations. While having higher adiposity and obesity prevalence, females are generally protected from obesity-related metabolic and cardiovascular complications. This protection is, at least in part, attributed to sex differences in metabolic effects of hormonal mediators such as the renin-angiotensin system (RAS). Previous literature has predominantly focused on the vasoconstrictor arm of the RAS and shown that, in contrast to male rodent models of obesity and diabetes, females are protected from metabolic and cardiovascular derangements produced by angiotensinogen, renin, and angiotensin II. A vasodilator arm of the RAS has more recently emerged which includes angiotensin-(1-7), angiotensin-converting enzyme 2 (ACE2), mas receptors, and alamandine. While accumulating evidence suggests that activation of components of this counter-regulatory axis produces positive effects on glucose homeostasis, lipid metabolism, and energy balance in male animal models, female comparison studies and clinical data related to metabolic outcomes are lacking. This review will summarize current knowledge of sex differences in metabolic effects of the RAS, focusing on interactions with gonadal hormones and potential clinical implications.


Subject(s)
Renin-Angiotensin System/physiology , Sex Characteristics , Angiotensins/metabolism , Animals , Female , Gonadal Steroid Hormones/metabolism , Humans , Male , Receptors, Angiotensin/metabolism , Renin/metabolism
11.
Nutrients ; 11(3)2019 Mar 13.
Article in English | MEDLINE | ID: mdl-30871233

ABSTRACT

Macronutrient composition of the diet influences the development of obesity and insulin resistance. The aim of this study was to assess the role of dietary fat quantity and fatty acid composition in body composition, insulin resistance, and insulin secretion. An open parallel randomized trial design was used. Overweight participants (n = 75) were randomized to follow a low-fat vegan (n = 38) or control diet (n = 37) for 16 weeks. Dual X-ray absorptiometry was used to measure body composition. Insulin resistance was assessed with the Homeostasis Model Assessment (HOMA-IR) index. Insulin secretion was assessed after stimulation with a liquid breakfast (Boost Plus, Nestle, Vevey, Switzerland). Self-reported 3-day diet records were used to assess dietary intake. A linear regression model was used to test the relationship between fat intake and body composition, insulin resistance, and insulin secretion. Changes in fat intake expressed as percent of total energy consumed correlated positively with changes in fat mass (r = 0.52; p < 0.001; and 0.347; p = 0.006, respectively), even after adjustment for changes in body-mass index (BMI) and energy intake (0.33; p = 0.01). Decreased intakes of C18:0 (r = 0.37, p = 0.004) and CLA-trans-10-cis12 (r = 0.40, p = 0.002), but increased intake of C18:2 (r = -0.40, p = 0.002) and C18:3 (p = -0.36, p = 0.006), were associated with a decrease in HOMA-IR, independent on changes in BMI and energy intake. The main fatty acids associated with changes in fasting insulin secretion were C12:0 (r = -0.31, p = 0.03), and TRANS 16:1 (r = -0.33, p = 0.02), both independent on changes in BMI and energy intake. Our findings demonstrate that, in the context of a low-fat vegan diet, decreased intake of saturated and trans fats and increased relative content of polyunsaturated fatty acids, particularly linoleic and α-linolenic acids, are associated with decreased fat mass and insulin resistance, and enhanced insulin secretion.


Subject(s)
Body Composition , Diet, Vegan , Dietary Fats/administration & dosage , Insulin Resistance , Adult , Aged , Exercise , Fatty Acids, Unsaturated , Female , Humans , Male , Middle Aged , Trans Fatty Acids
12.
Nutr Diabetes ; 8(1): 58, 2018 11 02.
Article in English | MEDLINE | ID: mdl-30405108

ABSTRACT

BACKGROUND AND OBJECTIVES: A plant-based diet is an effective strategy in the treatment of obesity. In this 16-week randomized clinical trial, we tested the effect of a plant-based diet on body composition and insulin resistance. As a part of this trial, we investigated the role of plant protein on these outcomes. SUBJECTS AND METHODS: Overweight participants (n = 75) were randomized to follow a plant-based (n = 38) or a control diet (n = 37). Dual X-ray Absorptiometry assessed body composition, Homeostasis Model Assessment (HOMA-IR) assessed insulin resistance, and a linear regression model was used to test the relationship between protein intake, body composition, and insulin resistance. RESULTS: The plant-based vegan diet proved to be superior to the control diet in improving body weight, fat mass, and insulin resistance markers. Only the vegan group showed significant reductions in body weight (treatment effect -6.5 [95% CI -8.9 to -4.1] kg; Gxt, p < 0.001), fat mass (treatment effect -4.3 [95% CI -5.4 to -3.2] kg; Gxt, p < 0.001), and HOMA-IR (treatment effect -1.0 [95% CI -1.2 to -0.8]; Gxt, p = 0.004). The decrease in fat mass was associated with an increased intake of plant protein and decreased intake of animal protein (r = -0.30, p = 0.011; and r = +0.39, p = 0.001, respectively). In particular, decreased % leucine intake was associated with a decrease in fat mass (r = +0.40; p < 0.001), in both unadjusted and adjusted models for changes in BMI and energy intake. In addition, decreased % histidine intake was associated with a decrease in insulin resistance (r = +0.38; p = 0.003), also independent of changes in BMI and energy intake. CONCLUSIONS: These findings provide evidence that plant protein, as a part of a plant-based diet, and the resulting limitation of leucine and histidine intake are associated with improvements in body composition and reductions in both body weight and insulin resistance.


Subject(s)
Body Weight/physiology , Diet, Vegan , Insulin Resistance/physiology , Overweight/diet therapy , Weight Loss/physiology , Adult , Aged , Body Composition/physiology , Female , Humans , Male , Middle Aged , Overweight/metabolism , Treatment Outcome
13.
J Electromyogr Kinesiol ; 41: 27-33, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29723799

ABSTRACT

Our ability to control movements is influenced by the developmental status of the neuromuscular system. Consequently, movement control improves from childhood to early adulthood but gradually declines thereafter. However, no study has compared movement accuracy between children and older adults. The purpose of this study was to compare endpoint accuracy during a fast goal-directed movement task in children and older adults. Ten pre-adolescent children (9.7 ±â€¯0.67 yrs) and 19 older adults (71.95 ±â€¯6.99 yrs) attempted to accurately match a peak displacement of the foot to a target (9° in 180 ms) with a dorsiflexion movement. We recorded electromyographic activity from the tibialis anterior (agonist) and soleus (antagonist) muscles. We quantified position error (i.e. spatial accuracy) as well as the coordination, magnitude, and variability of the antagonistic muscles. Children exhibited greater position error than older adults (36.4 ±â€¯13.4% vs. 27.0 ±â€¯9.8%). This age-related difference in spatial accuracy, was related to a more variable activation of the agonist muscle (R2: 0.358; P < 0.01). These results suggest that an immature neuromuscular system, compared to an aged one, affects the generation and refinement of the motor plan which increases the variability in the neural drive to the muscle and reduces spatial accuracy in children.


Subject(s)
Aging/physiology , Biological Variation, Individual , Foot/physiology , Movement , Muscle, Skeletal/physiology , Aged , Child , Female , Humans , Male , Muscle, Skeletal/growth & development , Postural Balance
SELECTION OF CITATIONS
SEARCH DETAIL
...