Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2025 Jul 1.
Published in final edited form as: Brain Behav Immun. 2024 Apr 16;119:681–692. doi: 10.1016/j.bbi.2024.04.016

Mediterranean Diet Protects Against a Neuroinflammatory Cortical Transcriptome: Associations with Brain Volumetrics, Peripheral Inflammation, Social Isolation, and Anxiety in Nonhuman Primates (Macaca fascicularis)

Brett M Frye 1,2,3,#, Jacob D Negrey 1,4,#, Corbin SC Johnson 5, Jeongchul Kim 6, Richard A Barcus 6, Samuel N Lockhart 3,7, Christopher T Whitlow 3,6, Kenneth L Chiou 8,9, Noah Snyder-Mackler 8,9,10, Thomas J Montine 11, Suzanne Craft 3,7, Carol A Shively 1,3,*,#, Thomas C Register 1,3,*,#
PMCID: PMC12051215  NIHMSID: NIHMS2077712  PMID: 38636565

Abstract

Mediterranean diets may be neuroprotective and prevent cognitive decline relative to Western diets, however the underlying biology is poorly understood. We assessed the effects of Western versus Mediterranean-like diets on RNAseq-generated transcriptional profiles in lateral temporal cortex and their relationships with longitudinal changes in neuroanatomy, circulating monocyte gene expression, and observations of social isolation and anxiety in 38 socially-housed, middle-aged female cynomolgus macaques (Macaca fascicularis). Diet resulted in differential expression of seven transcripts (FDR<0.05). Cyclin dependent kinase 14 (CDK14), a proinflammatory regulator, was lower in the Mediterranean group. The remaining six transcripts [i.e., “lunatic fringe” (LFNG), mannose receptor C type 2 (MRC2), solute carrier family 3 member 2 (SLCA32), butyrophilin subfamily 2 member A1 (BTN2A1), katanin regulatory subunit B1 (KATNB1), and transmembrane protein 268 (TMEM268)] were higher in cortex of the Mediterranean group and generally associated with anti-inflammatory/neuroprotective pathways. KATNB1 encodes a subcomponent of katanin, important in maintaining microtubule homeostasis. BTN2A1 is involved in immunomodulation of γδ T-cells which have anti-neuroinflammatory and neuroprotective effects. CDK14, LFNG, MRC2, and SLCA32 are associated with inflammatory pathways. The latter four differentially expressed cortex transcripts were associated with peripheral monocyte transcript levels, neuroanatomical changes determined by MRI, and with social isolation and anxiety. These results provide important insights into the potential mechanistic processes linking diet, peripheral and central inflammation, and behavior. Collectively, our results provide evidence that, relative to Western diets, Mediterranean diets confer protection against peripheral and central inflammation which is reflected in preserved brain structure and socioemotional behavior. Ultimately, such protective effects may confer resilience to the development of neuropathology and associated disease.

Keywords: Brain, temporal cortex, RNAseq, inflammation, Alzheimer’s disease, Mediterranean diet, nonhuman primate, socioemotional behavior

1. Introduction

An increasing literature suggests that diet composition may affect mental health, neurobiological aging, the rate of cognitive decline with aging, and the risk of neurodegenerative disorders such as Alzheimer’s disease (AD)13. The Mediterranean-style diet (hereafter, “Mediterranean diet”), which is largely plant-based and rich in healthy oils, complex carbohydrates, and dietary fiber, is associated with increased longevity4 and quality of life5, lower rates of psychiatric3, 6 and neurodegenerative disease7, and slower cognitive aging8, 9 as well as reduced risk of other chronic health conditions1013. Conversely, consumption of a so-called “Western” diet, which is high in saturated fats, salt, and simple sugars14, 15, has been shown to increase systemic inflammation16 and have detrimental neurobiological effects. Short term consumption of very high fat diets (60–75% fat) impairs attention and memory in clinical and preclinical studies17. Observational studies show associations of Western diet consumption with increased risk of depression, anxiety6, 18 and faster cognitive decline with aging19, 20. Furthermore, Western diet consumption is associated with brain-related changes, such as blood-brain barrier disruption and neuroinflammation, which could contribute to neurocognitive decline and neurodegenerative disease21, 22. Conflicting findings have also been reported. Of the ten most comprehensive trials of cognitive effects of the Mediterranean diet reviewed by Morris et al.23, only four reported clear protective effects against cognitive decline or dementia2427. A recent evaluation of the association between adherence to the MIND (Mediterranean-Dietary Approaches to Stop Hypertension Diet Intervention for Neurodegenerative Delay) diet and cognitive health in the UK Biobank reported no association overall with better cognitive test scores and lower dementia risk only in women28. These variable results may be related to differences in adherence, or how Mediterranean diet was defined or consumption was measured23. The majority of these data derive from observational studies that rely on self-reported diet consumption and may be confounded by other factors which correlate with diet consumption (e.g., activity levels29) and are known to impact neurocognitive outcomes. Consequently, experimental studies testing the neurobiological effects of diet composition are essential to determine whether and how diet patterns directly impact the brain.

Given their genetic, physiological, neuroanatomical, and social similarities to humans, nonhuman primates are useful models for studying the effects of diet across multiple physiologic systems, thereby providing important opportunities for uncovering the molecular mechanisms of disease30. Macaques (Macaca spp.) are especially relevant models of human health31 and aging32, and they exhibit neurobiological shifts and cognitive decline reminiscent of chronic neurodegenerative diseases like AD33, 34. Preclinical studies of nonhuman primates enable us to understand the effects of long-term exposure to dietary patterns, which more closely recapitulates the human condition, but are difficult to conduct in clinical studies due to high cost and variable adherence to prescribed diets. Additionally, given our knowledge of the potential deleterious effects of Western-style diets, randomized clinical trials that contrast the Western dietary pattern with alternatives face ethical constraints.

In this randomized preclinical trial, Mediterranean diet consumption by female macaques protected against overeating, obesity, hepatosteatosis, insulin resistance35, and increased hypothalamic-pituitary-adrenal and sympathetic nervous system activity36 compared to Western diet consumption. Longitudinal structural magnetic resonance imaging (MRI) measures of neuroanatomy demonstrated that brain volumes changed in response to Western but not Mediterranean diets37, with Western diet fed monkeys having increased total brain (TBV) and gray matter (GM) volumes and decreased volumes of cerebrospinal fluid (CSF) and white matter (WM), and increased cortical thickness in multiple regions in the temporal and parietal lobes relevant to AD37. Given these observations, we hypothesized that diet-related increases in total brain volume and GM thickness as well as decreases in WM volume were likely due to Western diet-induced neuroinflammation37. This hypothesis was spurred by clinical data from the Alzheimer’s Disease Neuroimaging Initiative (ADNI) trial indicating biphasic changes in brain volumes over time, with increased gray matter volumes and cortical thickening early in the Alzheimer’s disease process (asymptomatic amyloidosis) followed by reductions in these parameters along with AD pathology and neurodegeneration38. The investigators hypothesized that these increased volumes may reflect neuroinflammation3840, which, with time, results in cortical thinning, a predictor of increased AD risk41. Consistent with this hypothesis, in our NHP study, circulating monocytes in the Western group were polarized towards a pro-inflammatory phenotype, relative to those from the Mediterranean group, characterized by increased transcripts for IL6, IL1α, NFKB1, and NFKB2, key regulators of inflammatory pathways42. Likewise, a beneficial circulating inflammatory profile is associated with Mediterranean diet consumption in human studies43. Experimental diets induced rapid and persistent changes in a suite of NHP behaviors, as monkeys fed the Western diet spent more time alone and displayed more anxiety behavior, whereas those fed the Mediterranean diet spent more time resting, attentive, and in body contact with groupmates44. Finally, the indices of peripheral inflammation in this study were associated with social isolation and anxiety, behaviors thought to be associated with neuroinflammation44.

In this study we sought to identify molecular mechanisms underlying diet-induced neuroanatomical changes by testing if diet composition altered gene expression in cortical gray matter from the superior temporal gyrus in the lateral temporal cortex of socially housed, female cynomolgus macaques (Macaca fascicularis). We focused on females because there is a paucity of data from female animal models, and because many brain diseases are more prevalent in females than males. For example, two-thirds of the Alzheimer’s disease burden is in women45. We focused on middle-age because that is the earliest period in which the neuropathological trajectory that culminates in cognitive decline and neurodegeneration has been observed; thus, identification of targets for prevention is needed at this life stage. We focused on the temporal cortex because this region is among the first to exhibit aging-related neuropathology associated with neurodegenerative diseases like AD4648. We first identified differentially expressed genes (DEGs) between the two diets and then examined relationships between gene expression and changes in brain volumes and cortical thicknesses determined by MRI between baseline and after 31 months of experimental diet. Based on our prior findings3537, 42, 49, we hypothesized that monkeys fed a Western diet would exhibit transcriptomic signals of inflammation that would be associated with peripheral inflammation, changes in brain volumes and behavior, potentially illuminating molecular targets for intervention. We also examined associations of temporal cortex transcript levels with MRI derived changes in brain anatomy, peripheral monocyte transcripts, and levels of anxiety and social isolation.

2. Methods

2.1. Ethics Statement

All experimental procedures complied with National Institutes of Health Guide for the Care and Use of Laboratory Animals (NIH Publications No. 8023, revised 1978), state and federal laws, and were approved by the Animal Care and Use Committee of Wake Forest School of Medicine.

2.2. Subjects

Details of the experiment and subjects have been previously described3537, 50, 51. Briefly, adult female cynomolgus (a.k.a. long-tailed) macaques (Macaca fascicularis) aged 11 to 13 years (as estimated from dentition) were obtained from SNBL USA (Alice, Texas) and quarantined for one month in single-cages, during which time subjects had visual, auditory and olfactory contact with each other. Following quarantine, monkeys were transferred to social groups of n=4–5 each and housed in indoor enclosures (3m × 3m × 3m) with natural light exposure, 12h/12h light-dark cycles, and water available ad libitum. All individuals were fed monkey chow (LabDiet) during a 7-month baseline period prior to the experimental dietary intervention.

2.3. Experimental Procedure

After the 7-month baseline phase, the monkeys were assigned to one of two diet groups: Western or Mediterranean. The experimental groups were balanced for several relevant subject characteristics, including body composition (body weight and body mass index), circulating basal cortisol, and fasting triglyceride concentrations, using stratified randomization35. All members of an individual pen then received the same experimental diet. Experimental diets were developed and prepared at the Wake Forest School of Medicine Comparative Medicine Diet Lab and fed by animal care staff unassociated with the research project. Samples of all diet batches were kept. Batch analyses demonstrated consistency over time. Diets were balanced for macronutrients and cholesterol content, but differed in sources of protein, fat, and carbohydrates. Diet compositions are detailed in Supplementary Table 1 and the complete ingredient list may be found in35. The Western diet, which was designed to resemble that consumed by middle-aged women (40–49 years) living in the USA14, featured primarily animal-derived fats and proteins, and a relatively high content of simple sugars and salt15. In contrast, the Mediterranean diet featured plant-based fats and proteins, resulting in a diet high in monounsaturated fats, complex carbohydrates, and fiber, and low in sodium and refined sugars15, 52. Importantly for inflammation, the Western diet had relatively higher amounts of omega-6 fatty acids, whereas the Mediterranean diet was higher in omega-3 content. English walnut powder and extra virgin olive oil, as given to participants in the PREDIMED study53, were among the chief dietary components of the Mediterranean diet. Thirty-eight monkeys (21 Western, 17 Mediterranean) completed the experiment and were subsequently assessed in this study.

2.4. Social Isolation and Anxiety Behavior Observations

The behavioral characterization of these animals has been described in-depth44. Briefly, behavioral data were collected live weekly during two 10-min focal observations54, balanced for the time of day for 24 consecutive months beginning the third month of the treatment phase (approximately 200 behavior samples/monkey, mean = 31.0 h/monkey, and 1178 observation hours total). Inter-rater reliability was maintained at ≥93%, and no other research activities were ongoing during behavior observations. Observers were aware of the diet group of individual pens. Since we previously determined that the Western diet increased social isolation and anxiety, we focused on these behaviors here44. Social isolation was operationally defined as the percent of time a monkey spent alone, i.e., out of arm’s reach of another monkey. Anxiety was operationally defined as the rate (frequency per hour) of self-directed behaviors i.e. self-grooming and scratching5559.

2.5. Magnetic Resonance Imaging

Neuroimaging methods used in this study have been previously described37. Briefly, T1-weighted structural MRI scanning was performed at baseline and again at 31 months at the MRI facility at Wake Forest University School of Medicine using a 3T Siemens Skyra scanner (Siemens, Erlangen, Germany) with a 32-channel head coil (Litzcage, Doty Scientific, SC). Brain images were segmented into three main tissue types: gray matter, white matter, and cerebrospinal fluid. The tissue segmentations were created using segmentation priors created for this Study Specific Template, and were non-linearly warped to the native brains and used as prior inputs in via the AntsAtroposN4 algorithm60. We then used the parcellation map to determine volumes for the following regions of interest (ROIs): total gray matter (tGM), cortical gray matter (cGM), white matter (WM), cerebrospinal fluid (CSF), and total brain volume (TBV (tGM + WM)). The cortical thicknesses were determined using T1w MR images and the cortical GM tissue segmentation in the subject’s native space. We also generated volume- and thickness-based AD temporoparietal meta-ROIs based on the AD clinical literature39 using a previously described method37. Each meta-ROI represented the following ROIs (as illustrated in Figure 1) from the Neuromaps Macaque Atlas61: the angular gyrus, the inferior, middle, and superior temporal gyrus, entorhinal cortex, fusiform cortex, supramarginal gyrus, precuneus, and parahippocampus. A detailed description of imaging methods is available in the Supplementary Information.

Figure 1.

Figure 1.

Parcellation map of macaque brains illustrating regions included in Alzheimer’s disease meta-regions of interest. Individual regions making up this composite ROI include: the angular gyrus, the inferior, middle, and superior temporal gyrus, entorhinal cortex, fusiform cortex, supramarginal gyrus, precuneus, and parahippocampus. Images are provided in axial (panel A), coronal (panels B and C), and sagittal (panels D and E) cross-sections. The white box (panel B) indicates a box approximating the region from which gray matter was collected for transcriptomic profiling.

2.6. Peripheral Monocyte Transcriptomics

Details regarding isolation and purification of CD14+ monocytes, RNAseq analyses, and previous findings have been published42. Briefly, CD14+ monocytes were purified from PBMCs using positive selection with Miltenyi anti-CD14 magnetic beads. RNA was extracted from monocytes using the AllPrep DNA/RNA Mini Kit (Qiagen, Inc, Hilden, Germany), and quantified using a NanoDrop spectrophotometer and Agilent 2100 Bioanalyzer with RNA 6000 Nano chips (Agilent Technology, Inc, Santa Clara, CA). RNA libraries were prepared for sequencing by the Cancer Genomics Shared Resource (Wake Forest University School of Medicine, Winston-Salem, NC) using the TruSeq-stranded total RNA kit (Illumina), which includes a ribosomal depletion step. RNA-seq libraries were sequenced using single-end 76 bp reads on an Illumina NextSeq 500 to an average read depth of 34.5 million reads per sample (range 25.9–41.6 million reads). Reads were mapped to the M. fascicularis reference genome (Macaca_fascicularis_5.0, v 93, Ensembl using HiSat2 and then converted to a sample-by-gene read count matrix using featureCounts (Liao et al., 2014) (median = 38.0%; range 24.5–50.4% of reads mapped to exons). Sample processing order was randomized and where possible all samples were manipulated simultaneously so as to avoid introducing batch effects. Genes with low expression (median reads per kilobase per million reads mapped < 1) were removed, which resulted in 12,240 genes for downstream analyses. Read counts were normalized using the voom function of the R package limma (Ritchie et al., 2015). The final sample size was 35 monkeys (n=20 fed the Western diet, n=15 Mediterranean diet). To control for batch effects related to RNA quality and monocyte purity, we calculated the residual gene expression from a model of normalized gene expression as a function of CD14 expression, CD3 expression, RNA integrity, and RNA concentration. These residual gene expression values were used for all subsequent analyses.

2.7. Tissue Collection and Processing

Following the 31-month exposure to the experimental diets, monkeys were anesthetized with pentobarbital (30–50 mg/kg) to obtain a surgical plane of anesthesia, followed by exsanguination and perfusion with ice cold saline, according to the guidelines of the American Veterinary Medical Association’s Panel on Euthanasia. To provide high quality samples for transcriptomic analyses, we implemented short post-mortem intervals (i.e., from euthanasia to brain preservation). The brain was hemisected and blocked into coronal slabs 4 millimeters in thickness. We dissected 25 mg of gray matter from the superior temporal gyrus, at the level of the caudate, primarily lateral to the white matter shown in Figure 1B. Tissue collected for transcriptomic analyses was covered with finely crushed dry ice and frozen for at least 20 minutes, then placed in a vacuum-sealed sample bag and stored in a −80°C freezer. Detailed examination of brain showed generally normal brain architecture and absence of amyloid plaques or tauopathies, consistent with the age of the monkeys (data not shown). The lateral temporal cortical gray matter region sampled for this study is indicated in Figure 1B.

2.8. Temporal cerebral cortex transcriptomics

RNA isolation and sequencing were performed by the Wake Forest School of Medicine Cancer Genomics Shared Resource which were blinded to treatment. RNA was isolated from the lateral temporal cortical gray matter samples using the Zymo Quick-DNA/RNA Miniprep Plus Kit (Zymo, Irvine, CA, USA) and purified using the RNA Clean and Concentrator-5 kit (Zymo). All RNA integrity number (RIN) scores were > 7.7, indicating high integrity. Bulk RNA-seq was then performed using an Illumina NovaSeq 6000 (single end, 100bp). We subsequently aligned sequenced reads to the cynomolgus macaque genome (Macaca_fascicularis_6.0) using the STAR alignment software62.

Differential gene expression (DGE) analysis was performed in R version 4.1.163 using the pqlseq function in package “PQLseq”64, which controls for genetic relatedness between subjects while performing DGE analysis within the framework of a generalized linear mixed model – i.e., linear models for mircroarray data analysis (“Limma”)65, 66. We first filtered genes exhibiting low reads per kilobase of transcript per million mapped reads (RPKM), leaving 10,684 genes for DGE analysis. To determine relatedness, we used bcftools mpileup67 to infer SNP genotypes from RNA-seq reads, as previously described42. Based on this approach we found 1 full sib pair and 9 other pair combinations which were half-sibs. We modeled the expression of each gene as a function of diet (Western or Mediterranean) and RIN (to control for any effects of RNA quality on expression). Given that social status68, 69 and estradiol70 exert neurological effects in NHPs, we also included subject social status (relative dominance rank) and serum estradiol levels at time of necropsy as covariates. Genes were considered differentially expressed if they passed a Benjamini-Hochberg false discovery rate (FDR) threshold of 0.05.71

2.9. Identifying Biological Pathways and Causal Networks

Biological processes and regulatory networks exhibiting diet-specific activation patterns were investigated using Ingenuity Pathway Analysis (IPA; Qiagen, Hilden, Germany). Given the limited number of DEGs identified at a FDR cutoff of 0.05, for exploratory analyses we broadened our criteria to include genes with an FDR<0.2 (n=312), following prior work42. First, we extracted canonical pathways associated with the gene set, retaining all pathways for which p<0.05. We then analyzed causal networks to identify “master” regulators associated with the gene set; we retained only endogenous regulators, discarding all exogenous substances (e.g., drugs, chemical reagents). To determine whether causal networks and associated regulators were significantly activated or inhibited, we employed activation Z-scores, which indicate the confidence that a regulatory network is activated or inhibited based on transcript levels within a gene set. We used thresholds of >2 (activated) and <−2 (inhibited) and discarded regulators with non-significant Z-scores (i.e., Z-score between 2 and −2).

2.10. Correlations between Temporal Cortex Gene Expression, Brain Volumes, Peripheral Monocyte Transcripts, and Behaviors

We assessed relationships between transcript levels from genes that were differentially expressed by diet and circulating monocyte gene expression, changes in neuroanatomy, and behavior using the corr.test function in R. For more information, see the Supplementary Information.

3. Results

3.1. Temporal Cortex Gene Expression Varied by Diet

Seven genes were significantly differentially expressed between the two diets (FDR<0.05; Figure 2). Cyclin dependent kinase 14 (CDK14), was downregulated in the Mediterranean cohort relative to the Western cohort (β=−0.087, SE=0.021, p=2.85E-05, FDR=0.044). The remaining six genes were upregulated in the Mediterranean cohort relative to the Western cohort: butyrophilin subfamily 2 member A1 (BTN2A1; β=0.172, SE=0.041, p=2.45E-05, FDR= 0.044), katanin regulatory subunit B1 (KATNB1; β=0.118, SE=0.028, p=2.68E-05, FDR=0.044), beta-1,3-N-acetylglucosaminyltransferase lunatic fringe, or “Lunatic Fringe” (LFNG; β=0.253, SE=0.053, p=1.93E-06, FDR=0.021), mannose receptor C type 2 (MRC2; β=0.324, SE=0.073, p=9.58E-06, FDR=0.032), solute carrier family 3 member 2 (SLC3A2; β=0.119, SE=0.027, p=1.21E-05, FDR=0.032), and transmembrane protein 268 (TMEM268; β=0.157, SE=0.035, p=7.51E-06, FDR=0.032). Of these seven DEGs, Ingenuity Pathway Analysis identified four as having known causal relationships or associations with cytokines: CDK14, LFNG, MRC2, and SLC3A2 (Figure 3).

Figure 2.

Figure 2.

Transcript levels (TMM normalized) for seven genes differentially expressed by diet: Cyclin dependent kinase 14 (CDK14), butyrophilin subfamily 2 member A1 (BTN2A1), katanin regulatory subunit B1 (KATNB1), “Lunatic Fringe” (LFNG), mannose receptor C type 2 (MRC2), solute carrier family 3 member 2 (SLC3A2), and transmembrane protein 268 (TMEM268).

Figure 3.

Figure 3.

Associations between differentially expressed genes (bold, italicized font) and cytokines determined using Ingenuity Pathway Analysis. Solid and dashed lines indicate direct and indirect interactions, respectively. Arrows indicate directional relationships.

To determine whether diet effects were driven by differences in cell type proportions, we estimated cell type proportions represented in each transcriptional profile (Supplementary Information). Cell type biases for each sample are available in Supplementary Table 2. We did not find diet-based differences in the estimated proportion of any of six brain cell types (Supplementary Table 3). Furthermore, Wilcoxon rank sum tests of the 7 DEGs indicated that diet-based differences persisted even after correcting transcript levels for cell type proportions (Supplementary Table 4).

At a relaxed FDR threshold (0.2), we found 312 diet-associated DEGs (Supplementary Table 5). Pathways associated with these genes included: (1) DNA Double-Strand Break Repair by Non-Homologous End Joining, (2) Phosphatase and Tensin Homolog (PTEN) Signaling, (3) Ciliary Neurotrophic Factor (CNTF) Signaling, (4) Polyamine Regulation in Colon Cancer, and (5) Tumor Microenvironment Pathway. (All associated pathways are in Supplementary Table 6.) The top five master regulators identified by IPA included: (1) Sp3 Transcription Factor, (2) Chymase 1 (CMA1), (3) LXR ligand-LXR-Retinoic acid-RXR, (4) Transcriptional Repressor GATA Binding 1 (TRPS1), and (5) transforming growth factor beta 1 (TGFB1).

SP3 (Z-score=2.26), CMA1 (Z-score=3.46), LXR ligand-LXR-Retinoic acid-RXR (Z-score=2.24), and TGFB1 (Z-score=2.83) were more highly expressed in the Mediterranean cohort, whereas TRPS1 was lower (Z-score=−2.95). A complete list of causal networks with associated master regulators is available in Supplementary Table 7. IPA identified four of the seven DEGs to have known causal relationships or associations with cytokines (Figure 3). To address our central hypothesis that monkeys fed a Western diet would exhibit transcriptomic signals of inflammation that would be associated with peripheral inflammation, changes in brain volumes, and behavior, we focused on these four DEGS (i.e., CDK14, LFNG, MRC2, and SLC3A2) in the following correlational analyses with brain structure, circulating monocyte gene expression, and behavior.

3.2. Temporal Cortex Gene Expression was Associated with Circulating Monocyte Gene Expression

To explore potential relationships between peripheral and central gene expression, we assessed relationships of differentially expressed transcripts in temporal cortex at necropsy with circulating CD14+ monocyte transcript levels midway through the project. Patterns of gene expression observed in monocytes predicted patterns observed in the brain, with consistent anti-inflammatory effects of the Mediterranean Diet relative to the Western Diet (Figure 4). Monocyte transcripts associated with inflammation, including proinflammatory components of the family of genes represented in the constitutive transcriptional response to adversity (CTRA72), NFKB1, NFKB2, and IL6, were all positively associated with transcript levels of the proinflammatory gene CDK14 in the temporal cortex. In general, these genes were all negatively correlated with anti-inflammatory transcripts LFNG, SLC3A2, and MRC2 in the temporal cortex (11 of 12 transcripts p<0.10, 8 of 12 transcripts p<0.05, df=33).

Figure 4.

Figure 4.

Relationships of differentially expressed transcripts in temporal cortex at necropsy with circulating CD14+ monocyte transcript levels midway through the project.

3.3. Temporal Cortex Gene Expression was Associated with Changes in Brain Structure

To determine whether expression levels for the four DEGs known to have causal relationships or associations with inflammation reflected variation in brain structure, we ran a series of Pearson’s correlations of changes in global brain volumes and composite temporoparietal regions of interest (“meta-ROI”) with gene expression levels (Figure 5). CDK14 transcript levels were significantly positively correlated with percent change in total brain volume, total gray matter volume, cortical gray matter volume, thickness of the temporoparietal meta-ROI in both hemispheres, and volume of the temporoparietal meta-ROI in the right hemisphere, and negatively correlated with percent change in global white matter and CSF volumes (all p<0.05, df=36, Figure 5). In contrast, LFNG, MRC2, and SLC3A2 transcript levels were significantly negatively correlated with percent change in total brain volume, total gray matter volume, and thickness and volume of the temporoparietal meta-ROI in the right hemisphere, and positively correlated with percent change in global CSF volume (p<0.05, df=36, Figure 5). SLC3A2 transcript levels were also positively correlated with percent change in global white matter volume, and MRC2 and SLC3A2 were negatively correlated with thickness of the temporoparietal meta-ROI in the left hemisphere (Figure 5). We also analyzed each diet cohort independently to determine whether patterns were attributable to diet composition or upheld within each diet. When doing so, we found no significant correlations within the Mediterranean cohort (Supplementary Table 8) but did within the Western cohort; MRC2, in particular, was significantly correlated with seven of the nine MRI measures in the Western subset (Supplementary Table 9).

Figure 5.

Figure 5.

Scatterplots of transcript levels (TMM normalized; y axis) for genes with known inflammatory associations (CDK14, LFNG, SLC3A2, and MRC2) versus percent changes in brain tissue / fluid volumes (x axis) from baseline to end of experimental phase.

3.4. Temporal Cortex Gene Expression Was Associated with Behavior

Temporal cortex transcript levels of CDK14 were positively correlated with percent of time spent alone (R2=0.234; df=36; p=0.002) and with rates of anxious behavior (R2=0.237; df=36; p=0.002). In contrast, levels of the anti-inflammatory transcript for SLCA32 were negatively correlated with the percent of time spent alone, but this relationship did not reach statistical significance (R2=0.102; df=36; p=0.051) (Figure 6).

Figure 6.

Figure 6.

Lateral temporal neuroinflammatory transcriptome associations with anxiety and time spent alone. Temporal cortex CDK14 transcripts were positively associated with the rate of anxiety (frequency/hour) and the percentage of time spent alone, while cortex SLC3A2 was inversely associated time alone.

4. Discussion

We observed differences in transcript levels in the temporal cortex of macaques fed Western versus Mediterranean diets. Four of these DEGs, known to have causal relationships or associations with cytokines, showed consistent, significant correlations with circulating monocyte gene expression, changes in brain volume and temporoparietal meta-ROIs, and social isolation and anxious behavior. These data support our previous hypothesis that diet-related changes in neuroanatomy – i.e., increases in total brain volume and cortical thickness as well as decreases in white matter volume37—may reflect Western diet-induced inflammation37, whereas relative stability in such neuroanatomical profiles in the Mediterranean diet consumers may reflect resilience and heathy aging during middle-age. Furthermore, these data provide links between cortical inflammatory profiles, peripheral monocyte programming, and behavioral phenotypes as modeled in Figure 7. Understanding patterns during this stage of life may shed light on the etiology of healthy versus pathological aging trajectories in the brain.

Figure 7.

Figure 7.

Differences between Mediterranean (blue) and Western (red) diet groups in peripheral and central inflammation, neuroanatomical changes, and behavioral profiles. Consumption of the Western diet was associated with elevations of monocyte transcripts for NF-κB, a key central regulator of inflammation, as well as IL-6 and other proinflammatory genes involved in the CTRA130. Western diet may disrupt the blood-brain barrier, increasing infiltration of Western-diet induced cytokines from the periphery21. Peripheral inflammation may contribute to inflammation in the central nervous system (CNS) indicated by elevated transcription of CDK14 and suppression of anti-inflammatory transcripts (LFNG, SLC3A2, and MRC2). Western diet driven neuroinflammation may promote increases gray matter volumes and losses in white matter and cerebrospinal fluid. These neuroanatomical and inflammatory perturbations may promote behavioral shifts, including increased social isolation and anxiety. Central nervous system perturbations may also impact efferent pathways related to the hypothalamic-pituitary-adrenal and autonomic nervous system, which, in turn, promotes proinflammatory cascades. Mediterranean diets may exert neuroprotective effects via opposing pro-inflammatory pathways in the periphery and the brain. Conversely, anti-inflammatory effects of the Mediterranean diet may dampen systemic inflammation, thereby preserving brain homeostasis and promoting resilience to pathology.

Our transcriptomic results support the hypothesis that the Western diet promoted and the Mediterranean diet protected against neuroinflammation, as several DEGs identified in this study are explicitly associated with inflammation. The single gene found to be increased in the Western cohort, cyclin dependent kinase 14 (CDK14), regulates the Wnt signaling pathway73, an important inflammatory modulator74. In clinical applications, CDK14 has been targeted for its role in tumorigenesis75. CDK14 also contributes to cell cycle regulation76, and its inhibition effectively limits cell proliferation77. CDK14 has also been implicated in the progression of neurodegenerative disorders such as AD and Parkinson’s disease (PD)78, 79. In a multiomics study to identify potential targets for AD treatment, assessment of transcriptional profiles from 5 different studies of brains from AD patients and controls identified a set of common DEGs; CDK14, among others, was identified as being co-expressed with this set of genes80. Ex vivo Inhibition of CDK14 in human dopaminergic neurons of PD patients caused a decrease in the concentration of total alpha-synuclein, a presynaptic protein which is strongly linked to pathogenesis in PD81. Accumulation of alpha-synuclein in PD has been hypothesized to promote the activation of pro-inflammatory cascades, which may ultimately trigger exaggerated inflammatory responses that cause neuronal injury and neurodegeneration82.

Neuropathological or neuroinflammatory actions by CDK14 are further implicated in our dataset by its correlations with brain volumes: this gene was positively correlated with total brain volume, total gray matter volume, and temporoparietal meta-ROI thicknesses, and negatively correlated with white matter and CSF volumes, suggesting Western diet-induced inflammation in gray matter. Such inflammation in middle-age may predispose individuals for developing neurodegenerative disorders later in life8385, including reduced hippocampal and AD-signature region brain volumes86. Previously we showed that the indices of peripheral inflammation in this study were associated with social isolation and anxiety, behaviors thought to be associated with neuroinflammation44. Here we show that brain CDK14 transcript levels were positively associated with anxiety behavior and time spent alone, consistent with previous associations of Western diet consumption with increased risk of anxiety6.

The inflammation hypothesis is further supported by other transcripts which were significantly lower in the Western cohort. “Lunatic fringe” (LFNG) regulates Notch signaling87, which is in turn an indispensable regulator of neural progenitor cell renewal88, 89. Experimental inflammation stimulated by tumor necrosis factor alpha causes downregulation of LFNG in rodent brain endothelial cells90, which may represent an important target by which inflammatory mediators disrupt the blood-brain barrier91 and promote pathology of the central nervous system (CNS)90, 92. Furthermore, LFNG was negatively associated with total brain volume, total gray matter volume, and temporoparietal meta-ROI thicknesses, and positively associated with CSF volume.

Similarly, mannose receptor C type 2 (MRC2), which encodes a receptor expressed on the surface of fibroblasts, M2 macrophages and microglia93, 94, was also upregulated in the Mediterranean cohort. M2 macrophages are anti-inflammatory and promote tissue repair and neuroprotection, in contrast to the proinflammatory action of M1 macrophages95, 96.

Previously, we observed that circulating monocytes from the Western cohort exhibited upregulation of genes associated with M1 polarization, and a non-significant trend for downregulation of M242. Results from the present work are consistent with these findings, suggesting that the Western cohort are polarized more towards a proinflammatory phenotype.

This is further supported by the neuroanatomical data, as MRC2 transcript levels, markers of anti-inflammatory M2 macrophages, were negatively correlated with total brain volumes, gray matter volumes, and temporoparietal volumes and thicknesses, and positively associated with white matter and CSF volumes.

In addition, we observed alterations in circulating monocyte expression profiles which were correlated with transcript levels in the temporal cortex and consistent with anti-inflammatory effects of the Mediterranean Diet relative to the Western Diet, and with the hypothesis that peripheral inflammation may promote neuroinflammation. Thus, potential interventions to increase anti-inflammatory activity may have a role in prevention or treatment of systemic as well as neuroinflammation.

SLC3A2 was also lower in the Western cohort and showed negative relationships with time spent alone and percent change in cortical thickness, and positive associations with white matter volume. SLC3A2, which encodes SLC3A2 (the heavy subunit of cluster of differentiation 98, CD98), is an amino acid transporter and anti-apoptotic factor97. Although SLC3A2 and CD98 exhibit a complex relationship with inflammation (e.g., upregulated in intestinal epithelium in Crohn’s disease98), there is evidence that the action of this gene is downregulated in the CNS during proinflammatory states. In a study of microglia-derived extracellular vesicles, CD98 was present in control vesicles (i.e., those derived from un-activated microglia) but not in experimentally activated microglia, suggesting downregulation in the proinflammatory state99.

Three other transcripts which were significantly reduced in the temporal cortex of the Western diet consumers have no previous published direct associations with neuropathology. Butyrophilin subfamily 2 member A1 (BTN2A1) encodes an integral plasma membrane protein which plays a role in immunomodulation of γδ T-cells which have been shown to have important anti-neuroinflammatory and neuroprotective effects100, 101. Katanin regulatory subunit B1 (KATNB1) is a subcomponent of the katanin heterodimer which functions in the cleavage and disassembly of microtubules, and perhaps plays a role in microtubule homeostasis102, axonal growth, and neuronal migration103. Transmembrane protein 268 (TMEM268) is a relatively understudied transmembrane protein overexpressed in brain which appears to be important for cell adhesion, proliferation, and viability. TMEM268 has been shown to interact with integrin subunit β4 (ITGB4)104.

Our exploratory analyses of potential pathways yielded further evidence of biological networks associated with inflammation. Among the top canonical pathways identified by Ingenuity Pathway Analysis (IPA) were phosphatase and tensin homolog (PTEN) signaling as well as ciliary neurotrophic factor (CNTF) signaling, both of which exert distinct immunomodulatory (including inflammatory) effects105107 and which have been found to be associated with AD-related neuropathology108, 109. Furthermore, among the top master regulators identified by IPA were transcription factor Sp3 and chymase 1, both of which have been implicated in inflammatory regulation110, 111 and are present in higher levels in the brains of AD patients112, 113. Finally, transforming growth factor beta 1 (TGFB1), which was reduced in the Western cohort, is a pleiotropic cytokine with multifarious effects on both innate and adaptive immune function114 and has been variously described as pro- and anti-inflammatory115.

While our study provides compelling experimental evidence of diet-induced alterations in transcriptional regulation in the brain potentially associated with neuroinflammation, it also suggests important future directions for mechanistic research concerning neuroanatomical effects of diet composition. The potential mechanisms by which the Western diet promotes— and the Mediterranean diet limits—neuroinflammation are numerous. Metabolic and microbial disturbances induced by Western diet consumption likely promote inflammation116, 117, while the Mediterranean diet’s antioxidant-rich composition likely limits inflammation118. Relatedly, Western diets increase central nervous system activity and perturb hypothalamic-pituitary-adrenal (HPA) axis physiology36 which may promote inflammation119121 and instigate a cascade of pathologic processes (e.g., AD122 and PD123). Pinpointing the key mechanisms by which diet impacts the brain will prove critical for developing effective interventions for populations that do not adhere to a Mediterranean diet.

In addition, as indicated by our prior work37, diet-induced inflammation may be further compounded by social inequalities. In developed nations, individuals of low socioeconomic status are more likely to consume a stereotypical Western diet and experience greater psychological stress124, 125, which may further exacerbate neurocognitive decline. This project focused on female macaques, as there is a paucity of data from female animal models45, and sex is known to affect the etiologies of neuropsychiatric and neurodegenerative disorders, including depression, AD, and PD126, 127. Consequently, data from males are necessary for fully understanding diet-induced neuroanatomical effects and associated molecular mechanisms. In addition, our transcriptional data are restricted to a single area of the brain. Additional studies are planned to explore other brain areas of importance to cognitive and other functions. Finally, whether these brain changes specifically impact cognitive function remains to be determined.

In conclusion, our results provide transcriptomic, neuroanatomical, and behavioral support for the premise that the Mediterranean diet exerts anti-inflammatory effects and preserves brain homeostasis and resilience compared to a Western diet. Increased cortical thickness and decreased white matter in the Western cohort, combined with transcriptomic signatures of inflammation and blood-brain barrier disruption, and behavioral correlates of neuroinflammation, suggest that the monkeys fed a Western diet developed neuroinflammation, whereas the animals consuming the Mediterranean diet were protected. The neuroanatomical variation we identified has important clinical relevance: increases in gray matter volume or cortical thickness associated with neuroinflammation could provide a biomarker for heightened risk of neurodegeneration and associated disorders. Future work integrating data from a variety of physiological systems (e.g., the gut microbiome, cardiovascular system, immune function, HPA physiology, metabolic health, physical and cognitive function) will help map the causal pathways by which diet precipitates neuroinflammation.

The impact of diet composition on neuroinflammation in NHPs suggests that changes in diet composition could be an effective intervention in humans to reduce the risk of neurodegenerative processes in aging. Population level diet modification in humans has been shown to be feasible as evidenced by: (1) the National Cholesterol Education Program which led to decreases in dietary fat and cholesterol intake and circulating cholesterol concentrations128; and (2) the FDA requirement to list trans fats on food labels and reduce trans fats in foods which reduced circulating levels of trans fats by more than 50%129. Our findings suggest that population-wide adoption of a Mediterranean-like diet pattern may provide a cost-effective intervention on brain aging with the potential for widespread efficacy.

Supplementary Material

1
2

Highlights.

  • Mediterranean diet is associated with lower expression of proinflammatory pathways

  • Mediterranean diet is associated with higher expression of neuroprotective pathways

  • Mediterranean diets may confer protection against peripheral & central inflammation

  • Mediterranean diet may help to preserve brain structure and socioemotional behavior

  • Nonhuman primates are useful models for studying diet effects on the body & brain

Funding:

This work was supported by the National Institutes of Health (R01-HL087103, RF1-AG058829, R01-HL122393, and U24-DK097748), an Intramural Grant from the Department of Pathology, the Wake Forest Alzheimer’s Disease Research Center (P30-AG049638, P30-AG072947), the Wake Forest Claude D. Pepper Older Americans for Independence Center (P30 AG21332), the Wake Forest Clinical and Translational Science Institute (UL1TR001420), and the National Cancer Institute’s Cancer Center Support Grant (P30CA012197, Wake Forest Baptist Comprehensive Cancer Center Cancer Genomics Shared Resource).

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Conflicts of Interest: The authors declare no conflicts of interest.

References

  • 1.Yassine HN, Samieri C, Livingston G, et al. Nutrition state of science and dementia prevention: recommendations of the Nutrition for Dementia Prevention Working Group. Lancet Healthy Longev 2022;3:e501–e512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Fekete M, Szarvas Z, Fazekas-Pongor V, et al. Nutrition Strategies Promoting Healthy Aging: From Improvement of Cardiovascular and Brain Health to Prevention of Age-Associated Diseases. Nutrients 2022;15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Muscaritoli M The Impact of Nutrients on Mental Health and Well-Being: Insights From the Literature. Front Nutr 2021;8:656290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Campanella A, Misciagna G, Mirizzi A, et al. The effect of the Mediterranean Diet on lifespan: a treatment-effect survival analysis of a population-based prospective cohort study in Southern Italy. Int J Epidemiol 2020;50:245–255. [DOI] [PubMed] [Google Scholar]
  • 5.Henríquez Sánchez P, Ruano C, de Irala J, Ruiz-Canela M, Martínez-González MA, Sánchez-Villegas A. Adherence to the Mediterranean diet and quality of life in the SUN Project. European Journal of Clinical Nutrition 2012;66:360–368. [DOI] [PubMed] [Google Scholar]
  • 6.Jacka FN, Pasco JA, Mykletun A, et al. Association of Western and traditional diets with depression and anxiety in women. The American journal of psychiatry 2010;167:305–311. [DOI] [PubMed] [Google Scholar]
  • 7.Hoscheidt S, Sanderlin AH, Baker LD, et al. Mediterranean and Western diet effects on Alzheimer’s disease biomarkers, cerebral perfusion, and cognition in mid-life: A randomized trial. Alzheimer’s & dementia : the journal of the Alzheimer’s Association 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Lourida I, Soni M, Thompson-Coon J, et al. Mediterranean diet, cognitive function, and dementia: a systematic review. Epidemiology 2013;24:479–489. [DOI] [PubMed] [Google Scholar]
  • 9.Dominguez LJ, Barbagallo M. Nutritional prevention of cognitive decline and dementia. Acta Biomed 2018;89:276–290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Jennings A, Berendsen AM, de Groot LCPGM, et al. Mediterranean-style diet improves systolic blood pressure and arterial stiffness in older adults. Hypertension 2019;73:578–586. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Salas-Salvadó J, Bulló M, Babio N, et al. Reduction in the incidence of Type 2 diabetes with the Mediterranean Diet: results of the PREDIMED-Reus nutrition intervention randomized trial. Diabetes Care 2010;34:14–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Toledo E, Salas-Salvadó J, Donat-Vargas C, et al. Mediterranean diet and invasive breast cancer risk among women at high cardiovascular risk in the PREDIMED trial: a randomized clinical trial. JAMA Intern Med 2015;175:1752–1760. [DOI] [PubMed] [Google Scholar]
  • 13.Sköldstam L, Hagfors L, Johansson G. An experimental study of a Mediterranean diet intervention for patients with rheumatoid arthritis. Ann Rheum Dis 2003;62:208–214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.US Department of Agriculture. Energy intakes: percentages of energy from protein, carbohydrate, fat, and alcohol, by gender and age, what we eat in America, NHANES 2013–2014. US Department of Agriculture, Agricultural Research Service Washington, DC, 2016. [Google Scholar]
  • 15.Cordain L, Eaton SB, Sebastian A, et al. Origins and evolution of the Western diet: health implications for the 21st century. The American Journal of Clinical Nutrition 2005;81:341–354. [DOI] [PubMed] [Google Scholar]
  • 16.Patel D, Evanchuk J, Wang R, Dunbar CL, Munhoz J, Field CJ. Regulation of immune function in healthy adults: one-stop guide on the role of dietary fatty acids, gut microbiota-derived short chain fatty acids, and select micronutrients in combination with physical activity. Appl Physiol Nutr Metab 2023;48:554–568. [DOI] [PubMed] [Google Scholar]
  • 17.Kendig MD, Leigh S-J, Morris MJ. Unravelling the impacts of western-style diets on brain, gut microbiota and cognition. Neuroscience & Biobehavioral Reviews 2021;128:233–243. [DOI] [PubMed] [Google Scholar]
  • 18.Matison AP, Mather KA, Flood VM, Reppermund S. Associations between nutrition and the incidence of depression in middle-aged and older adults: A systematic review and meta-analysis of prospective observational population-based studies. Ageing Res Rev 2021;70:101403. [DOI] [PubMed] [Google Scholar]
  • 19.Shakersain B, Santoni G, Larsson SC, et al. Prudent diet may attenuate the adverse effects of Western diet on cognitive decline. Alzheimer’s & Dementia 2016;12:100–109. [DOI] [PubMed] [Google Scholar]
  • 20.Harriden B, D’Cunha NM, Kellett J, Isbel S, Panagiotakos DB, Naumovski N. Are dietary patterns becoming more processed? The effects of different dietary patterns on cognition: A review. Nutr Health 2022;28:341–356. [DOI] [PubMed] [Google Scholar]
  • 21.Hsu TM, Kanoski SE. Blood-brain barrier disruption: Mechanistic links between Western diet consumption and dementia. Front Aging Neurosci 2014;6:88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Więckowska-Gacek A, Mietelska-Porowska A, Wydrych M, Wojda U. Western diet as a trigger of Alzheimer’s disease: From metabolic syndrome and systemic inflammation to neuroinflammation and neurodegeneration. Ageing Research Reviews 2021;70:101397. [DOI] [PubMed] [Google Scholar]
  • 23.Morris MC. Nutrition and risk of dementia: overview and methodological issues. Ann N Y Acad Sci 2016;1367:31–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Scarmeas N, Stern Y, Tang MX, Mayeux R, Luchsinger JA. Mediterranean diet and risk for Alzheimer’s disease. Ann Neurol 2006;59:912–921. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Tangney CC, Kwasny MJ, Li H, Wilson RS, Evans DA, Morris MC. Adherence to a Mediterranean-type dietary pattern and cognitive decline in a community population. Am J Clin Nutr 2011;93:601–607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Morris MC, Tangney CC, Wang Y, Sacks FM, Bennett DA, Aggarwal NT. MIND diet associated with reduced incidence of Alzheimer’s disease. Alzheimers Dement 2015;11:1007–1014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Tsivgoulis G, Judd S, Letter AJ, et al. Adherence to a Mediterranean diet and risk of incident cognitive impairment. Neurology 2013;80:1684–1692. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Cornelis MC, Agarwal P, Holland TM, van Dam RM. MIND Dietary Pattern and Its Association with Cognition and Incident Dementia in the UK Biobank. Nutrients 2022;15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Pribisalić A, Popović R, Salvatore FP, et al. The role of socioeconomic status in adherence to the Mediterranean diet and body mass index change: a follow-up study in the general population of southern Croatia. Nutrients 2021;13:3802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Wood EK, Sullivan EL. The Influence of Diet on Metabolism and Health Across the Lifespan in Nonhuman Primates. Curr Opin Endocr Metab Res 2022;24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Register TC. Primate models in women’s health: inflammation and atherogenesis in female cynomolgus macaques (Macaca fascicularis). American Journal of Primatology 2009;71:766–775. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Chiou KL, Montague MJ, Goldman EA, et al. Rhesus macaques as a tractable physiological model of human ageing. Philosophical Transactions of the Royal Society B: Biological Sciences 2020;375:20190612. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Stonebarger GA, Bimonte-Nelson HA, Urbanski HF. The rhesus macaque as a translational model for neurodegeneration and Alzheimer’s disease. Frontiers in Aging Neuroscience 2021;13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Arnsten AFT, Datta D, Leslie S, Yang S-T, Wang M, Nairn AC. Alzheimer’s-like pathology in aging rhesus macaques: Unique opportunity to study the etiology and treatment of Alzheimer’s disease. Proceedings of the National Academy of Sciences 2019;116:26230–26238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Shively CA, Appt SE, Vitolins MZ, et al. Mediterranean versus Western diet effects on caloric intake, obesity, metabolism, and hepatosteatosis in nonhuman primates. Obesity (Silver Spring) 2019;27:777–784. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Shively CA, Appt SE, Chen H, et al. Mediterranean diet, stress resilience, and aging in nonhuman primates. Neurobiology of Stress 2020;13:100254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Frye BM, Craft S, Register TC, et al. Diet, psychosocial stress, and Alzheimer’s disease–related neuroanatomy in female nonhuman primates. Alzheimer’s & Dementia 2021;17:733–744. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Pegueroles J, Vilaplana E, Montal V, et al. Longitudinal brain structural changes in preclinical Alzheimer’s disease. Alzheimer’s & Dementia 2017;13:499–509. [DOI] [PubMed] [Google Scholar]
  • 39.Batzu L, Westman E, Pereira JB. Cerebrospinal fluid progranulin is associated with increased cortical thickness in early stages of Alzheimer’s disease. Neurobiology of Aging 2020;88:61–70. [DOI] [PubMed] [Google Scholar]
  • 40.Gispert JD, Suárez-Calvet M, Monté GC, et al. Cerebrospinal fluid sTREM2 levels are associated with gray matter volume increases and reduced diffusivity in early Alzheimer’s disease. Alzheimer’s & Dementia 2016;12:1259–1272. [DOI] [PubMed] [Google Scholar]
  • 41.Eskildsen SF, Coupé P, García-Lorenzo D, Fonov V, Pruessner JC, Collins DL. Prediction of Alzheimer’s disease in subjects with mild cognitive impairment from the ADNI cohort using patterns of cortical thinning. Neuroimage 2013;65:511–521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Johnson CSC, Shively C, Michalson KT, et al. Contrasting effects of Western vs Mediterranean diets on monocyte inflammatory gene expression and social behavior in a primate model. eLife 2021;10:e68293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Schwingshackl L, Hoffmann G. Mediterranean dietary pattern, inflammation and endothelial function: a systematic review and meta-analysis of intervention trials. Nutrition, metabolism, and cardiovascular diseases : NMCD 2014;24:929–939. [DOI] [PubMed] [Google Scholar]
  • 44.Johnson CSC, Frye BM, Register TC, Snyder-Mackler N, Shively CA. Mediterranean Diet Reduces Social Isolation and Anxiety in Adult Female Nonhuman Primates. Nutrients 2022;14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.The Lancet N Sex, gender, and the cost of neurological disorders. The Lancet Neurology 2023;22:367. [DOI] [PubMed] [Google Scholar]
  • 46.Grothe MJ, Barthel H, Sepulcre J, Dyrba M, Sabri O, Teipel SJ. In vivo staging of regional amyloid deposition. Neurology 2017;89:2031–2038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Insel PS, Mormino EC, Aisen PS, Thompson WK, Donohue MC. Neuroanatomical spread of amyloid β and tau in Alzheimer’s disease: implications for primary prevention. Brain Commun 2020;2:fcaa007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Yang H, Xu H, Li Q, et al. Study of brain morphology change in Alzheimer’s disease and amnestic mild cognitive impairment compared with normal controls. Gen Psychiatr 2019;32:e100005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Silverstein-Metzler MG, Frye BM, Justice JN, et al. Psychosocial stress increases risk for type 2 diabetes in female cynomolgus macaques consuming a western diet. Psychoneuroendocrinology 2022;139:105706. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Shively CA, Register TC, Appt SE, et al. Consumption of Mediterranean versus Western Diet Leads to Distinct Mammary Gland Microbiome Populations. Cell Rep 2018;25:47–56.e43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Shively CA, Frye BM, Negrey JD, et al. The interactive effects of psychosocial stress and diet composition on health in primates. Neuroscience & Biobehavioral Reviews 2023;152:105320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Kafatos A, Verhagen H, Moschandreas J, Apostolaki I, Westerop JJMV. Mediterranean diet of Crete: foods and nutrient content. Journal of the American Dietetic Association 2000;100:1487–1493. [DOI] [PubMed] [Google Scholar]
  • 53.Estruch R, Ros E, Salas-Salvadó J, et al. Primary prevention of cardiovascular disease with a Mediterranean diet supplemented with extra-virgin olive oil or nuts. New England Journal of Medicine 2018;378:e34. [DOI] [PubMed] [Google Scholar]
  • 54.Altmann J Observational Study of Behavior: Sampling Methods. Behaviour 1974;49:227–267. [DOI] [PubMed] [Google Scholar]
  • 55.Maestripieri D, Schino G, Aureli F, Troisi A. A Modest Proposal - Displacement Activities as an Indicator of Emotions in Primates. Anim Behav 1992;44:967–979. [Google Scholar]
  • 56.Schino G, Perretta G, Taglioni AM, Monaco V, Troisi A. Primate displacement activities as an ethopharmacological model of anxiety. Anxiety 1996;2:186–191. [DOI] [PubMed] [Google Scholar]
  • 57.Shively CA, Register TC, Appt SE, Clarkson TB. Effects of Long-Term Sertraline Treatment and Depression on Coronary Artery Atherosclerosis in Premenopausal Female Primates. Psychosom Med 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Troisi A, Belsanti S, Bucci AR, Mosco C, Sinti F, Verucci M. Affect regulation in alexithymia: an ethological study of displacement behavior during psychiatric interviews. The Journal of nervous and mental disease 2000;188:13–18. [DOI] [PubMed] [Google Scholar]
  • 59.Troisi A Displacement activities as a behavioral measure of stress in nonhuman primates and human subjects. Stress 2002;5:47–54. [DOI] [PubMed] [Google Scholar]
  • 60.Avants BB, Tustison NJ, Wu J, Cook PA, Gee JC. An open source multivariate framework for n-tissue segmentation with evaluation on public data. Neuroinformatics 2011;9:381–400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Rohlfing T, Kroenke CD, Sullivan EV, et al. The INIA19 Template and NeuroMaps Atlas for Primate Brain Image Parcellation and Spatial Normalization. Front Neuroinform 2012;6:27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Dobin A, Davis CA, Schlesinger F, et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 2012;29:15–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.R: A language and environment for statistical computing [computer program]. Vienna, Austria: R Foundation for Statistical Computing, 2021. [Google Scholar]
  • 64.Sun S, Zhu J, Mozaffari S, Ober C, Chen M, Zhou X. Heritability estimation and differential analysis of count data with generalized linear mixed models in genomic sequencing studies. Bioinformatics 2018;35:487–496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Ritchie ME, Phipson B, Wu D, et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic acids research 2015;43:e47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Law CW, Alhamdoosh M, Su S, et al. RNA-seq analysis is easy as 1-2-3 with limma, Glimma and edgeR. F1000Res 2016;5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Li H, Handsaker B, Wysoker A, et al. The sequence alignment/map format and SAMtools. Bioinformatics 2009;25:2078–2079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Willard SL, Shively CA. Social Status and the Non-human Primate Brain. In: Shively CA, Wilson ME, eds. Social Inequalities in Health in Nonhuman Primates: The Biology of the Gradient. Cham: Springer International Publishing, 2016: 117–139. [Google Scholar]
  • 69.Shively CA, Day SM. Social inequalities in health in nonhuman primates. Neurobiology of Stress 2015;1:156–163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Cervera-Juanes R, Darakjian P, Ball M, Kohama SG, Urbanski HF. Effects of estradiol supplementation on the brain transcriptome of old rhesus macaques maintained on an obesogenic diet. GeroScience 2022;44:229–252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J Roy Statist Soc Ser B (Methodological) 1995;57:289–300. [Google Scholar]
  • 72.Cole SW, Levine ME, Arevalo JM, Ma J, Weir DR, Crimmins EM. Loneliness, eudaimonia, and the human conserved transcriptional response to adversity. Psychoneuroendocrinology 2015;62:11–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Hisamoto N, Sakai Y, Ohta K, et al. CDK14 promotes axon regeneration by regulating the noncanonical Wnt signaling pathway in a kinase-independent manner. J Neurosci 2021;41:8309–8320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Jridi I, Canté-Barrett K, Pike-Overzet K, Staal FJT. Inflammation and Wnt signaling: target for immunomodulatory therapy? Front Cell Dev Biol 2021;8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Chen L, Wang Y, Jiang W, Ni R, Wang Y, Ni S. CDK14 involvement in proliferation migration and invasion of esophageal cancer. Ann Transl Med 2019;7:681. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Ferguson FM, Doctor ZM, Ficarro SB, et al. Discovery of covalent CDK14 inhibitors with pan-TAIRE family specificity. Cell Chem Biol 2019;26:804–817.e812. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Li Q, Zhou L, Wang M, et al. MicroRNA-613 impedes the proliferation and invasion of glioma cells by targeting cyclin-dependent kinase 14. Biomed Pharmacother 2018;98:636–642. [DOI] [PubMed] [Google Scholar]
  • 78.Bai B, Wang X, Li Y, et al. Deep Multilayer Brain Proteomics Identifies Molecular Networks in Alzheimer’s Disease Progression. Neuron 2020;105:975–991.e977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Kelly J, Moyeed R, Carroll C, Albani D, Li X. Gene expression meta-analysis of Parkinson’s disease and its relationship with Alzheimer’s disease. Molecular Brain 2019;12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Wu ATH, Lawal B, Wei L, Wen YT, Tzeng DTW, Lo WC. Multiomics identification of potential targets for Alzheimer sisease and antrocin as a therapeutic candidate. Pharmaceutics 2021;13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Parmasad J-LA, Ricke KM, Stykel MG, et al. Genetic and pharmacological reduction of CDK14 mitigates α-synuclein pathology in human neurons and in rodent models of Parkinson’s disease. bioRxiv 2023:2022.2005.2002.490309. [Google Scholar]
  • 82.Allen Reish HE, Standaert DG. Role of α-Synuclein in Inducing Innate and Adaptive Immunity in Parkinson Disease. Journal of Parkinson’s Disease 2015;5:1–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Perry VH. The influence of systemic inflammation on inflammation in the brain: implications for chronic neurodegenerative disease. Brain Behav Immun 2004;18:407–413. [DOI] [PubMed] [Google Scholar]
  • 84.Perry VH, Holmes C. Microglial priming in neurodegenerative disease. Nature Reviews Neurology 2014;10:217–224. [DOI] [PubMed] [Google Scholar]
  • 85.Vauzour D, Camprubi-Robles M, Miquel-Kergoat S, et al. Nutrition for the ageing brain: Towards evidence for an optimal diet. Ageing Res Rev 2017;35:222–240. [DOI] [PubMed] [Google Scholar]
  • 86.Walker KA, Hoogeveen RC, Folsom AR, et al. Midlife systemic inflammatory markers are associated with late-life brain volume: The ARIC study. Neurology 2017;89:2262–2270. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Kato TM, Kawaguchi A, Kosodo Y, Niwa H, Matsuzaki F. Lunatic fringe potentiates Notch signaling in the developing brain. Mol Cell Neurosci 2010;45:12–25. [DOI] [PubMed] [Google Scholar]
  • 88.Pompa de la JL, Wakeham A, Correia KM, et al. Conservation of the Notch signalling pathway in mammalian neurogenesis. Development 1997;124:1139–1148. [DOI] [PubMed] [Google Scholar]
  • 89.Imayoshi I, Sakamoto M, Yamaguchi M, Mori K, Kageyama R. Essential roles of notch signaling in maintenance of neural stem cells in developing and adult brains. J Neurosci 2010;30:3489–3498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Derada Troletti C, Lopes Pinheiro MA, Charabati M, et al. Notch signaling is impaired during inflammation in a Lunatic Fringe-dependent manner. Brain Behav Immun 2018;69:48–56. [DOI] [PubMed] [Google Scholar]
  • 91.Varatharaj A, Galea I. The blood-brain barrier in systemic inflammation. Brain Behav Immun 2017;60:1–12. [DOI] [PubMed] [Google Scholar]
  • 92.Glabinski AR, Ransohoff RM. Chemokines and chemokine receptors in CNS pathology. J Neurovirol 1999;5:3–12. [DOI] [PubMed] [Google Scholar]
  • 93.López-Guisa JM, Cai X, Collins SJ, et al. Mannose receptor 2 attenuates renal fibrosis. J Am Soc Nephrol 2012;23:236–251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.DePaula-Silva AB, Gorbea C, Doty DJ, et al. Differential transcriptional profiles identify microglial- and macrophage-specific gene markers expressed during virus-induced neuroinflammation. J Neuroinflammation 2019;16:152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Mosser DM, Edwards JP. Exploring the full spectrum of macrophage activation. Nat Rev Immunol 2008;8:958–969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Burke NN, Kerr DM, Moriarty O, Finn DP, Roche M. Minocycline modulates neuropathic pain behaviour and cortical M1–M2 microglial gene expression in a rat model of depression. Brain Behav Immun 2014;42:147–156. [DOI] [PubMed] [Google Scholar]
  • 97.Fotiadis D, Kanai Y, Palacín M. The SLC3 and SLC7 families of amino acid transporters. Mol Aspects Med 2013;34:139–158. [DOI] [PubMed] [Google Scholar]
  • 98.Nguyen HTT, Dalmasso G, Yan Y, et al. MicroRNA-7 modulates CD98 expression during intestinal epithelial cell differentiation. J Biol Chem 2010;285:1479–1489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Yang Y, Boza-Serrano A, Dunning CJR, Clausen BH, Lambertsen KL, Deierborg T. Inflammation leads to distinct populations of extracellular vesicles from microglia. J Neuroinflammation 2018;15:168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Herrmann T, Karunakaran MM. Butyrophilins: γδ T Cell Receptor Ligands, Immunomodulators and More. Front Immunol 2022;13:876493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Su X, Yang S, Li Y, et al. γδ T cells recruitment and local proliferation in brain parenchyma benefit anti-neuroinflammation after cerebral microbleeds. Front Immunol 2023;14:1139601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Andrés-Benito P, Delgado-Morales R, Ferrer I. Altered Regulation of KIAA0566, and Katanin Signaling Expression in the Locus Coeruleus With Neurofibrillary Tangle Pathology. Frontiers in cellular neuroscience 2018;12:131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.McNally KP, Bazirgan OA, McNally FJ. Two domains of p80 katanin regulate microtubule severing and spindle pole targeting by p60 katanin. J Cell Sci 2000;113 (Pt 9):1623–1633. [DOI] [PubMed] [Google Scholar]
  • 104.Hong D, Zhang X, Li R, et al. Deletion of TMEM268 inhibits growth of gastric cancer cells by downregulating the ITGB4 signaling pathway. Cell Death Differ 2019;26:1453–1466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Günzl P, Schabbauer G. Recent advances in the genetic analysis of PTEN and PI3K innate immune properties. Immunobiology 2008;213:759–765. [DOI] [PubMed] [Google Scholar]
  • 106.Taylor H, Laurence ADJ, Uhlig HH. The role of PTEN in innate and adaptive immunity. Cold Spring Harbor perspectives in medicine 2019;9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Meazza C, Di Marco A, Fruscella P, et al. Centrally mediated inhibition of local inflammation by ciliary neurotrophic factor. Neuroimmunomodulation 1997;4:271–276. [DOI] [PubMed] [Google Scholar]
  • 108.Benetatos J, Bennett RE, Evans HT, et al. PTEN activation contributes to neuronal and synaptic engulfment by microglia in tauopathy. Acta Neuropathol 2020;140:7–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Garcia P, Youssef I, Utvik JK, et al. Ciliary neurotrophic factor cell-based delivery prevents synaptic impairment and improves memory in mouse models of Alzheimer’s disease. J Neurosci 2010;30:7516–7527. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Wan D, Feng J, Wang P, Yang Z, Sun T. Hypoxia- and inflammation-related transcription factor SP3 may be involved in platelet activation and inflammation in intracranial hemorrhage. Frontiers in Neurology 2022;13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Caughey GH. Mast cell tryptases and chymases in inflammation and host defense. Immunol Rev 2007;217:141–154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Boutillier S, Lannes B, Buée L, et al. Sp3 and Sp4 transcription factor levels are increased in brains of patients with Alzheimer’s disease. Neurodegener Dis 2007;4:413–423. [DOI] [PubMed] [Google Scholar]
  • 113.Jones MK, Nair A, Gupta M. Mast cells in neurodegenerative disease. Front Cell Neurosci 2019;13:171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Travis MA, Sheppard D. TGF-β activation and function in immunity. Ann Rev Immunol 2014;32:51–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Sanjabi S, Zenewicz LA, Kamanaka M, Flavell RA. Anti-inflammatory and proinflammatory roles of TGF-β, IL-10, and IL-22 in immunity and autoimmunity. Curr Opin Pharmacol 2009;9:447–453. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Zinöcker MK, Lindseth IA. The Western diet–microbiome-host interaction and its role in metabolic disease. Nutrients 2018;10:365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Zhang L, Bruce-Keller AJ, Dasuri K, Nguyen A, Liu Y, Keller JN. Diet-induced metabolic disturbances as modulators of brain homeostasis. Biochim Biophys Acta 2009;1792:417–422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Román GC, Jackson RE, Gadhia R, Román AN, Reis J. Mediterranean diet: The role of long-chain ω-3 fatty acids in fish; polyphenols in fruits, vegetables, cereals, coffee, tea, cacao and wine; probiotics and vitamins in prevention of stroke, age-related cognitive decline, and Alzheimer disease. Rev Neurol 2019;175:724–741. [DOI] [PubMed] [Google Scholar]
  • 119.Zhang B, Zhang Y, Wu W, et al. Chronic glucocorticoid exposure activates BK-NLRP1 signal involving in hippocampal neuron damage. Journal of Neuroinflammation 2017;14:139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Pongratz G, Straub RH. The sympathetic nervous response in inflammation. Arthritis Research & Therapy 2014;16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Toral M, Robles-Vera I, de la Visitación N, et al. Critical Role of the Interaction Gut Microbiota - Sympathetic Nervous System in the Regulation of Blood Pressure. Front Physiol 2019;10:231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Milligan Armstrong A, Porter T, Quek H, et al. Chronic stress and Alzheimer’s disease: the interplay between the hypothalamic–pituitary–adrenal axis, genetics and microglia. Biological Reviews 2021;96:2209–2228. [DOI] [PubMed] [Google Scholar]
  • 123.Herrero MT, Estrada C, Maatouk L, Vyas S. Inflammation in Parkinson’s disease: role of glucocorticoids. Front Neuroanat 2015;9:32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Cohen S, Doyle WJ, Baum A. Socioeconomic status is associated with stress hormones. Psychosomatic Medicine 2006;68. [DOI] [PubMed] [Google Scholar]
  • 125.Vliegenthart J, Noppe G, van Rossum EFC, Koper JW, Raat H, van den Akker ELT. Socioeconomic status in children is associated with hair cortisol levels as a biological measure of chronic stress. Psychoneuroendocrinology 2016;65:9–14. [DOI] [PubMed] [Google Scholar]
  • 126.Ullah MF, Ahmad A, Bhat SH, Abu-Duhier FM, Barreto GE, Ashraf GM. Impact of sex differences and gender specificity on behavioral characteristics and pathophysiology of neurodegenerative disorders. Neuroscience & Biobehavioral Reviews 2019;102:95–105. [DOI] [PubMed] [Google Scholar]
  • 127.Pinares-Garcia P, Stratikopoulos M, Zagato A, Loke H, Lee J. Sex: A Significant Risk Factor for Neurodevelopmental and Neurodegenerative Disorders. Brain Sci 2018;8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Cleeman JI, Lenfant C. The National Cholesterol Education Program: progress and prospects. Jama 1998;280:2099–2104. [DOI] [PubMed] [Google Scholar]
  • 129.Li H, Zhang Q, Song J, et al. Plasma trans-fatty acids levels and mortality: a cohort study based on 1999–2000 National Health and Nutrition Examination Survey (NHANES). Lipids Health Dis 2017;16:176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Johnson CS, Shively C, Michalson KT, et al. Contrasting effects of Western vs Mediterranean diets on monocyte inflammatory gene expression and social behavior in a primate model. eLife 2021;10. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

1
2

RESOURCES